Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Carcinogenesis ; 39(9): 1165-1175, 2018 09 21.
Article in English | MEDLINE | ID: mdl-29893810

ABSTRACT

The NF2 gene encodes the tumor and metastasis suppressor protein Merlin. Merlin exerts its tumor suppressive role by inhibiting proliferation and inducing contact-growth inhibition and apoptosis. In the current investigation, we determined that loss of Merlin in breast cancer tissues is concordant with the loss of the inhibitory SMAD, SMAD7, of the TGF-ß pathway. This was reflected as dysregulated activation of TGF-ß signaling that co-operatively engaged with effectors of the Hippo pathway (YAP/TAZ/TEAD). As a consequence, the loss of Merlin in breast cancer resulted in a significant metabolic and bioenergetic adaptation of cells characterized by increased aerobic glycolysis and decreased oxygen consumption. Mechanistically, we determined that the co-operative activity of the Hippo and TGF-ß transcription effectors caused upregulation of the long non-coding RNA Urothelial Cancer-Associated 1 (UCA1) that disengaged Merlin's check on STAT3 activity. The consequent upregulation of Hexokinase 2 (HK2) enabled a metabolic shift towards aerobic glycolysis. In fact, Merlin deficiency engendered cellular dependence on this metabolic adaptation, endorsing a critical role for Merlin in regulating cellular metabolism. This is the first report of Merlin functioning as a molecular restraint on cellular metabolism. Thus, breast cancer patients whose tumors demonstrate concordant loss of Merlin and SMAD7 may benefit from an approach of incorporating STAT3 inhibitors.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Glycolysis/genetics , Neurofibromin 2/genetics , Protein Serine-Threonine Kinases/genetics , Smad7 Protein/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Contact Inhibition/genetics , Genes, Tumor Suppressor , Hexokinase/biosynthesis , Hippo Signaling Pathway , Humans , MCF-7 Cells , Neurofibromin 2/deficiency , Oxygen Consumption/genetics , RNA, Long Noncoding/biosynthesis , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
2.
Redox Biol ; 9: 57-66, 2016 10.
Article in English | MEDLINE | ID: mdl-27393890

ABSTRACT

Metabolic control of cellular function is significant in the context of inflammation-induced metabolic dysregulation in immune cells. Generation of reactive oxygen species (ROS) such as hydrogen peroxide and superoxide are one of the critical events that modulate the immune response in neutrophils. When activated, neutrophil NADPH oxidases consume large quantities of oxygen to rapidly generate ROS, a process that is referred to as the oxidative burst. These ROS are required for the efficient removal of phagocytized cellular debris and pathogens. In chronic inflammatory diseases, neutrophils are exposed to increased levels of oxidants and pro-inflammatory cytokines that can further prime oxidative burst responses and generate lipid oxidation products such as 4-hydroxynonenal (4-HNE). In this study we hypothesized that since 4-HNE can target glycolysis then this could modify the oxidative burst. To address this the oxidative burst was determined in freshly isolated healthy subject neutrophils using 13-phorbol myristate acetate (PMA) and the extracellular flux analyzer. Neutrophils pretreated with 4-HNE exhibited a significant decrease in the oxidative burst response and phagocytosis. Mass spectrometric analysis of alkyne-HNE treated neutrophils followed by click chemistry detected modification of a number of cytoskeletal, metabolic, redox and signaling proteins that are critical for the NADPH oxidase mediated oxidative burst. These modifications were confirmed using a candidate immunoblot approach for critical proteins of the active NADPH oxidase enzyme complex (Nox2 gp91phox subunit and Rac1 of the NADPH oxidase) and glyceraldehyde phosphate dehydrogenase, a critical enzyme in the metabolic regulation of oxidative burst. Taken together, these data suggest that 4-HNE-induces a pleiotropic mechanism to inhibit neutrophil function. These mechanisms may contribute to the immune dysregulation associated with chronic pathological conditions where 4-HNE is generated.


Subject(s)
Aldehydes/pharmacology , Neutrophils/drug effects , Neutrophils/physiology , Phagocytosis/drug effects , Respiratory Burst/drug effects , Adult , Cytoskeletal Proteins/metabolism , Glycolysis/drug effects , Healthy Volunteers , Humans , Lipid Peroxidation/drug effects , Middle Aged , NADPH Oxidases/metabolism , Phagocytosis/immunology , Reactive Oxygen Species/metabolism , Respiratory Burst/immunology
3.
Redox Biol ; 8: 136-48, 2016 08.
Article in English | MEDLINE | ID: mdl-26774751

ABSTRACT

Many cancer cells follow an aberrant metabolic program to maintain energy for rapid cell proliferation. Metabolic reprogramming often involves the upregulation of glutaminolysis to generate reducing equivalents for the electron transport chain and amino acids for protein synthesis. Critical enzymes involved in metabolism possess a reactive thiolate group, which can be modified by certain oxidants. In the current study, we show that modification of mitochondrial protein thiols by a model compound, iodobutyl triphenylphosphonium (IBTP), decreased mitochondrial metabolism and ATP in MDA-MB 231 (MB231) breast adenocarcinoma cells up to 6 days after an initial 24h treatment. Mitochondrial thiol modification also depressed oxygen consumption rates (OCR) in a dose-dependent manner to a greater extent than a non-thiol modifying analog, suggesting that thiol reactivity is an important factor in the inhibition of cancer cell metabolism. In non-tumorigenic MCF-10A cells, IBTP also decreased OCR; however the extracellular acidification rate was significantly increased at all but the highest concentration (10µM) of IBTP indicating that thiol modification can have significantly different effects on bioenergetics in tumorigenic versus non-tumorigenic cells. ATP and other adenonucleotide levels were also decreased by thiol modification up to 6 days post-treatment, indicating a decreased overall energetic state in MB231 cells. Cellular proliferation of MB231 cells was also inhibited up to 6 days post-treatment with little change to cell viability. Targeted metabolomic analyses revealed that thiol modification caused depletion of both Krebs cycle and glutaminolysis intermediates. Further experiments revealed that the activity of the Krebs cycle enzyme, aconitase, was attenuated in response to thiol modification. Additionally, the inhibition of glutaminolysis corresponded to decreased glutaminase C (GAC) protein levels, although other protein levels were unaffected. This study demonstrates for the first time that mitochondrial thiol modification inhibits metabolism via inhibition of both aconitase and GAC in a breast cancer cell model.


Subject(s)
Breast Neoplasms/metabolism , Energy Metabolism , Mitochondria/metabolism , Sulfhydryl Compounds/metabolism , Adenosine Triphosphate/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Glutaminase/metabolism , Humans , Metabolome , Metabolomics/methods , Stress, Physiological
4.
Oncotarget ; 6(40): 43065-80, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26515588

ABSTRACT

Mitochondrial function is influenced by alterations in oncogenes and tumor suppressor genes and changes in the microenvironment occurring during tumorigenesis. Therefore, we hypothesized that mitochondrial function will be stably and dynamically altered at each stage of the prostate tumor development. We tested this hypothesis in RWPE-1 cells and its tumorigenic clones with progressive malignant characteristics (RWPE-1 < WPE-NA22 < WPE-NB14 < WPE-NB11 < WPE-NB26) using high-throughput respirometry. Our studies demonstrate that mitochondrial content do not change with increasing malignancy. In premalignant cells (WPE-NA22 and WPE-NB14), OXPHOS is elevated in presence of glucose or glutamine alone or in combination compared to RWPE-1 cells and decreases with increasing malignancy. Glutamine maintained higher OXPHOS than glucose and suggests that it may be an important substrate for the growth and proliferation of prostate epithelial cells. Glycolysis significantly increases with malignancy and follow a classical Warburg phenomenon. Fatty acid oxidation (FAO) is significantly lower in tumorigenic clones and invasive WPE-NB26 does not utilize FAO at all. In this paper, we introduce for the first time the mitochondrial oncobioenergetic index (MOBI), a mathematical representation of oncobioenergetic profile of a cancer cell, which increases significantly upon transformation into localized premalignant form and rapidly falls below the normal as they become aggressive in prostate tumorigenesis. We have validated this in five prostate cancer cell lines and MOBI appears to be not related to androgen dependence or mitochondrial content, but rather dependent on the stage of the cancer. Altogether, we propose that MOBI could be a potential biomarker to distinguish aggressive cancer from that of indolent disease.


Subject(s)
Biomarkers, Tumor/analysis , Mitochondria/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Disease Progression , Energy Metabolism/physiology , Humans , Male
5.
J Biol Chem ; 290(31): 19055-66, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26085096

ABSTRACT

Peroxynitrite production and tyrosine nitration are present in several pathological conditions, including neurodegeneration, stroke, aging, and cancer. Nitration of the pro-survival chaperone heat shock protein 90 (Hsp90) in position 33 and 56 induces motor neuron death through a toxic gain-of-function. Here we show that nitrated Hsp90 regulates mitochondrial metabolism independently of the induction of cell death. In PC12 cells, a small fraction of nitrated Hsp90 was located on the mitochondrial outer membrane and down-regulated mitochondrial membrane potential, oxygen consumption, and ATP production. Neither endogenous Hsp90 present in the homogenate nor unmodified and fully active recombinant Hsp90 was able to compete with the nitrated protein for the binding to mitochondria. Moreover, endogenous or recombinant Hsp90 did not prevent the decrease in mitochondrial activity but supported nitrated Hsp90 mitochondrial gain-of-function. Nitrotyrosine in position 33, but not in any of the other four tyrosine residues prone to nitration in Hsp90, was sufficient to down-regulate mitochondrial activity. Thus, in addition to induction of cell death, nitrated Hsp90 can also regulate mitochondrial metabolism, suggesting that depending on the cell type, distinct Hsp90 nitration states regulate different aspects of cellular metabolism. This regulation of mitochondrial homeostasis by nitrated Hsp90 could be of particular relevance in cancer cells.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Protein Processing, Post-Translational , Tyrosine/analogs & derivatives , Adenosine Triphosphate/biosynthesis , Animals , Energy Metabolism , PC12 Cells , Protein Transport , Rats , Tyrosine/metabolism
6.
PLoS One ; 10(3): e0120460, 2015.
Article in English | MEDLINE | ID: mdl-25785718

ABSTRACT

Despite advances in screening and treatment over the past several years, breast cancer remains a leading cause of cancer-related death among women in the United States. A major goal in breast cancer treatment is to develop safe and clinically useful therapeutic agents that will prevent the recurrence of breast cancers after front-line therapeutics have failed. Ideally, these agents would have relatively low toxicity against normal cells, and will specifically inhibit the growth and proliferation of cancer cells. Our group and others have previously demonstrated that breast cancer cells exhibit increased mitochondrial oxygen consumption compared with non-tumorigenic breast epithelial cells. This suggests that it may be possible to deliver redox active compounds to the mitochondria to selectively inhibit cancer cell metabolism. To demonstrate proof-of-principle, a series of mitochondria-targeted soft electrophiles (MTSEs) has been designed which selectively accumulate within the mitochondria of highly energetic breast cancer cells and modify mitochondrial proteins. A prototype MTSE, IBTP, significantly inhibits mitochondrial oxidative phosphorylation, resulting in decreased breast cancer cell proliferation, cell attachment, and migration in vitro. These results suggest MTSEs may represent a novel class of anti-cancer agents that prevent cancer cell growth by modification of specific mitochondrial proteins.


Subject(s)
Breast Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Electron Transport/drug effects , Energy Metabolism/drug effects , Humans , Organophosphorus Compounds/pharmacology , Protein Processing, Post-Translational/drug effects
7.
Redox Biol ; 2: 1038-47, 2014.
Article in English | MEDLINE | ID: mdl-25454745

ABSTRACT

Chronic ethanol-mediated oxidative stress and lipid peroxidation increases the levels of various reactive lipid species including 4-hydroxynonenal (4-HNE), which can subsequently modify proteins in the liver. It has been proposed that 4-HNE modification adversely affects the structure and/or function of mitochondrial proteins, thereby impairing mitochondrial metabolism. To determine whether chronic ethanol consumption increases levels of 4-HNE modified proteins in mitochondria, male rats were fed control and ethanol-containing diets for 5 weeks and mitochondrial samples were analyzed using complementary proteomic methods. Five protein bands (approx. 35, 45, 50, 70, and 90kDa) showed strong immunoreactivity for 4-HNE modified proteins in liver mitochondria from control and ethanol-fed rats when proteins were separated by standard 1D SDS-PAGE. Using high-resolution proteomic methods (2D IEF/SDS-PAGE and BN-PAGE) we identified several mitochondrial proteins immunoreactive for 4-HNE, which included mitofilin, dimethylglycine dehydrogenase, choline dehydrogenase, electron transfer flavoprotein α, cytochrome c1, enoyl CoA hydratase, and cytochrome c. The electron transfer flavoprotein α consistently showed increased 4-HNE immunoreactivity in mitochondria from ethanol-fed rats as compared to mitochondria from the control group. Increased 4-HNE reactivity was also detected for dimethylglycine dehydrogenase, enoyl CoA hydratase, and cytochrome c in ethanol samples when mitochondria were analyzed by BN-PAGE. In summary, this work identifies new targets of 4-HNE modification in mitochondria and provides useful information needed to better understand the molecular mechanisms underpinning chronic ethanol-induced mitochondrial dysfunction and liver injury.


Subject(s)
Aldehydes/metabolism , Ethanol/toxicity , Liver Diseases, Alcoholic/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Proteomics , Animals , Chronic Disease , Liver Diseases, Alcoholic/pathology , Male , Mitochondria, Liver/pathology , Rats , Rats, Sprague-Dawley
8.
Biochim Biophys Acta ; 1840(2): 913-22, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24021887

ABSTRACT

BACKGROUND: Redox signaling is an important emerging mechanism of cellular function. Dysfunctional redox signaling is increasingly implicated in numerous pathologies, including atherosclerosis, diabetes, and cancer. The molecular messengers in this type of signaling are reactive species which can mediate the post-translational modification of specific groups of proteins, thereby effecting functional changes in the modified proteins. Electrophilic compounds comprise one class of reactive species which can participate in redox signaling. Electrophiles modulate cell function via formation of covalent adducts with proteins, particularly cysteine residues. SCOPE OF REVIEW: This review will discuss the commonly used methods of detection for electrophile-sensitive proteins, and will highlight the importance of identifying these proteins for studying redox signaling and developing novel therapeutics. MAJOR CONCLUSIONS: There are several methods which can be used to detect electrophile-sensitive proteins. These include the use of tagged model electrophiles, as well as derivatization of endogenous electrophile-protein adducts. GENERAL SIGNIFICANCE: In order to understand the mechanisms by which electrophiles mediate redox signaling, it is necessary to identify electrophile-sensitive proteins and quantitatively assess adduct formation. Strengths and limitations of these methods will be discussed. This article is part of a Special Issue entitled Current methods to study reactive oxygen species - pros and cons and biophysics of membrane proteins. Guest Editor: Christine Winterbourn.


Subject(s)
Proteins/analysis , Proteins/chemistry , Reactive Oxygen Species/metabolism , Animals , Humans , Oxidation-Reduction
9.
Cancer Res ; 74(3): 954-63, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24351292

ABSTRACT

Cancer cells tend to utilize aerobic glycolysis even under normoxic conditions, commonly called the "Warburg effect." Aerobic glycolysis often directly correlates with malignancy, but its purpose, if any, in metastasis remains unclear. When wild-type KISS1 metastasis suppressor is expressed, aerobic glycolysis decreases and oxidative phosphorylation predominates. However, when KISS1 is missing the secretion signal peptide (ΔSS), invasion and metastasis are no longer suppressed and cells continue to metabolize using aerobic glycolysis. KISS1-expressing cells have 30% to 50% more mitochondrial mass than ΔSS-expressing cells, which are accompanied by correspondingly increased mitochondrial gene expression and higher expression of PGC1α, a master coactivator that regulates mitochondrial mass and metabolism. PGC1α-mediated downstream pathways (i.e., fatty acid synthesis and ß-oxidation) are differentially regulated by KISS1, apparently reliant upon direct KISS1 interaction with NRF1, a major transcription factor involved in mitochondrial biogenesis. Since the downstream effects could be reversed using short hairpin RNA to KISS1 or PGC1α, these data appear to directly connect changes in mitochondria mass, cellular glucose metabolism, and metastasis.


Subject(s)
Kisspeptins/genetics , Mitochondria/genetics , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Extracellular Space/metabolism , Female , Gene Expression , Glucose/metabolism , Glycolysis , Humans , Hydrogen-Ion Concentration , Kisspeptins/metabolism , Lactic Acid/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Redox Biol ; 1: 381-6, 2013.
Article in English | MEDLINE | ID: mdl-24024175

ABSTRACT

Hypercholesterolemia is a risk factor for the development of hypertrophic cardiomyopathy. Nevertheless, there are few studies aimed at determining the effects of dietary compounds on early or mild cardiac hypertrophy associated with dyslipidemia. Here we describe left ventricular (LV) hypertrophy in 12 week-old Apo E(-/-) hypercholesterolemic mice. The LV end diastolic posterior wall thickness and overall LV mass were significantly increased in Apo E(-/-) mice compared with wild type (WT) controls. Fractional shortening, LV end diastolic diameter, and hemodynamic parameters were unchanged from WT mice. Oral low dose quercetin (QCN; 0.1 µmol QCN/kg body weight for 6 weeks) significantly reduced total cholesterol and very low density lipoprotein in the plasma of Apo E(-/-) mice. QCN treatment also significantly decreased LV posterior wall thickness and LV mass in Apo E(-/-) mice. Myocardial geometry and function were unaffected in WT mice by QCN treatment. These data suggest that dietary polyphenolic compounds such as QCN may be effective modulators of plasma cholesterol and could prevent maladaptive myocardial remodeling.


Subject(s)
Antioxidants/administration & dosage , Apolipoproteins E/genetics , Hypercholesterolemia/diet therapy , Hypertrophy, Left Ventricular/diet therapy , Quercetin/administration & dosage , Animals , Antioxidants/therapeutic use , Cholesterol/blood , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Hypertrophy, Left Ventricular/complications , Hypertrophy, Left Ventricular/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Quercetin/therapeutic use , Ventricular Remodeling/drug effects
12.
PLoS One ; 8(6): e68348, 2013.
Article in English | MEDLINE | ID: mdl-23840849

ABSTRACT

Solid tumors are characterized by regions of low oxygen tension (OT), which play a central role in tumor progression and resistance to therapy. Low OT affects mitochondrial function and for the cells to survive, mitochondria must functionally adapt to low OT to maintain the cellular bioenergetics. In this study, a novel experimental approach was developed to examine the real-time bioenergetic changes in breast cancer cells (BCCs) during adaptation to OT (from 20% to <1% oxygen) using sensitive extracellular flux technology. Oxygen was gradually removed from the medium, and the bioenergetics of metastatic BCCs (MDA-MB-231 and MCF10CA clones) was compared with non-tumorigenic (MCF10A) cells. BCCs, but not MCF10A, rapidly responded to low OT by stabilizing HIF-1α and increasing HIF-1α responsive gene expression and glucose uptake. BCCs also increased extracellular acidification rate (ECAR), which was markedly lower in MCF10A. Interestingly, BCCs exhibited a biphasic response in basal respiration as the OT was reduced from 20% to <1%. The initial stimulation of oxygen consumption is found to be due to increased mitochondrial respiration. This effect was HIF-1α-dependent, as silencing HIF-1α abolished the biphasic response. During hypoxia and reoxygenation, BCCs also maintained oxygen consumption rates at specific OT; however, HIF-1α silenced BCC were less responsive to changes in OT. Our results suggest that HIF-1α provides a high degree of bioenergetic flexibility under different OT which may confer an adaptive advantage for BCC survival in the tumor microenvironment and during invasion and metastasis. This study thus provides direct evidence for the cross-talk between HIF-1α and mitochondria during adaptation to low OT by BCCs and may be useful in identifying novel therapeutic agents that target the bioenergetics of BCCs in response to low OT.


Subject(s)
Breast Neoplasms/physiopathology , Energy Metabolism/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mitochondria/pathology , Mitochondria/physiology , Neoplasm Metastasis/physiopathology , Oxygen/metabolism , Adaptation, Biological/genetics , Adaptation, Biological/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Respiration/genetics , Cell Respiration/physiology , Energy Metabolism/genetics , Female , Glucose/metabolism , Glycolysis/genetics , Glycolysis/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Neoplasm Metastasis/genetics , Oxygen Consumption/genetics , Oxygen Consumption/physiology
13.
Am J Physiol Renal Physiol ; 305(3): F255-64, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23720344

ABSTRACT

Mitochondria are both a source and target of the actions of reactive oxygen species and possess a complex system of inter-related antioxidants that control redox signaling and protect against oxidative stress. Interestingly, the antioxidant enzyme heme oxygenase-1 (HO-1) is not present in the mitochondria despite the fact that the organelle is the site of heme synthesis and contains multiple heme proteins. Detoxification of heme is an important protective mechanism since the reaction of heme with hydrogen peroxide generates pro-oxidant ferryl species capable of propagating oxidative stress and ultimately cell death. We therefore hypothesized that a mitochondrially localized HO-1 would be cytoprotective. To test this, we generated a mitochondria-targeted HO-1 cell line by transfecting HEK293 cells with a plasmid construct containing the manganese superoxide dismutase mitochondria leader sequence fused to HO-1 cDNA (Mito-HO-1). Nontargeted HO-1-overexpressing cells were generated by transfecting HO-1 cDNA (HO-1) or empty vector (Vector). Mitochondrial localization of HO-1 with increased HO activity in the mitochondrial fraction of Mito-HO-1 cells was observed, but a significant decrease in the expression of heme-containing proteins occurred in these cells. Both cytosolic HO-1- and Mito-HO-1-expressing cells were protected against hypoxia-dependent cell death and loss of mitochondrial membrane potential, but these effects were more pronounced with Mito-HO-1. Furthermore, decrement in production of tricarboxylic acid cycle intermediates following hypoxia was significantly mitigated in Mito-HO-1 cells. These data suggest that specific mitochondrially targeted HO-1 under acute pathological conditions may have beneficial effects, but the selective advantage of long-term expression is constrained by a negative impact on the synthesis of heme-containing mitochondrial proteins.


Subject(s)
Epithelial Cells/metabolism , Heme Oxygenase-1/metabolism , Kidney/metabolism , Mitochondria/enzymology , Aerobiosis/physiology , Blotting, Western , Cell Survival/drug effects , Citrate (si)-Synthase/metabolism , Citric Acid Cycle/physiology , Cytochromes c/metabolism , Electron Transport Complex IV/metabolism , Epithelial Cells/enzymology , HEK293 Cells , Heme Oxygenase-1/physiology , Humans , Immunohistochemistry , Kidney/cytology , Kidney/enzymology , Membrane Potential, Mitochondrial/physiology , Oxidative Stress/physiology , Plasmids/genetics , Plasmids/physiology , Voltage-Dependent Anion Channels/metabolism
14.
Front Physiol ; 3: 369, 2012.
Article in English | MEDLINE | ID: mdl-23049513

ABSTRACT

There are a wide variety of reactive species which can affect cell function, including reactive oxygen, nitrogen, and lipid species. Some are formed endogenously through enzymatic or non-enzymatic pathways, and others are introduced through diet or environmental exposure. Many of these reactive species can interact with biomolecules and can result in oxidative post-translational modification of proteins. It is well documented that some oxidative modifications cause macromolecular damage and cell death. However, a growing body of evidence suggests that certain classes of reactive species initiate cell signaling by reacting with specific side chains of peptide residues without causing cell death. This process is generally termed "redox signaling," and its role in physiological and pathological processes is a subject of active investigation. This review will give an overview of oxidative protein modification as a mechanism of redox signaling, including types of reactive species and how they modify proteins, examples of modified proteins, and a discussion about the current concepts in this area.

15.
Biochem J ; 442(3): 453-64, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22364280

ABSTRACT

The process of lipid peroxidation is widespread in biology and is mediated through both enzymatic and non-enzymatic pathways. A significant proportion of the oxidized lipid products are electrophilic in nature, the RLS (reactive lipid species), and react with cellular nucleophiles such as the amino acids cysteine, lysine and histidine. Cell signalling by electrophiles appears to be limited to the modification of cysteine residues in proteins, whereas non-specific toxic effects involve modification of other nucleophiles. RLS have been found to participate in several physiological pathways including resolution of inflammation, cell death and induction of cellular antioxidants through the modification of specific signalling proteins. The covalent modification of proteins endows some unique features to this signalling mechanism which we have termed the 'covalent advantage'. For example, covalent modification of signalling proteins allows for the accumulation of a signal over time. The activation of cell signalling pathways by electrophiles is hierarchical and depends on a complex interaction of factors such as the intrinsic chemical reactivity of the electrophile, the intracellular domain to which it is exposed and steric factors. This introduces the concept of electrophilic signalling domains in which the production of the lipid electrophile is in close proximity to the thiol-containing signalling protein. In addition, we propose that the role of glutathione and associated enzymes is to insulate the signalling domain from uncontrolled electrophilic stress. The persistence of the signal is in turn regulated by the proteasomal pathway which may itself be subject to redox regulation by RLS. Cell death mediated by RLS is associated with bioenergetic dysfunction, and the damaged proteins are probably removed by the lysosome-autophagy pathway.


Subject(s)
Signal Transduction , Animals , Autophagy , Cell Death , Humans , Lipid Metabolism , Lipid Peroxidation , Oxidation-Reduction , Stress, Physiological
16.
Antioxid Redox Signal ; 17(11): 1580-9, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22352679

ABSTRACT

SIGNIFICANCE: The process of lipid peroxidation is emerging as an important mechanism that mediates the post-translational modification of proteins. Through advanced analytical techniques, lipidomics is now emerging as a critical factor in our understanding of the pathology of a broad range of diseases. RECENT ADVANCES: During enzymatic or nonenzymatic lipid peroxidation, the simple structure of an unsaturated fatty acid is converted to an oxylipidome, many members of which are electrophilic and form the reactive lipid species (RLS). This aspect of lipid biology is particularly important, as it directly connects lipidomics with proteomics through the post-translational modification of a sub-proteome in the cell. This arises, because the electrophilic members of the oxylipidome react with proteins at nucleophilic amino-acid residues and so change their structure and function to form electrophile-responsive proteomes (ERP). CRITICAL ISSUES: Biological systems have relatively few but well-defined and mechanistically distinct pro-oxidant pathways generating RLS. Defining the ERPs and the mechanisms underlying their formation and action has been a major focus for the field of lipidomics and redox signaling. FUTURE DIRECTIONS: We propose that a unique oxylipidome can be defined for specific oxidants and will predict the biological responses through the reaction with proteins to form a specific ERP. In this review, we will describe the ERPs that modulate antioxidant and anti-inflammatory protective pathways, including the activation of Keap1/Nrf2 and the promotion of cell death through interactions with mitochondria.


Subject(s)
Lipid Peroxidation , Mitochondria/metabolism , Protein Processing, Post-Translational , Proteins , Antioxidants/metabolism , Cell Death , Fatty Acids, Unsaturated/metabolism , Humans , Oxidation-Reduction , Proteins/analysis , Proteins/metabolism , Proteome , Signal Transduction
17.
Am J Physiol Heart Circ Physiol ; 302(7): H1394-409, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22245770

ABSTRACT

The hemolysis of red blood cells and muscle damage results in the release of the heme proteins myoglobin, hemoglobin, and free heme into the vasculature. The mechanisms of heme toxicity are not clear but may involve lipid peroxidation, which we hypothesized would result in mitochondrial damage in endothelial cells. To test this, we used bovine aortic endothelial cells (BAEC) in culture and exposed them to hemin. Hemin led to mitochondrial dysfunction, activation of autophagy, mitophagy, and, at high concentrations, apoptosis. To detect whether hemin induced lipid peroxidation and damaged proteins, we used derivatives of arachidonic acid tagged with biotin or Bodipy (Bt-AA, BD-AA). We found that in cells treated with hemin, Bt-AA was oxidized and formed adducts with proteins, which were inhibited by α-tocopherol. Hemin-dependent mitochondrial dysfunction was also attenuated by α-tocopherol. Protein thiol modification and carbonyl formation occurred on exposure and was not inhibited by α-tocopherol. Supporting a protective role of autophagy, the inhibitor 3-methyladenine potentiated cell death. These data demonstrate that hemin mediates cytotoxicity through a mechanism which involves protein modification by oxidized lipids and other oxidants, decreased respiratory capacity, and a protective role for the autophagic process. Attenuation of lipid peroxidation may be able to preserve mitochondrial function in the endothelium and protect cells from heme-dependent toxicity.


Subject(s)
Autophagy/physiology , Endothelial Cells/drug effects , Hemin/pharmacology , Lipid Peroxidation/drug effects , Mitochondrial Myopathies/chemically induced , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Blotting, Western , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Dogs , Energy Metabolism/drug effects , Extracellular Fluid/metabolism , Fluorescent Dyes , Green Fluorescent Proteins/metabolism , Indicators and Reagents , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondrial Myopathies/pathology , Protein Processing, Post-Translational/physiology
18.
FEBS Lett ; 586(1): 27-31, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22119728

ABSTRACT

Gap junctional intercellular communication (GJIC) regulates cellular homeostasis by propagating signaling molecules, exchanging cellular metabolites, and coupling electrical signals. In cancer, cells exhibit altered rates of GJIC which may play a role in neoplastic progression. K(ATP) channels help maintain membrane polarity and linkages between K(ATP) channel activity and rates of GJIC have been established. The mechanistic relationship has not been fully elucidated. We report the effects of treatment with multiple K(ATP) antagonist compounds on GJIC in metastatic cell lines demonstrating an increase in communication rates following treatment with compounds possessing specificities towards the SUR2 subunit of K(ATP). These effects remained consistent using cell lines with different expression levels of SUR1 and SUR2, suggesting possible off target effects on GJIC by these compounds.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , KATP Channels/antagonists & inhibitors , Mediator Complex/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , Cell Communication/drug effects , Cell Line, Tumor/pathology , Gap Junctions/metabolism , Humans , Neoplasm Metastasis , Sulfonylurea Receptors
19.
Biochim Biophys Acta ; 1807(12): 1573-82, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21971515

ABSTRACT

Chronic alcohol consumption results in hepatotoxicity, steatosis, hypoxia, increased expression of inducible nitric oxide synthase (iNOS) and decreased activities of mitochondrial respiratory enzymes. The impact of these changes on cellular respiration and their interaction in a cellular setting is not well understood. In the present study we tested the hypothesis that nitric oxide (NO)-dependent modulation of cellular respiration and the sensitivity to hypoxic stress is increased following chronic alcohol consumption. This is important since NO has been shown to regulate mitochondrial function through its interaction with cytochrome c oxidase, although at higher concentrations, and in combination with reactive oxygen species, can result in mitochondrial dysfunction. We found that hepatocytes isolated from alcohol-fed rats had decreased mitochondrial bioenergetic reserve capacity and were more sensitive to NO-dependent inhibition of respiration under room air and hypoxic conditions. We reasoned that this would result in greater hypoxic stress in vivo, and to test this, wild-type and iNOS(-/-) mice were administered alcohol-containing diets. Chronic alcohol consumption resulted in liver hypoxia in the wild-type mice and increased levels of hypoxia-inducible factor 1 α in the peri-venular region of the liver lobule. These effects were attenuated in the alcohol-fed iNOS(-/-) mice suggesting that increased mitochondrial sensitivity to NO and reactive nitrogen species in hepatocytes and iNOS plays a critical role in determining the response to hypoxic stress in vivo. These data support the concept that the combined effects of NO and ethanol contribute to an increased susceptibility to hypoxia and the deleterious effects of alcohol consumption on liver.


Subject(s)
Ethanol/pharmacology , Hepatocytes/metabolism , Hypoxia/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/physiology , Nitric Oxide/metabolism , Animals , Cell Respiration/drug effects , Cell Respiration/physiology , Diet , Energy Metabolism/drug effects , Ethanol/administration & dosage , Hepatocytes/cytology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley
20.
Free Radic Biol Med ; 51(9): 1621-35, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21872656

ABSTRACT

It is now clear that mitochondria are an important target for oxidative stress in a broad range of pathologies, including cardiovascular disease, diabetes, neurodegeneration, and cancer. Methods for assessing the impact of reactive species on isolated mitochondria are well established but constrained by the need for large amounts of material to prepare intact mitochondria for polarographic measurements. With the availability of high-resolution polarography and fluorescence techniques for the measurement of oxygen concentration in solution, measurements of mitochondrial function in intact cells can be made. Recently, the development of extracellular flux methods to monitor changes in oxygen concentration and pH in cultures of adherent cells in multiple-sample wells simultaneously has greatly enhanced the ability to measure bioenergetic function in response to oxidative stress. Here we describe these methods in detail using representative cell types from renal, cardiovascular, nervous, and tumorigenic model systems while illustrating the application of three protocols to analyze the bioenergetic response of cells to oxidative stress.


Subject(s)
Energy Metabolism , Mitochondria/metabolism , Oxidative Stress , Animals , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Oxygen/analysis , Oxygen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL