Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Redox Biol ; 64: 102794, 2023 08.
Article in English | MEDLINE | ID: mdl-37402332

ABSTRACT

Continued oxidant production during chronic inflammation generates host tissue damage, with this being associated with pathologies including atherosclerosis. Atherosclerotic plaques contain modified proteins that may contribute to disease development, including plaque rupture, the major cause of heart attacks and strokes. Versican, a large extracellular matrix (ECM) chondroitin-sulfate proteoglycan, accumulates during atherogenesis, where it interacts with other ECM proteins, receptors and hyaluronan, and promotes inflammation. As activated leukocytes produce oxidants including peroxynitrite/peroxynitrous acid (ONOO-/ONOOH) at sites of inflammation, we hypothesized that versican is an oxidant target, with this resulting in structural and functional changes that may exacerbate plaque development. The recombinant human V3 isoform of versican becomes aggregated on exposure to ONOO-/ONOOH. Both reagent ONOO-/ONOOH and SIN-1 (a thermal source of ONOO-/ONOOH) modified Tyr, Trp and Met residues. ONOO-/ONOOH mainly favors nitration of Tyr, whereas SIN-1 mostly induced hydroxylation of Tyr, and oxidation of Trp and Met. Peptide mass mapping indicated 26 sites with modifications (15 Tyr, 5 Trp, 6 Met), with the extent of modification quantified at 16. Multiple modifications, including the most extensively nitrated residue (Tyr161), are within the hyaluronan-binding region, and associated with decreased hyaluronan binding. ONOO-/ONOOH modification also resulted in decreased cell adhesion and increased proliferation of human coronary artery smooth muscle cells. Evidence is also presented for colocalization of versican and 3-nitrotyrosine epitopes in advanced (type II-III) human atherosclerotic plaques. In conclusion, versican is readily modified by ONOO-/ONOOH, resulting in chemical and structural modifications that affect protein function, including hyaluronan binding and cell interactions.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Oxidants/metabolism , Peroxynitrous Acid/metabolism , Versicans/genetics , Versicans/metabolism , Hyaluronic Acid/metabolism , Plaque, Atherosclerotic/metabolism , Extracellular Matrix/metabolism , Atherosclerosis/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Inflammation/metabolism
2.
Antioxidants (Basel) ; 12(2)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36829979

ABSTRACT

Atherosclerosis is a chronic inflammatory disease and a leading cause of mortality. It is characterized by arterial wall plaques that contain high levels of cholesterol and other lipids and activated leukocytes covered by a fibrous cap of extracellular matrix (ECM). The ECM undergoes remodelling during atherogenesis, with increased expression of aggrecan, a proteoglycan that binds low-density-lipoproteins (LDL). Aggrecan levels are regulated by proteases, including a disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1). Activated leukocytes release myeloperoxidase (MPO) extracellularly, where it binds to proteins and proteoglycans. Aggrecan may therefore mediate colocalization of MPO and LDL. MPO generates hypochlorous acid (HOCl) and chloramines (RNHCl species, from reaction of HOCl with amines on amino acids and proteins) that damage LDL and proteins, but effects on aggrecan have not been examined. The present study demonstrates that HOCl cleaves truncated (G1-IGD-G2) recombinant human aggrecan at specific sites within the IGD domain, with these being different from those induced by ADAMTS1 which also cleaves within this region. Irreversible protein cross-links are also formed dose-dependently. These effects are limited by the HOCl scavenger methionine. Chloramines including those formed on amino acids, proteins, and ECM materials induce similar damage. HOCl and taurine chloramines inactivate ADAMTS1 consistent with a switch from proteolytic to oxidative aggrecan fragmentation. Evidence is also presented for colocalization of aggrecan and HOCl-generated epitopes in advanced human atherosclerotic plaques. Overall, these data show that HOCl and chloramines can induce specific modifications on aggrecan, and that these effects are distinct from those of ADAMTS1.

3.
Int J Mol Sci ; 21(23)2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33287422

ABSTRACT

Sepsis is a major cause of mortality in critically ill patients and associated with cardiac dysfunction, a complication linked to immunological and metabolic aberrations. Cardiac neutrophil infiltration and subsequent release of myeloperoxidase (MPO) leads to the formation of the oxidant hypochlorous acid (HOCl) that is able to chemically modify plasmalogens (ether-phospholipids) abundantly present in the heart. This reaction gives rise to the formation of reactive lipid species including aldehydes and chlorinated fatty acids. During the present study, we tested whether endotoxemia increases MPO-dependent lipid oxidation/modification in the mouse heart. In hearts of lipopolysaccharide-injected mice, we observed significantly higher infiltration of MPO-positive cells, increased fatty acid content, and formation of 2-chlorohexadecanal (2-ClHDA), an MPO-derived plasmalogen modification product. Using murine HL-1 cardiomyocytes as in vitro model, we show that exogenously added HOCl attacks the cellular plasmalogen pool and gives rise to the formation of 2-ClHDA. Addition of 2-ClHDA to HL-1 cardiomyocytes resulted in conversion to 2-chlorohexadecanoic acid and 2-chlorohexadecanol, indicating fatty aldehyde dehydrogenase-mediated redox metabolism. However, a recovery of only 40% indicated the formation of non-extractable (protein) adducts. To identify protein targets, we used a clickable alkynyl analog, 2-chlorohexadec-15-yn-1-al (2-ClHDyA). After Huisgen 1,3-dipolar cycloaddition of 5-tetramethylrhodamine azide (N3-TAMRA) and two dimensional-gel electrophoresis (2D-GE), we were able to identify 51 proteins that form adducts with 2-ClHDyA. Gene ontology enrichment analyses revealed an overrepresentation of heat shock and chaperone, energy metabolism, and cytoskeletal proteins as major targets. Our observations in a murine endotoxemia model demonstrate formation of HOCl-modified lipids in the heart, while pathway analysis in vitro revealed that the chlorinated aldehyde targets specific protein subsets, which are central to cardiac function.


Subject(s)
Aldehydes/metabolism , Endotoxemia/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Peroxidase/metabolism , Animals , Biomarkers , Click Chemistry , Endotoxemia/etiology , Fatty Acids/metabolism , Hypochlorous Acid/metabolism , Lipopolysaccharides/administration & dosage , Mice , Proteome , Proteomics/methods , Reactive Oxygen Species/metabolism
4.
Free Radic Biol Med ; 136: 118-134, 2019 05 20.
Article in English | MEDLINE | ID: mdl-30959171

ABSTRACT

Dysfunction of endothelial cells of the artery wall is an early event in cardiovascular disease and atherosclerosis. The cause(s) of this dysfunction are unresolved, but accumulating evidence suggests that oxidants arising from chronic low-grade inflammation are contributory agents, with increasing data implicating myeloperoxidase (MPO, released by activated leukocytes), and the oxidants it generates (e.g. HOCl and HOSCN). As these are formed extracellularly and react rapidly with proteins, we hypothesized that MPO-mediated damage to the matrix glycoprotein fibronectin (FN) would modulate FN structure and function, and its interactions with human coronary artery endothelial cells (HCAEC). Exposure of human plasma FN to HOCl resulted in modifications to FN and its functional epitopes. A dose-dependent loss of methionine and tryptophan residues, together with increasing concentrations of methionine sulfoxide, and modification of the cell-binding fragment (CBF) and heparin-binding fragment (HBF) domains was detected with HOCl, but not HOSCN. FN modification resulted in a loss of HCAEC adhesion, impaired cell spreading and reduced cell proliferation. Exposure to HCAEC to HOCl-treated FN altered the expression of HCAEC genes associated with extracellular matrix (ECM) synthesis and adhesion. Modifications were detected on HCAEC-derived ECM pre-treated with HOCl, but not HOSCN, with a loss of antibody recognition of the CBF, HBF and extra-domain A. Co-localization of epitopes arising from MPO-generated HOCl and cell-derived FN was detected in human atherosclerotic lesions. Damage was also detected on FN extracted from lesions. These data support the hypothesis that HOCl, but not HOSCN, targets and modifies FN resulting in arterial wall endothelial cell dysfunction.


Subject(s)
Atherosclerosis , Endothelial Cells/metabolism , Fibronectins/metabolism , Inflammation/metabolism , Oxidants/toxicity , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Fibronectins/drug effects , Humans , Inflammation/pathology , Inflammation/physiopathology , Oxidation-Reduction , Peroxidase/toxicity
5.
Redox Biol ; 20: 496-513, 2019 01.
Article in English | MEDLINE | ID: mdl-30476874

ABSTRACT

Basement membranes are specialized extracellular matrices that underlie arterial wall endothelial cells, with laminin being a key structural and biologically-active component. Hypochlorous acid (HOCl), a potent oxidizing and chlorinating agent, is formed in vivo at sites of inflammation via the enzymatic action of myeloperoxidase (MPO), released by activated leukocytes. Considerable data supports a role for MPO-derived oxidants in cardiovascular disease and particularly atherosclerosis. These effects may be mediated via extracellular matrix damage to which MPO binds. Herein we detect and quantify sites of oxidation and chlorination on isolated laminin-111, and laminin in basement membrane extracts (BME), by use of mass spectrometry. Increased modification was detected with increasing oxidant exposure. Mass mapping indicated selectivity in the sites and extent of damage; Met residues were most heavily modified. Fewer modifications were detected with BME, possibly due to the shielding effects. HOCl oxidised 30 (of 56 total) Met and 7 (of 24) Trp residues, and chlorinated 33 (of 99) Tyr residues; 3 Tyr were dichlorinated. An additional 8 Met and 10 Trp oxidations, 14 chlorinations, and 18 dichlorinations were detected with the MPO/H2O2/Cl- system when compared to reagent HOCl. Interestingly, chlorination was detected at Tyr2415 in the integrin-binding region; this may decrease cellular adhesion. Co-localization of MPO-damaged epitopes and laminin was detected in human atherosclerotic lesions. These data indicate that laminin is extensively modified by MPO-derived oxidants, with structural and functional changes. These modifications, and compromised cell-matrix interactions, may promote endothelial cell dysfunction, weaken the structure of atherosclerotic lesions, and enhance lesion rupture.


Subject(s)
Basement Membrane/metabolism , Chlorine/metabolism , Hypochlorous Acid/metabolism , Laminin/metabolism , Oxidation-Reduction , Peroxidase/metabolism , Amino Acids/metabolism , Amino Acids, Diamino/metabolism , Animals , Extracellular Matrix Proteins , Humans , Hydrogen Peroxide/metabolism , Mice
6.
Redox Biol ; 15: 441-451, 2018 05.
Article in English | MEDLINE | ID: mdl-29413957

ABSTRACT

Peripheral leukocytes induce blood-brain barrier (BBB) dysfunction through the release of cytotoxic mediators. These include hypochlorous acid (HOCl) that is formed via the myeloperoxidase-H2O2-chloride system of activated phagocytes. HOCl targets the endogenous pool of ether phospholipids (plasmalogens) generating chlorinated inflammatory mediators like e.g. 2-chlorohexadecanal and its conversion product 2-chlorohexadecanoic acid (2-ClHA). In the cerebrovasculature these compounds inflict damage to brain microvascular endothelial cells (BMVEC) that form the morphological basis of the BBB. To follow subcellular trafficking of 2-ClHA we synthesized a 'clickable' alkyne derivative (2-ClHyA) that phenocopied the biological activity of the parent compound. Confocal and superresolution structured illumination microscopy revealed accumulation of 2-ClHyA in the endoplasmic reticulum (ER) and mitochondria of human BMVEC (hCMEC/D3 cell line). 2-ClHA and its alkyne analogue interfered with protein palmitoylation, induced ER-stress markers, reduced the ER ATP content, and activated transcription and secretion of interleukin (IL)-6 as well as IL-8. 2-ClHA disrupted the mitochondrial membrane potential and induced procaspase-3 and PARP cleavage. The protein kinase R-like ER kinase (PERK) inhibitor GSK2606414 suppressed 2-ClHA-mediated activating transcription factor 4 synthesis and IL-6/8 secretion, but showed no effect on endothelial barrier dysfunction and cleavage of procaspase-3. Our data indicate that 2-ClHA induces potent lipotoxic responses in brain endothelial cells and could have implications in inflammation-induced BBB dysfunction.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Mitochondria/drug effects , Palmitic Acids/adverse effects , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/pathology , Cell Line , Endoplasmic Reticulum Stress/drug effects , Humans , Indoles/pharmacology , Interleukin-8/genetics , Leukocytes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/pathology , Palmitic Acids/metabolism , Peroxidase/metabolism , eIF-2 Kinase/genetics
7.
Free Radic Biol Med ; 115: 219-231, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29191462

ABSTRACT

Elastin is an abundant extracellular matrix protein in elastic tissues, including the lungs, skin and arteries, and comprises 30-57% of the aorta by dry mass. The monomeric precursor, tropoelastin (TE), undergoes complex processing during elastogenesis to form mature elastic fibres. Peroxynitrous acid (ONOOH), a potent oxidising and nitrating agent, is formed in vivo from superoxide and nitric oxide radicals. Considerable evidence supports ONOOH formation in the inflamed artery wall, and a role for this species in the development of human atherosclerotic lesions, with ONOOH-damaged extracellular matrix implicated in lesion rupture. We demonstrate that TE is highly sensitive to ONOOH, with this resulting in extensive dimerization, fragmentation and nitration of Tyr residues to give 3-nitrotyrosine (3-nitroTyr). This occurs with equimolar or greater levels of oxidant and increases in a dose-dependent manner. Quantification of Tyr loss and 3-nitroTyr formation indicates extensive Tyr modification with up to two modified Tyr per protein molecule, and up to 8% conversion of initial ONOOH to 3-nitroTyr. These effects were modulated by bicarbonate, an alternative target for ONOOH. Inter- and intra-protein di-tyrosine cross-links have been characterized by mass spectrometry. Examination of human atherosclerotic lesions shows colocalization of 3-nitroTyr with elastin epitopes, consistent with TE or elastin modification in vivo, and also an association of 3-nitroTyr containing proteins and elastin with lipid deposits. These data suggest that exposure of TE to ONOOH gives marked chemical and structural changes to TE and altered matrix assembly, and that such damage accumulates in human arterial tissue during the development of atherosclerosis.


Subject(s)
Arteries/physiology , Atherosclerosis/metabolism , Inflammation/metabolism , Macrophages/physiology , Peroxynitrous Acid/metabolism , Tropoelastin/metabolism , Tyrosine/metabolism , Atherosclerosis/pathology , Cadaver , Elastin/metabolism , Extracellular Matrix/metabolism , Humans , Immunohistochemistry , Inflammation/pathology , Nitro Compounds/metabolism , Oxidation-Reduction , Protein Conformation , Tyrosine/analogs & derivatives , Unfolded Protein Response
8.
Biochem Pharmacol ; 145: 64-80, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28859968

ABSTRACT

Saxagliptin treatment has been associated with increased rate of hospitalization for heart failure in type 2 diabetic patients, though the underlying mechanism(s) remain elusive. To address this, we assessed the effects of saxagliptin on human atrial trabeculae, guinea pig hearts and cardiomyocytes. We found that the primary target of saxagliptin, dipeptidyl peptidase-4, is absent in cardiomyocytes, yet saxagliptin internalized into cardiomyocytes and impaired cardiac contractility via inhibition of the Ca2+/calmodulin-dependent protein kinase II-phospholamban-sarcoplasmic reticulum Ca2+-ATPase 2a axis and Na+-Ca2+ exchanger function in Ca2+ extrusion. This resulted in reduced sarcoplasmic reticulum Ca2+ content, diastolic Ca2+ overload, systolic dysfunction and impaired contractile force. Furthermore, saxagliptin reduced protein kinase C-mediated delayed rectifier K+ current that prolonged action potential duration and consequently QTc interval. Importantly, saxagliptin aggravated pre-existing cardiac dysfunction induced by ischemia/reperfusion injury. In conclusion, our novel results provide mechanisms for the off-target deleterious effects of saxagliptin on cardiac function and support the outcome of SAVOR-TIMI 53 trial that linked saxagliptin with the risk of heart failure.


Subject(s)
Adamantane/analogs & derivatives , Dipeptides/toxicity , Dipeptidyl Peptidase 4/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Heart Atria/cytology , Myocytes, Cardiac/drug effects , Adamantane/toxicity , Aged , Animals , Cell Line , Dipeptidyl Peptidase 4/genetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic/physiology , Heart Diseases/metabolism , Humans , Male , Mice , Middle Aged , Myocardial Contraction/drug effects , Myocytes, Cardiac/enzymology
9.
J Neuroinflammation ; 13(1): 205, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27565558

ABSTRACT

BACKGROUND: Microglia, the immunocompetent cells of the CNS, rapidly respond to brain injury and disease by altering their morphology and phenotype to adopt an activated state. Microglia can exist broadly between two different states, namely the classical (M1) and the alternative (M2) phenotype. The first is characterized by the production of pro-inflammatory cytokines/chemokines and reactive oxygen and/or nitrogen species. In contrast, alternatively activated microglia are typified by an anti-inflammatory phenotype supporting wound healing and debris clearance. The objective of the present study was to determine the outcome of lysophosphatidic acid (LPA)-mediated signaling events on microglia polarization. METHODS: LPA receptor expression and cyto-/chemokine mRNA levels in BV-2 and primary murine microglia (PMM) were determined by qPCR. M1/M2 marker expression was analyzed by Western blotting, immunofluorescence microscopy, or flow cytometry. Cyto-/chemokine secretion was quantitated by ELISA. RESULTS: BV-2 cells express LPA receptor 2 (LPA2), 3, 5, and 6, whereas PMM express LPA1, 2, 4, 5, and 6. We show that LPA treatment of BV-2 and PMM leads to a shift towards a pro-inflammatory M1-like phenotype. LPA treatment increased CD40 and CD86 (M1 markers) and reduced CD206 (M2 marker) expression. LPA increased inducible nitric oxide synthase (iNOS) and COX-2 levels (both M1), while the M2 marker Arginase-1 was suppressed in BV-2 cells. Immunofluorescence studies (iNOS, COX-2, Arginase-1, and RELMα) extended these findings to PMM. Upregulation of M1 markers in BV-2 and PMM was accompanied by increased cyto-/chemokine transcription and secretion (IL-1ß, TNFα, IL-6, CCL5, and CXCL2). The pharmacological LPA5 antagonist TCLPA5 blunted most of these pro-inflammatory responses. CONCLUSIONS: LPA drives BV-2 and PMM towards a pro-inflammatory M1-like phenotype. Suppression by TCLPA5 indicates that the LPA/LPA5 signaling axis could represent a potential pharmacological target to interfere with microglia polarization in disease.


Subject(s)
Cell Polarity/drug effects , Lysophospholipids/pharmacology , Microglia/classification , Microglia/drug effects , Actins/metabolism , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Time Factors
10.
Free Radic Biol Med ; 97: 602-615, 2016 08.
Article in English | MEDLINE | ID: mdl-27396946

ABSTRACT

Fibronectin is a large dimeric glycoprotein present in both human plasma and in basement membranes. The latter are specialized extracellular matrices underlying endothelial cells in the artery wall. Peroxynitrous acid (ONOOH) a potent oxidizing and nitrating agent, is formed in vivo from superoxide and nitric oxide radicals by stimulated macrophages and other cells. Considerable evidence supports ONOOH involvement in human atherosclerotic lesion development and rupture, possibly via extracellular matrix damage. Here we demonstrate that Tyr and Trp residues on human plasma fibronectin are highly sensitive to ONOOH with this resulting in the formation of 3-nitrotyrosine, 6-nitrotryptophan and dityrosine as well as protein aggregation and fragmentation. This occurs with equimolar or greater levels of oxidant, and in a dose-dependent manner. Modification of Tyr was quantitatively more significant than Trp (9.1% versus 1.5% conversion with 500µM ONOOH) after accounting for parent amino acid abundance, but only accounts for a small percentage of the total oxidant added. LC-MS studies identified 28 nitration sites (24 Tyr, 4 Trp) with many of these present within domains critical to protein function, including the cell-binding and anastellin domains. Human coronary artery endothelial cells showed decreased adherence and cell-spreading on ONOOH-modified fibronectin compared to control, consistent with cellular dysfunction induced by the modified matrix. Studies on human atherosclerotic lesions have provided evidence for co-localization of 3-nitrotyrosine and fibronectin. ONOOH-mediated fibronectin modification and compromised cell-matrix interactions, may contribute to endothelial cell dysfunction, a weakening of the fibrous cap of atherosclerotic lesions, and an increased propensity to rupture.


Subject(s)
Fibronectins/chemistry , Oxidants/chemistry , Peroxynitrous Acid/chemistry , Amino Acid Sequence , Atherosclerosis/metabolism , Atherosclerosis/pathology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Cell Adhesion , Cells, Cultured , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fibronectins/metabolism , Humans , Nitrates/chemistry , Oxidation-Reduction
11.
Free Radic Biol Med ; 89: 721-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453917

ABSTRACT

Basement membranes (BM) are specialized extracellular matrices underlying endothelial cells in the artery wall. Laminin, the most abundant BM glycoprotein, is a structural and biologically active component. Peroxynitrous acid (ONOOH), a potent oxidizing and nitrating agent, is formed in vivo at sites of inflammation from superoxide and nitric oxide radicals. Considerable data supports ONOOH formation in human atherosclerotic lesions, and an involvement of this oxidant in atherosclerosis development and lesion rupture. These effects may be mediated, at least in part, via extracellular matrix damage. In this study we demonstrate co-localization of 3-nitrotyrosine (a product of tyrosine damage by ONOOH) and laminin in human atherosclerotic lesions. ONOOH-induced damage to BM was characterized for isolated murine BM, and purified murine laminin-111. Exposure of laminin-111 to ONOOH resulted in dose-dependent loss of protein tyrosine and tryptophan residues, and formation of 3-nitrotyrosine, 6-nitrotryptophan and the cross-linked material di-tyrosine, as detected by amino acid analysis and Western blotting. These changes were accompanied by protein aggregation and fragmentation as detected by SDS-PAGE. Endothelial cell adhesion to isolated laminin-111 exposed to 10 µM or higher levels of ONOOH was significantly decreased (~25%) compared to untreated controls. These data indicate that laminin is oxidized by equimolar or greater concentrations of ONOOH, with this resulting in structural and functional changes. These modifications, and resulting compromised cell-matrix interactions, may contribute to endothelial cell dysfunction, a weakening of the structure of atherosclerotic lesions, and an increased propensity to rupture.


Subject(s)
Atherosclerosis/pathology , Basement Membrane/pathology , Endothelium, Vascular/pathology , Laminin/metabolism , Peroxynitrous Acid/metabolism , Animals , Atherosclerosis/metabolism , Blotting, Western , Cells, Cultured , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix , Humans , Immunohistochemistry , Mice , Microscopy, Confocal , Oxidants/metabolism , Oxidants/pharmacology , Oxidation-Reduction , Peroxynitrous Acid/pharmacology
12.
Eur J Immunol ; 45(5): 1548-59, 2015 May.
Article in English | MEDLINE | ID: mdl-25645675

ABSTRACT

Prostaglandin E2 (PGE2 ) protects against allergic responses via binding to prostanoid receptor EP4, which inhibits eosinophil migration in a PI3K/PKC-dependent fashion. The phosphoinositide-dependent protein kinase 1 (PDK1) is known to act as a downstream effector in PI3K signaling and has been implicated in the regulation of neutrophil migration. Thus, here we elucidate whether PDK1 mediates inhibitory effects of E-type prostanoid receptor 4 (EP4) receptors on eosinophil function. Therefore, eosinophils were isolated from human peripheral blood or differentiated from mouse BM. PDK1 signaling was investigated in shape change, chemotaxis, CD11b, respiratory burst, and Ca(2+) mobilization assays. The specific PDK1 inhibitors BX-912 and GSK2334470 prevented the inhibition by prostaglandin E2 and the EP4 agonist ONO-AE1-329. Depending on the cellular function, PDK1 seemed to act through PI3K-dependent or PI3K-independent mechanisms. Stimulation of EP4 receptors caused PDK1 phosphorylation at Ser396 and induced PI3K-dependent nuclear translocation of PDK1. EP4-induced inhibition of shape change and chemotaxis was effectively reversed by the Akt inhibitor triciribine. In support of this finding, ONO-AE1-329 induced a PI3K/PDK1-dependent increase in Akt phosphorylation. In conclusion, our data illustrate a critical role for PDK1 in transducing inhibitory signals on eosinophil effector function. Thus, our results suggest that PDK1 might serve as a novel therapeutic target in diseases involving eosinophilic inflammation.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Eosinophils/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , Active Transport, Cell Nucleus , Animals , CD11b Antigen/metabolism , Calcium Signaling , Cell Shape , Dinoprostone/metabolism , Eosinophils/drug effects , Eosinophils/immunology , Humans , Indazoles/pharmacology , Methyl Ethers/pharmacology , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Respiratory Burst , Ribonucleosides/pharmacology , Signal Transduction/drug effects
13.
Biochem J ; 459(2): 313-22, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24517414

ABSTRACT

ECM (extracellular matrix) materials, such as laminin, perlecan, type IV collagen and fibronectin, play a key role in determining the structure of the arterial wall and the properties of cells that interact with the ECM. The aim of the present study was to investigate the effect of peroxynitrous acid, an oxidant generated by activated macrophages, on the structure and function of the ECM laid down by HCAECs (human coronary artery endothelial cells) in vitro and in vivo. We show that exposure of HCAEC-derived native matrix components to peroxynitrous acid (but not decomposed oxidant) at concentrations >1 µM results in a loss of antibody recognition of perlecan, collagen IV, and cell-binding sites on laminin and fibronectin. Loss of recognition was accompanied by decreased HCAEC adhesion. Real-time PCR showed up-regulation of inflammation-associated genes, including MMP7 (matrix metalloproteinase 7) and MMP13, as well as down-regulation of the laminin α2 chain, in HCAECs cultured on peroxynitrous acid-treated matrix compared with native matrix. Immunohistochemical studies provided evidence of co-localization of laminin with 3-nitrotyrosine, a biomarker of peroxynitrous acid damage, in type II-III/IV human atherosclerotic lesions, consistent with matrix damage occurring during disease development in vivo. The results of the present study suggest a mechanism through which peroxynitrous acid modifies endothelial cell-derived native ECM proteins of the arterial basement membrane in atherosclerotic lesions. These changes to ECM and particularly perlecan and laminin may be important in inducing cellular dysfunction and contribute to atherogenesis.


Subject(s)
Coronary Vessels/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Peroxynitrous Acid/pharmacology , Atherosclerosis/metabolism , Cells, Cultured , Extracellular Matrix Proteins/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Oxidants/pharmacology , Oxidation-Reduction
14.
Neuro Oncol ; 16(7): 933-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24463355

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is a highly aggressive tumor of the central nervous system with a dismal prognosis for affected patients. Aberrant protein kinase C (PKC) signaling has been implicated in gliomagenesis, and a member of the PKC-activated protein kinase D (PRKD) family, PRKD2, was identified as mediator of GBM growth in vitro and in vivo. METHODS: The outcome of PRKD2 silencing and pharmacological inhibition on glioma cell proliferation was established with different glioma cell lines. Western blotting, senescence assays, co-immunoprecipitation, fluorescence activated cell sorting, quantitative PCR, and immunofluorescence microscopy were utilized to analyze downstream signaling. RESULTS: RNA-interference (21-mer siRNA) and pharmacological inhibition (CRT0066101) of PRKD2 profoundly inhibited proliferation of p53(wt) (U87MG, A172, and primary GBM2), and p53(mut) (GM133, T98G, U251, and primary Gli25) glioma cells. In a xenograft experiment, PRKD2 silencing significantly delayed tumor growth of U87MG cells. PRKD2 silencing in p53(wt) and p53(mut) cells was associated with typical hallmarks of senescence and cell cycle arrest in G1. Attenuated AKT/PKB phosphorylation in response to PRKD2 silencing was a common observation made in p53(wt) and p53(mut) GBM cells. PRKD2 knockdown in p53(wt) cells induced upregulation of p53, p21, and p27 expression, decreased phosphorylation of CDK2 and/or CDK4, hypophosphorylation of retinoblastoma protein (pRb), and reduced transcription of E2F1. In p53(mut) GM133 and primary Gli25 cells, PRKD2 silencing increased p27 and p15 and reduced E2F1 transcription but did not affect pRb phosphorylation. CONCLUSIONS: PRKD2 silencing induces glioma cell senescence via p53-dependent and -independent pathways.


Subject(s)
Brain Neoplasms/metabolism , Cellular Senescence/physiology , Glioma/metabolism , Protein Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Gene Silencing , Heterografts , Humans , Immunoprecipitation , Mice , Microscopy, Fluorescence , Protein Kinase D2 , RNA Interference , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Transfection
15.
Biochim Biophys Acta ; 1831(12): 1665-78, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973266

ABSTRACT

Neuronal sphingolipids (SL) play important roles during axonal extension, neurotrophic receptor signaling and neurotransmitter release. Many of these signaling pathways depend on the presence of specialized membrane microdomains termed lipid rafts. Sphingomyelin (SM), one of the main raft constituents, can be formed de novo or supplied from exogenous sources. The present study aimed to characterize fluorescently-labeled SL turnover in a murine neuronal cell line (CATH.a). Our results demonstrate that at 4°C exogenously added BODIPY-SM accumulates exclusively at the plasma membrane. Treatment of cells with bacterial sphingomyelinase (SMase) and back-exchange experiments revealed that 55-67% of BODIPY-SM resides in the outer leaflet of the plasma membrane. Endocytosis of BODIPY-SM occurs via caveolae with part of internalized BODIPY-fluorescence ending up in the Golgi and the ER. Following endocytosis BODIPY-SM undergoes hydrolysis, a reaction substantially faster than BODIPY-SM synthesis from BODIPY-ceramide. RNAi demonstrated that both, acid (a)SMase and neutral (n)SMases contribute to BODIPY-SM hydrolysis. Finally, high-density lipoprotein (HDL)-associated BODIPY-SM was efficiently taken up by CATH.a cells. Our findings indicate that endocytosis of exogenous SM occurs almost exclusively via caveolin-dependent pathways, that both, a- and nSMases equally contribute to neuronal SM turnover and that HDL-like particles might represent physiological SM carriers/donors in the brain.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Membrane Microdomains/metabolism , Neurons/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Animals , Boron Compounds , Caveolins/genetics , Caveolins/metabolism , Cell Line , Endocytosis , Endoplasmic Reticulum/drug effects , Fluorescent Dyes , Gene Expression Regulation , Golgi Apparatus/drug effects , Hydrolysis , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Lipoproteins, HDL/metabolism , Membrane Microdomains/drug effects , Mice , Neurons/cytology , Neurons/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelins/pharmacology , Temperature
16.
Cell Cycle ; 12(11): 1704-12, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23656787

ABSTRACT

Following microbial pathogen invasion, the human immune system of activated phagocytes generates and releases the potent oxidant hypochlorous acid (HOCl), which contributes to the killing of menacing microorganisms. Though tightly controlled, HOCl generation by the myeloperoxidase-hydrogen peroxide-chloride system of neutrophils/monocytes may occur in excess and lead to tissue damage. It is thus of marked importance to delineate the molecular pathways underlying HOCl cytotoxicity in both microbial and human cells. Here, we show that HOCl induces the generation of reactive oxygen species (ROS), apoptotic cell death and the formation of specific HOCl-modified epitopes in the budding yeast Saccharomyces cerevisiae. Interestingly, HOCl cytotoxicity can be prevented by treatment with ROS scavengers, suggesting oxidative stress to mediate the lethal effect. The executing pathway involves the pro-apoptotic protease Kex1p, since its absence diminishes HOCl-induced production of ROS, apoptosis and protein modification. By characterizing HOCl-induced cell death in yeast and identifying a corresponding central executor, these results pave the way for the use of Saccharomyces cerevisiae in HOCl research, not least given that it combines both being a microorganism as well as a model for programmed cell death in higher eukaryotes.


Subject(s)
Apoptosis/drug effects , Carboxypeptidases/metabolism , Hypochlorous Acid/toxicity , Saccharomyces cerevisiae/metabolism , Epitopes/chemistry , Epitopes/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism
17.
PLoS One ; 8(5): e64034, 2013.
Article in English | MEDLINE | ID: mdl-23691142

ABSTRACT

Peripheral leukocytes can exacerbate brain damage by release of cytotoxic mediators that disrupt blood-brain barrier (BBB) function. One of the oxidants released by activated leukocytes is hypochlorous acid (HOCl) formed via the myeloperoxidase (MPO)-H2O2-Cl(-) system. In the present study we examined the role of leukocyte activation, leukocyte-derived MPO and MPO-generated oxidants on BBB function in vitro and in vivo. In a mouse model of lipopolysaccharide (LPS)-induced systemic inflammation, neutrophils that had become adherent released MPO into the cerebrovasculature. In vivo, LPS-induced BBB dysfunction was significantly lower in MPO-deficient mice as compared to wild-type littermates. Both, fMLP-activated leukocytes and the MPO-H2O2-Cl(-) system inflicted barrier dysfunction of primary brain microvascular endothelial cells (BMVEC) that was partially rescued with the MPO inhibitor 4-aminobenzoic acid hydrazide. BMVEC treatment with the MPO-H2O2-Cl(-) system or activated neutrophils resulted in the formation of plasmalogen-derived chlorinated fatty aldehydes. 2-chlorohexadecanal (2-ClHDA) severely compromised BMVEC barrier function and induced morphological alterations in tight and adherens junctions. In situ perfusion of rat brain with 2-ClHDA increased BBB permeability in vivo. 2-ClHDA potently activated the MAPK cascade at physiological concentrations. An ERK1/2 and JNK antagonist (PD098059 and SP600125, respectively) protected against 2-ClHDA-induced barrier dysfunction in vitro. The current data provide evidence that interference with the MPO pathway could protect against BBB dysfunction under (neuro)inflammatory conditions.


Subject(s)
Blood-Brain Barrier/metabolism , Oxidants/metabolism , Peroxidase/metabolism , Aldehydes/pharmacology , Animals , Blood-Brain Barrier/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fatty Acids/biosynthesis , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Microvessels/cytology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Peroxidase/deficiency , Plasmalogens/metabolism , Rats , Swine
18.
Exp Cell Res ; 319(12): 1828-1838, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23541792

ABSTRACT

Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor and is invariably fatal to affected patients. Oxysterols belong to a class of bioactive lipids that are implicated in neurological disease and are associated with various types of cancer. Here, we investigated expression and transcriptional regulation of cholesterol 25-hydroxylase (CH25H) in human U87MG and GM133 glioblastoma cell lines. We demonstrate that in both cell lines transcription and translation of CH25H are increased in response to TNFα and IL1ß. In parallel, both cell lines upregulate 25-hydroxycholesterol (25-OHC) synthesis and secretion to levels comparable to bone marrow-derived mouse macrophages under inflammatory conditions. To determine whether 25-OHC acts as chemoattractant for tumor-associated macrophages, the human THP-1 monoblastic leukemia cell line was treated with varying amounts of the oxysterol. Experiments revealed that 25-OHC and lipid extracts isolated from GM133-conditioned medium (containing 7-fold higher 25-OHC concentrations than U87MG medium) induce chemotactic migration of THP-1 cells. Of note, 25-OHC also induced the migration of primary human peripheral blood monocytes. In response to exogenously added 25-OHC, THP-1 cells reorganized intermediate filament-associated vimentin to more cortical and polarized structures. Chemotactic migration of monocytes in response to 25-OHC was pertussis toxin-sensitive, indicating the involvement of G protein-coupled receptors. Using RNA interference we demonstrated that G protein-coupled receptor 183 (EBI2) contributes to 25-OHC-mediated chemotactic migration of THP-1 cells. These in vitro data indicate that GBM-derived and secreted 25-OHC may be involved in the recruitment of immune-competent cells to a tumor via EBI2.


Subject(s)
Brain Neoplasms/metabolism , Chemotaxis/drug effects , Glioblastoma/metabolism , Hydroxycholesterols/metabolism , Monocytes/physiology , Cell Line, Tumor , Humans , Protein Biosynthesis , RNA, Small Interfering , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Sterols/pharmacology , Transcription, Genetic , Up-Regulation , Vimentin/metabolism
19.
J Reprod Immunol ; 90(1): 21-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21641660

ABSTRACT

In investigating the immunological regulation of trophoblast invasion, emphasis is frequently placed on the constitution of the implantation site and its specific characteristics. However trophoblast cells are able of invading into not only the uterine tissue, but in the case of ectopic pregnancies also invade into other tissues, where they are actively involved in the creation of a beneficial local environment by expressing a range of membrane bound and soluble factors. The similarities and differences in trophoblast invasion in uterine and tubal pregnancies concerning trophoblast cells and maternal leukocytes and their interaction with each other are discussed in this review.


Subject(s)
Embryo Implantation/physiology , Trophoblasts/immunology , Apoptosis , Female , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/immunology , Humans , Placentation/physiology , Pregnancy , Pregnancy, Ectopic/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism
20.
Biochem Biophys Res Commun ; 410(4): 895-900, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21708126

ABSTRACT

Myeloperoxidase is abundantly present in inflammatory diseases where activation of monocytes/macrophages and T-cell-mediated immune response occurs. The potent oxidant hypochlorous acid (HOCl), generated by the myeloperoxidase-H(2)O(2)-chloride system of activated phagocytes, converts low-density lipoprotein (LDL) into a proinflammatory lipoprotein particle. Here, we investigated the apoptotic effect of HOCl-LDL, an in vivo occurring LDL modification, on human T-cell lymphoblast-like Jurkat cells. Experiments revealed that HOCl-LDL, depending on the oxidant:lipoprotein molar ratio, induces apoptosis via activation of caspase-3, PARP cleavage and accumulation of reactive oxygen species. The absence of Fas-associated protein with death domain or caspase-8 in mutant cells did not prevent HOCl-LDL induced apoptosis. In contrast, overexpression of the anti-apoptotic Bcl-2 protein protects Jurkat cells against HOCl-LDL-induced apoptosis and prevents accumulation of reactive oxygen species. We conclude that HOCl-LDL-mediated apoptosis in Jurkat cells follows predominantly the intrinsic, mitochondrial pathway. Insitu experiments revealed that an antibody raised against HOCl-LDL recognized epitopes that colocalize both with myeloperoxidase and CD3-positive T-cells in human decidual tissue where local stimulation of the immune system occurs. We provide convincing evidence that formation of HOCl-modified (lipo)proteins generated by the myeloperoxidase-H(2)O(2)-chloride system contributes to apoptosis in T-cells.


Subject(s)
Apoptosis/physiology , Hypochlorous Acid/metabolism , Lipoproteins, LDL/metabolism , T-Lymphocytes/physiology , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/genetics , Fas-Associated Death Domain Protein/genetics , Humans , Hypochlorous Acid/chemistry , Hypochlorous Acid/pharmacology , Jurkat Cells , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/pharmacology , Peroxidase/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...