Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Rep ; 40(12): 111364, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36130504

ABSTRACT

Mitochondria are dynamic organelles essential for cell survival whose structural and functional integrity rely on selective and regulated transport of lipids from/to the endoplasmic reticulum (ER) and across the mitochondrial intermembrane space. As they are not connected by vesicular transport, the exchange of lipids between ER and mitochondria occurs at membrane contact sites. However, the mechanisms and proteins involved in these processes are only beginning to emerge. Here, we show that the main physiological localization of the lipid transfer proteins ORP5 and ORP8 is at mitochondria-associated ER membrane (MAM) subdomains, physically linked to the mitochondrial intermembrane space bridging (MIB)/mitochondrial contact sites and cristae junction organizing system (MICOS) complexes that bridge the two mitochondrial membranes. We also show that ORP5/ORP8 mediate non-vesicular transport of phosphatidylserine (PS) lipids from the ER to mitochondria by cooperating with the MIB/MICOS complexes. Overall our study reveals a physical and functional link between ER-mitochondria contacts involved in lipid transfer and intra-mitochondrial membrane contacts maintained by the MIB/MICOS complexes.


Subject(s)
Mitochondrial Proteins , Phosphatidylserines , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Phosphatidylserines/metabolism
2.
Cell Mol Life Sci ; 79(4): 220, 2022 Apr 04.
Article in English | MEDLINE | ID: mdl-35368213

ABSTRACT

During angiogenesis, endothelial cells form protrusive sprouts and migrate towards the angiogenic stimulus. In this study, we investigate the role of the endoplasmic reticulum (ER)-anchored protein, Protrudin, in endothelial cell protrusion, migration and angiogenesis. Our results demonstrate that Protrudin regulates angiogenic tube formation in primary endothelial cells, Human umbilical vein endothelial cells (HUVECs). Analysis of RNA sequencing data and its experimental validation revealed cell migration as a prominent cellular function affected in HUVECs subjected to Protrudin knockdown. Further, our results demonstrate that knockdown of Protrudin inhibits focal adhesion kinase (FAK) activation in HUVECs and human aortic endothelial cells (HAECs). This is associated with a loss of polarized phospho-FAK distribution upon Protrudin knockdown as compared to Protrudin expressing HUVECs. Reduction of Protrudin also results in a perinuclear accumulation of mTOR and a decrease in VEGF-mediated S6K activation. However, further experiments suggest that the observed inhibition of angiogenesis in Protrudin knockdown cells is not affected by mTOR disturbance. Therefore, our findings suggest that defects in FAK activation and its abnormal subcellular distribution upon Protrudin knockdown are associated with a detrimental effect on endothelial cell migration and angiogenesis. Furthermore, mice with global Protrudin deletion demonstrate reduced retinal vascular progression. To conclude, our results provide evidence for a novel key role of Protrudin in endothelial cell migration and angiogenesis.


Subject(s)
Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Cell Movement/genetics , Focal Adhesion Protein-Tyrosine Kinases/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Vesicular Transport Proteins
3.
FASEB J ; 34(11): 14671-14694, 2020 11.
Article in English | MEDLINE | ID: mdl-32914503

ABSTRACT

Oxysterol-binding protein-related protein 2 (ORP2), a cholesterol-PI(4,5)P2 countercurrent transporter, was recently identified as a novel regulator of plasma membrane (PM) cholesterol and PI(4,5)P2 content in HeLa cells. Here, we investigate the role of ORP2 in endothelial cell (EC) cholesterol and PI(4,5)P2 distribution, angiogenic signaling, and angiogenesis. We show that ORP2 knock-down modifies the distribution of cholesterol accessible to a D4H probe, between late endosomes and the PM. Depletion of ORP2 from ECs inhibits their angiogenic tube formation capacity, alters the gene expression of angiogenic signaling pathways such as VEGFR2, Akt, mTOR, eNOS, and Notch, and reduces EC migration, proliferation, and cell viability. We show that ORP2 regulates the integrity of VEGFR2 at the PM in a cholesterol-dependent manner, the depletion of ORP2 resulting in proteolytic cleavage by matrix metalloproteinases, and reduced activity of VEGFR2 and its downstream signaling. We demonstrate that ORP2 depletion increases the PM PI(4,5)P2 coincident with altered F-actin morphology, and reduces both VEGFR2 and cholesterol in buoyant raft membranes. Moreover, ORP2 knock-down suppresses the expression of the lipid raft-associated proteins VE-cadherin and caveolin-1. Analysis of the retinal microvasculature in ORP2 knock-out mice generated during this study demonstrates the subtle alterations of morphology characterized by reduced vessel length and increased density of tip cells and perpendicular sprouts. Gene expression changes in the retina suggest disturbance of sterol homeostasis, downregulation of VE-cadherin, and a putative disturbance of Notch signaling. Our data identifies ORP2 as a novel regulator of EC cholesterol and PI(4,5)P2 homeostasis and cholesterol-dependent angiogenic signaling.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptors, Steroid/metabolism , Signal Transduction , Actins/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Caveolins/metabolism , Cell Membrane/metabolism , Cell Movement , Endosomes/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Matrix Metalloproteinases/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , Receptors, Steroid/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , TOR Serine-Threonine Kinases/metabolism
4.
Biochimie ; 158: 90-101, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30590084

ABSTRACT

ORP2 is a sterol-binding protein with documented functions in lipid and glucose metabolism, Akt signaling, steroidogenesis, cell adhesion, migration and proliferation. Here we investigate the interactions of ORP2 with phosphoinositides (PIPs) by surface plasmon resonance (SPR), its affinity for cholesterol with a pull-down assay, and its capacity to transfer sterol in vitro. Moreover, we determine the effects of wild-type (wt) ORP2 and a mutant with attenuated PIP binding, ORP2(mHHK), on the subcellular distribution of cholesterol, and analyze the interaction of ORP2 with the related cholesterol transporter ORP1L. ORP2 showed specific affinity for PI(4,5)P2, PI(3,4,5)P3 and PI(4)P, with suggestive Kd values in the µM range. Also binding of cholesterol by ORP2 was detectable, but a Kd could not be determined. Wt ORP2 was in HeLa cells mainly detected in the cytosol, ER, late endosomes, and occasionally on lipid droplets (LDs), while ORP2(mHHK) displayed an enhanced LD localization. Overexpression of wt ORP2 shifted the D4H cholesterol probe away from endosomes, while ORP2(mHHK) caused endosomal accumulation of the probe. Although ORP2 failed to transfer dehydroergosterol in an in vitro assay where OSBP is active, its knock-down resulted in the accumulation of cholesterol in late endocytic compartments, as detected by both D4H and filipin probes. Interestingly, ORP2 was shown to interact and partially co-localize on late endosomes with ORP1L, a cholesterol transporter/sensor at ER-late endosome junctions. Our data demonstrates that ORP2 binds several phosphoinositides, both PI(4)P and multiply phosphorylated species. ORP2 regulates the subcellular distribution of cholesterol dependent on its PIP-binding capacity. The interaction of ORP2 with ORP1L suggests a concerted action of the two ORPs.


Subject(s)
Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Lipid Droplets/metabolism , Phosphatidylinositols/metabolism , Receptors, Steroid/metabolism , Cholesterol/genetics , Endoplasmic Reticulum/genetics , Endosomes/genetics , HeLa Cells , Humans , Phosphatidylinositols/genetics , Receptors, Steroid/genetics
5.
FASEB J ; 32(3): 1281-1295, 2018 03.
Article in English | MEDLINE | ID: mdl-29092904

ABSTRACT

ORP2 is implicated in cholesterol transport, triglyceride metabolism, and adrenocortical steroid hormone production. We addressed ORP2 function in hepatocytes by generating ORP2-knockout (KO) HuH7 cells by CRISPR-Cas9 gene editing, followed by analyses of transcriptome, F-actin morphology, migration, adhesion, and proliferation. RNA sequencing of ORP2-KO cells revealed >2-fold changes in 579 mRNAs. The Ingenuity Pathway Analysis (IPA) uncovered alterations in the following functional categories: cellular movement, cell-cell signaling and interaction, cellular development, cellular function and maintenance, cellular growth and proliferation, and cell morphology. Many pathways in these categories involved actin cytoskeleton, cell migration, adhesion, or proliferation. Analysis of the ORP2 interactome uncovered 109 putative new partners. Their IPA analysis revealed Ras homolog A (RhoA) signaling as the most significant pathway. Interactions of ORP2 with SEPT9, MLC12, and ARHGAP12 were validated by independent assays. ORP2-KO resulted in abnormal F-actin morphology characterized by impaired capacity to form lamellipodia, migration defect, and impaired adhesion and proliferation. Rescue of the migration phenotype and generation of typical cell surface morphology required an intact ORP2 phosphoinositide binding site, suggesting that ORP2 function involves phosphoinositide binding and transport. The results point at a novel function of ORP2 as a lipid-sensing regulator of the actin cytoskeleton, with impacts on hepatocellular migration, adhesion, and proliferation.-Kentala, H., Koponen, A., Kivelä, A. M., Andrews, R., Li, C., Zhou, Y., Olkkonen, V. M. Analysis of ORP2-knockout hepatocytes uncovers a novel function in actin cytoskeletal regulation.


Subject(s)
Actin Cytoskeleton/physiology , Cell Movement , Hepatocytes/physiology , Receptors, Steroid/metabolism , CRISPR-Cas Systems , Cell Adhesion , Cell Proliferation , Gene Knockout Techniques , Hepatocytes/cytology , Humans , Receptors, Steroid/genetics , Signal Transduction
6.
Expert Opin Biol Ther ; 15(12): 1699-712, 2015.
Article in English | MEDLINE | ID: mdl-26328616

ABSTRACT

INTRODUCTION: Despite considerable improvements in therapies, atherosclerotic cardiovascular diseases remain the leading cause of death worldwide. Therefore, in addition to current treatment options, new therapeutic approaches are still needed. AREAS COVERED: In this review, novel gene and RNA interference-based therapy approaches and promising target genes for treating atherosclerosis are addressed. In addition, relevant animal models for the demonstration of the efficacy of different gene therapy applications, and current progress toward more efficient, targeted and safer gene transfer vectors are reviewed. EXPERT OPINION: Atherosclerosis represents a complex multifactorial disease that is dependent on the interplay between lipoprotein metabolism, cellular reactions and inflammation. Recent advances and novel targets, especially in the field of RNA interference-based therapies, are very promising. However, it should be noted that the modulation of a particular gene is not as clearly associated with a complex polygenic disease as it is in the case of monogenic diseases. A deeper understanding of molecular mechanisms of atherosclerosis, further progress in vector development and the demonstration of treatment efficacy in relevant animal models will be required before gene therapy of atherosclerosis meets its clinical reality.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/therapy , Genetic Therapy/trends , Animals , Atherosclerosis/diagnosis , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , RNA Interference/physiology , Treatment Outcome
7.
J Lipid Res ; 56(6): 1206-21, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25861792

ABSTRACT

Lipolytic modification of LDL particles by SMase generates LDL aggregates with a strong affinity for human arterial proteoglycans and may so enhance LDL retention in the arterial wall. Here, we evaluated the effects of apoA-I mimetic peptide 4F on structural and functional properties of the SMase-modified LDL particles. LDL particles with and without 4F were incubated with SMase, after which their aggregation, structure, and proteoglycan binding were analyzed. At a molar ratio of L-4F to apoB-100 of 2.5 to 20:1, 4F dose-dependently inhibited SMase-induced LDL aggregation. At a molar ratio of 20:1, SMase-induced aggregation was fully blocked. Binding of 4F to LDL particles inhibited SMase-induced hydrolysis of LDL by 10% and prevented SMase-induced LDL aggregation. In addition, the binding of the SMase-modified LDL particles to human aortic proteoglycans was dose-dependently inhibited by pretreating LDL with 4F. The 4F stabilized apoB-100 conformation and inhibited SMase-induced conformational changes of apoB-100. Molecular dynamic simulations showed that upon binding to protein-free LDL surface, 4F locally alters membrane order and fluidity and induces structural changes to the lipid layer. Collectively, 4F stabilizes LDL particles by preventing the SMase-induced conformational changes in apoB-100 and so blocks SMase-induced LDL aggregation and the resulting increase in LDL retention.


Subject(s)
Apolipoprotein A-I/pharmacology , Apolipoprotein B-100/metabolism , Lipoproteins, LDL/metabolism , Peptides/pharmacology , Sphingomyelin Phosphodiesterase/metabolism , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoprotein A-I/metabolism , Biomimetics , Humans , Lipolysis/drug effects , Peptides/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors
8.
Nucleic Acids Res ; 42(20): 12570-84, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25352550

ABSTRACT

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


Subject(s)
Transcriptional Activation , Vascular Endothelial Growth Factor A/pharmacology , Cell Compartmentation , Cells, Cultured , Chromatin/metabolism , Enhancer Elements, Genetic , Genome, Human , Humans , Phenotype , Transcription Factors/metabolism , Transcription, Genetic/drug effects
9.
Cardiovasc Res ; 99(4): 716-23, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23756254

ABSTRACT

AIMS: The role of vascular endothelial growth factor (VEGF-A) in atherogenesis has remained controversial. We addressed this by comparing the effects of adenoviral VEGF-A gene transfer on atherosclerosis and lipoproteins in ApoE(-/-), LDLR(-/-), LDLR(-/-)ApoE(-/-), and LDLR(-/-)ApoB(100/100) mice. METHODS AND RESULTS: After 4 weeks on western diet, systemic adenoviral gene transfer was performed with hVEGF-A or control vectors. Effects on atherosclerotic lesion area and composition, lipoprotein profiles, and plasma lipoprotein lipase (LPL) activity were examined. On day 4, VEGF-A induced alterations in lipoprotein profiles and a significant negative correlation was observed between plasma LPL activity and VEGF-A levels. One month after gene transfer, no changes in atherosclerosis were observed in LDLR(-/-) and LDLR(-/-)ApoB(100/100) models, whereas both ApoE(-/-) models displayed increased en face lesion areas in thoracic and abdominal aortas. VEGF-A also reduced LPL mRNA in heart and white adipose tissue, whereas Angptl4 was increased, potentially providing further mechanistic explanation for the findings. CONCLUSION: VEGF-A gene transfer induced pro-atherogenic changes in lipoprotein profiles in all models. As a novel finding, VEGF-A also reduced LPL activity, which might underlie the observed changes in lipid profiles. However, VEGF-A was observed to increase atherosclerosis only in the ApoE(-/-) background, clearly indicating some mouse model-specific effects.


Subject(s)
Atherosclerosis/etiology , Genetic Therapy , Hyperlipidemias/therapy , Lipoprotein Lipase/metabolism , Lipoproteins/blood , Vascular Endothelial Growth Factor A/genetics , Adenoviridae/genetics , Animals , Apolipoproteins B/deficiency , Apolipoproteins B/physiology , Disease Models, Animal , Female , Hyperlipidemias/blood , Hyperlipidemias/pathology , Lipids/blood , Lipoprotein Lipase/genetics , Male , Mice , Mice, Inbred C57BL , Receptors, LDL/physiology
10.
Atherosclerosis ; 225(2): 335-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23102786

ABSTRACT

OBJECTIVE: Endothelial lipase (EL) regulates HDL cholesterol levels and in inflammatory states, like atherosclerosis, EL expression is increased contributing to low HDL cholesterol. The regulation of EL expression is poorly understood and has mainly been attributed to inflammatory stimuli. As sterol regulatory element binding proteins (SREBPs) are regulators of genes involved in lipid metabolism, we hypothesized that EL is regulated by SREBPs and that EL expression is modified by the SREBP activator vascular endothelial growth factor A (VEGF-A). METHODS: and results: Quantitative PCR and Western blot results demonstrated that starvation increased EL expression in human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs). Also, 25-hydroxycholesterol (25HC), an inhibitor of SREBP activation inhibited EL expression. With siRNA-mediated inhibition of SREBPs the effect of starvation was shown to be SREBP-2 dependent. VEGF-A decreased EL expression in both endothelial cell lines used, most likely via inhibition of SREBP-2 binding determined by chromatin immunoprecipitation (ChIP). Furthermore, in atherosclerosis prone LDLR(-/-)ApoB(100/100) mice, systemic adenoviral gene transfer with human VEGF-A decreased EL mRNA in peripheral tissues and increased plasma HDL cholesterol. CONCLUSIONS: These results identify SREBPs as novel regulators of EL expression. VEGF-A as an endogenous EL inhibitor could be therapeutically relevant in atherosclerosis by increasing systemic HDL cholesterol levels.


Subject(s)
Cholesterol, HDL/blood , Endothelial Cells/enzymology , Lipase/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae/genetics , Animals , Apolipoprotein B-100/deficiency , Apolipoprotein B-100/genetics , Atherosclerosis/blood , Atherosclerosis/enzymology , Atherosclerosis/genetics , Binding Sites , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Disease Models, Animal , Endothelial Cells/drug effects , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , Genetic Vectors , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hydroxycholesterols/pharmacology , Lipase/genetics , Mice , Mice, Knockout , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 2/genetics , Time Factors , Transfection , Vascular Endothelial Growth Factor A/genetics
11.
Atherosclerosis ; 213(1): 122-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20688330

ABSTRACT

OBJECTIVE: Endothelial lipase (EL) is a new member of triacylglycerol lipase family that has been shown to decrease high-density lipoprotein (HDL) cholesterol levels leading to increased risk of atherosclerosis. Its expression is increased during inflammation and by inflammatory cytokines. Sulforaphane (SFN) is a naturally occurring isothiocyanate present in cruciferous vegetables that has antioxidant and anti-inflammatory effects. Nuclear factor (NF)-κB is one of the molecular targets for SFN-mediated protective effects. Our aim was therefore to assess whether SFN could impact on EL expression via modulation of NF-κB pathway. METHODS AND RESULTS: Quantitative PCR and Western blot results demonstrated that SFN inhibited tumor necrosis factor (TNF)-α-mediated induction of EL in human umbilical vein endothelial cells (HUVEC). Lentiviral transduction of HUVEC with mutated form of IκB-α (IκBM) as well as silencing of NF-κB subunit p65 using RNA interference revealed that TNF-α-mediated induction of EL is mediated through NF-κB pathway. In addition, a total of five NF-κB binding sites were found in LIPG gene, which encodes EL. SFN inhibited binding of NF-κB to these sites analyzed by chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA). SFN also inhibited TNF-α mediated phosphorylation of IκB kinase (IKK) 1/2 and IκB-α. CONCLUSIONS: Collectively, these results indicate that SFN inhibits EL expression via inhibition of NF-κB which may have a beneficial effect on HDL cholesterol levels.


Subject(s)
Endothelial Cells/cytology , Gene Expression Regulation, Enzymologic , Lipase/antagonists & inhibitors , NF-kappa B/metabolism , Thiocyanates/pharmacology , Antioxidants/chemistry , Atherosclerosis/metabolism , Cells, Cultured , Humans , I-kappa B Kinase/metabolism , Isothiocyanates/chemistry , Lipase/biosynthesis , Models, Biological , Protein Binding , Sulfoxides , Umbilical Veins/cytology , Vegetables
12.
J Biol Chem ; 284(48): 33233-41, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19808663

ABSTRACT

Electrophilic fatty acid derivatives, including nitrolinoleic acid and nitro-oleic acid (OA-NO(2)), can mediate anti-inflammatory and pro-survival signaling reactions. The transcription factor Nrf2, activated by electrophilic fatty acids, suppresses redox-sensitive pro-inflammatory gene expression and protects against vascular endothelial oxidative injury. It was therefore postulated that activation of Nrf2 by OA-NO(2) accounts in part for its anti-inflammatory actions, motivating the characterization of Nrf2-dependent and -independent effects of OA-NO(2) on gene expression using genome-wide transcriptional profiling. Control and Nrf2-small interfering RNA-transfected human endothelial cells were treated with vehicle, oleic acid, or OA-NO(2), and differential gene expression profiles were determined. Although OA-NO(2) significantly induced the expression of Nrf2-dependent genes, including heme oxygenase-1 and glutamate-cysteine ligase modifier subunit, the majority of OA-NO(2)-regulated genes were regulated by Nrf2-independent pathways. Moreover, gene set enrichment analysis revealed that the heat shock response is the major pathway activated by OA-NO(2), with robust induction of a number of heat shock genes regulated by the heat shock transcription factor. Inasmuch as the heat shock response mediates anti-inflammatory and cytoprotective actions, this mechanism is proposed to contribute to the protective cell signaling functions of nitro-fatty acids and other electrophilic fatty acid derivatives.


Subject(s)
Endothelial Cells/drug effects , Linoleic Acids/pharmacology , NF-E2-Related Factor 2/genetics , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Blotting, Western , Cell Line , Cells, Cultured , Cluster Analysis , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hot Temperature , Humans , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
13.
Free Radic Biol Med ; 47(9): 1310-7, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19573595

ABSTRACT

The J series of cyclopentenone prostaglandins (PGs) such as 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)) are electrophilic lipid signaling mediators derived from the nonenzymatic dehydration of PGD(2), a major product of the cyclooxygenase pathway. The biological actions of 15d-PGJ(2) are attributed to its ability to form covalent adducts with thiol residues within specific signaling proteins, thus triggering redox-sensitive cell signaling pathways. One of the signaling pathways potently activated by 15d-PGJ(2) is the Keap1-Nrf2-ARE system, which has a well-appreciated role in protecting cells from endogenous and exogenous stresses as well as anti-inflammatory effects. In this review, we give an overview of the mechanisms by which 15d-PGJ(2) activates the Keap1-Nrf2-ARE system, focusing particularly on the role of Keap1 in sensing electrophilic stress. In addition, the Nrf2-dependent anti-inflammatory and cytoprotective effects of 15d-PGJ(2) are discussed.


Subject(s)
Gene Expression Regulation/physiology , NF-E2-Related Factor 2/biosynthesis , Prostaglandin D2/analogs & derivatives , Signal Transduction/physiology , Animals , Humans , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Prostaglandin D2/metabolism
14.
Circ Res ; 103(1): e1-9, 2008 Jul 03.
Article in English | MEDLINE | ID: mdl-18535259

ABSTRACT

Besides their well-characterized proinflammatory and proatherogenic effects, oxidized phospholipids, such as oxPAPC (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphocholine) have been shown to have beneficial responses in vascular cells via induction of antioxidant enzymes such as heme oxygenase-1. We therefore hypothesized that oxPAPC could evoke a general cytoprotective response via activation of antioxidative transcription factor Nrf2. Here, we show that oxPAPC increases nuclear accumulation of Nrf2. Using the small interfering RNA approach, we demonstrate that Nrf2 is critical in mediating the induction of glutamate-cysteine ligase modifier subunit (GCLM) and NAD(P)H quinone oxidoreductase-1 (NQO1) by oxPAPC in human endothelial cells, whereas the contribution to the induction of heme oxygenase-1 was less significant. The induction of GCLM and NQO1 was attenuated by reduction of electrophilic groups with sodium borohydrate, as well as treatment with thiol antioxidant N-acetylcysteine, suggesting that the thiol reactivity of oxPAPC is largely mediating its effect on Nrf2-responsive genes. Moreover, we show that oxidized phospholipid having a highly electrophilic isoprostane ring in its sn-2 position is a potent inducer of Nrf2 target genes. Finally, we demonstrate that the oxPAPC-inducible expression of heme oxygenase-1, GCLM, and NQO1 is lower in Nrf2-null than wild-type mouse carotid arteries in vivo. We suggest that the activation of Nrf2 by oxidized phospholipids provides a mechanism by which their deleterious effects are limited in the vasculature.


Subject(s)
Antioxidants/metabolism , Carotid Arteries/enzymology , Cell Nucleus/metabolism , Endothelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , NF-E2-Related Factor 2/metabolism , Phosphatidylcholines/pharmacokinetics , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Carotid Arteries/cytology , Cell Nucleus/genetics , Endothelial Cells/cytology , Gene Expression Regulation, Enzymologic/physiology , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/genetics , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Mice , Mice, Mutant Strains , NAD(P)H Dehydrogenase (Quinone) , NADPH Dehydrogenase/biosynthesis , NADPH Dehydrogenase/genetics , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Oxidation-Reduction/drug effects , Phosphatidylcholines/metabolism , RNA, Small Interfering/genetics
15.
J Nutr ; 138(7): 1263-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18567745

ABSTRACT

Enterolactone is a lignan formed by enterobacteria from precursors in plant foods. Due to its phenolic structure, it can act as an antioxidant, e.g. via direct scavenging of hydroxyl radical. Moreover, many, but not all, phenolic compounds can have indirect antioxidative effects through induction of heme oxygenase-1 (HO-1), which has antiinflammatory functions via production of antioxidants bilirubin and biliverdin as well as carbon monoxide, thereby contributing to cardiovascular health. Our aim was therefore to assess whether enterolactone has indirect antioxidative effects via induction of HO-1 in endothelial cells. The effect of enterolactone on HO-1 mRNA and protein expression in human umbilical vein endothelial cells (HUVEC) was analyzed by quantitative real-time PCR and western blot. The role of nuclear factor-E2-related factor 2 (Nrf2) in HO-1 induction by enterolactone was studied using small interfering RNA (siRNA) and chromatin immunoprecipitation (ChIP) methods. Our results showed that enterolactone induced HO-1 in HUVEC in a time- and concentration-dependent manner. The induction appeared to be mediated via the transcription factor Nrf2, as Nrf2 siRNA abolished the HO-1 induction by enterolactone. We also showed using ChIP that exposure to enterolactone increased the binding of Nrf2 to the promoter region of HO-1. In conclusion, enterolactone increases the expression of HO-1 via Nrf2, which may contribute to its vasculoprotective effects.


Subject(s)
4-Butyrolactone/analogs & derivatives , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Lignans/pharmacology , NF-E2-Related Factor 2/metabolism , 4-Butyrolactone/pharmacology , Antioxidants/pharmacology , Base Sequence , Binding Sites/genetics , Cells, Cultured , Chromatin Immunoprecipitation , DNA Primers/genetics , Enzyme Induction/drug effects , Gene Expression/drug effects , Gene Silencing , Humans , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...