Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Circulation ; 148(13): 1035-1038, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37747956

Subject(s)
Blood Pressure , Humans
2.
Front Physiol ; 12: 693702, 2021.
Article in English | MEDLINE | ID: mdl-34456745

ABSTRACT

Oral treatment of apolipoprotein E-knockout (ApoE-KO) mice with the putative sirtuin 1 (SIRT1) activator resveratrol led to a reduction of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity in the heart. In contrast, the SIRT1 inhibitor EX527 enhanced the superoxide production in isolated human polymorphonuclear granulocytes. In human monocytic THP-1 cells, phorbol ester-stimulated superoxide production was enhanced by inhibitors of histone deacetylases (HDACs; including quisinostat, trichostatin A (TSA), PCI34051, and tubastatin A) and decreased by inhibitors of histone acetyltransferases [such as garcinol, curcumin, and histone acetyltransferase (HAT) Inhibitor II]. These results indicate that protein acetylation and deacetylation may represent crucial mechanisms regulating NADPH oxidase-mediated superoxide production. In cell-free systems, incubation of recombinant Rac1 with SIRT1 resulted in decreased Rac1 acetylation. Mass spectrometry analyses identified lysine 166 (K166) in Rac1 as a residue targeted by SIRT1. Deacetylation of Rac1 by SIRT1 markedly reduced the interaction of Rac1 with p67phox in in vitro assays. Computational modeling analyses revealed that K166 deacetylation of Rac1 led to a 5-fold reduction in its binding affinity to guanosine-5'-triphosphate, and a 21-fold decrease in its binding potential to p67phox. The latter is crucial for Rac1-mediated recruitment of p67phox to the membrane and for p67phox activation. In conclusion, both SIRT1 and non-sirtuin deacetylases play a role in regulating NADPH oxidase activity. Rac1 can be directly deacetylated by SIRT1 in a cell-free system, leading to an inhibition of Rac1-p67phox interaction. The downstream targets of non-sirtuin deacetylases are still unknown. The in vivo significance of these findings needs to be investigated in future studies.

3.
J Ethnopharmacol ; 251: 112534, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-31893533

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Red fruit (Pandanus conoideus Lam) oil (RFO) is utilized by inhabitants of the Papua Island to treat diseases such as infections, cancer, and cardiovascular disease, but the mechanism of action is unknown. AIM OF THE STUDY: We have recently shown that RFO stimulates nitric oxide (NO) production in endothelial cells. The present study was conducted to investigate the molecular mechanism of endothelial NO synthase (eNOS) activation by RFO. MATERIALS AND METHODS: NO production by endothelial cells was determined with electron paramagnetic resonance. The vascular function of isolated mouse aorta was examined using a wire myograph system. Phosphorylation of eNOS was studied with Western blot analyses. RESULTS: RFO induced concentration-dependent vasodilation in isolated mouse aorta. The vasodilator effect of RFO was lost in endothelium-denuded aorta and in aorta from mice deficient in eNOS. Treatment of human EA.hy 926 endothelial cells with RFO led to an enhancement of eNOS phosphorylation at serine 1177 and NO production. The RFO-induced eNOS phosphorylation and NO production were reduced by inhibitors of Akt or AMPK, but not by an inhibitor of CaMKII. The effects of RFO were decreased by pharmacological inhibition of PI3K, indicating an involvement of the PI3K-Akt pathway. Moreover, acetone-soluble fractions and oily fractions of RFO showed higher efficacies than the RFO polar fraction in activating eNOS. CONCLUSIONS: RFO contains highly active compounds that enhance NO production through Akt- or AMPK-mediated eNOS phosphorylation. The increase in endothelial NO production is likely to represent one of the molecular mechanisms responsible for the therapeutic effects of RFO.


Subject(s)
Endothelial Cells/drug effects , Fruit , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Pandanaceae , Plant Oils/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Cell Line , Endothelial Cells/metabolism , Humans , Mice, Inbred C57BL , Phosphorylation , Vasodilator Agents/pharmacology
4.
Ann N Y Acad Sci ; 1403(1): 132-141, 2017 09.
Article in English | MEDLINE | ID: mdl-28672425

ABSTRACT

Under physiological conditions, nitric oxide (NO) is produced in the vasculature mainly by the endothelial NO synthase (eNOS). Experiments using gene-disrupted mice have demonstrated that eNOS has antihypertensive, antithrombotic, and antiatherosclerotic effects. Recent studies show that eNOS is expressed not only in the endothelium but also in the perivascular adipose tissue (PVAT). Resveratrol prevents eNOS uncoupling and upregulates eNOS expression and activity. These effects of resveratrol are well established for the eNOS enzyme in the endothelium. Interestingly, resveratrol also improves PVAT function. However, a causal role for eNOS in the effects of resveratrol on PVAT function has not yet been verified and needs to be studied in the future.


Subject(s)
Adipose Tissue/drug effects , Endothelium, Vascular/drug effects , Nitric Oxide Synthase Type III/metabolism , Stilbenes/pharmacology , Adipose Tissue/metabolism , Animals , Endothelium, Vascular/metabolism , Mice , Resveratrol
5.
Redox Biol ; 13: 94-162, 2017 10.
Article in English | MEDLINE | ID: mdl-28577489

ABSTRACT

The European Cooperation in Science and Technology (COST) provides an ideal framework to establish multi-disciplinary research networks. COST Action BM1203 (EU-ROS) represents a consortium of researchers from different disciplines who are dedicated to providing new insights and tools for better understanding redox biology and medicine and, in the long run, to finding new therapeutic strategies to target dysregulated redox processes in various diseases. This report highlights the major achievements of EU-ROS as well as research updates and new perspectives arising from its members. The EU-ROS consortium comprised more than 140 active members who worked together for four years on the topics briefly described below. The formation of reactive oxygen and nitrogen species (RONS) is an established hallmark of our aerobic environment and metabolism but RONS also act as messengers via redox regulation of essential cellular processes. The fact that many diseases have been found to be associated with oxidative stress established the theory of oxidative stress as a trigger of diseases that can be corrected by antioxidant therapy. However, while experimental studies support this thesis, clinical studies still generate controversial results, due to complex pathophysiology of oxidative stress in humans. For future improvement of antioxidant therapy and better understanding of redox-associated disease progression detailed knowledge on the sources and targets of RONS formation and discrimination of their detrimental or beneficial roles is required. In order to advance this important area of biology and medicine, highly synergistic approaches combining a variety of diverse and contrasting disciplines are needed.


Subject(s)
International Cooperation , Reactive Oxygen Species/metabolism , Animals , European Union , Humans , Molecular Biology/organization & administration , Molecular Biology/trends , Oxidation-Reduction , Reactive Oxygen Species/chemistry , Signal Transduction , Societies, Scientific
6.
Circ Res ; 120(4): 713-735, 2017 Feb 17.
Article in English | MEDLINE | ID: mdl-28209797

ABSTRACT

Major reactive oxygen species (ROS)-producing systems in vascular wall include NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, xanthine oxidase, the mitochondrial electron transport chain, and uncoupled endothelial nitric oxide (NO) synthase. ROS at moderate concentrations have important signaling roles under physiological conditions. Excessive or sustained ROS production, however, when exceeding the available antioxidant defense systems, leads to oxidative stress. Animal studies have provided compelling evidence demonstrating the roles of vascular oxidative stress and NO in atherosclerosis. All established cardiovascular risk factors such as hypercholesterolemia, hypertension, diabetes mellitus, and smoking enhance ROS generation and decrease endothelial NO production. Key molecular events in atherogenesis such as oxidative modification of lipoproteins and phospholipids, endothelial cell activation, and macrophage infiltration/activation are facilitated by vascular oxidative stress and inhibited by endothelial NO. Atherosclerosis develops preferentially in vascular regions with disturbed blood flow (arches, branches, and bifurcations). The fact that these sites are associated with enhanced oxidative stress and reduced endothelial NO production is a further indication for the roles of ROS and NO in atherosclerosis. Therefore, prevention of vascular oxidative stress and improvement of endothelial NO production represent reasonable therapeutic strategies in addition to the treatment of established risk factors (hypercholesterolemia, hypertension, and diabetes mellitus).


Subject(s)
Atherosclerosis/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide/physiology , Oxidative Stress/physiology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Endothelium, Vascular/pathology , Humans , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism , Species Specificity
7.
Br J Pharmacol ; 174(20): 3443-3453, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28055105

ABSTRACT

BACKGROUND AND PURPOSE: We have recently shown that a reduced function of endothelial nitric oxide synthase (eNOS) in the perivascular adipose tissue (PVAT) contributes crucially to obesity-induced vascular dysfunction in mice. The current study was conducted to test the hypothesis that vascular dysfunction in obesity can be reversed by in vivo improvement of PVAT eNOS activity. EXPERIMENTAL APPROACH: Male C57BL/6J mice were fed a high-fat diet (HFD) for 22 weeks to induce obesity. During the last 4 weeks of HFD feeding, the obese mice were treated p.o. with the standardized Crataegus extract WS® 1442, which has been shown previously to improve eNOS activity. KEY RESULTS: Diet-induced obesity in mice markedly reduced the vasodilator response of thoracic aorta to acetylcholine in wire myograph experiments. Strikingly, this vascular dysfunction was only evident in PVAT-containing aorta but not in PVAT-free aorta. In vivo treatment of obese mice with WS® 1442 had no effect on body weight or epididymal fat mass, but completely restored the vascular function of PVAT-containing aorta. Feeding a HFD led to a reduced phosphorylation and an enhanced acetylation of PVAT eNOS, both effects were reversed by WS® 1442 treatment. CONCLUSION AND IMPLICATIONS: PVAT plays a key role in vascular dysfunction in diet-induced obese mice. Not obesity itself, but a PVAT dysfunction is responsible for obesity-induced vascular disorders. Improving PVAT function by pharmacological means (e.g. with WS® 1442) can ameliorate vascular function even without reducing body weight or fat mass. LINKED ARTICLES: This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.


Subject(s)
Adipose Tissue/physiology , Aorta, Thoracic/physiology , Diet, High-Fat , Obesity/physiopathology , Acetylation/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Aorta, Thoracic/drug effects , Body Weight , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Flavonoids/pharmacology , Lipids/blood , Male , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects
8.
Br J Pharmacol ; 174(12): 1633-1646, 2017 06.
Article in English | MEDLINE | ID: mdl-27058985

ABSTRACT

The antioxidant effects of resveratrol (3,5,4'-trihydroxy-trans-stilbene) contribute substantially to the health benefits of this compound. Resveratrol has been shown to be a scavenger of a number of free radicals. However, the direct scavenging activities of resveratrol are relatively poor. The antioxidant properties of resveratrol in vivo are more likely to be attributable to its effect as a gene regulator. Resveratrol inhibits NADPH oxidase-mediated production of ROS by down-regulating the expression and activity of the oxidase. This polyphenolic compound reduces mitochondrial superoxide generation by stimulating mitochondria biogenesis. Resveratrol prevents superoxide production from uncoupled endothelial nitric oxide synthase by up-regulating the tetrahydrobiopterin-synthesizing enzyme GTP cyclohydrolase I. In addition, resveratrol increases the expression of various antioxidant enzymes. Some of the gene-regulating effects of resveratrol are mediated by the histone/protein deacetylase sirtuin 1 or by the nuclear factor-E2-related factor-2. In this review article, we have also summarized the cardiovascular effects of resveratrol observed in clinical trials. LINKED ARTICLES: This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.


Subject(s)
Antioxidants/pharmacology , Cardiovascular System/drug effects , Stilbenes/pharmacology , Animals , Cardiovascular System/metabolism , Humans , Resveratrol
9.
Molecules ; 21(8)2016 Jul 25.
Article in English | MEDLINE | ID: mdl-27463705

ABSTRACT

Betulinic acid (BA) is a naturally occurring pentacyclic triterpenoid with anti-inflammatory, antiviral and anti-cancer properties. Beneficial cardiovascular effects such as increased nitric oxide (NO) production through enhancement of endothelial NO synthase (eNOS) activity and upregulation of eNOS expression have been demonstrated for this compound. In the present study, immortalized human EA.hy 926 endothelial cells were incubated for up to 1 h with 1-100 µM BA and with the phosphatidylinositol-3-kinase (PI3K) inhibitors LY294002 and wortmannin, or the estrogen receptor (ER) antagonist ICI 182,780. Phosphorylation status of eNOS and total eNOS protein were analyzed by Western blotting using a serine 1177 phosphosite-specific antibody. Bioactive NO production was assessed by determination of cGMP content in rat lung fibroblasts (RFL-6) reporter cells. Short-term incubation of EA.hy 926 cells with BA resulted in eNOS phosphorylation at the serine 1177 residue in a concentration- and time-dependent manner with a half-maximal effective concentration of 0.57 µM. This was associated with an enhanced production of NO. BA-induced eNOS phosphorylation and NO production was completely blocked by pretreatment with ICI 182,780, and was attenuated by pretreatment with the PI3K inhibitors wortmannin and LY294002. These results indicate that fast non-genomic effects of ER with downstream signaling through the PI3K/Akt pathway and consecutive eNOS phosphorylation at serine 1177 are involved in BA-induced eNOS activation.


Subject(s)
Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Triterpenes/pharmacology , Androstadienes/pharmacology , Animals , Cell Line , Chromones/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fulvestrant , Gene Expression Regulation/drug effects , Humans , Lung/cytology , Lung/metabolism , Morpholines/pharmacology , Pentacyclic Triterpenes , Phosphorylation/drug effects , Rats , Signal Transduction/drug effects , Wortmannin , Betulinic Acid
10.
Arterioscler Thromb Vasc Biol ; 36(1): 78-85, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586660

ABSTRACT

OBJECTIVE: The present study was conducted to investigate the contribution of perivascular adipose tissue (PVAT) to vascular dysfunction in a mouse model of diet-induced obesity. APPROACH AND RESULTS: Obesity was induced in male C57BL/6J mice with a high-fat diet for 20 weeks, and vascular function was studied with myograph. In PVAT-free aortas isolated from obese mice, the endothelium-dependent, nitric oxide-mediated vasodilator response to acetylcholine remained normal. In contrast, a clear reduction in the vasodilator response to acetylcholine was observed in aortas from obese mice when PVAT was left in place. Adipocytes in PVAT were clearly positive in endothelial nitric oxide synthase (eNOS) staining, and PVAT nitric oxide production was significantly reduced in obese mice. High-fat diet had no effect on eNOS expression but led to eNOS uncoupling, evidenced by diminished superoxide production in PVAT after eNOS inhibition. As mechanisms for eNOS uncoupling, arginase induction and l-arginine deficiency were observed in PVAT. Obesity-induced vascular dysfunction could be reversed by ex vivo l-arginine treatment and arginase inhibition. CONCLUSIONS: Diet-induced obesity leads to l-arginine deficiency and eNOS uncoupling in PVAT. The combination therapy with l-arginine and arginase inhibitors may represent a novel therapeutic strategy for obesity-induced vascular disease.


Subject(s)
Adipose Tissue/enzymology , Aorta, Thoracic/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Obesity/enzymology , Vasodilation , Adipocytes/enzymology , Adipokines/metabolism , Adipose Tissue/physiopathology , Adiposity , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Arginase/antagonists & inhibitors , Arginase/metabolism , Arginine/deficiency , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/antagonists & inhibitors , Obesity/physiopathology , Paracrine Communication , Phosphorylation , Signal Transduction , Superoxides/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
11.
J Geriatr Cardiol ; 12(5): 528-39, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26512245

ABSTRACT

OBJECTIVE: To find out whether dexamethasone induces an uncoupling of the endothelial nitric oxide synthase (eNOS). METHODS & RESULTS: A major cause of eNOS uncoupling is a deficiency of its cofactor tetrahydrobiopterin (BH4). Treatment of human EA.hy 926 endothelial cells with dexamethasone decreased mRNA and protein expression of both BH4-synthesizing enzymes: GTP cyclohydrolase I and dihydrofolate reductase. Consistently, a concentration- and time-dependent reduction of BH4, dihydrobiopterin (BH2) as well as BH4: BH2 ratio was observed in dexamethasone-treated cells. Surprisingly, no evidence for eNOS uncoupling was found. We then analyzed the expression and phosphorylation of the eNOS enzyme. Dexamethasone treatment led to a down-regulation of eNOS protein and a reduction of eNOS phosphorylation at serine 1177. A reduction of eNOS expression may lead to a relatively normal BH4: eNOS molar ratio in dexamethasone-treated cells. Because the BH4-eNOS stoichiometry rather than the absolute BH4 amount is the key determinant of eNOS functionality (i.e., coupled or uncoupled), the down-regulation of eNOS may represent an explanation for the absence of eNOS uncoupling. Phosphorylation of eNOS at serine 1177 is needed for both the NO-producing activity of the coupled eNOS and the superoxide-producing activity of the uncoupled eNOS. Thus, a reduction of serine 1177 phosphorylation may render a potentially uncoupled eNOS hardly detectable. CONCLUSIONS: Although dexamethasone reduces BH4 levels in endothelial cells, eNOS uncoupling is not evident. The reduction of NO production in dexamethasone-treated endothelial cells is mainly attributable to reduced eNOS expression and decreased eNOS phosphorylation at serine 1177.

12.
Pharmacology ; 96(1-2): 1-10, 2015.
Article in English | MEDLINE | ID: mdl-26021525

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a neurotrophin best characterized for its survival and differentiative effects on neurons. Recent studies demonstrated that BDNF and its receptors are also expressed in the peripheral vasculature, where it stimulates angiogenesis and promotes the survival of endothelial cells. This study was designed to investigate the angiogenic effects of BDNF and its expressional regulation by tumor necrosis factor (TNF-α) and protein kinase C (PKC) in endothelial cells. In the Matrigel angiogenesis assay, BDNF-stimulated vascular tube formation of human umbilical vein endothelial cells (HUVEC) was completely blocked by an inhibition of the TrkB receptor, but only partially inhibited by the inhibition of the p75(NTR) signaling. Treatment of HUVEC and HUVEC-derived EA.hy 926 cells with TNF-α resulted in a downregulation of BDNF expression, which could be prevented by the TNFR1 antagonist WP9QY. BDNF downregulation by TNF-α was associated with decreased angiogenic activity of HUVEC. The effect of TNF-α on BDNF expression could not be abolished by the inhibition of PKC. Treatment of HUVEC and EA.hy 926 cells with PKC-activating phorbol esters (phorbol-12-myristate-13-acetate, PMA or phorbol-12,13-dibutyrate) resulted in a downregulation of BDNF expression, whereas the inactive 4α-phorbol-12,13-didecanoate was without effect. PMA had no significant effect on BDNF mRNA stability and the downregulation of BDNF mRNA expression by PKC activation was likely a transcriptional event. BDNF downregulation by PMA could be prevented by PKC inhibitors Gö 6983 and rottlerin, but not by Gö 6976. Thus, a Gö 6983/rottlerin-sensitive PKC isoform is likely to be responsible for PMA-induced BDNF downregulation.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Down-Regulation , Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Protein Kinase C/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acetophenones/pharmacology , Benzopyrans/pharmacology , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/pharmacology , Carbazoles/pharmacology , Down-Regulation/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Human Umbilical Vein Endothelial Cells , Humans , Indoles/pharmacology , Maleimides/pharmacology , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Phorbol Esters/pharmacology , Protein Kinase C/antagonists & inhibitors , Receptor, trkB/antagonists & inhibitors , Receptors, Nerve Growth Factor/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
13.
Hypertension ; 65(1): 232-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385760

ABSTRACT

Pentaerythritol tetranitrate is devoid of nitrate tolerance and shows no reproductive or developmental toxicity in animal studies. Recently, pentaerythritol tetranitrate has been demonstrated to reduce the risk of intrauterine growth restriction and the risk of preterm birth in women with abnormal placental perfusion. This study was conducted to test the perinatal programming effect of pentaerythritol tetranitrate in spontaneously hypertensive rats, a rat model of genetic hypertension. Parental spontaneously hypertensive rats were treated with pentaerythritol tetranitrate (50 mg/kg per day) during pregnancy and lactation periods; the offspring received standard chow without pentaerythritol tetranitrate after weaning. Maternal treatment with pentaerythritol tetranitrate had no effect on blood pressure in male offspring. In the female offspring, however, a persistent reduction in blood pressure was observed at 6 and 8 months. This long-lasting effect was accompanied by an upregulation of endothelial nitric oxide synthase, mitochondrial superoxide dismutase, glutathione peroxidase 1, and heme oxygenase 1 in the aorta of 8-month-old female offspring, which was likely to result from epigenetic changes (enhanced histone 3 lysine 27 acetylation and histone 3 lysine 4 trimethylation) and transcriptional activation (enhanced binding of DNA-directed RNA polymerase II to the transcription start site of the genes). In organ chamber experiments, the endothelium-dependent, nitric oxide-mediated vasodilation to acetylcholine was enhanced in aorta from female offspring of the pentaerythritol tetranitrate-treated parental spontaneously hypertensive rats. In conclusion, maternal pentaerythritol tetranitrate treatment leads to epigenetic modifications, gene expression changes, an improvement of endothelial function and a persistent blood pressure reduction in the female offspring.


Subject(s)
Blood Pressure/drug effects , Hypertension/drug therapy , Maternal Exposure , Pentaerythritol Tetranitrate/pharmacology , Pregnancy, Animal , Vasodilation/drug effects , Animals , Animals, Newborn , DNA/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Female , Gene Expression Regulation, Developmental , Hypertension/genetics , Hypertension/physiopathology , Male , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Pregnancy , Rats , Rats, Inbred SHR , Vasodilator Agents/pharmacology
14.
Nucleic Acids Res ; 42(20): 12555-69, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25352548

ABSTRACT

Resveratrol shows beneficial effects in inflammation-based diseases like cancer, cardiovascular and chronic inflammatory diseases. Therefore, the molecular mechanisms of the anti-inflammatory resveratrol effects deserve more attention. In human epithelial DLD-1 and monocytic Mono Mac 6 cells resveratrol decreased the expression of iNOS, IL-8 and TNF-α by reducing mRNA stability without inhibition of the promoter activity. Shown by pharmacological and siRNA-mediated inhibition, the observed effects are SIRT1-independent. Target-fishing and drug responsive target stability experiments showed selective binding of resveratrol to the RNA-binding protein KSRP, a central post-transcriptional regulator of pro-inflammatory gene expression. Knockdown of KSRP expression prevented resveratrol-induced mRNA destabilization in human and murine cells. Resveratrol did not change KSRP expression, but immunoprecipitation experiments indicated that resveratrol reduces the p38 MAPK-related inhibitory KSRP threonine phosphorylation, without blocking p38 MAPK activation or activity. Mutation of the p38 MAPK target site in KSRP blocked the resveratrol effect on pro-inflammatory gene expression. In addition, resveratrol incubation enhanced KSRP-exosome interaction, which is important for mRNA degradation. Finally, resveratrol incubation enhanced its intra-cellular binding to the IL-8, iNOS and TNF-α mRNA. Therefore, modulation of KSRP mRNA binding activity and, thereby, enhancement of mRNA degradation seems to be the common denominator of many anti-inflammatory effects of resveratrol.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation Mediators/metabolism , RNA Stability/drug effects , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Stilbenes/pharmacology , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Exosome Multienzyme Ribonuclease Complex/metabolism , Gene Expression/drug effects , Humans , Mice , Mice, Knockout , Mutation , RNA-Binding Proteins/genetics , Resveratrol , Trans-Activators/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Molecules ; 19(10): 16102-21, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25302702

ABSTRACT

Nitric oxide (NO) derived from the endothelial NO synthase (eNOS) has antihypertensive, antithrombotic, anti-atherosclerotic and antiobesogenic properties. Resveratrol is a polyphenol phytoalexin with multiple cardiovascular and metabolic effects. Part of the beneficial effects of resveratrol are mediated by eNOS. Resveratrol stimulates NO production from eNOS by a number of mechanisms, including upregulation of eNOS expression, stimulation of eNOS enzymatic activity and reversal of eNOS uncoupling. In addition, by reducing oxidative stress, resveratrol prevents oxidative NO inactivation by superoxide thereby enhancing NO bioavailability. Molecular pathways underlying these effects of resveratrol involve SIRT1, AMPK, Nrf2 and estrogen receptors.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nitric Oxide/metabolism , Stilbenes/pharmacology , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Nitric Oxide Synthase Type III/metabolism , Protein Binding , Resveratrol , Stilbenes/metabolism
17.
Atherosclerosis ; 237(1): 208-19, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25244505

ABSTRACT

In the vascular wall, reactive oxygen species (ROS) are produced by several enzyme systems including NADPH oxidase, xanthine oxidase, uncoupled endothelial nitric oxide synthase (eNOS) and the mitochondrial electron transport chain. On the other hand, the vasculature is protected by antioxidant enzyme systems, including superoxide dismutases, catalase, glutathione peroxidases and paraoxonases, which detoxify ROS. Cardiovascular risk factors such as hypercholesterolemia, hypertension, and diabetes mellitus enhance ROS generation, resulting in oxidative stress. This leads to oxidative modification of lipoproteins and phospholipids, mechanisms that contribute to atherogenesis. In addition, oxidation of tetrahydrobiopterin may cause eNOS uncoupling and thus potentiation of oxidative stress and reduction of eNOS-derived NO, which is a protective principle in the vasculature. This review summarizes the latest advances in the role of ROS-producing enzymes, antioxidative enzymes as well as NO synthases in the initiation and development of atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Cardiovascular System , Oxidative Stress , Superoxides/chemistry , Animals , Antioxidants/metabolism , Aryldialkylphosphatase/metabolism , Atherosclerosis/metabolism , Catalase/metabolism , Glutathione Peroxidase/metabolism , Humans , Mice , Mice, Transgenic , Mitochondria/metabolism , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Xanthine Oxidase/metabolism
18.
Pharmacology ; 94(1-2): 13-20, 2014.
Article in English | MEDLINE | ID: mdl-25171190

ABSTRACT

BACKGROUND/AIM: It has been demonstrated that dexamethasone-induced hypertension can be prevented by the NADPH oxidase inhibitor apocynin. The effect of dexamethasone on NADPH oxidase, however, is unknown. The present study was conducted to investigate the effect of dexamethasone on the gene expression of Nox1, the major NADPH oxidase isoform in vascular smooth muscle cells. RESULTS: Oral treatment of Wistar-Kyoto rats with dexamethasone (0.03 mg/kg/day) for 12 days led to an upregulation of Nox1 mRNA expression in the aorta. In cultured A7r5 rat aortic smooth muscle cells, dexamethasone increased Nox1 mRNA expression in a concentration- and time-dependent manner. The upregulation of Nox1 mRNA expression was completely prevented by the glucocorticoid receptor antagonist mifepristone. The effect of dexamethasone on Nox1 expression was likely to be indirect as it could be largely blocked by cycloheximide, an inhibitor of protein biosynthesis. Dexamethasone increased Nox1 mRNA stability as well as Nox1 transcription. The dexamethasone-induced Nox1 expression was completely prevented by scriptaid, a pan-inhibitor of histone deacetylases (HDAC), indicating a crucial role for HDAC enzymes. In total, A7r5 cells expressed 8 HDAC isoforms, with HDAC1, 5, 6 and 7 being the most abundant ones. Knockdown of these 4 individual HDAC enzymes did not prevent the effect of dexamethasone on Nox1 expression, although HDAC5 knockdown markedly reduced basal Nox1 expression. CONCLUSION: Dexamethasone upregulates Nox1 expression in vascular smooth muscle cells. This effect involves the glucocorticoid receptor and HDAC enzymes.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , NADH, NADPH Oxidoreductases/genetics , Up-Regulation/drug effects , Animals , Aorta/cytology , Aorta/drug effects , Dexamethasone/administration & dosage , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Glucocorticoids/administration & dosage , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , NADPH Oxidase 1 , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Time Factors
19.
Curr Pharm Biotechnol ; 15(4): 401-8, 2014.
Article in English | MEDLINE | ID: mdl-25022271

ABSTRACT

Resveratrol (3,5,4'-trihydroxy-trans-stilbene) possesses multiple protective properties in the vasculature, including anti-oxidative and anti-inflammatory effects and improvement of endothelial function. A substantial part of these effects is attributable to gene expression changes induced by the compound. Resveratrol can activate the NAD-dependent deacetylase sirtuin 1 (SIRT1), leading to deacetylation of SIRT1 target molecules such as NF-kB and forkhead box O (FOXO) transcription factors. The inhibition of NF-kB by resveratrol reduces the expression of inflammation mediators. FOXO factors are implicated in the upregulation of antioxidant enzymes and the endothelial-type nitric oxide synthase. In addition, resveratrol upregulates a number of antioxidant enzymes by activating nuclear factor-E2-related factor-2 (Nrf2) and downregulates NADPH oxidases through yet known mechanisms.


Subject(s)
Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Stilbenes/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Endothelium, Vascular/metabolism , Humans , Reactive Oxygen Species/metabolism , Resveratrol , Sirtuin 1/metabolism
20.
Molecules ; 19(3): 3654-68, 2014 Mar 24.
Article in English | MEDLINE | ID: mdl-24662080

ABSTRACT

Artichoke (Cynara scolymus L.) is one of the world's oldest medicinal plants with multiple health benefits. We have previously shown that artichoke leaf extracts and artichoke flavonoids upregulate the gene expression of endothelial-type nitric oxide synthase (eNOS) in human endothelial cells. Whereas NO produced by the eNOS is a vasoprotective molecule, NO derived from the inducible iNOS plays a pro-inflammatory role in the vasculature. The present study was aimed to investigate the effects of artichoke on iNOS expression in human coronary artery smooth muscle cells (HCASMC). Incubation of HCASMC with a cytokine mixture led to an induction of iNOS mRNA expression. This iNOS induction was concentration- and time-dependently inhibited by an artichoke leaf extract (1-100 µg/mL, 6 h or 24 h). Consistently, the artichoke leaf extract also reduced cytokine-induced iNOS promoter activation and iNOS protein expression. In addition, treatment of HCASMC with four well-known artichoke compounds (cynarin > cyanidin > luteolin ≈ cynaroside) led to a downregulation iNOS mRNA and protein expression, with cynarin being the most potent one. In conclusion, artichoke contains both eNOS-upregulating and iNOS-downregulating compounds. Such compounds may contribute to the beneficial effects of artichoke and may per se have therapeutic potentials.


Subject(s)
Anthocyanins/pharmacology , Cinnamates/pharmacology , Coronary Vessels , Cynara scolymus/chemistry , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Synthase Type II/genetics , Cells, Cultured , Down-Regulation/drug effects , Humans , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...