Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.918
Filter
2.
J Neuroimmune Pharmacol ; 15(2): 292-308, 2020 06.
Article in English | MEDLINE | ID: mdl-31897976

ABSTRACT

Cellular arachidonic acid (AA), an unsaturated fatty acid found ubiquitously in plasma membranes, is metabolized to different prostanoids, such as prostacyclin (PGI2) and prostaglandin E2 (PGE2), by the three-step reactions coupling the upstream cyclooxygenase (COX) isoforms (COX-1 and COX-2) with the corresponding individual downstream synthases. While the vascular actions of these prostanoids are well-characterized, their specific roles in the hippocampus, a major brain area for memory, are poorly understood. The major obstacle for its understanding in the brain was to mimic the biosynthesis of each prostanoid. To solve the problem, we utilized Single-Chain Hybrid Enzyme Complexes (SCHECs), which could successfully control cellular AA metabolites to the desired PGI2 or PGE2. Our in vitro studies suggested that neurons with higher PGI2 content and lower PGE2 content exhibited survival protection and resistance to Amyloid-ß-induced neurotoxicity. Further extending to an in vivo model, the hybrid of PGI2-producing transgenic mice and Alzheimer's disease (AD) mice showed restored long-term memory. These findings suggested that the vascular prostanoids, PGI2 and PGE2, exerted significant regulatory influences on neuronal protection (by PGI2), or damage (by PGE2) in the hippocampus, and raised a concern that the wide uses of aspirin in cardiovascular diseases may exert negative impacts on neurodegenerative protection. Graphic Abstract Our study intended to understand the crosstalk of prostanoids in the hippocampus, a major brain area impacted in AD, by using hybrid enzymes to redirect the synthesis of prostanoids to PGE2 and PGI2, respectively. Our data indicated that during inflammation, the vascular mediators, PGI2 and PGE2, exerted significant regulatory influences on neuronal protection (by PGI2), or damage (by PGE2) in the hippocampus. These findings also raised a concern that the widely uses of non-steroidal anti-inflammatory drugs in cardiovascular diseases may exert negative impacts on neurodegenerative protection.


Subject(s)
Epoprostenol/biosynthesis , Hippocampus/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Up-Regulation/physiology , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Epoprostenol/genetics , Hippocampus/drug effects , Hippocampus/pathology , Iloprost/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Up-Regulation/drug effects
3.
FASEB J ; 33(1): 1510-1521, 2019 01.
Article in English | MEDLINE | ID: mdl-30156911

ABSTRACT

Despite its well-known antithrombotic properties, the effect of aspirin on blood pressure (BP) and hypertension pathology is unclear. The hugely varying doses used clinically have contributed to this confusion, with high-dose aspirin still commonly used due to concerns about the efficacy of low-dose aspirin. Because prostaglandins have been shown to both promote and inhibit T-cell activation, we also explored the immunomodulatory properties of aspirin in hypertension. Although the common preclinical high dose of 100 mg/kg/d improved vascular dysfunction and cardiac hypertrophy, this effect was accompanied by indices of elevated adaptive immunity, renal T-cell infiltration, renal fibrosis, and BP elevation in stroke-prone spontaneously hypertensive rats and in angiotensin II-induced hypertensive mice. The cardioprotective effects of aspirin were conserved with a lower dose (10 mg/kg/d) while circumventing heightened adaptive immunity and elevated BP. We also show that low-dose aspirin improves renal fibrosis. Differential inhibition of the COX-2 isoform may underlie the disparate effects of the 2 doses. Our results demonstrate the efficacy of low-dose aspirin in treating a vast array of cardiovascular parameters and suggest modulation of adaptive immunity as a novel mechanism underlying adverse cardiovascular profiles associated with COX-2 inhibitors. Clinical studies should identify the dose of aspirin that achieves maximal cardioprotection with a new awareness that higher doses of aspirin could trigger undesired autoimmunity in hypertensive individuals. This work also warrants an evaluation of high-dose aspirin and COX-2 inhibitor therapy in sufferers of inflammatory conditions who are already at increased risk for cardiovascular disease.-Khan, S. I., Shihata, W. A., Andrews, K. L., Lee, M. K. S., Moore, X.-L., Jefferis, A.-M., Vinh, A., Gaspari, T., Dragoljevic, D., Jennings, G. L., Murphy, A. J., Chin-Dusting, J. P. F. Effects of high- and low-dose aspirin on adaptive immunity and hypertension in the stroke-prone spontaneously hypertensive rat.


Subject(s)
Adaptive Immunity/drug effects , Aspirin/pharmacology , Hypertension/drug therapy , Stroke/complications , Stroke/immunology , Angiotensin II/pharmacology , Animals , Aspirin/administration & dosage , Aspirin/therapeutic use , Biomarkers/blood , Blood Pressure/drug effects , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/physiopathology , Cardiomegaly/drug therapy , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cytokines/blood , Disease Susceptibility , Dose-Response Relationship, Drug , Epoprostenol/biosynthesis , Hypertension/chemically induced , Kidney/drug effects , Kidney/enzymology , Kidney/pathology , Mice , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Real-Time Polymerase Chain Reaction , Systole , T-Lymphocytes/immunology , Thromboxanes/blood
4.
Biol Reprod ; 100(1): 162-174, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30481284

ABSTRACT

The prostacyclin (prostaglandin I2) signaling system is an essential regulator of vascular homeostasis. Since the corpus luteum is a highly vascularized gland, prostacyclin seems to be crucial for luteal development and function. Although progress has been made in understanding the luteotropic action of prostacyclin in mammals, its role in the porcine corpus luteum remains to be determined. Therefore, studies were conducted to (1) determine profiles of prostacyclin synthase expression and prostacyclin metabolite concentration, as well as prostacyclin G-protein-coupled receptor expression in porcine luteal tissue on days 2 to 16 of the estrous cycle and days 10 to 30 of pregnancy using real-time PCR, western blot, or enzyme immunoassay; and (2) examine the effect of prostacyclin on progesterone synthesis in vitro. To accomplish the second aim, luteal cells were treated with prostacyclin analogs, iloprost and carbaprostacyclin, in the presence or absence of prostacyclin receptor antagonists. The mRNA expression of cytochrome P450 family 11 subfamily A member 1 and hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1 was analyzed using real-time PCR, while progesterone concentration in culture medium was assessed by radioimmunoassay.Dynamic changes of prostacyclin synthase and prostacyclin receptor expression were observed in porcine luteal tissue during the estrous cycle and early pregnancy. Moreover, prostacyclin stimulated progesterone production and this effect was abolished by the addition of prostacyclin receptor antagonists. Our findings provide strong evidence that prostacyclin and its signaling system are present in corpus luteum of the pig and may directly promote luteotropic activity through upregulation of progesterone synthesis.


Subject(s)
Corpus Luteum/metabolism , Epoprostenol/biosynthesis , Luteal Cells/metabolism , Receptors, Epoprostenol/genetics , Animals , Cells, Cultured , Corpus Luteum/cytology , Female , Gene Expression , Pregnancy , Receptors, Epoprostenol/metabolism , Swine
5.
PLoS One ; 12(12): e0189658, 2017.
Article in English | MEDLINE | ID: mdl-29261735

ABSTRACT

INTRODUCTION: Long chain n-3 fatty acid supplementation may modulate septic shock-induced host response to pathogen-induced sepsis. The composition of lipid emulsions for parenteral nutrition however remains a real challenge in intensive care, depending on their fatty acid content. Because they have not been assessed yet, we aimed at determining the respective effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) during septic shock-induced vascular dysfunction. METHODS: In a peritonitis-induced septic shock model, rats were infused with EPA, DHA, an EPA/DHA mixture or 5% dextrose (D5) during 22 hours. From H18, rats were resuscitated and monitored during 4 hours. At H22, plasma, aorta and mesenteric resistance arteries were collected to perform ex vivo experiments. RESULTS: We have shown that septic rats needed an active resuscitation with fluid challenge and norepinephrine treatment, while SHAM rats did not. In septic rats, norepinephrine requirements were significantly decreased in DHA and EPA/DHA groups (10.6±12.0 and 3.7±8.0 µg/kg/min respectively versus 17.4±19.3 µg/kg/min in D5 group, p<0.05) and DHA infusion significantly improved contractile response to phenylephrine through nitric oxide pathway inhibition. DHA moreover significantly reduced vascular oxidative stress and nitric oxide production, phosphorylated IκB expression and vasodilative prostaglandin production. DHA also significantly decreased polyunsaturated fatty acid pro-inflammatory mediators and significantly increased several anti-inflammatory metabolites. CONCLUSIONS: DHA infusion in septic rats improved hemodynamic dysfunction through decreased vascular oxidative stress and inflammation, while EPA infusion did not have beneficial effects.


Subject(s)
Arteries/pathology , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Shock, Septic/complications , Vascular Diseases/drug therapy , Animals , Epoprostenol/biosynthesis , Male , Nitric Oxide/biosynthesis , Norepinephrine/administration & dosage , Oxidative Stress , Rats , Rats, Wistar , Vascular Diseases/etiology
6.
Bull Exp Biol Med ; 164(2): 123-126, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29181669

ABSTRACT

We studied the effect of IFNα-2b and IFNß-1a on phasic and tonic contractions of isolated bovine mesenteric lymphatic vessels and nodes. IFNα-2b and IFNß-1a in concentrations of 250-1000 U/ml produced dose-dependent negative chronotropic and inotropic effects on spontaneous phasic contractions and tonus of lymphatic vessels and nodes. In de-endothelialized lymphatic vessels and nodes, IFNα-2b and IFNß-1a in the same concentrations had less pronounced inhibitory effect on spontaneous contraction and tonus. L-NAME (100 µM) and charybdotoxin (0.1 µM with 0.5 µM apamine) significantly attenuated the inhibitory effect of IFNα-2b on phasic and tonic contractions of lymph nodes. L-NAME (100 µM) and indomethacin (10 µM) significantly reduced the IFNα-2b-induced inhibitory effect on phasic and tonic contractions of lymph node. These results indicate that IFNα-2b and IFNß-1a have a pronounced inhibitory effect on the phasic and tonic contractions of bovine mesenteric lymphatic vessels and nodes. The responses are endothelium-dependent and are determined by production of NO and endothelium-dependent hyperpolarizing factor by endotheliocytes in lymphatic vessels and by production of NO and prostacyclin by endotheliocytes in the lymphatic nodes.


Subject(s)
Interferon beta-1a/pharmacology , Interferon-alpha/pharmacology , Lymph Nodes/drug effects , Lymphatic Vessels/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Animals , Apamin/pharmacology , Cattle , Charybdotoxin/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epoprostenol/biosynthesis , Epoprostenol/metabolism , Indomethacin/pharmacology , Interferon alpha-2 , Interferon beta-1a/antagonists & inhibitors , Interferon-alpha/antagonists & inhibitors , Lymph Nodes/cytology , Lymph Nodes/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mesentery/cytology , Mesentery/drug effects , Mesentery/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Recombinant Proteins/pharmacology , Tissue Culture Techniques
7.
Eur J Pharmacol ; 814: 87-94, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28780058

ABSTRACT

In this work, we hypothesized that cyclooxygenase (COX) activity can be regulated by nitric oxide (NO) and hydrogen peroxide (H2O2). In the renal hypertension (2K-1C), phenylephrine (PE)-induced contraction was lower than in normotensive (2K) rat aortas. This impaired contraction is due to NO/H2O2- induced vasodilation. We evaluated the effects of H2O2 on the activity of COX and endothelial NO-Synthase (eNOS) in 2K-1C rat aortas stimulated with PE. Responses for PE or H2O2 were evaluated in 2K-1C and 2K rat aortas, without or with inhibitors for COX (Indomethacin) or eNOS (L-NAME). COX isoforms expression was evaluated by Western blotting. eNOS inhibition was tested on thromboxane A2 (TXA2) and prostacyclin (PGI2) production. PE-induced contraction was lower in 2K-1C than in 2K. Indomethacin reduced PE-induced contraction in 2K, but it had no effect in 2K-1C. L-NAME reversed indomethacin-induced effect in 2K and it normalized PE-induced contraction in 2K-1C to the normotensive levels. COX-1 and COX-2 expression, TXA2 and PGI2 production were higher in 2K-1C than in 2K. eNOS inhibition did no modify TXA2/PGI2 production. In low concentrations, H2O2 induced relaxation only in 2K that was abolished by L-NAME while the contractions induced by high concentrations were abolished by indomethacin in both 2K and 2K-1C. The activity/expression of COX, and TXA2/PGI2 production were increased in 2K-1C, which were not modified by eNOS. High levels of H2O2 increased the endothelial COX activity, which induced contraction. Therefore, an high increase in H2O2 production may increase COX-induced vasoconstriction rather than eNOS-induced relaxation, which might contribute to aggravate hypertension.


Subject(s)
Aorta/drug effects , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Hydrogen Peroxide/pharmacology , Hypertension, Renal/metabolism , Nitric Oxide Synthase Type III/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Aorta/metabolism , Aorta/physiopathology , Blood Pressure/drug effects , Cyclooxygenase Inhibitors/pharmacology , Enzyme Activation/drug effects , Epoprostenol/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Hypertension, Renal/physiopathology , Male , Nitric Oxide Synthase Type III/antagonists & inhibitors , Rats , Thromboxane A2/biosynthesis , Vasoconstriction/drug effects , Vasodilation/drug effects
8.
BMC Surg ; 17(1): 90, 2017 Aug 11.
Article in English | MEDLINE | ID: mdl-28800765

ABSTRACT

BACKGROUND: This study aimed to reveal the appropriate timing for the intravenous administration of flurbiprofen axetil for preventing mesenteric traction syndrome (MTS), caused by prostacyclin release. METHODS: In this prospective, randomized, clinical study, forty-five patients who were undergoing elective surgery for colorectal cancer via laparotomy were enrolled. Patients were randomly divided into 3 groups: a preoperative group (n = 16) receiving flurbiprofen axetil directly before surgery; a post-MTS group (n = 14) receiving following MTS onset; and a control group (n = 15) who were not administered flurbiprofen axetil. 6-keto-PGF1α, a stable metabolite of prostacyclin, levels were measured and mean blood pressures were recorded. RESULTS: In the preoperative group, 6-keto-PGF1α levels did not increase, blood pressure levels did not decrease, and no facial flushing was observed. In both the post-MTS and control groups, 6-keto-PGF1α levels increased markedly after mesenteric traction and blood pressure decreased significantly. The post-MTS group exhibited a faster decreasing trend in 6-keto-PGF1α levels and quick restore of the mean blood pressure, and the use of vasopressors and phenylephrine were lower than that in the control group. CONCLUSIONS: Even therapeutic administration of flurbiprofen axetil after the onset of MTS has also effects on MTS by suppressing prostacyclin production. TRIAL REGISTRATION: Clinical trial number: UMIN000009111 . (Registered 14 October 2012).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Flurbiprofen/analogs & derivatives , Flushing/drug therapy , Hemodynamics/drug effects , Hypotension/drug therapy , Intraoperative Complications/drug therapy , Tachycardia/drug therapy , 6-Ketoprostaglandin F1 alpha/blood , Aged , Blood Pressure/drug effects , Colorectal Neoplasms/surgery , Epoprostenol/antagonists & inhibitors , Epoprostenol/biosynthesis , Female , Flurbiprofen/administration & dosage , Flushing/prevention & control , Humans , Hypotension/prevention & control , Infusions, Intravenous , Intraoperative Complications/prevention & control , Laparotomy , Male , Middle Aged , Prospective Studies , Syndrome , Tachycardia/prevention & control
9.
Clin Exp Hypertens ; 39(3): 210-219, 2017.
Article in English | MEDLINE | ID: mdl-28448188

ABSTRACT

In endothelium-denuded abdominal (but not thoracic) aortas of rats, the nonselective cyclooxygenase (COX) inhibitor, indomethacin, suppressed contractions evoked by α-adrenergic agonists hypothetically mediated by prostanoids. We aimed to identify these non-endothelial-derived contractile prostanoids released by α-adrenergic receptors activation. Endothelium-denuded abdominal and thoracic aortas of Wistar rats were used for biochemical and functional analyses. Western blot analysis showed that COX-1 and COX-2 protein levels were respectively equivalent in endothelium-denuded abdominal and thoracic aortas. Enzyme immunoassay data supported direct evidence of phenylephrine-stimulated release of prostanoids (PGI2, PGE2, and PGF2α) by thoracic and abdominal aortas without endothelium, and their almost complete inhibition by 1 µM indomethacin. Isometric force measurements established that 10 µM indomethacin-but no lower concentrations-inhibited the contractions evoked by phenylephrine in endothelium-denuded abdominal aorta. In this preparation, 10 µM indomethacin also depressed the contractions provoked by angiotensin II and high K+ (80 mM). In fact, indomethacin (up to 1 mM) caused concentration-dependent reductions in all abovementioned contractile responses. In endothelium-denuded thoracic aortas, however, only 1 mM indomethacin significantly depressed the contractile activity stimulated by either phenylephrine, angiotensin II, or high K+. Hence, there was a clear quantitative difference in response to indomethacin between abdominal and thoracic aortas without endothelium. Altogether, the results indicate that prostanoids induced by phenylephrine in abdominal and thoracic aortas were derived from non-endothelial COX-mediated metabolism; notably, the decrease in prostanoid synthesis could not account for the inhibition of vasoconstrictor responses by indomethacin: Through COX-independent actions, indomethacin inhibited aortic smooth muscle contractility.


Subject(s)
Aorta, Abdominal/drug effects , Aorta, Thoracic/drug effects , Biosynthetic Pathways/drug effects , Cyclooxygenase Inhibitors/pharmacology , Indomethacin/pharmacology , Muscle Contraction/drug effects , Angiotensin II/pharmacology , Animals , Aorta, Abdominal/metabolism , Aorta, Thoracic/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Epoprostenol/biosynthesis , In Vitro Techniques , Male , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Phenylephrine/pharmacology , Potassium/pharmacology , Rats , Rats, Wistar , Vasoconstrictor Agents/pharmacology
10.
J Cell Mol Med ; 21(8): 1584-1592, 2017 08.
Article in English | MEDLINE | ID: mdl-28244682

ABSTRACT

Extracellular histones are mediators of inflammation, tissue injury and organ dysfunction. Interactions between circulating histones and vascular endothelial cells are key events in histone-mediated pathologies. Our aim was to investigate the implication of extracellular histones in the production of the major vasoactive compounds released by human endothelial cells (HUVECs), prostanoids and nitric oxide (NO). HUVEC exposed to increasing concentrations of histones (0.001 to 100 µg/ml) for 4 hrs induced prostacyclin (PGI2) production in a dose-dependent manner and decreased thromboxane A2 (TXA2) release at 100 µg/ml. Extracellular histones raised cyclooxygenase-2 (COX-2) and prostacyclin synthase (PGIS) mRNA and protein expression, decreased COX-1 mRNA levels and did not change thromboxane A2 synthase (TXAS) expression. Moreover, extracellular histones decreased both, eNOS expression and NO production in HUVEC. The impaired NO production was related to COX-2 activity and superoxide production since was reversed after celecoxib (10 µmol/l) and tempol (100 µmol/l) treatments, respectively. In conclusion, our findings suggest that extracellular histones stimulate the release of endothelial-dependent mediators through an up-regulation in COX-2-PGIS-PGI2 pathway which involves a COX-2-dependent superoxide production that decreases the activity of eNOS and the NO production. These effects may contribute to the endothelial cell dysfunction observed in histone-mediated pathologies.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Epoprostenol/agonists , Histones/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/metabolism , Thromboxane A2/antagonists & inhibitors , Celecoxib/pharmacology , Cyclic N-Oxides/pharmacology , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Epoprostenol/biosynthesis , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/genetics , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Spin Labels , Superoxides/antagonists & inhibitors , Superoxides/metabolism , Thromboxane A2/biosynthesis , Thromboxane-A Synthase/genetics , Thromboxane-A Synthase/metabolism
11.
Eur J Pharmacol ; 802: 7-19, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28228357

ABSTRACT

Mammalian target of rapamycin (mTOR), a serine/threonine kinase plays an important role in various pathophysiological processes including cancer, metabolic diseases, and inflammation. Although mTOR participates in Toll-like receptor 4 signalling in different cell types, the role of this enzyme in sepsis pathogenesis and its effects on hypotension and inflammation in endotoxemic rats remains unclear. In this study we investigated the effects of mTOR inhibition on lipopolysaccharide (LPS)-induced changes on expressions and/or activities of ribosomal protein S6 (rpS6), an mTOR substrate, nuclear factor-κB (NF-κB) p65, inhibitor κB (IκB)-α, inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2 with production of nitric oxide, peroxynitrite, prostacyclin, and tumor necrosis factor (TNF)-α and activity of myeloperoxidase (MPO), which results in hypotension and inflammation. Injection of LPS (10mg/kg, i.p.) to male Wistar rats decreased blood pressure and increased heart rate that were associated with elevated nitrotyrosine, 6-keto-PGF1α, and TNF-α levels and MPO activity, and increased expressions and/or activities of rpS6, NF-κB p65, iNOS, and COX-2 and decreased expression of IκB-α in renal, cardiac, and vascular tissues. LPS also increased serum and tissue nitrite levels. Rapamycin (1mg/kg, i.p.) given one h after injection of LPS reversed these effects of LPS. These data suggest that the activation of mTOR/IκB-α/NF-κB pathway associated with vasodilator and proinflammatory mediator formation contributes to LPS-induced hypotension and inflammation.


Subject(s)
Hypotension/chemically induced , Hypotension/pathology , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Arterial Pressure/drug effects , Cyclooxygenase 2/metabolism , Epoprostenol/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Heart Rate/drug effects , Hypotension/metabolism , Hypotension/physiopathology , Inflammation/chemically induced , Inflammation/pathology , Male , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Peroxynitrous Acid/biosynthesis , Rats , Rats, Wistar , Ribosomal Protein S6/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tyrosine/analogs & derivatives , Tyrosine/metabolism
12.
Am J Physiol Heart Circ Physiol ; 312(3): H485-H500, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28011582

ABSTRACT

Vascular endothelial cells play an important role in the regulation of vascular function in response to mechanical stimuli in both healthy and diseased states. Prostaglandin I2 (PGI2) is an important antiatherogenic prostanoid and vasodilator produced in endothelial cells through the action of the cyclooxygenase (COX) isoenzymes COX-1 and COX-2. However, the mechanisms involved in sustained, shear-induced production of COX-2 and PGI2 have not been elucidated but are determined in the present study. We used cultured endothelial cells exposed to steady fluid shear stress (FSS) of 10 dyn/cm2 for 5 h to examine shear stress-induced induction of COX-2/PGI2 Our results demonstrate the relationship between the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1) and the intracellular mechanoresponsive molecules phosphatidylinositol 3-kinase (PI3K), focal adhesion kinase (FAK), and mitogen-activated protein kinase p38 in the FSS induction of COX-2 expression and PGI2 release. Knockdown of PECAM-1 (small interference RNA) expression inhibited FSS-induced activation of α5ß1-integrin, upregulation of COX-2, and release of PGI2 in both bovine aortic endothelial cells (BAECs) and human umbilical vein endothelial cells (HUVECs). Furthermore, inhibition of the PI3K pathway (LY294002) substantially inhibited FSS activation of α5ß1-integrin, upregulation of COX-2 gene and protein expression, and release of PGI2 in BAECs. Inhibition of integrin-associated FAK (PF573228) and MAPK p38 (SB203580) also inhibited the shear-induced upregulation of COX-2. Finally, a PECAM-1-/- mouse model was characterized by reduced COX-2 immunostaining in the aorta and reduced plasma PGI2 levels compared with wild-type mice, as well as complete inhibition of acute flow-induced PGI2 release compared with wild-type animals.NEW & NOTEWORTHY In this study we determined the major mechanotransduction pathway by which blood flow-driven shear stress activates cyclooxygenase-2 (COX-2) and prostaglandin I2 (PGI2) release in endothelial cells. Our work has demonstrated for the first time that COX-2/PGI2 mechanotransduction is mediated by the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1).


Subject(s)
Cyclooxygenase 2/biosynthesis , Endothelial Cells/metabolism , Epoprostenol/biosynthesis , Stress, Mechanical , Animals , Cattle , Cell Line , Cilia/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Integrins/metabolism , Peptides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Signal Transduction/physiology , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Am J Cardiol ; 118(12): 1941-1947, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27769511

ABSTRACT

High platelet reactivity and high platelet turnover have been implicated in incomplete platelet inhibition during immediate-release acetylsalicylic acid therapy in patients with type 2 diabetes mellitus (DM). An extended-release acetylsalicylic acid (ER-ASA; Durlaza) formulation was developed to provide 24-hour antithrombotic effects with once-daily dosing. The objective of the study was to evaluate the antiplatelet effects of ER-ASA in patients with DM. In this open-label, single-center study, patients with DM (n = 40) and multiple cardiovascular risk factors received ER-ASA 162.5 mg/day for 14 ± 4 days. Multiple platelet function tests, serum and urinary thromboxane B2 metabolites, prostacyclin metabolite, and high-sensitive C-reactive protein levels were assessed at 1, 12, 16, and 24 hours post-dose. Patients with high platelet turnover and/or high platelet reactivity were treated with ER-ASA 325 mg/day for 14 ± 4 days, and laboratory analyses were repeated. All patients responded to ER-ASA 162.5 mg/day as measured by arachidonic acid-induced aggregation, and there was no loss of the platelet inhibitory effect of ER-ASA 162.5 mg/day over 24 hours post-dose (p = not significant). The antiplatelet effect was sustained over 24 hours for all platelet function measurements. Mean 1- to 24-hour serum thromboxane B2 levels were low with both doses and were lower with ER-ASA 325 mg/day compared with 162.5 mg/day therapy (p = 0.002). In conclusion, ER-ASA 162.5 mg daily dose provided sustained antiplatelet effects over 24 hours in patients with type 2 DM and multiple cardiovascular risk factors and had a favorable tolerability profile.


Subject(s)
Aspirin/administration & dosage , Cardiovascular Diseases/prevention & control , Coronary Artery Disease/drug therapy , Peripheral Vascular Diseases/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Aged , Aspirin/therapeutic use , C-Reactive Protein/metabolism , Coronary Artery Disease/complications , Delayed-Action Preparations , Diabetes Mellitus, Type 2/complications , Epoprostenol/biosynthesis , Epoprostenol/urine , Female , Humans , Male , Manometry , Mean Platelet Volume , Middle Aged , Peripheral Vascular Diseases/complications , Platelet Aggregation , Platelet Aggregation Inhibitors/therapeutic use , Thrombelastography , Thromboxane B2/analogs & derivatives , Thromboxane B2/blood , Thromboxane B2/urine , Vasodilation
14.
J Chin Med Assoc ; 79(11): 583-588, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27580512

ABSTRACT

BACKGROUND: Hepatic encephalopathy (HE) is a complex neuropsychiatric syndrome secondary to acute or chronic liver failure. However, its pathophysiology remains obscure. Recently, we found that the inhibition of cyclooxygenase by indomethacin aggravated HE in rats with thioacetamide-induced acute hepatic failure, suggesting a pivotal role of cyclooxygenase in HE. This study was aimed at surveying the roles of cyclooxygenase isoforms responsible for prostaglandins synthesis, cyclooxygenase-1 (COX1) and COX2, in cirrhotic rats with HE. METHODS: Liver cirrhosis was induced (using formalin) in male Sprague-Dawley rats with bile duct ligation (FBDL). Sham-operated rats served as the surgical controls. The severity of HE was assessed by motor activity counts. Plasma 6-keto-prostaglandin-F1α [6-keto-PGF1α; a relatively stable metabolite of prostacyclin (PGI2)], alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALK-P), and total bilirubin were measured. Hepatic mRNA expressions of COX1 and COX2 were tested. RESULTS: The FBDL group showed lower motor activity counts than the sham group in total (1472 ± 156 vs. 2174 ± 262 counts/30 min, p = 0.034), ambulatory (824 ± 99 vs. 1443 ± 206 counts/30 min, p = 0.014), and vertical movement (431 ± 69 vs. 849 ± 145 counts/30 min, p = 0.018). The mRNA expression of hepatic COX2 was significantly higher in the FBDL group. Plasma ALK-P and bilirubin levels were negatively correlated with total movements, respectively (both p < 0.05). In addition, hepatic COX2 mRNA expression was positively correlated with AST, ALK-P, total bilirubin, and 6-keto-PGF1α (all p < 0.05), but not correlated with total movements. CONCLUSION: Hepatic COX2 expression and PGI2 production are enhanced in cirrhotic rats, but the correlation with HE is not remarkable. Cyclooxygenase and PGI2 may not play important roles in HE in the setting of chronic liver failure.


Subject(s)
Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Hepatic Encephalopathy/etiology , Liver Cirrhosis/complications , 6-Ketoprostaglandin F1 alpha/blood , Animals , Epoprostenol/biosynthesis , Liver/enzymology , Male , Motor Activity , Rats , Rats, Sprague-Dawley
15.
J Biol Chem ; 291(13): 7070-86, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26841868

ABSTRACT

In Candida albicans-infected resident peritoneal macrophages, activation of group IVA cytosolic phospholipase A2(cPLA2α) by calcium- and mitogen-activated protein kinases triggers the rapid production of prostaglandins I2 and E2 through cyclooxygenase (COX)-1 and regulates gene expression by increasing cAMP. InC. albicans-infected cPLA2α(-/-)or COX-1(-/-)macrophages, expression ofI l10,Nr4a2, and Ptgs2 was lower, and expression ofTnfα was higher, than in wild type macrophages. Expression was reconstituted with 8-bromo-cAMP, the PKA activator 6-benzoyl-cAMP, and agonists for prostaglandin receptors IP, EP2, and EP4 in infected but not uninfected cPLA2α(-/-)or COX-1(-/-)macrophages. InC. albicans-infected cPLA2α(+/+)macrophages, COX-2 expression was blocked by IP, EP2, and EP4 receptor antagonists, indicating a role for both prostaglandin I2 and E2 Activation of ERKs and p38, but not JNKs, by C. albicansacted synergistically with prostaglandins to induce expression of Il10,Nr4a2, and Ptgs2. Tnfα expression required activation of ERKs and p38 but was suppressed by cAMP. Results using cAMP analogues that activate PKA or Epacs suggested that cAMP regulates gene expression through PKA. However, phosphorylation of cAMP-response element-binding protein (CREB), the cAMP-regulated transcription factor involved inIl10,Nr4a2,Ptgs2, andTnfα expression, was not mediated by cAMP/PKA because it was similar inC. albicans-infected wild type and cPLA2α(-/-)or COX-1(-/-)macrophages. CREB phosphorylation was blocked by p38 inhibitors and induced by the p38 activator anisomycin but not by the PKA activator 6-benzoyl-cAMP. Therefore, MAPK activation inC. albicans-infected macrophages plays a dual role by promoting the cPLA2α/prostaglandin/cAMP/PKA pathway and CREB phosphorylation that coordinately regulate immediate early gene expression.


Subject(s)
Candida albicans/physiology , Cyclooxygenase 1/immunology , Gene Expression Regulation , Group IV Phospholipases A2/immunology , Host-Pathogen Interactions , Macrophages, Peritoneal/immunology , Membrane Proteins/immunology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Dinoprostone/biosynthesis , Epoprostenol/biosynthesis , Group IV Phospholipases A2/deficiency , Group IV Phospholipases A2/genetics , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/microbiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/immunology , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
16.
Adv Med Sci ; 61(1): 154-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26773760

ABSTRACT

PURPOSE: Obstructive sleep apnea increases the risk of cardiovascular diseases. Alternations in prostacyclin and thromboxane concentrations and balance could constitute one of mechanisms linking sleep apnea and cardiovascular events. Thus we aimed to assess the concentrations of 6-keto-prostaglandin F1α (6-keto-PGF1α) (metabolite of prostacyclin) and thromboxane B2 (TXB2) (metabolite of thromboxane A2) in urine and blood of obstructive sleep apnea patients and controls (snoring subjects with otherwise normal polysomnogram). MATERIAL AND METHODS: Overnight urine and morning blood samples were taken from subjects and controls at baseline and in sleep apnea group during continuous positive airway pressure (CPAP) treatment. Samples were analyzed using mass chromatography/gas spectrometry. RESULTS: We analyzed data from 26 obstructive sleep apnea subjects (mean apnea-hypopnea index 45.4±17.3) and 22 well-matched controls. At baseline sleep apnea patients, when compared to controls, have higher 6-keto-PGF1α in urine (0.89±0.15 vs 0.34±0.06, p=0.01) and blood (24.49±1.54 vs 19.70±1.77, p=0.04). TXB2 levels in urine and blood were not different across groups. CPAP treatment significantly decreased 6-keto-PGF1α in urine (0.92±0.17 vs 0.22±0.10, p=0.04), but not in blood. TXB2 levels during CPAP treatment did not change significantly. CONCLUSIONS: These results suggest augmented systemic prostacyclin production in obstructive sleep apnea patients, which potentially could constitute a protective mechanism against detrimental effects of sleep apnea.


Subject(s)
Epoprostenol/biosynthesis , Sleep Apnea, Obstructive/metabolism , Thromboxanes/biosynthesis , 6-Ketoprostaglandin F1 alpha/blood , 6-Ketoprostaglandin F1 alpha/urine , Adult , Aged , Case-Control Studies , Continuous Positive Airway Pressure , Female , Humans , Male , Middle Aged , Sleep Apnea, Obstructive/blood , Sleep Apnea, Obstructive/urine , Thromboxane B2/blood , Thromboxane B2/urine
18.
Diabetes ; 65(2): 503-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26470782

ABSTRACT

Platelet activation is persistently enhanced, and its inhibition by low-dose aspirin is impaired in type 2 diabetes mellitus. We investigated in vivo thromboxane (TX) and prostacyclin (PGI2) biosynthesis and their determinants, as well as aspirin responsiveness, in young adult subjects with type 1 diabetes mellitus (T1DM) without overt cardiovascular disease and stable glycemic control. The biosynthesis of TXA2 was persistently increased in subjects with T1DM versus matched healthy subjects, with females showing higher urinary TX metabolite (TXM) excretion than male subjects with T1DM. Microalbuminuria and urinary 8-iso-PGF2α, an index of in vivo oxidative stress, independently predicted TXM excretion in T1DM. No homeostatic increase in PGI2 biosynthesis was detected. Platelet COX-1 suppression by low-dose aspirin and the kinetics of its recovery after drug withdrawal were similar in patients and control subjects and were unaffected by glucose variability. We conclude that patients with T1DM and stable glycemic control display enhanced platelet activation correlating with female sex and microvascular and oxidative damages. Moreover, aspirin responsiveness is unimpaired in T1DM, suggesting that the metabolic disturbance per se is unrelated to altered pharmacodynamics. The efficacy and safety of low-dose aspirin in T1DM warrant further clinical investigation.


Subject(s)
Aspirin/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Epoprostenol/biosynthesis , Platelet Activation/physiology , Platelet Aggregation Inhibitors/pharmacology , Thromboxane A2/biosynthesis , Adult , Albuminuria/urine , Aspirin/administration & dosage , Blood Glucose/analysis , Cross-Sectional Studies , Cyclooxygenase 1/drug effects , Diabetes Mellitus, Type 1/metabolism , Dinoprost/analogs & derivatives , Dinoprost/urine , Female , Humans , Male , Matched-Pair Analysis , Microvessels/physiopathology , Middle Aged , Oxidative Stress/physiology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Sex Factors , Thromboxane A2/urine
19.
Clin Pharmacol Ther ; 99(3): 274-84, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26351780

ABSTRACT

To assess the safety, tolerability, and pharmacology of LY3023703, a microsomal prostaglandin E synthase 1 (mPGES1) inhibitor, a multiple ascending dose study was conducted. Forty-eight subjects received LY3023703, celecoxib (400 mg), or placebo once daily for 28 days. Compared with placebo, LY3023703 inhibited ex vivo lipopolysaccharide-stimulated prostaglandin E2 (PGE2 ) synthesis 91% and 97% on days 1 and 28, respectively, after 30-mg dosing, comparable to celecoxib's effect (82% inhibition compared to placebo). Unlike celecoxib, which also inhibited prostacyclin synthesis by 44%, LY3023703 demonstrated a maximal increase in prostacyclin synthesis of 115%. Transient elevations of serum aminotransferase were observed in one subject after 30-mg LY3023703 dosing (10× upper limit of normal (ULN)), and one subject after 15-mg dosing (about 1.5× ULN). Results from this study suggest that mPGES1 inhibits inducible PGE synthesis without suppressing prostacyclin generation and presents a novel target for inflammatory pain.


Subject(s)
Celecoxib/pharmacology , Celecoxib/pharmacokinetics , Intramolecular Oxidoreductases/antagonists & inhibitors , Adult , Celecoxib/administration & dosage , Celecoxib/blood , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Epoprostenol/biosynthesis , Female , Humans , Male , Middle Aged , Prostaglandin-E Synthases , Young Adult
20.
Clin Exp Pharmacol Physiol ; 43(1): 67-74, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26444418

ABSTRACT

The objective of this study was to determine the role of cyclooxygenase (COX)-1 or -2 in endothelium-dependent contraction under atherosclerotic conditions. Atherosclerosis was induced in apoE knockout (apoE(-/-)) mice and those with COX-1(-/-) (apoE(-/-)-COX-1(-/-)) by feeding with high fat and cholesterol food. Aortas (abdominal or the whole section) were isolated for functional and/or biochemical analyses. As in non-atherosclerotic conditions, the muscarinic receptor agonist acetylcholine (ACh) evoked an endothelium-dependent, COX-mediated contraction following NO synthase (NOS) inhibition in abdominal aortic rings from atherosclerotic apoE(-/-) mice. Interestingly, COX-1 inhibition not only abolished such a contraction in rings showing normal appearance, but also diminished that in rings with plaques. Accordingly, only a minor contraction (<30% that of apoE(-/-) counterparts) was evoked by ACh (following NOS inhibition) in abdominal aortic rings of atherosclerotic apoE(-/-)-COX-1(-/-) mice with plaques, and none was evoked in those showing normal appearance. Also, the contraction evoked by ACh in apoE(-/-)-COX-1(-/-) abdominal aortic rings with plaques was abolished by non-selective COX inhibition, thromboxane-prostanoid (TP) receptor antagonism, or endothelial denudation. Moreover, it was noted that ACh evoked a predominant production of the prostacyclin (PGI2, which mediates abdominal aortic contraction via TP receptors in mice) metabolite 6-keto-PGF1α, which was again sensitive to COX-1 inhibition or COX-1(-/-). Therefore, in atherosclerotic mouse abdominal aortas, COX-1 can still be the major isoform mediating endothelium-dependent contraction, which probably results largely from PGI2 synthesis as in non-atherosclerotic conditions. In contrast, COX-2 may have only a minor role in such response limited to areas of plaques under the same pathological condition.


Subject(s)
Aorta, Abdominal/physiopathology , Atherosclerosis/physiopathology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Endothelium, Vascular/metabolism , Vasoconstriction , Acetylcholine/pharmacology , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/biosynthesis , Endothelium, Vascular/drug effects , Epoprostenol/biosynthesis , Epoprostenol/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Muscarinic Agonists/pharmacology , Nitric Oxide/metabolism , Receptors, Muscarinic/metabolism , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...