Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.917
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Blood ; 125(4): 710-9, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25339356

ABSTRACT

The precise mechanism for reduced thrombosis in prekallikrein null mice (Klkb1(-/-)) is unknown. Klkb1(-/-) mice have delayed carotid artery occlusion times on the rose bengal and ferric chloride thrombosis models. Klkb1(-/-) plasmas have long-activated partial thromboplastin times and defective contact activation-induced thrombin generation that partially corrects upon prolonged incubation. However, in contact activation-induced pulmonary thromboembolism by collagen/epinephrine or long-chain polyphosphate, Klkb1(-/-) mice, unlike F12(-/-) mice, do not have survival advantage. Klkb1(-/-) mice have reduced plasma BK levels and renal B2R mRNA. They also have increased expression of the renal receptor Mas and plasma prostacyclin. Increased prostacyclin is associated with elevated aortic vasculoprotective transcription factors Sirt1 and KLF4. Treatment of Klkb1(-/-) mice with the Mas antagonist A-779, COX-2 inhibitor nimesulide, or Sirt1 inhibitor splitomicin lowers plasma prostacyclin and normalizes arterial thrombosis times. Treatment of normal mice with the Mas agonist AVE0991 reduces thrombosis. Klkb1(-/-) mice have reduced aortic tissue factor (TF) mRNA, antigen, and activity. In sum, Klkb1(-/-) mice have a novel mechanism for thrombosis protection in addition to reduced contact activation. This pathway arises when bradykinin delivery to vasculature is compromised and mediated by increased receptor Mas, prostacyclin, Sirt1, and KLF4, leading to reduced vascular TF.


Subject(s)
Carotid Artery Thrombosis , Epoprostenol , Kruppel-Like Transcription Factors , Prekallikrein , Proto-Oncogene Proteins , Receptors, G-Protein-Coupled , Thromboplastin , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Carotid Artery Thrombosis/chemically induced , Carotid Artery Thrombosis/genetics , Carotid Artery Thrombosis/metabolism , Carotid Artery Thrombosis/pathology , Epoprostenol/biosynthesis , Epoprostenol/genetics , Imidazoles/pharmacology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Naphthalenes/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Partial Thromboplastin Time , Peptide Fragments/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pyrones/pharmacology , RNA, Messenger , Receptor, Bradykinin B2/biosynthesis , Receptor, Bradykinin B2/genetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/biosynthesis , Sirtuin 1/genetics , Sulfonamides/pharmacology , Synaptotagmins/biosynthesis , Synaptotagmins/genetics , Thromboplastin/antagonists & inhibitors , Thromboplastin/biosynthesis , Thromboplastin/genetics
7.
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
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.
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
10.
FASEB J ; 29(11): 4568-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26183771

ABSTRACT

Eicosanoids are important vascular regulators, but the phospholipase A2 (PLA2) isoforms supporting their production within the cardiovascular system are not fully understood. To address this, we have studied platelets, endothelial cells, and leukocytes from 2 siblings with a homozygous loss-of-function mutation in group IVA cytosolic phospholipase A2 (cPLA2α). Chromatography/mass spectrometry was used to determine levels of a broad range of eicosanoids produced by isolated vascular cells, and in plasma and urine. Eicosanoid release data were paired with studies of cellular function. Absence of cPLA2α almost abolished eicosanoid synthesis in platelets (e.g., thromboxane A2, control 20.5 ± 1.4 ng/ml vs. patient 0.1 ng/ml) and leukocytes [e.g., prostaglandin E2 (PGE2), control 21.9 ± 7.4 ng/ml vs. patient 1.9 ng/ml], and this was associated with impaired platelet activation and enhanced inflammatory responses. cPLA2α-deficient endothelial cells showed reduced, but not absent, formation of prostaglandin I2 (prostacyclin; control 956 ± 422 pg/ml vs. patient 196 pg/ml) and were primed for inflammation. In the urine, prostaglandin metabolites were selectively influenced by cPLA2α deficiency. For example, prostacyclin metabolites were strongly reduced (18.4% of control) in patients lacking cPLA2α, whereas PGE2 metabolites (77.8% of control) were similar to healthy volunteer levels. These studies constitute a definitive account, demonstrating the fundamental role of cPLA2α to eicosanoid formation and cellular responses within the human circulation.


Subject(s)
Antigens, Human Platelet/genetics , Blood Platelets/enzymology , Dinoprostone/genetics , Endothelial Cells/enzymology , Epoprostenol/genetics , Leukocytes/enzymology , Mutation , Adult , Blood Platelets/pathology , Dinoprostone/biosynthesis , Endothelial Cells/pathology , Epoprostenol/biosynthesis , Female , Humans , Leukocytes/pathology , Male , Platelet Activation/genetics
11.
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
12.
Circulation ; 129(17): 1761-9, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24519928

ABSTRACT

BACKGROUND: Placebo-controlled trials of nonsteroidal anti-inflammatory drugs selective for inhibition of cyclooxygenase-2 (COX-2) reveal an emergent cardiovascular hazard in patients selected for low risk of heart disease. Postnatal global deletion of COX-2 accelerates atherogenesis in hyperlipidemic mice, a process delayed by selective enzyme deletion in macrophages. METHODS AND RESULTS: In the present study, selective depletion of COX-2 in vascular smooth muscle cells and endothelial cells depressed biosynthesis of prostaglandin I2 and prostaglandin E2, elevated blood pressure, and accelerated atherogenesis in Ldlr knockout mice. Deletion of COX-2 in vascular smooth muscle cells and endothelial cells coincided with an increase in COX-2 expression in lesional macrophages and increased biosynthesis of thromboxane. Increased accumulation of less organized intimal collagen, laminin, α-smooth muscle actin, and matrix-rich fibrosis was also apparent in lesions of the mutants. CONCLUSIONS: Although atherogenesis is accelerated in global COX-2 knockouts, consistent with evidence of risk transformation during chronic nonsteroidal anti-inflammatory drug administration, this masks the contrasting effects of enzyme depletion in macrophages versus vascular smooth muscle cells and endothelial cells. Targeting delivery of COX-2 inhibitors to macrophages may conserve their efficacy while limiting cardiovascular risk.


Subject(s)
Atherosclerosis/metabolism , Cyclooxygenase 2/genetics , Endothelium, Vascular/enzymology , Hyperlipidemias/metabolism , Muscle, Smooth, Vascular/enzymology , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/pathology , Atherosclerosis/epidemiology , Atherosclerosis/pathology , Blood Pressure/physiology , Cyclooxygenase 2/metabolism , Diet, Atherogenic , Dietary Fats/pharmacology , Dinoprostone/biosynthesis , Endothelium, Vascular/pathology , Epoprostenol/biosynthesis , Female , Hyperlipidemias/epidemiology , Hyperlipidemias/pathology , Macrophages/enzymology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Receptors, LDL/genetics , Risk Factors
13.
Behav Brain Funct ; 11: 6, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-25888780

ABSTRACT

BACKGROUND: Aluminum overload can cause severe brain injury and neurodegeneration. Previous studies suggest that prostacyclin synthase (PGIS) expression and prostacyclin receptor (IP) activation are beneficial for treatment of acute traumatic and ischemic brain injury. However, the potential value of PGIS/IP signaling pathway to chronic brain injury is still unclear. In this study, we investigated the change of PGIS/IP signaling pathway and the effect of beraprost sodium (BPS) on chronic brain injury in chronic aluminum-overload rats. METHODS: Rat model of chronic cerebral injury was established by chronic intragastric administration of aluminum gluconate(Al3+ 200 mg/kg per day,5d a week for 20 weeks). The methods of ELISA, qRT-PCR and Western blotting were used to detect the PGI2 level and the PGIS and IP mRNA and protein levels in hippocampi of chronic aluminum-overload rats, respectively. Rat hippocampal superoxide dismutase (SOD) activity and malondialdehyde (MDA) content also were measured. The effects of BPS (6, 12 and 24 µg⋅kg(-1)) on brain injury in chronic aluminum-overload rats were evaluated. RESULTS: Compared with the control group, PGIS mRNA expression, PGI2 level, and the IP mRNA and protein expressions significantly increased in hippocampi of chronic aluminum-overload rats. Administration of BPS significantly improved spatial learning and memory function impairment and hippocampal neuron injury induced by chronic aluminum overload in rats. Meanwhile, administration of BPS resulted in a decrease of PGI2 level and downregulation of PGIS and IP expressions in a dose-dependent manner. Aluminum overload also caused a decrease of SOD activity and an increase of MDA content. Administration of BPS significantly blunted the decrease of SOD activity and the increase of MDA content induced by aluminum overload in rats. CONCLUSIONS: BPS has a significant neuroprotective effect on chronic brain injury induced by aluminum overload in rats. Remodeling the balance of PGIS/IP signaling pathway and inhibition of oxidative stress involve in the neuroprotective mechanism of BPS in aluminum-overload rats. The PGIS/IP signaling pathway is a potential therapeutic strategy for chronic brain injury patients.


Subject(s)
Aluminum Compounds , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Damage, Chronic/chemically induced , Brain Damage, Chronic/prevention & control , Epoprostenol/analogs & derivatives , Neuroprotective Agents/therapeutic use , Aluminum/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/biosynthesis , Epoprostenol/therapeutic use , Gluconates , Hippocampus/metabolism , Hippocampus/pathology , Intramolecular Oxidoreductases/metabolism , Male , Malondialdehyde/metabolism , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/prevention & control , Memory Disorders/psychology , Nerve Tissue Proteins/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Epoprostenol/drug effects , Superoxide Dismutase/metabolism
14.
Proc Natl Acad Sci U S A ; 109(43): 17597-602, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23045674

ABSTRACT

Prostacyclin is an antithrombotic hormone produced by the endothelium, whose production is dependent on cyclooxygenase (COX) enzymes of which two isoforms exist. It is widely believed that COX-2 drives prostacyclin production and that this explains the cardiovascular toxicity associated with COX-2 inhibition, yet the evidence for this relies on indirect evidence from urinary metabolites. Here we have used a range of experimental approaches to explore which isoform drives the production of prostacyclin in vitro and in vivo. Our data show unequivocally that under physiological conditions it is COX-1 and not COX-2 that drives prostacyclin production in the cardiovascular system, and that urinary metabolites do not reflect prostacyclin production in the systemic circulation. With the idea that COX-2 in endothelium drives prostacyclin production in healthy individuals removed, we must seek new answers to why COX-2 inhibitors increase the risk of cardiovascular events to move forward with drug discovery and to enable more informed prescribing advice.


Subject(s)
Cardiovascular System/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Epoprostenol/biosynthesis , Animals , Blotting, Western , Cells, Cultured , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Pharmacol Rev ; 64(3): 540-82, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22679221

ABSTRACT

Prostacyclin (PGI(2)) is a member of the prostanoid group of eicosanoids that regulate homeostasis, hemostasis, smooth muscle function and inflammation. Prostanoids are derived from arachidonic acid by the sequential actions of phospholipase A(2), cyclooxygenase (COX), and specific prostaglandin (PG) synthases. There are two major COX enzymes, COX1 and COX2, that differ in structure, tissue distribution, subcellular localization, and function. COX1 is largely constitutively expressed, whereas COX2 is induced at sites of inflammation and vascular injury. PGI(2) is produced by endothelial cells and influences many cardiovascular processes. PGI(2) acts mainly on the prostacyclin (IP) receptor, but because of receptor homology, PGI(2) analogs such as iloprost may act on other prostanoid receptors with variable affinities. PGI(2)/IP interaction stimulates G protein-coupled increase in cAMP and protein kinase A, resulting in decreased [Ca(2+)](i), and could also cause inhibition of Rho kinase, leading to vascular smooth muscle relaxation. In addition, PGI(2) intracrine signaling may target nuclear peroxisome proliferator-activated receptors and regulate gene transcription. PGI(2) counteracts the vasoconstrictor and platelet aggregation effects of thromboxane A(2) (TXA(2)), and both prostanoids create an important balance in cardiovascular homeostasis. The PGI(2)/TXA(2) balance is particularly critical in the regulation of maternal and fetal vascular function during pregnancy and in the newborn. A decrease in PGI(2)/TXA(2) ratio in the maternal, fetal, and neonatal circulation may contribute to preeclampsia, intrauterine growth restriction, and persistent pulmonary hypertension of the newborn (PPHN), respectively. On the other hand, increased PGI(2) activity may contribute to patent ductus arteriosus (PDA) and intraventricular hemorrhage in premature newborns. These observations have raised interest in the use of COX inhibitors and PGI(2) analogs in the management of pregnancy-associated and neonatal vascular disorders. The use of aspirin to decrease TXA(2) synthesis has shown little benefit in preeclampsia, whereas indomethacin and ibuprofen are used effectively to close PDA in the premature newborn. PGI(2) analogs have been used effectively in primary pulmonary hypertension in adults and have shown promise in PPHN. Careful examination of PGI(2) metabolism and the complex interplay with other prostanoids will help design specific modulators of the PGI(2)-dependent pathways for the management of pregnancy-related and neonatal vascular disorders.


Subject(s)
Adaptation, Physiological , Epoprostenol/biosynthesis , Pregnancy Complications, Cardiovascular/metabolism , Receptors, Epoprostenol/metabolism , Signal Transduction , Vascular Diseases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Female , Humans , Infant, Newborn , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Ligands , Pregnancy , Pregnancy Complications, Cardiovascular/enzymology , Pregnancy Complications, Cardiovascular/prevention & control , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Epoprostenol/agonists , Receptors, Epoprostenol/antagonists & inhibitors , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxane-A Synthase/metabolism , Vascular Diseases/enzymology , Vascular Diseases/prevention & control , Vasodilation/drug effects
16.
Biochem Biophys Res Commun ; 448(1): 83-8, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24780398

ABSTRACT

We show that prostacyclin production is increased in bone and osteocytes from sclerostin (Sost) knockout mice which have greatly increased bone mass. The addition of prostacyclin or a prostacyclin analog to bone forming osteoblasts enhances differentiation and matrix mineralization of osteoblasts. The increase in prostacyclin synthesis is linked to increases in ß-catenin concentrations and activity as shown by enhanced binding of lymphoid enhancer factor, Lef1, to promoter elements within the prostacyclin synthase promoter. Blockade of Wnt signaling reduces prostacyclin production in osteocytes. Increased prostacyclin production by osteocytes from sclerostin deficient mice could potentially contribute to the increased bone formation seen in this condition.


Subject(s)
Epoprostenol/biosynthesis , Glycoproteins/deficiency , Osteocytes/metabolism , Wnt Signaling Pathway/genetics , Adaptor Proteins, Signal Transducing , Animals , Bone and Bones/metabolism , Intercellular Signaling Peptides and Proteins , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Mice , Mice, Knockout , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
17.
FASEB J ; 27(2): 656-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23109673

ABSTRACT

Accumulating evidence suggests that sphingosine kinase 1 (SphK1) plays a key role in carcinogenesis by regulating cyclooxygenase-2 (COX-2) expression. Recent clinical studies have revealed that COX-2 inhibitors cause adverse cardiovascular side effects, likely due to inhibition of prostacyclin (PGI(2)). In this work, we investigated the roles of SphK1 inhibition on blood pressure (BP). The results show that lack of SphK1 expression did not exacerbate angiotensin II (Ang II)-induced acute hypertension, whereas celecoxib, a COX-2 inhibitor, augmented and sustained higher BP in mice. Interestingly, SphK1-knockout mice inhibited prostaglandin E(2) (PGE(2)) but not PGI(2) production in response to Ang II, whereas celecoxib blocked both PGE(2) and PGI(2) production. Mechanistically, SphK1 down-regulation by siRNA in human umbilical vein endothelial cells decreased cytokine-induced PGE(2) production primarily through inhibition of microsomal PGE synthase-1 (mPGES-1), not COX-2. SphK1 down-regulation also decreased MKK6 expression, which phosphorylates and activates P38 MAPK, which, in turn, regulates early growth response-1 (Egr-1), a transcription factor of mPGES-1. Together, these data indicate that SphK1 regulates PGE(2) production by mPGES-1 expression via the p38 MAPK pathway, independent of COX-2 signaling, in endothelial cells, suggesting that SphK1 inhibition may be a promising strategy for cancer chemoprevention with lack of the adverse cardiovascular side effects associated with coxibs.


Subject(s)
Blood Pressure/physiology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Base Sequence , Celecoxib , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/biosynthesis , Epoprostenol/biosynthesis , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Hypertension/drug therapy , Hypertension/physiopathology , Intramolecular Oxidoreductases/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mitochondrial Proteins , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , Prostaglandin-E Synthases , Pyrazoles/pharmacology , RNA, Small Interfering/genetics , Sulfonamides/pharmacology
18.
Am J Physiol Renal Physiol ; 305(9): F1315-22, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23986518

ABSTRACT

This study was to determine whether prostacyclin [prostaglandin I2 (PGI2)] evokes mouse renal vasoconstriction and, if so, the underlying mechanism(s) and how its synthesis via cyclooxygenase-1 (COX-1) influences local vasomotor reaction. Experiments were performed on vessels from C57BL/6 mice and/or those with COX-1 deficiency (COX-1(-/-)). Results showed that in renal arteries PGI2 evoked contraction more potently than in carotid arteries, where COX-1 is suggested to mediate prominent endothelium-dependent contraction. A similar result was observed with the thromboxane-prostanoid (TP) receptor agonist U46619. However, in renal arteries TP receptor antagonism, which inhibited the contraction, did not result in any relaxation in response to PGI2. Moreover, we noted that the endothelial muscarinic receptor agonist ACh evoked an increase in the production of the PGI2 metabolite 6-keto-PGF1α, which was prevented by endothelial denudation or COX-1(-/-). Interestingly, COX-1(-/-) was further found to abolish a force development that was sensitive to TP receptor antagonism and result in enhanced relaxation evoked by ACh following NO synthase inhibition. Also, in renal arteries the COX substrate arachidonic acid evoked a vasoconstrictor response, which was again abolished by COX-1(-/-). Meanwhile, nonselective COX inhibition did not show any effect in vessels from COX-1(-/-) mice. Thus, in mouse renal arteries, high expression of TP receptors together with little functional involvement from the vasodilator PGI2 receptors results in a potent vasoconstrictor effect evoked by PGI2. Also, our data imply that endogenous COX-1-mediated PGI2 synthesis leads to vasoconstrictor activity and this could be an integral part of endothelium-derived mechanisms in regulating local renal vascular function.


Subject(s)
Cyclooxygenase 1/physiology , Epoprostenol/biosynthesis , Receptors, Epoprostenol/metabolism , Renal Artery/enzymology , Vasoconstriction , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Cyclooxygenase 1/genetics , Cytochrome P-450 Enzyme System/metabolism , Female , Intramolecular Oxidoreductases/metabolism , Male , Mice , Mice, Inbred C57BL , Vasomotor System/physiology
19.
Circ J ; 77(11): 2823-30, 2013.
Article in English | MEDLINE | ID: mdl-23883876

ABSTRACT

BACKGROUND: Linoleic acid (LA) promotes monocyte chemotaxis and cell adhesion molecules such as MCP-1 and VCAM-1, which contribute to atherosclerogenesis. These molecules are restrained by endothelium-derived relaxing factors (EDRFs), such as nitric oxide (NO) and prostaglandin I2 (PGI2). Hence, the expressions of MCP-1 and VCAM-1 upregulated by LA may be partly attributable to decreased EDRF production. However, effect of LA on EDRF production remains controversial. METHODS AND RESULTS: The present study aimed to examine the effects of LA and other free fatty acids on EDRF production and the endothelial Ca(2+) responses that mediate EDRF production, using primary cultured porcine aortic endothelial cells (PAECs). LA at 0.1-5 µmol/L attenuated bradykinin (BK)-induced NO and PGI2 production while suppressing the BK-induced Ca(2+) response dose-dependently. The inhibitory effect of LA on the Ca(2+) response was eliminated by adenylate cyclase inhibitor SQ22536, boosted by cAMP-hydrolyzing phosphodiesterase (PDE) inhibitor, rolipram, and mimicked by plasma membrane permeable 8-bromo-cAMP. Moreover, LA was confirmed to dose-dependently increase intracellular cAMP levels and selectively inhibit cAMP-hydrolyzing PDE activity in vitro. In contrast, none of palmitic, stearic, or oleic acid affected BK-induced EDRF production or Ca(2+) responses, or induced intracellular cAMP accumulation. CONCLUSIONS: LA induced intracellular cAMP accumulation by inhibiting cAMP-hydrolyzing PDE activity, thus resulting in attenuation of Ca(2+) responses and EDRF production in PAECs.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/biosynthesis , Calcium Signaling/drug effects , Cyclic AMP/metabolism , Endothelial Cells/metabolism , Endothelium-Dependent Relaxing Factors/metabolism , Linoleic Acid/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Calcium Signaling/physiology , Cells, Cultured , Chemokine CCL2/biosynthesis , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Enzyme Inhibitors/pharmacology , Epoprostenol/biosynthesis , Nitric Oxide/biosynthesis , Swine , Vascular Cell Adhesion Molecule-1/biosynthesis
20.
J Immunol ; 186(7): 4361-6, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21357261

ABSTRACT

In addition to the well-described role of platelets in thrombosis, a growing body of evidence implicates platelets in diverse inflammatory responses. We recently showed platelets can contribute to the pathophysiology of inflammatory arthritis via IL-1- containing microparticles. In this study, we demonstrate that platelets, and not platelet microparticles, actively contribute to synovitis via production of proinflammatory prostacyclin in an autoimmune arthritis model. Using both genetic and pharmacologic approaches, we establish that paracrine production of prostacyclin proceeds in the absence of cyclooxygenase-2. Furthermore, we also demonstrate that prostacyclin generation can arise via transcellular collaboration between platelets and fibroblast-like synoviocytes. In addition to shedding light on an unappreciated pathway of lipid synthesis in arthritis, we further delineate a novel effector activity by which platelets can contribute to inflammatory disease.


Subject(s)
Blood Platelets/enzymology , Blood Platelets/pathology , Cell-Derived Microparticles/enzymology , Cyclooxygenase 1/physiology , Epoprostenol/biosynthesis , Synovitis/blood , Animals , Arthritis, Experimental/blood , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Blood Platelets/metabolism , Bone Marrow/enzymology , Bone Marrow/metabolism , Bone Marrow/pathology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Epoprostenol/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Synovitis/enzymology , Synovitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL