Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 793
Filter
1.
Adv Pharmacol ; 97: 133-165, 2023.
Article in English | MEDLINE | ID: mdl-37236757

ABSTRACT

Cyclooxygenase (COX) isozymes, i.e., COX-1 and COX-2, are encoded by separate genes and are involved in the generation of the same products, prostaglandin (PG)G2 and PGH2 from arachidonic acid (AA) by the COX and peroxidase activities of the enzymes, respectively. PGH2 is then transformed into prostanoids in a tissue-dependent fashion due to the different expression of downstream synthases. Platelets present almost exclusively COX-1, which generates large amounts of thromboxane (TX)A2, a proaggregatory and vasoconstrictor mediator. This prostanoid plays a central role in atherothrombosis, as shown by the benefit of the antiplatelet agent low-dose aspirin, a preferential inhibitor of platelet COX-1. Recent findings have shown the relevant role played by platelets and TXA2 in developing chronic inflammation associated with several diseases, including tissue fibrosis and cancer. COX-2 is induced in response to inflammatory and mitogenic stimuli to generate PGE2 and PGI2 (prostacyclin), in inflammatory cells. However, PGI2 is constitutively expressed in vascular cells in vivo and plays a crucial role in protecting the cardiovascular systems due to its antiplatelet and vasodilator effects. Here, platelets' role in regulating COX-2 expression in cells of the inflammatory microenvironment is described. Thus, the selective inhibition of platelet COX-1-dependent TXA2 by low-dose aspirin prevents COX-2 induction in stromal cells leading to antifibrotic and antitumor effects. The biosynthesis and functions of other prostanoids, such as PGD2, and isoprostanes, are reported. In addition to aspirin, which inhibits platelet COX-1 activity, possible strategies to affect platelet functions by influencing platelet prostanoid receptors or synthases are discussed.


Subject(s)
Aspirin , Prostaglandins , Humans , Cyclooxygenase 2 , Aspirin/pharmacology , Aspirin/therapeutic use , Thromboxane A2/physiology , Prostaglandin H2
3.
J Biol Regul Homeost Agents ; 34(5): 1623-1627, 2020.
Article in English | MEDLINE | ID: mdl-32744052

ABSTRACT

IL-1 induces a significant number of metabolic and hematological changes. In experimental animals, IL-1 treatments cause hypotension due to rapid reduction of systemic blood pressure, reduced vascular resistance, increased heart rate and leukocyte aggregations. IL-1 causes endothelial dysfunction, the triggering factor of which may be of a different nature including pathogen infection. This dysfunction, which includes macrophage intervention and increased protein permeability, can be mediated by several factors including cytokines and arachidonic acid products. These effects are caused by the induction of IL-1 in various pathologies, including those caused by pathogenic viral infections, including SARS-CoV-2 which provokes COVID-19. Activation of macrophages by coronavirus-19 leads to the release of pro-inflammatory cytokines, metalloproteinases and other proteolytic enzymes that can cause thrombi formation and severe respiratory dysfunction. Patients with COVID-19, seriously ill and hospitalized in intensive care, present systemic inflammation, intravascular coagulopathy with high risk of thrombotic complications, and venous thromboembolism, effects mostly mediated by IL-1. In these patients the lungs are the most critical target organ as it can present an increase in the degradation products of fibrin, fibrinogen and D-dimer, with organ lesions and respiratory failure. It is well known that IL-1 induces itself and another very important pro-inflammatory cytokine, TNF, which also participates in hemodynamic states, including shock syndrome in COVID-19. Both IL-1 and TNF cause pulmonary edema, thrombosis and bleeding. In addition to hypotension and resistance of systemic blood pressure, IL-1 causes leukopenia and thrombocytopenia. The formation of thrombi is the main complication of the circulatory system and functionality of the organ, and represents an important cause of morbidity and mortality. IL-1 causes platelet vascular thrombogenicity also on non-endothelial cells by stimulating the formation of thromboxane A2 which is released into the inflamed environment. IL-1 is the most important immune molecule in inducing fever, since it is involved in the metabolism of arachidonic acid which increases from vascular endothelial organs of the hypothalamus. The pathogenesis of thrombosis, vascular inflammation and angigenesis involves the mediation of the activation of the prostanoid thromboxane A2 receptor. In 1986, in an interesting article (Conti P, Reale M, Fiore S, Cancelli A, Angeletti PU, Dinarello CA. In vitro enhanced thromboxane B2 release by polymorphonuclear leukocytes and macrophages after treatment with human recombinant interleukin 1. Prostaglandins. 1986 Jul;32(1):111-5), we reported for the first time that IL-1 induces thromboxane B2 (TxB2) releases in activated neutrophils and macrophages. An increase in thromboxane can induce leukocyte aggregation and systemic inflammation, which would account for the dramatic thrombi formation and organ dysfunction. Hence, IL-1 stimulates endothelial cell-leukocyte adhesion, and TxB2 production. All these events are supported by the large increase in neutrophils that adhere to the lung and the decrease in lymphocytes. Therefore, ecosanoids such as TxA2 (detected as TxB2) have a powerful action on vascular inflammation and platelet aggregation, mediating the formation of thrombi. The thrombogenesis that occurs in COVID-19 includes platelet and cell aggregation with clotting abnormalities, and anti-clotting inhibitor agents are used in the prevention and therapy of thrombotic diseases. Prevention of or induction of TxA2 avoids thrombi formation induced by IL-1. However, in some serious vascular events where TxA2 increases significantly, it is difficult to inhibit, therefore, it would be much better to prevent its induction and generation by blocking its inductors including IL-1. The inhibition or lack of formation of IL-1 avoids all the above pathological events which can lead to death of the patient. The treatment of innate immune cells producing IL-1 with IL-1 receptor antagonist (IL-1Ra) can avoid hemodynamic changes, septic shock and organ inflammation by carrying out a new therapeutic efficacy on COVID-19 induced by SARS-CoV-2.


Subject(s)
Coronavirus Infections/pathology , Inflammation/virology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1/physiology , Pneumonia, Viral/pathology , Thrombosis/virology , Thromboxane A2/physiology , Animals , Betacoronavirus , COVID-19 , Humans , Pandemics , Receptors, Interleukin-1 , SARS-CoV-2
4.
J Agric Food Chem ; 66(23): 5853-5859, 2018 Jun 13.
Article in English | MEDLINE | ID: mdl-29771124

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is now a public health issue worldwide, but no drug has yet received approval. Genistein, an isoflavonoid derived from soybean, ameliorates high-fat-diet-induced NAFLD in mice, but the molecular underpinnings remain largely elusive. Arachidonic acid (AA) is a major ingredient of animal fats, and the AA cascade has been implicated in chronic inflammation. In this study, we investigated whether genistein was against NAFLD by targeting the AA cascade. Using a mouse model, we showed that genistein supplementation improved high-fat-diet-induced NAFLD by normalizing hepatomegaly, liver steatosis, aminotransferase abnormalities, and glucose tolerance. The thromboxane A2 (TXA2) pathway was aberrantly active in NAFLD, evidenced by an elevation of circulating TXA2 and hepatic thromboxane A2 receptor expression. Mechanistically, we found that genistein directly targeted cyclooxygenase-1 activity as well as its downstream TXA2 biosynthesis, while the TXA2 pathway might mediate NAFLD progression by impairing insulin sensitivity. Taken together, our study revealed a crucial pathophysiological role of the TXA2 pathway in NAFLD and provided an explanation as to how genistein was against NAFLD progression.


Subject(s)
Genistein/administration & dosage , Non-alcoholic Fatty Liver Disease/drug therapy , Thromboxane A2/physiology , Animals , Aspirin/administration & dosage , Cyclooxygenase Inhibitors , Diet, High-Fat , Disease Models, Animal , Disease Progression , Hep G2 Cells , Humans , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/analysis , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Receptors, Thromboxane A2, Prostaglandin H2/physiology , Thromboxane A2/blood
5.
J Cereb Blood Flow Metab ; 37(3): 1060-1068, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27178425

ABSTRACT

Cortical spreading depression (CSD) is considered a significant phenomenon for human neurological conditions and one of its key signatures is the development of persistent cortical oligemia. The factors underlying this reduction in cerebral blood flow (CBF) remain incompletely understood but may involve locally elaborated vasoconstricting eicosanoids. We employed laser Doppler flowmetry in urethane-anesthetized rats, together with a local pharmacological blockade approach, to test the relative contribution of cyclooxygenase (COX)-derived prostanoids to the oligemic response following CSD. Administration of the non-selective COX inhibitor naproxen completely inhibited the oligemic response. Selective inhibition of COX-1 with SC-560 preferentially reduced the early reduction in CBF while selective COX-2 inhibition with NS-398 affected only the later response. Blocking the action of thromboxane A2 (TXA2), using the selective thromboxane synthase inhibitor ozagrel, reduced only the initial CBF decrease, while inhibition of prostaglandin F2alpha action, using the selective FP receptor antagonist AL-8810, blocked the later phase of the oligemia. Our results suggest that the long-lasting oligemia following CSD consists of at least two distinct temporal phases, mediated by preferential actions of COX-1- and COX-2-derived prostanoids: an initial phase mediated by COX-1 that involves TXA2 followed by a later phase, mediated by COX-2 and PGF2alpha.


Subject(s)
Cerebrovascular Circulation/drug effects , Cortical Spreading Depression/drug effects , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Prostaglandins/physiology , Animals , Cyclooxygenase Inhibitors/pharmacology , Dinoprost/physiology , Laser-Doppler Flowmetry , Rats , Thromboxane A2/physiology , Time Factors
6.
Naunyn Schmiedebergs Arch Pharmacol ; 389(1): 1-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26531833

ABSTRACT

Remote hind limb preconditioning (RIPC) is a protective strategy in which short episodes of ischemia and reperfusion in a remote organ (hind limb) protects the target organ (heart) against sustained ischemic reperfusion injury. The present study was designed to investigate the possible role of thromboxane A2 in RIPC-induced cardioprotection in rats. Remote hind limb preconditioning was performed by four episodes of 5 min of inflation and 5 min of deflation of pressure cuff. Occlusion of the hind limb with blood pressure cuff is most feasible, non-invasive, clinically relevant, and safe method for inducing RIPC. Isolated rat hearts were perfused on Langendorff apparatus and were subjected to global ischemia for 30 min followed by 120-min reperfusion. The levels of lactate dehydrogenase (LDH) and creatine kinase (CK) were measured in coronary effluent to assess the degree of myocardial injury. The extent of myocardial infarct size along with the functional parameters including left ventricular developed pressure (LVDP), dp/dtmax, and dp/dtmin were also measured. Ozagrel (thromboxane synthase inhibitor) and seratrodast (thromboxane A2 receptor antagonist) were employed as pharmacological modulators of thromboxane A2. Remote hind limb preconditioning significantly attenuated ischemia/reperfusion-induced myocardial injury and produced cardioprotective effects. However, administration of ozagrel and seratrodast completely abolished the cardioprotective effects of RIPC suggesting the key role of thromboxane A2 in RIPC-induced cardioprotection. It may be concluded that brief episodes of preconditioning ischemia and reperfusion activates the thromboxane synthase enzyme that produces thromboxane A2, which may elicit cardioprotection either involving humoral or neurogenic pathway.


Subject(s)
Hindlimb/blood supply , Ischemic Preconditioning , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Thromboxane A2/physiology , Animals , Benzoquinones/pharmacology , Creatine Kinase/metabolism , Female , Heart , Heptanoic Acids/pharmacology , L-Lactate Dehydrogenase/metabolism , Male , Methacrylates/pharmacology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Rats, Wistar , Thromboxane-A Synthase/antagonists & inhibitors
7.
Zhongguo Dang Dai Er Ke Za Zhi ; 17(9): 956-60, 2015 Sep.
Article in Chinese | MEDLINE | ID: mdl-26412178

ABSTRACT

OBJECTIVE: To investigate the effects of high-volume hemofiltration (HVHF) on hemodynamics, vasoactive factors, and vascular endothelial permeability in children with septic shock by a comparative analysis. METHODS: Thirty-six children who were diagnosed with septic shock between January 2013 and September 2014 were randomly divided into control and observation groups (n=18 each). Children in the control group were treated with the standard-volume hemofiltration (SVHF), while children in the observation group were treated with HVHF. The hemodynamic indices and levels of vasoactive factors including 6-keto-prostaglandin F1α (6-keto-PGF1α), thromboxane B2 (TXB2), soluble E-selectin (sE-selectin), and endothelium-derived relaxing factor (EDRF) were determined before and after treatment. In addition, the effects of ultrafiltrate on endothelial cell permeability were assessed. RESULTS: Compared with the control group, the observation group had significantly higher mean arterial pressure, significantly higher blood oxygen saturation, and a significantly lower heart rate after treatment (P<0.05). The levels of TXB2 and sE-selectin were significantly lower in the observation group than in the control group (P<0.05), while the levels of 6-keto-PGF1α and EDRF were significantly higher in the observation group than in the control group (P<0.05). Compared with the control group, the ultrafiltrate significantly attenuated the transepithelial electrical resistance in the observation group (P<0.05). CONCLUSIONS: Compared with SVHF, HVHF is a more effective approach for improving the hemodynamics and levels of vasoactive factors and reducing the vascular endothelial permeability in children with septic shock.


Subject(s)
Capillary Permeability , Hemodynamics , Hemofiltration , Shock, Septic/physiopathology , Child , Child, Preschool , Epoprostenol/physiology , Female , Humans , Infant , Male , Thromboxane A2/physiology
9.
Blood ; 124(10): 1610-21, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25030064

ABSTRACT

Cyclooxygenases (COXs) and their prostanoid products play important roles in a diverse range of physiological processes, including in the immune system. Here, we provide evidence that COX-1 is an essential regulator in early stages of B-cell development. COX-1-deficient mice displayed systematic reduction in total B cells, which was attributed to the arrest of early B-cell development from pro-B to pre-B stage. We further demonstrated that this defect was mediated through downregulation of the Janus kinase/signal transducer and activator of transcription 5 (JAK/STAT5) signaling and its target genes, including Pax5, in COX-1(-/-) mice. Mechanistic studies revealed that COX-1-derived thromboxane A2 (TxA2) could regulate JAK3/STAT5 signaling through the cyclic adenosine monophosphate-protein kinase A pathway, via binding with its receptor thromboxane A2 receptor (TP). Administration of the TP agonist could rescue the defective B-cell development and JAK/STAT5 signaling activity in COX-1-deficient mice. Moreover, administration of low-dose aspirin caused a significant reduction in total B cells in peripheral blood of healthy human volunteers, coincidentally with reduced TxA2 production and downregulation of JAK/STAT5 signaling. Taken together, our results demonstrate that COX-1-derived TxA2 plays a critical role in the stage transition of early B-cell development through regulation of JAK/STAT5 signaling and indicate a potential immune-suppressive effect of low-dose aspirin in humans.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation , Cyclooxygenase 1/metabolism , Membrane Proteins/metabolism , Thromboxane A2/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Cyclooxygenase 1/genetics , Humans , Janus Kinases/metabolism , Leukopoiesis/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics , Thromboxane A2/metabolism
10.
Biochem Biophys Res Commun ; 441(2): 393-8, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24161392

ABSTRACT

The prothrombotic mediator thromboxane A2 is derived from arachidonic acid metabolism through the cyclooxygenase and thromboxane synthase pathways, and transduces its effect through the thromboxane prostanoid (TP) receptor. The aim of this study was to determine the effect of the TP receptor antagonist and thromboxane synthase inhibitor EV-077 on inflammatory markers in human umbilical vein endothelial cells and on human coronary artery smooth muscle cell proliferation. To this end, mRNA levels of different proinflammatory mediators were studied by real time quantitative PCR, supernatants were analyzed by enzyme immune assay, and cell proliferation was assessed using WST-1. EV-077 significantly decreased mRNA levels of ICAM-1 and PTX3 after TNFα incubation, whereas concentrations of 6-keto PGF1α in supernatants of endothelial cells incubated with TNFα were significantly increased after EV-077 treatment. Although U46619 did not alter coronary artery smooth muscle cell proliferation, this thromboxane mimetic enhanced the proliferation induced by serum, insulin and growth factors, which was significantly inhibited by EV-077. In conclusion, EV-077 inhibited TNFα-induced endothelial inflammation and reduced the enhancement of smooth muscle cell proliferation induced by a thromboxane mimetic, supporting that the thromboxane pathway may be associated with early atherosclerosis in terms of endothelial dysfunction and vascular hypertrophy.


Subject(s)
Endothelium, Vascular/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, Thromboxane/antagonists & inhibitors , Thromboxane-A Synthase/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , C-Reactive Protein/antagonists & inhibitors , C-Reactive Protein/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Gene Expression/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Prostaglandins F/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Serum Amyloid P-Component/antagonists & inhibitors , Serum Amyloid P-Component/biosynthesis , Thromboxane A2/physiology , Tumor Necrosis Factor-alpha/pharmacology
11.
Bull Acad Natl Med ; 197(2): 361-73, 2013 Feb.
Article in French | MEDLINE | ID: mdl-24919366

ABSTRACT

The main role of blood platelets is to ensure vascular integrity and hemostasis in case of vascular damage. The platelet functions involved in these physiological processes are also at work in arterial thrombosis, which is a dramatic complication of atherosclerosis that may lead to vascular occlusion. These functions of platelets include their ability to adhere to the injured vessel wall, to be activated by contact with various substrates and soluble activators, and to form aggregates stabilized by a fibrin network. Platelets are also involved in metastasis, various inflammatory processes, innate and adaptive immune defenses, and embryonic development. These roles are supported by multiple molecular mechanisms, some of which are common to several functions while others are distinct. Defects in one or other of these mechanisms do not necessarily disrupt all platelet functions.


Subject(s)
Platelet Activation/physiology , Humans , Platelet Adhesiveness/physiology , Receptors, Purinergic P2Y1/physiology , Receptors, Purinergic P2Y12/physiology , Thrombin/physiology , Thromboxane A2/physiology
12.
Pharmacol Res ; 66(5): 392-400, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22898325

ABSTRACT

Diabetes is associated with increased prevalence of hypertension, cardiovascular and renal disease. Atrial natriuretic peptide (ANP) plays an important role in cardiovascular pathophysiology and is claimed to have cardioprotective and renoprotective effect in diabetic patients. The working hypothesis was that alloxan-induced diabetes might modify the vascular effects of ANP in isolated rabbit renal arteries and the mechanisms involved in such actions. Plasma ANP levels were higher in diabetic rabbits than in control rabbits. ANP (10(-12)-10(-7)M) induced a relaxation of precontracted renal arteries, which was lower in diabetic than in control rabbits. In arteries from both groups of animals, endothelium removal decreased the ANP-induced relaxation but inhibition of NO-synthesis did not modify ANP-induced relaxations. In KCl-depolarised arteries, relaxation to ANP was almost abolished both in control and diabetic rabbits. Tetraethylammonium (TEA) partly inhibited the relaxation to ANP in control rabbits but did not modify it in diabetic rabbits. Glibenclamide and 4-aminopyridine inhibited the relaxation to ANP, and these inhibitions were lower in diabetic than in control rabbits. Indomethacin potentiated the relaxation to ANP, more in control than in diabetic rabbits. In the presence of ANP the renal artery released thromboxane A(2) and prostacyclin, and the release of prostacyclin resulted decreased in diabetic rabbits. The present results suggest that diabetes produces hyporeactivity of the rabbit renal artery to ANP by mechanisms that at least include the reduced modulation by prostacyclin and a lower participation of ATP-sensitive K(+) channel (K(ATP)), voltage-sensitive K(+) channels (K(V)) and TEA-sensitive K(+) channels (K(Ca)).


Subject(s)
Atrial Natriuretic Factor/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Epoprostenol/physiology , Renal Artery/drug effects , Vasodilation/drug effects , Animals , Atrial Natriuretic Factor/blood , Endothelium, Vascular/physiology , In Vitro Techniques , Male , Potassium Channels/physiology , Rabbits , Renal Artery/physiology , Tetraethylammonium/pharmacology , Thromboxane A2/physiology , Vasodilation/physiology
13.
Br J Anaesth ; 106(1): 31-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20935003

ABSTRACT

BACKGROUND: The roles of arachidonic acid (AA) metabolites in hypoxia-induced pulmonary vasoconstriction (HPV), a critical physiological mechanism that prevents ventilation/perfusion mismatch, are still incompletely understood. METHODS: Pulmonary arterial pressure was measured in ventilated/perfused rat lungs. Isometric tones of rat intralobar pulmonary arteries were also measured, using a myograph. RESULTS: Hypoxia (Po2, 3%)-induced pulmonary arterial pressure increases (ΔPAP(hypox)) were stable with blood-mixed perfusate, but decayed spontaneously. ΔPAP(hypox) was inhibited by 29%, 16%, and 28% by the thromboxane A2 (TXA2) antagonist SQ-29548, the 5-lipoxygenase inhibitor, MK886, and the leukotriene D4 antagonist, LY-171883, respectively. The prostacyclin synthase inhibitor tranylcypromine augmented ΔPAP(hypox) by 5%, whereas inhibition of cytochrome P450 did not affect ΔPAP(hypox). Consistently, the TXA2 analogue U46619 increased ΔPAP(hypox) whereas prostacyclin abolished ΔPAP(hypox). However, leukotriene D4 had no direct effect on ΔPAP(hypox). In the isolated pulmonary arteries, pretreatment with U46619 was essential to demonstrate hypoxia-induced contraction. CONCLUSIONS: The above results suggest that TXA2 and cysteinyl leukotrienes, other than leukotriene D4, are endogenous factors that facilitate HPV in rats. The indispensable role of TXA2-induced pretone in the HPV of isolated pulmonary arteries indicates that the signal from thromboxane receptors might be a critical component of oxygen sensation mechanisms.


Subject(s)
Arachidonic Acid/metabolism , Hypoxia/physiopathology , Pulmonary Artery/physiopathology , Vasoconstriction/physiology , Animals , Cyclooxygenase Inhibitors/pharmacology , Hypoxia/metabolism , Leukotrienes/physiology , Male , Pulmonary Artery/drug effects , Rats , Thromboxane A2/physiology , Vasoconstriction/drug effects
14.
Gut ; 59(6): 827-36, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20551467

ABSTRACT

OBJECTIVE: This study aimed to investigate the effect of amiloride on portal hypertension. Amiloride is known to inhibit Na(+)/H(+) exchangers on activated hepatic stellate cells. METHODS: Liver cirrhosis in rats was induced by bile duct ligation (BDL) or thioacetamide (TAA) administration. The effects of zymosan for Kupffer cell (KC) activation or a thromboxane (TX) analogue (U46619) were tested in isolated perfused livers of cirrhotic rats and in vivo. Downstream mechanisms were investigated using Rho kinase inhibitor (Y-27632) or amiloride. Acute and chronic effects of amiloride and canrenoate on portal pressure were compared in perfused livers and in vivo. TXB(2) efflux was measured by ELISA. The phosphorylation state of moesin (p-moesin) as an indicator of Rho kinase activity and expression of the thromboxane synthase were assessed by western blot analyses. The activity of hepatic stellate cells was analysed by western blot and staining for alpha-smooth muscle actin (alpha-SMA). RESULTS: In BDL rats, KC activation via zymosan increased portal pressure. This was attenuated by the Rho kinase inhibitor Y-27632. Increased thromboxane efflux following zymosan infusion remained unaltered by Y-27632. The infusion of amiloride attenuated zymosan- and U46619-induced increases in portal perfusion pressure. In vivo, direct administration of amiloride, but not of canrenoate, lowered portal pressure. In TAA and BDL rats, treatment with amiloride for 3 days reduced basal portal pressure and KC-induced increases in portal pressure whereas canrenoate had no effect. In livers of amiloride-treated animals, the phosphorylation state of moesin and the number of alpha-SMA positive cells were reduced. CONCLUSIONS: Amiloride lowers portal pressure in rat liver cirrhosis by inhibition of intrahepatic vasocontraction. Therefore, patients with cirrhosis and portal hypertension may benefit from amiloride therapy.


Subject(s)
Amiloride/therapeutic use , Antihypertensive Agents/therapeutic use , Hypertension, Portal/drug therapy , Liver Cirrhosis, Experimental/complications , Amiloride/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Canrenoic Acid/administration & dosage , Canrenoic Acid/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Hypertension, Portal/etiology , Hypertension, Portal/physiopathology , Kupffer Cells/physiology , Liver Cirrhosis, Experimental/physiopathology , Male , Portal Pressure/drug effects , Portal Pressure/physiology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/therapeutic use , Thromboxane A2/physiology , rho-Associated Kinases/physiology
15.
Clin Sci (Lond) ; 119(7): 283-92, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20459396

ABSTRACT

Our present study examines, in mesenteric resistance arteries, possible vasodilation alterations, and the role of NO and COX (cyclo-oxygenase) derivatives, in cirrhosis. The vasodilator response to acetylcholine was analysed in segments from control and cirrhotic rats. The effects of the non-specific COX inhibitor indomethacin, the specific COX-1 inhibitor SC-560 and the specific COX-2 inhibitor NS-398 were analysed in segments from both groups of rats. NO release was measured, and eNOS [endothelial NOS (NO synthase)], phospho-eNOS, iNOS (inducible NOS), COX-1 and COX-2 protein expression was also analysed. The effects of the TP receptor [TXA2 (thromboxane A(2)) receptor] antagonist SQ 29548, the TXA(2) synthesis inhibitor furegrelate, the PGI(2) (prostaglandin I(2)) synthesis inhibitor TCP (tranylcypromine) or TCP+furegrelate were only determined in segments from cirrhotic rats. The vasodilator response to acetylcholine was higher in segments from cirrhotic rats. Indomethacin, SC-560 and NS-398 did not modify the vasodilator response in control rats; however, indomethacin, NS-398 and TCP+furegrelate increased, whereas SC-560 did not modify and SQ 29548, furegrelate or TCP decreased, the vasodilator response to acetylcholine in cirrhotic rats. NO release was higher in cirrhotic rats. Furegrelate decreased, whereas TCP+furegrelate increased, the NO release in segments from cirrhotic rats. eNOS and COX-1 protein expression was not modified, whereas phosho-eNOS, iNOS and COX-2 protein expression was higher in cirrhotic rats. Therefore the increase in iNOS expression and eNOS activity may mediate increases in endothelial NO release. The COX-2 derivatives TXA(2) and PGI(2) may act simultaneously, producing a compensatory effect that reduces NO release and may limit the hyperdynamic circulation.


Subject(s)
Liver Cirrhosis, Experimental/metabolism , Mesenteric Arteries/metabolism , Nitric Oxide/metabolism , Prostaglandins A/physiology , Thromboxane A2/physiology , Animals , Blood Pressure/physiology , Body Weight/physiology , C-Reactive Protein/metabolism , Lipid Metabolism/physiology , Liver/pathology , Liver Cirrhosis, Experimental/physiopathology , Male , Organ Size/physiology , Prostaglandins/metabolism , Prostaglandins A/biosynthesis , Rats , Rats, Sprague-Dawley , Spleen/pathology , Superoxides/metabolism , Thromboxane A2/biosynthesis , Vasodilation/physiology
16.
Atherosclerosis ; 208(1): 43-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19615686

ABSTRACT

OBJECTIVE: Hyperhomocysteinemia (HHcy) has been shown to impair the endothelial function of arterial vessels and promote thrombosis. There are no studies, however, assessing the effects of HHcy on the vasomotor function of venules. We hypothesized that HHcy activates pathophysiological mechanisms impairing flow/shear stress-dependent responses of venules. METHODS AND RESULTS: Changes in diameter of isolated gracilis muscle venules (diameter: approximately 250 microm at 10 mmHg) of control and HHcy rats (induced by methionine diet for 5 weeks) to increases in intraluminal flow were measured. Increases in flow elicited dilations in control (at max.: 14+/-1%), but induced constrictions in HHcy venules (at max.: -24+/-4%). Flow-induced constrictions in HHcy venules were converted to dilations in the presence of the thromboxane A(2) (TxA(2)) receptor (TP) antagonist SQ 29,548, which were then abolished by the simultaneous administration of nitric oxide (NO) synthase inhibitor, L-NAME and non-selective cyclooxygenase (COX) blocker, indomethacin. In addition, the selective COX-2 inhibitor NS 398 reversed flow-induced constrictions to dilations, which were significantly decreased by additional COX-1 inhibitor, SC 560. Also, as compared to controls, a SOD/CAT sensitive increased ethidium bromide fluorescence was detected in HHcy small veins, indicating substantial production of reactive oxygen species (ROS) in HHcy. Correspondingly, SOD/CAT diminished flow-induced constrictions in venules of HHcy rats. CONCLUSIONS: In hyperhomocysteinemia increases in flow/shear stress increases the production of COX-2-derived TxA(2), and reactive oxygen species--that overcome the dilator effects of NO and prostaglandins--eliciting constrictions in skeletal muscle venules; changes which can increase vascular resistance and favor thrombus formation in the venular circulation.


Subject(s)
Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/physiopathology , Reactive Oxygen Species , Thromboxane A2/physiology , Vasoconstriction , Venules/physiopathology , Animals , Cyclooxygenase 2/metabolism , Male , Rats , Rats, Wistar , Regional Blood Flow , Thromboxane A2/metabolism
17.
Atherosclerosis ; 209(2): 415-21, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19932480

ABSTRACT

OBJECTIVE: The plasma level of soluble CD40 ligand (sCD40L), which induces pro-inflammatory and pro-atherogenic responses, is known to be elevated in atherosclerotic patients. In this study, we investigated the mechanism of sCD40L release from human platelets, focusing on the involvement of thromboxane (TX) A(2). METHODS: We measured sCD40L release and TXA(2) production induced by ristocetin, an activator of GPIb/IX/V, from human platelets in vitro. Moreover, plasma sCD40L and TXA(2) levels in the 10 patients with severe carotid artery stenosis who were not taking any anti-platelet medicines were measured and compared with those obtained from non-atherosclerotic controls. RESULTS: Ristocetin significantly promoted sCD40L release and TXA(2) generation from platelets in vitro. Aspirin and SC-560, a cyclooxygenase-1 inhibitor, suppressed the ristocetin-induced sCD40L release from platelets in parallel with TXA(2) production. Ozagrel, a TXA(2) synthase inhibitor and PTXA(2), a thromboxane receptor (TP) antagonist also suppressed sCD40L release. U46619, a TP agonist, reversed the suppressive effect of aspirin on sCD40L release. In vivo, plasma levels of sCD40L and TXA(2) in the patients were significantly higher than those in controls. Elevated plasma levels of TXA(2) and sCD40L in the patients were markedly diminished after 7 days of 100mg aspirin administration. CONCLUSION: These results strongly suggest that GPIb/IX/V activation induces sCD40L release via TXA(2) from human platelets, and that sCD40L release via TXA(2) generation from platelets in atherosclerotic patients are up-regulated.


Subject(s)
Blood Platelets/physiology , CD40 Ligand/blood , Thromboxane A2/biosynthesis , Thromboxane A2/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Aged , Aspirin/pharmacology , Atherosclerosis/blood , Blood Platelets/drug effects , Carotid Artery Diseases/blood , Female , Humans , MAP Kinase Kinase 4/metabolism , Male , Methacrylates/pharmacology , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Pyrazoles , Ristocetin/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Pharmacol Exp Ther ; 331(3): 917-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19741149

ABSTRACT

We have recently reported that left atrial injections of the thromboxane A(2) (TXA(2)) mimetic, (5Z)-7-[(1R,4S,5S,6R)-6-[(1E,3S)-3-hydroxy-1-octenyl]-2 -oxabicyclo[2.2.1]hept-5-yl]-5-heptenoic acid (U46619), induced ventricular arrhythmias in the anesthetized rabbit. Data from this study led us to hypothesize that TXA(2) may be inducing direct actions on the myocardium to induce these arrhythmias. The aim of this study was to further elucidate the mechanism responsible for these arrhythmias. We report that TXA(2)R is expressed at both the gene and protein levels in atrial and ventricular samples of adult rabbits. In addition, TXA(2)R mRNA was identified in single, isolated ventricular cardiac myocytes. Furthermore, treatment of isolated cardiac myocytes with U46619 increased intracellular calcium in a dose-dependent manner and these increases were blocked by the specific TXA(2)R antagonist, 7-(3-((2-((phenylamino)carbonyl)hydrazino)methyl)-7-oxabicyclo(2.2.1)hept-2-yl)-5-heptenoic acid (SQ29548). Pretreatment of myocytes with an inhibitor of inositol trisphosphate (IP(3)) formation, gentamicin, or with an inhibitor of IP(3) receptors, 2-aminoethoxydiphenylborate (2-APB), blocked the increase in intracellular calcium. In vivo pretreatment of anesthetized rabbits with either gentamicin or 2-APB subsequently inhibited the formation of ventricular arrhythmias elicited by U46619. These data support the hypothesis that TXA(2) can induce arrhythmias via a direct action on cardiac myocytes. Furthermore, these arrhythmogenic actions were blocked by inhibitors of the IP(3) pathway. In summary, this study provides novel evidence for direct TXA(2)-induced cardiac arrhythmias and provides a rationale for IP(3) as a potential target for the treatment of TXA(2)-mediated arrhythmias.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Inositol 1,4,5-Trisphosphate/antagonists & inhibitors , Myocytes, Cardiac/metabolism , Thromboxane A2/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Arrhythmias, Cardiac/prevention & control , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic , Cells, Cultured , Fatty Acids, Unsaturated , Heart Atria/metabolism , Heart Ventricles/metabolism , Hydrazines/pharmacology , Inositol 1,4,5-Trisphosphate/biosynthesis , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/biosynthesis , Male , Myocytes, Cardiac/drug effects , Rabbits , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Receptors, Thromboxane A2, Prostaglandin H2/biosynthesis , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thromboxane A2/agonists , Thromboxane A2/antagonists & inhibitors , Vasoconstrictor Agents/pharmacology
19.
Crit Care Med ; 37(7 Suppl): S186-202, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19535947

ABSTRACT

BACKGROUND: Mild to moderate hypothermia (32-35 degrees C) is the first treatment with proven efficacy for postischemic neurological injury. In recent years important insights have been gained into the mechanisms underlying hypothermia's protective effects; in addition, physiological and pathophysiological changes associated with cooling have become better understood. OBJECTIVE: To discuss hypothermia's mechanisms of action, to review (patho)physiological changes associated with cooling, and to discuss potential side effects. DESIGN: Review article. INTERVENTIONS: None. MAIN RESULTS: A myriad of destructive processes unfold in injured tissue following ischemia-reperfusion. These include excitotoxicty, neuroinflammation, apoptosis, free radical production, seizure activity, blood-brain barrier disruption, blood vessel leakage, cerebral thermopooling, and numerous others. The severity of this destructive cascade determines whether injured cells will survive or die. Hypothermia can inhibit or mitigate all of these mechanisms, while stimulating protective systems such as early gene activation. Hypothermia is also effective in mitigating intracranial hypertension and reducing brain edema. Side effects include immunosuppression with increased infection risk, cold diuresis and hypovolemia, electrolyte disorders, insulin resistance, impaired drug clearance, and mild coagulopathy. Targeted interventions are required to effectively manage these side effects. Hypothermia does not decrease myocardial contractility or induce hypotension if hypovolemia is corrected, and preliminary evidence suggests that it can be safely used in patients with cardiac shock. Cardiac output will decrease due to hypothermia-induced bradycardia, but given that metabolic rate also decreases the balance between supply and demand, is usually maintained or improved. In contrast to deep hypothermia (

Subject(s)
Brain Ischemia/complications , Hypothermia, Induced/adverse effects , Hypothermia, Induced/methods , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Acidosis/etiology , Apoptosis/physiology , Body Temperature Regulation/physiology , Brain Edema/etiology , Calpain/physiology , Critical Care/methods , Critical Care/trends , Epilepsy/etiology , Free Radicals/adverse effects , Genes, Immediate-Early/physiology , Humans , Hypothermia, Induced/trends , Infections/etiology , Inflammation , Ion Pumps/physiology , Mitochondria/physiology , Reperfusion Injury/etiology , Reperfusion Injury/physiopathology , Thrombosis/etiology , Thromboxane A2/physiology
20.
Endocrinology ; 150(7): 3267-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19325001

ABSTRACT

Recent studies suggested an involvement of thromboxane A2 in cyclooxygenase-2-dependent inhibition of steroidogenic acute regulatory (StAR) gene expression. The present study further investigated the role of thromboxane A2 receptor in StAR gene expression and steroidogenesis in testicular Leydig cells. The thromboxane A2 receptor was detected in several Leydig cell lines. Blocking thromboxane A2 binding to the receptor using specific antagonist SQ29548 or BM567 resulted in dose-dependent increases in StAR protein and steroid production in MA-10 mouse Leydig cells. The results were confirmed with Leydig cells isolated from rats. StAR promoter activity and StAR mRNA level in the cells were also increased after the treatments, suggesting an involvement of the thromboxane A2 receptor in StAR gene transcription. Furthermore study indicated that blocking the thromboxane A2 receptor reduced dosage sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1 protein, a transcriptional repressor of StAR gene expression. Specific binding of the antagonists to the receptors on cellular membrane was demonstrated by binding assays using (3)H-SQ29548 and binding competition between (3)H-SQ29548 and BM567. Whereas SQ29548 enhanced cAMP-induced StAR gene expression, in the absence of cAMP, it was unable to increase StAR protein and steroidogenesis. However, when the receptor was blocked by the antagonist, subthreshold levels of cAMP were able to induce maximal levels of StAR protein expression, suggesting that blocking the thromboxane A2 receptor increase sensitivity of MA-10 cells to cAMP stimulation. Taken together, the results from the present and previous studies suggest an autocrine loop, involving cyclooxygenase-2, thromboxane A synthase, and thromboxane A2 and its receptor, in cyclooxygenase-2-dependent inhibition of StAR gene expression.


Subject(s)
Leydig Cells/metabolism , Phosphoproteins/genetics , Receptors, Thromboxane A2, Prostaglandin H2/physiology , Animals , Bridged Bicyclo Compounds, Heterocyclic , Cell Line , Cyclooxygenase 2/genetics , Fatty Acids, Unsaturated , Gene Expression Regulation , Hydrazines/pharmacology , Male , Mice , Rats , Sulfonylurea Compounds/pharmacology , Thromboxane A2/genetics , Thromboxane A2/physiology , Thromboxane-A Synthase/genetics , Thromboxane-A Synthase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL