Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Int J Exp Pathol ; 103(1): 4-12, 2022 02.
Article in English | MEDLINE | ID: mdl-34655121

ABSTRACT

The process of gastric ulcer healing includes cell migration, proliferation, angiogenesis and re-epithelialization. Platelets contain angiogenesis stimulating factors that induce angiogenesis. Thromboxane A2 (TXA2 ) not only induces platelet activity but also angiogenesis. This study investigated the role of TXA2 in gastric ulcer healing using TXA2 receptor knockout (TPKO) mice. Gastric ulcer healing was suppressed by treatment with the TXA2 synthase inhibitor OKY-046 and the TXA2 receptor antagonist S-1452 compared with vehicle-treated mice. TPKO showed delayed gastric ulcer healing compared with wild-type mice (WT). The number of microvessels and CD31 expression were lower in TPKO than in WT mice, and TPKO suppressed the expression of transforming growth factor beta (TGF-ß) and vascular endothelial growth factor A (VEGF-A) in areas around gastric ulcers. Immunofluorescence assays showed that TGF-ß and VEGF-A co-localized with platelets. Gastric ulcer healing was significantly reduced in WT mice transplanted with TPKO compared with WT bone marrow. These results suggested that TP signalling on platelets facilitates gastric ulcer healing through TGF-ß and VEGF-A.


Subject(s)
Neovascularization, Pathologic/metabolism , Stomach Ulcer/drug therapy , Thromboxanes/pharmacology , Wound Healing/drug effects , Animals , Mice, Inbred C57BL , Platelet Activation/drug effects , Prostaglandins/pharmacology , Receptors, Thromboxane/drug effects , Signal Transduction/drug effects , Stomach Ulcer/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
Life Sci ; 286: 120073, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34688694

ABSTRACT

AIMS: Thromboxane (TxA2) is synthesized from arachidonic acid (AA) via thromboxane synthase (TxS) enzyme and induces vasoconstriction via TP receptor. Our aim is to compare the effects of aspirin, TxS inhibitor and TP receptor antagonist on vascular reactivity of bypass grafts (saphenous vein and internal mammary artery). MAIN METHODS: Using isolated organ bath, saphenous vein and internal mammary artery preparations were incubated with TP receptor antagonist, TxS inhibitor, aspirin, IP or EP4 receptor antagonist. Then prostaglandin (PG)E2, PGF2α, phenylephrine and AA were administered in concentration-dependent manner. The expression of prostanoid receptor and PGI2 synthase (PGIS) enzyme was determined by Western Blot. KEY FINDINGS: TP receptor antagonist inhibited the contraction induced by PGE2, PGF2α, and AA but not that induced by phenylephrine in both types of vessels. Aspirin increased phenylephrine-induced contraction only in internal mammary artery and decreased AA-induced contraction in saphenous vein. TxS inhibitor decreased both PGE2 and AA-induced contraction in both types of vessels. This decrease was reversed by co-incubation of TxS inhibitor and IP/EP4 receptor antagonists. The expressions of EP3 receptor and PGIS enzyme were greater in internal mammary artery compared to saphenous vein while IP and TP receptors expressed at similar levels. SIGNIFICANCE: TP receptor antagonist and TxS inhibitor are more effective to reduce contraction induced by different spasmogens in comparison to aspirin. Our results suggest that TP receptor antagonist and TxS inhibitor might have an advantage over aspirin due to their preventive effect on increased vascular reactivity observed in post-operative period of coronary artery bypass grafting.


Subject(s)
Mammary Arteries/drug effects , Saphenous Vein/drug effects , Arachidonic Acid/metabolism , Aspirin/pharmacology , Benzofurans/pharmacology , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Mammary Arteries/metabolism , Muscle, Smooth, Vascular/metabolism , Phenylephrine/pharmacology , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/drug effects , Receptors, Thromboxane/metabolism , Saphenous Vein/metabolism , Sulfonamides/pharmacology , Thromboxane A2/pharmacology , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxane-A Synthase/drug effects , Thromboxane-A Synthase/metabolism , Thromboxanes/antagonists & inhibitors , Thromboxanes/metabolism , Vasoconstriction/drug effects
3.
Am J Physiol Renal Physiol ; 320(4): F537-F547, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33491563

ABSTRACT

Local or systemic inflammation can severely impair urinary bladder functions and contribute to the development of voiding disorders in millions of people worldwide. Isoprostanes are inflammatory lipid mediators that are upregulated in the blood and urine by oxidative stress and may potentially induce detrusor overactivity. The aim of the present study was to investigate the effects and signal transduction of isoprostanes in human and murine urinary bladders in order to provide potential pharmacological targets in detrusor overactivity. Contraction force was measured with a myograph in murine and human urinary bladder smooth muscle (UBSM) ex vivo. Isoprostane 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the murine UBSM, which was abolished in mice deficient in the thromboxane prostanoid (TP) receptor. The responses remained unaltered after removal of the mucosa or incubation with tetrodotoxin. Smooth muscle-specific deletion of Gα12/13 protein or inhibition of Rho kinase by Y-27632 decreased the contractions. In Gαq/11-knockout mice, responses were reduced and in the presence of Y-27632 abolished completely. In human UBSM, the TP agonist U-46619 evoked dose-dependent contractions. Neither atropine nor the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid decreased the effect, indicating that TP receptors directly mediate detrusor muscle contraction. 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the human UBSM, and these responses were abolished by the TP antagonist SQ-29548 and were decreased by Y-27632. Our results indicate that isoprostanes evoke contraction in murine and human urinary bladders, an effect mediated by the TP receptor. The G12/13-Rho-Rho kinase pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target in detrusor overactivity.NEW & NOTEWORTHY Voiding disorders affect millions of people worldwide. Inflammation can impair urinary bladder functions and contribute to the development of detrusor overactivity. The effects and signal transduction of inflammatory lipid mediator isoprostanes were studied in human and murine urinary bladders ex vivo. We found that isoprostanes evoke contraction, an effect mediated by thromboxane prostanoid receptors. The G12/13-Rho-Rho kinase signaling pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target.


Subject(s)
Isoprostanes/pharmacology , Muscle, Smooth, Vascular/drug effects , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin/drug effects , Receptors, Thromboxane/drug effects , Animals , Humans , Prostaglandins/pharmacology , Receptors, Thromboxane/physiology
4.
FASEB J ; 34(2): 2568-2578, 2020 02.
Article in English | MEDLINE | ID: mdl-31908041

ABSTRACT

Although recognized to have an in vivo vasodepressor effect blunted by the vasoconstrictor effect of E-prostanoid receptor-3 (EP3), prostaglandin E2 (PGE2 ) evokes contractions of many vascular beds that are sensitive to antagonizing the thromboxane prostanoid receptor (TP). This study aimed to determine the direct effect of PGE2 on renal arteries and/or the whole renal vasculature and how each of these two receptors is involved in the responses. Experiments were performed on isolated vessels and perfused kidneys of wild-type mice and/or mice with deficiency in TP (TP-/- ), EP3 (EP3-/- ), or both TP and EP3 (TP-/- /EP3-/- ). Here we show that PGE2 (0.001-30 µM) evoked not only contraction of main renal arteries, but also a decrease of flow in perfused kidneys. EP3-/- diminished the response to 0.001-0.3 µM PGE2 , while TP-/- reduced that to the prostanoid of higher concentrations. In TP-/- /EP3-/- vessels and perfused kidneys, PGE2 did not evoke contraction but instead resulted in vasodilator responses. These results demonstrate that PGE2 functions as an overall direct vasoconstrictor of the mouse renal vasculature with an effect reflecting the vasoconstrictor activities outweighing that of dilation. Also, our results suggest that EP3 dominates the vasoconstrictor effect of PGE2 of low concentrations (≤0.001-0.3 µM), but its effect is further added by that of TP, which has a higher efficacy, although activated by higher concentrations (from 0.01 µM) of the same prostanoid PGE2 .


Subject(s)
Dinoprostone/pharmacology , Receptors, Prostaglandin E, EP3 Subtype/drug effects , Receptors, Thromboxane/drug effects , Vasoconstriction/drug effects , Animals , Dinoprost/pharmacology , Kidney/drug effects , Mice, Inbred C57BL , Prostaglandins/pharmacology , Receptors, Prostaglandin/drug effects , Thromboxanes/pharmacology , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
5.
Crit Care ; 23(1): 42, 2019 Feb 11.
Article in English | MEDLINE | ID: mdl-30744667

ABSTRACT

BACKGROUND: Several lipid metabolites in cerebrospinal fluid are correlated with poor outcomes in aneurysmal subarachnoid haemorrhage. Most of these metabolites bind to ubiquitous thromboxane-prostaglandin (TP) receptors, causing vasoconstriction and inflammation. Here, we evaluated terutroban (TBN), a specific TP receptor antagonist, for the prevention of post-haemorrhage blood-brain barrier disruption, neuronal apoptosis and delayed cerebral hypoperfusion. METHODS: The rat double subarachnoid haemorrhage model was produced by twice injecting (days 1 and 2) autologous blood into the cisterna magna. Seventy-eight male Sprague-Dawley rats were assigned to experimental groups. Rats exposed to subarachnoid haemorrhage were allocated to no treatment (SAH group) or TBN treatment by gastric gavage during the first 5 days after haemorrhage (SAH+TBN group). Control rats received artificial cerebrospinal fluid injections (CSF group). Sham-operated rats with or without TBN administration were also studied. Body weight and Garcia neurological scores were assessed on day 2 and day 5. We used nanoscale single-photon emission computed tomography (nanoSPECT) to measure brain uptake of three radiolabelled agents: 99mTechnetium-diethylenetriaminepentacetate (99mTc-DTPA), which indicated blood-brain barrier permeability on day 3, 99mTechnetium-annexin V-128 (99mTc-Anx-V128), which indicated apoptosis on day 4, and 99mTechnetium-hexamethylpropyleneamineoxime (99mTc-HMPAO), which indicated cerebral perfusion on day 5. Basilar artery narrowing was verified histologically, and cerebral TP receptor agonists were quantified. RESULTS: 99mTc-DTPA uptake unveiled blood-brain barrier disruption in the SAH group. TBN mitigated this disruption in the brainstem area. 99mTc-Anx-V128 uptake was increased in the SAH group and TBN diminished this effect in the cerebellum. 99mTc-HMPAO uptake revealed a global decreased perfusion on day 5 in the SAH group that was significantly counteracted by TBN. TBN also mitigated basilar artery vasoconstriction, neurological deficits (on day 2), body weight loss (on day 5) and cerebral production of vasoconstrictors such as Thromboxane B2 and Prostaglandin F2α. CONCLUSIONS: Based on in vivo nanoscale imaging, we demonstrated that TBN protected against blood-brain barrier disruption, exerted an anti-apoptotic effect and improved cerebral perfusion. Thus, TP receptor antagonists showed promising results in treating post-haemorrhage neurovascular events.


Subject(s)
Naphthalenes/pharmacokinetics , Propionates/pharmacokinetics , Receptors, Thromboxane/drug effects , Subarachnoid Hemorrhage/drug therapy , Animals , Basilar Artery/pathology , Male , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Physical Functional Performance , Propionates/pharmacology , Propionates/therapeutic use , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/prevention & control , Treatment Outcome
6.
Am J Physiol Cell Physiol ; 307(6): C571-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25080488

ABSTRACT

A multitude of environmental signaling molecules influence monocyte and macrophage innate and adaptive immune responses, including ATP and prostanoids. Interestingly, purinergic (P2) and eicosanoid receptor signaling interact such that the activation of P2 receptors leads to prostanoid production, which can then interfere with P2Y-mediated macrophage migration. Recent studies suggest that blockade of 5-lipoxygenase (5-LOX) in macrophages can activate a permeation pathway involved in the influx of dye and the release of ATP. Here, we provide evidence that pannexin1 (Panx1) is a component of this pathway and present the intracellular signaling molecules linking the thromboxane (TP) receptor to Panx1-mediated dye influx and ATP release. Using pharmacological tools and transgenic mice deficient in Panx1, we show that two 5-LOX pathway inhibitors induce ATP release and influx of dye in a Panx1-dependent manner. Electrophysiological recordings performed in wild-type and Panx1-deficient macrophages confirmed that these 5-LOX pathway inhibitors activate currents characteristic of Panx1 channels. We found that the mechanism by which Panx1 channels are activated under this condition involves activation of the TP receptor that is mediated by the cAMP/PKA pathway. This is to our knowledge the first evidence for the involvement of Panx1 in the TP receptor signaling pathway. Future studies aimed to clarify the contribution of this TP-Panx1 signaling network to macrophage immune responses are likely to be important for targeting inflammatory and autoimmune diseases.


Subject(s)
Connexins/metabolism , Lipoxygenase Inhibitors/pharmacology , Macrophages, Peritoneal/drug effects , Nerve Tissue Proteins/metabolism , Receptors, Thromboxane/drug effects , Adenosine Triphosphate/metabolism , Animals , Cell Line , Connexins/deficiency , Connexins/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Macrophages, Peritoneal/metabolism , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptors, Thromboxane/metabolism , Signal Transduction/drug effects
7.
Am J Physiol Renal Physiol ; 305(6): F830-8, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23884143

ABSTRACT

The present renal hemodynamic study tested the hypothesis that CD38 and superoxide anion (O2(·-)) participate in the vasoconstriction produced by activation of thromboxane prostanoid (TP) receptors in the mouse kidney. CD38 is the major mammalian ADP-ribosyl cyclase contributing to vasomotor tone through the generation of cADP-ribose, a second messenger that activates ryanodine receptors to release Ca(2+) from the sarcoplasmic reticulum in vascular smooth muscle cells. We evaluated whether the stable thromboxane mimetic U-46619 causes less pronounced renal vasoconstriction in CD38-deficient mice and the involvement of O2(·-) in U-46619-induced renal vasoconstriction. Our results indicate that U-46619 activation of TP receptors causes renal vasoconstriction in part by activating cADP-ribose signaling in renal resistance arterioles. Based on maximal renal blood flow and renal vascular resistance responses to bolus injections of U-46619, CD38 contributes 30-40% of the TP receptor-induced vasoconstriction. We also found that the antioxidant SOD mimetic tempol attenuated the magnitude of vasoconstriction by U-46619 in both groups of mice, suggesting mediation by O2(·-). The degree of tempol blockage of U-46619-induced renal vasoconstriction was greater in wild-type mice, attenuating renal vasoconstriction by 40% compared with 30% in CD38-null mice. In other experiments, U-46619 rapidly stimulated O2(·-) production (dihydroethidium fluorescence) in isolated mouse afferent arterioles, an effect abolished by tempol. These observations provide the first in vivo demonstration of CD38 and O2(·-) involvement in the vasoconstrictor effects of TP receptor activation in the kidney and in vitro evidence for TP receptor stimulation of O2(·-) production by the afferent arteriole.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Kidney/blood supply , Membrane Glycoproteins/physiology , Superoxides/pharmacology , Vasoconstriction/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , ADP-ribosyl Cyclase 1/deficiency , Animals , Arterioles/drug effects , Cyclic N-Oxides/pharmacology , Kidney/metabolism , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Receptors, Thromboxane/drug effects , Receptors, Thromboxane/physiology , Spin Labels
8.
Arterioscler Thromb Vasc Biol ; 32(10): 2468-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22879580

ABSTRACT

OBJECTIVE: Thromboxane A(2) and prostacyclin are thromboregulatory prostaglandins. The inflammatory C-reactive protein (CRP) promotes thrombosis after vascular injury, presumably via potentiation of thromboxane activity. Using a genetic approach, we investigated the role of thromboxane receptor (TP) pathway in CRP-induced thrombosis. METHODS AND RESULTS: Four genetically engineered mice strains were used: C57BL/6 wild-type, human CRP transgenic (CRPtg), thromboxane receptor-deficient (Tp(-/-)), and CRPtgTp(-/-) mice. CRP and TP expression were correlated, and suppression of CRP expression using small interfering RNA/CRP led to reduction in TP expression. Platelet-endothelial adherence was increased in CRPtg and suppressed in CRPtgTP(-/-)and CRPtg cells that were suppressed with TP small interfering RNA. TP deficiency in both platelets and endothelial cells was synergistic in affecting platelet-endothelial interactions. Time until arterial occlusion, measured after photochemical injury, was significantly shorter in CRPtg and prolonged in CRPtgTp(-/-) compared with controls (n=10-15, 35±3.4, 136±13.8, and 67±8.9 minutes, respectively; P<0.05). CONCLUSIONS: TP pathway is of major importance in CRP-induced thrombosis. The expression of TP is increased in CRPtg endothelial cells, and its blockade significantly suppresses the prothrombotic effect of CRP.


Subject(s)
C-Reactive Protein/physiology , Receptors, Thromboxane/physiology , Signal Transduction/physiology , Thrombosis/physiopathology , Adult , Animals , Blood Platelets/pathology , Blood Platelets/physiology , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Cell Adhesion/physiology , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Small Interfering/pharmacology , Receptors, Thromboxane/deficiency , Receptors, Thromboxane/drug effects , Thrombosis/pathology , Transfection
9.
Exp Physiol ; 97(7): 895-904, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22447972

ABSTRACT

This study aimed to determine whether cyclo-oxygenase-1 (COX-1) mediates dilatation of mouse arteries via synthesis of prostacyclin (PGI(2)) and, if so, how PGI(2) (IP) receptors contribute and whether thromboxane prostanoid (TP) receptors are implicated in the process. Mesenteric arteries were isolated from wild-type mice or mice with COX-1 deficiency (COX-1(-/-)). The vasomotor reaction to the COX substrate arachidonic acid (AA) was determined with isometric force measurement, while the in vitro production or the plasma level of the PGI(2) metabolite 6-keto-PGF(1α) was analysed with high-performance liquid chromatography-mass spectroscopy or enzyme immunoassay, respectively. Results showed that AA, which evoked endothelium-dependent 6-keto-PGF(1α) production, elicited relaxation that was inhibited or enhanced by antagonizing IP or TP receptors, respectively. Also, IP receptor blockade resulted in contraction in response to AA (following NO synthase inhibition), which was prevented by a concomitant TP receptor antagonism. Meanwhile, COX-1(-/-) or COX-1 inhibition abolished the in vitro 6-keto-PGF(1α) production and reduced the relaxation or contraction observed with AA. Real-time PCR showed that whereas TP receptor mRNAs were detected at similar levels, IP receptor mRNAs were present at higher levels in the branches than in the main stem of the mesenteric artery. In addition, antagonizing the IP receptors enhanced the contraction evoked by PGI(2) in the carotid artery. Also, we noted that COX-1(-/-) mice had a reduced basal plasma 6-keto-PGF(1α) level. These results demonstrate an explicit vasodilator role for COX-1-mediated endothelial PGI(2) synthesis and suggest that the functionally opposing IP and TP receptors concomitantly mediate the vasomotor reaction to PGI(2), with the dilator activity of IP receptors being compromised by the vasoconstrictor effect of TP receptors and vice versa.


Subject(s)
Arachidonic Acid/pharmacology , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Arachidonic Acid/metabolism , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/metabolism , Epoprostenol/biosynthesis , Epoprostenol/metabolism , Male , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Receptors, Epoprostenol/drug effects , Receptors, Prostaglandin/drug effects , Receptors, Thromboxane/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects
10.
Eur J Pharmacol ; 644(1-3): 128-37, 2010 Oct 10.
Article in English | MEDLINE | ID: mdl-20615400

ABSTRACT

Organ culture is an in vitro method for investigating cellular mechanisms involved in upregulation of vasocontractile G-protein coupled receptors. We hypothesize that mitogen-activated-protein kinase (MEK) and/or extracellular-signal-regulated kinase (ERK) specific inhibitors will attenuate the G-protein coupled receptor expression following organ culture. Rat cerebral arteries were incubated 48h in the presence of MEK/ERK specific inhibitors U0126, PD98059, SL327, or AG126 for different time periods. Contractile responses by activation of endothelin receptor type A and type B, serotonin receptor 5-HT(1B), prostanoid TP receptor, and angiotensin II receptor type 1 and type 2 were investigated. Results were verified by measurement of mRNA with real time PCR and by protein immunohistochemistry. Organ culture induced transcriptional upregulation of endothelin ET(B) receptor and of serotonin 5-HT(1B) receptor on translational level and increased respective contractions. The prostanoid TP receptor mediated contraction curve was left-wards shifted by organ culture. Organ culture was associated with elevated pERK1/2 in the vascular smooth muscle cells: the MEK1/2 inhibitor U0126 attenuated the endothelin ET(B) receptor mediated contraction at post-translational level or by changing the receptor affinities. The serotonin 5-HT(1B) receptor and prostanoid TP receptor mediated contractions were abolished by U0126. Administration of U0126 6h after start of incubation blocked the receptor upregulation. In conclusion, MEK specific inhibitor U0126 is a potent inhibitor of G-protein coupled receptor alteration seen during organ culture. Given the ability to inhibit G-protein coupled receptor alteration at the clinically relevant time-point 6h post incubation makes it an attractive therapeutic agent for in vivo studies.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Receptor, Endothelin B/drug effects , Receptor, Serotonin, 5-HT1B/drug effects , Animals , Male , Middle Cerebral Artery/drug effects , Middle Cerebral Artery/metabolism , Organ Culture Techniques , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Endothelin B/genetics , Receptor, Serotonin, 5-HT1B/genetics , Receptors, Thromboxane/drug effects , Receptors, Thromboxane/metabolism , Time Factors , Up-Regulation/drug effects
11.
Br J Pharmacol ; 158(7): 1787-95, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19891662

ABSTRACT

BACKGROUND AND PURPOSE: Prostacyclin (PGI(2)) is usually described as an endothelium-derived vasodilator, but it can also induce vasoconstriction. We studied the vasomotor responses to PGI(2) in resistance arteries and the role of thromboxane (TP) and prostaglandin E(2) (EP) receptors in this effect. EXPERIMENTAL APPROACH: Mesenteric resistance arteries were obtained from Sprague-Dawley rats. Vasomotion to PGI(2) was studied in segments of these arteries with and without endothelium and in presence of the nitric oxide (NO) synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME), the potassium channel blockers apamin plus charybdotoxin, the non-selective EP receptor antagonist AH6809, the selective TP receptor antagonist SQ29548 or the EP(1) receptor antagonist SC19220. PGI(2)-induced NO release was analysed in the absence or presence of SQ29548, AH6809 or SC19220. KEY RESULTS: PGI(2) caused contractions in arterial segments that were increased by endothelium removal, L-NAME or L-NAME plus apamin plus charybdotoxin and abolished by SQ29548. In segments with endothelium, AH6809 or SC19220 almost abolished the contractions to PGI(2); this effect was prevented by L-NAME, L-NAME plus apamin plus charybdotoxin or by endothelium removal. PGI(2) induced NO release that was inhibited by the prostacyclin receptor (IP receptor) antagonist, RO1138452, and increased by SQ29548, SC19220 and AH6809. The increase in NO release induced by these separate drugs was inhibited by RO1138452. CONCLUSIONS AND IMPLICATIONS: PGI(2) activated the TP receptor in mesenteric resistance arteries and produced vasoconstriction, which the endothelium modulated through TP and EP(1) receptors. PGI(2) also released endothelium-derived hyperpolarizing factor and, through IP receptor activation, induced NO release, which in turn, was antagonized by TP and EP(1) receptor activation.


Subject(s)
Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Thromboxane/metabolism , Animals , Biological Factors/metabolism , Epoprostenol/pharmacology , Male , Mesenteric Arteries/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Thromboxane/drug effects , Vasoconstriction/physiology
14.
J Pharmacol Exp Ther ; 328(1): 231-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18836067

ABSTRACT

Epoxy- and dihydroxy-eicosatrienoic acids (EETs and DHETs) are vasoactive cytochrome P450 metabolites of arachidonic acid. Interestingly, however, the mechanism(s) by which EETs/DHETs mediate smooth muscle relaxation remains unclear. In contrast to previous reports, where dilation was purportedly large-conductance Ca(2+)-activated K(+) (BK(Ca)) and/or transient receptor potential cation channel, subfamily V, member 4 (TRPV4) channel-mediated, 14,15-EET-induced vasodilation [reversal of contractile tone established with the thromboxane receptor (TP) agonist 15-hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic acid (U-46619)] was unaltered in BK(Ca) and TRPV4 knockout mouse isolated aortae compared with wild-type controls, indicating a significant BK(Ca)/TRPV4-resistant mechanism. Whereas all EET and DHET regioisomers reversed U-46619 contraction in rat aortae and mouse mesenteric resistance arteries, these eicosanoids failed to alter phenylephrine-induced contraction, suggesting that they mediated dilation via a "TP-selective" mechanism. Competitive TP antagonism was also observed in nonvascular tissue, including rat fundus and tertiary bronchus, indicating that the effect is not specific to blood vessels. Such effects were TP-selective because 14,15-EET failed to inhibit "non-TP" prostanoid receptor-mediated function in multiple cell/tissue-based assays (K(b) > 10 microM). In accordance, 14,15-EET inhibited specific [(3)H]7-(3-((2-((phenylamino)carbonyl)hydrazino)-methyl)-7-oxabicyclo(2.2.1)hept-2-yl)-5-heptenoic acid (SQ-29548) binding to human recombinant TP receptor, with a K(i) value of 3.2 microM, and it showed weaker affinity for non-TP prostanoid receptors, including DP, FP, EP(1-4), and IP receptors (K(i) values of 6.1, 5.3, 42.6, 19.7, 13.2, 20.2, and >25 microM, respectively) and no appreciable affinity (K(i) values >10 microM) for a diverse array of pharmacologically distinct receptors, including the leukotriene receptors Cys-LT(1/2) and BLT(1). As such, EETs/DHETs represent a unique class of "endogenous" G protein-coupled receptor competitive antagonists, inducing vasodilation via direct TP inhibition. Thus, EETs/DHETs represent novel autoregulatory agents, directly modulating the actions of cyclooxygenase-derived eicosanoids following arachidonic acid mobilization.


Subject(s)
8,11,14-Eicosatrienoic Acid/pharmacology , Aorta, Thoracic/physiology , Vasodilation/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Animals , Aorta, Thoracic/drug effects , Arachidonic Acids/pharmacology , Guinea Pigs , Hydroxyeicosatetraenoic Acids/pharmacology , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, Thromboxane/agonists , Receptors, Thromboxane/drug effects , Receptors, Thromboxane/physiology , Splanchnic Circulation/drug effects , Splanchnic Circulation/physiology , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics , TRPV Cation Channels/physiology , Trachea/drug effects , Trachea/physiology , Vascular Resistance/drug effects , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects
15.
Vasc Health Risk Manag ; 4(3): 637-45, 2008.
Article in English | MEDLINE | ID: mdl-18827913

ABSTRACT

A common pathophysiological course in vascular diseases is an overwhelming activation and aggregation of blood platelets, which results in atherothrombosis. By causing the last decisive step of cerebral, coronary, or peripheral arterial ischemia thrombotic complications of atherosclerotic disease represent a major player in death cause statistics of most western countries. The development of novel therapies against platelet-dependent thrombosis and the concurrent improvement of existing therapeutic strategies thus is a paramount focus of pharmaceutical research. Currently, efficiency, dosing and indications of established antiplatelet substances are being re-evaluated, whilst new, so far unrecognized molecular targets for inhibition of platelet activity come up front. This not only allows for interesting new therapeutical options, but also widens our insight into the role platelets play in atherosclerosis in general. This article summarizes the relevant pathophysiology of platelet activation, presents current concepts in antiplatelet drug therapy, and highlights the role of platelets in vascular diseases apart from atherothrombosis.


Subject(s)
Atherosclerosis/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Thrombosis/drug therapy , Atherosclerosis/physiopathology , Clopidogrel , Drug-Eluting Stents , Humans , Piperazines/administration & dosage , Platelet Activation/physiology , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Prasugrel Hydrochloride , Purinergic P2 Receptor Antagonists , Pyridines/therapeutic use , Receptors, Thromboxane/drug effects , Signal Transduction/physiology , Thiophenes/administration & dosage , Thrombosis/physiopathology , Ticlopidine/administration & dosage , Ticlopidine/analogs & derivatives , Ticlopidine/therapeutic use
16.
Thromb Haemost ; 100(1): 83-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18612542

ABSTRACT

Aspirin 'resistance' (AR) is a phenomenon of uncertain etiology describing decreased platelet inhibition by aspirin. We studied whether (i) platelets in AR demonstrate increased basal sensitivity to a lower degree of stimulation and (ii) platelet aggregation with submaximal stimulation could predict responses to aspirin. Serum thromboxane B(2) (TxB(2)) levels and platelet aggregation with light transmission aggregometry (LTA) were measured at baseline and 24 hours after 325 mg aspirin administration in 58 healthy subjects. AR was defined as the upper sixth of LTA (> or = 12%) to 1.5 mM AA. Baseline platelet aggregation with submaximal concentrations of agonists [ADP 2 microM, arachidonic acid (AA) 0.75 mM, collagen 0.375 and 0.5 microg/ml] was greater in AR subjects compared with non-AR subjects, but not with higher concentrations (ADP 5 microM and 20 microM, AA 1.5 mM and collagen 1 microg/ml). Post-aspirin platelet aggregation was elevated in AR subjects with both submaximal and maximal stimulation. Baseline and post-aspirin serum TxB(2) were higher in AR subjects and decreased further with ex-vivo COX-1 inhibition, suggesting incompletely suppressed COX-1 activity. Pre-aspirin platelet aggregation to 0.75 AA demonstrated a dichotomous response with 29/58 subjects having aggregation < or = 15% and 29/58 subjects having aggregation > or = 75%. In the high aggregation group 28% had AR compared to 6% in the non-AR group (p = 0.04). In conclusion, platelets in AR subjects demonstrate increased basal sensitivity to submaximal stimulation, which could predict responses to antiplatelet therapy.


Subject(s)
Arachidonic Acid , Aspirin/pharmacology , Blood Platelets/drug effects , Drug Resistance , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Adenosine Diphosphate , Adult , Aspirin/therapeutic use , Blood Platelets/metabolism , Collagen , Cyclooxygenase 1/blood , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Male , Platelet Aggregation Inhibitors/therapeutic use , Platelet Function Tests , Predictive Value of Tests , Receptors, Thromboxane/blood , Receptors, Thromboxane/drug effects , Reference Values , Thromboxane B2/blood , Treatment Failure
17.
Eur J Pharmacol ; 592(1-3): 81-6, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18647601

ABSTRACT

Neuromedin U is a hypothalamic peptide involved in energy homeostasis and stress responses. The peptide, when administered intracerebroventricularly (i.c.v.), decreases food intake and body weight while increasing body temperature and heat production. We examined the effect of i.c.v. administered neuromedin U on plasma catecholamines with regard to the brain prostanoid using anesthetized rats. Neuromedin U (0.1, 0.5 and 1 nmol/animal, i.c.v.) effectively elevated plasma adrenaline (a maximal response was obtained at 0.5 nmol/animal), but had little effect on plasma noradrenaline. However, intravenously administered neuromedin U (0.5 nmol/animal) had no effect on plasma catecholamines. Neuromedin U (0.5 nmol/animal, i.c.v.)-induced elevation of plasma adrenaline was effectively reduced by intracerebroventricular pretreatments with indomethacin (an inhibitor of cyclooxygenase) (0.6 and 1.2 micromol/animal), furegrelate (an inhibitor of thromboxane A2 synthase) (0.9 and 1.8 micromol/animal) and (+)-S-145 (a blocker of prostanoid TP receptors) (250 and 625 nmol/animal), respectively. The neuromedin U-induced adrenaline response was also abolished by acute bilateral adrenalectomy. These results suggest that centrally administered neuromedin U evokes the secretion of adrenaline from the adrenal medulla by brain prostanoid TP receptor-mediated mechanisms in rats.


Subject(s)
Epinephrine/blood , Neuropeptides/pharmacology , Receptors, Thromboxane/drug effects , Anesthesia , Anesthetics, Inhalation , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzofurans/pharmacology , Brain Chemistry/drug effects , Bridged Bicyclo Compounds/pharmacology , Catecholamines/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Indomethacin/pharmacology , Injections, Intraventricular , Male , Neuropeptides/administration & dosage , Prostaglandin Antagonists/pharmacology , Rats , Rats, Wistar , Thromboxane-A Synthase/antagonists & inhibitors , Urethane
18.
J Thorac Cardiovasc Surg ; 135(1): 131-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18179929

ABSTRACT

OBJECTIVES: Radial artery vasospasm remains a potential cause of early graft failure after coronary bypass graft surgery, despite pretreatment with alpha-adrenergic or calcium channel blockers. We examined the roles of isoprostanes and prostanoid receptors selective for thromboxane A2 in the vasoconstriction of human radial arteries. METHODS: Human radial arterial segments were pretreated intraoperatively with verapamil/papaverine or nitroglycerine/phenoxybenzamine, or not treated. In the laboratory, we measured isometric contractions in ring segments, vasoconstriction in pressurized segments, and changes in [Ca2+] and K+ currents in single cells. RESULTS: Although phenoxybenzamine eliminated adrenergic responses, the isoprostane 15-F(2t)-IsoP and 2 closely related E-ring molecules (15-E(1t)-IsoP and 15-E(2t)-IsoP) still evoked powerful contractions; 15-E(2t)-IsoP was approximately 10-fold more potent than the other 2 agents. Responses were mediated through thromboxane receptors because they were sensitive to ICI-192605. Furthermore, they were sensitive to the Rho-kinase inhibitors Y-27632 or H-1152 (both 10(-5) mol/L) or to cyclopiazonic acid (which depletes the internal Ca2+ pool), but not to nifedipine. In single cells, 15-E(2t)-IsoP elevated [Ca2+]i and suppressed K+ current. CONCLUSIONS: Isoprostanes accumulate after coronary artery bypass graft surgery, yet none of the currently available antispasm treatments for radial artery grafts is effective against isoprostane-induced vasoconstriction. It is imperative that more specific treatment strategies be developed. We found that isoprostane responses in radial arteries are mediated by prostanoid receptors selective for thromboxane A2 with activation of Rho-kinase and release of Ca2+. Pretreatment of radial artery grafts with Rho-associated kinase inhibitors may potentially reduce postoperative graft spasm. Clinical studies to test this are indicated.


Subject(s)
Isoprostanes/pharmacology , Radial Artery/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Calcium/metabolism , Humans , In Vitro Techniques , Radial Artery/physiopathology , Receptors, Thromboxane/drug effects , Receptors, Thromboxane A2, Prostaglandin H2 , Signal Transduction , Tissue and Organ Harvesting , rhoA GTP-Binding Protein/metabolism
19.
Eur J Pharmacol ; 562(3): 227-35, 2007 May 21.
Article in English | MEDLINE | ID: mdl-17362923

ABSTRACT

Considering the potential physiological, pharmacological and therapeutic relevance of synergistic interaction of thromboxane A(2) with adrenaline at postjunctional receptor sites, we examined whether sub-threshold concentrations of thromboxane A(2) mimetic U-46619 (9,11-dideoxy-9alpha, 11alpha-methanoepoxy prostaglandin F(2alpha)) could amplify adrenaline-induced contraction in human umbilical vein. The receptor involved in U-46619-induced potentiation of adrenaline contractility was also investigated. Umbilical cords (n=125) from healthy patients after full-term vaginal or caesarean deliveries were employed. The vein was dissected out of cords and rings used for isolated organ bath experiments or reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. Presence of endothelium did not modify U-46619-induced contraction in human umbilical vein. Prostanoid TP-selective receptor antagonist, SQ-29548 (7-[3-[[2-[(phenylamino)carbonyl]hydrazino]methyl]-7-oxabicyclo[2.2.1]hept-2-yl]-[1S(1alpha,2alpha(Z),3alpha,4alpha)]-5-Heptenoic acid), inhibited U-46619-induced contraction (pA(2)=8.22+/-0.11). U-46619 sub-threshold concentrations (0.1-0.3 nM) potentiated adrenaline-vasoconstriction response in a concentration-dependent manner. SQ-29548 (0.1 microM) abolished this potentiation. Using RT-PCR, we found that human umbilical vein rings with or without endothelium express the prostanoid TP(alpha), but not the prostanoid TP(beta) receptor isoform. Western blot allowed the identification of proteins with an electrophoretic mobility (47- and 55-kDa) indistinguishable from human platelet prostanoid TP receptor, a rich source of prostanoid TP(alpha) receptor isoform. Collectively, present results demonstrate that prostanoid TP(alpha) is the major receptor isoform localized on smooth muscle cells which participate in both direct vasoconstriction and potentiating effects of U-46619 on adrenaline contractions in human umbilical vein. These results suggest that thromboxane A(2) may interact synergistically with adrenaline in pathophysiological situations that lead to an increase of its umbilical venous levels (e.g. preeclampsia associated with fetal distress) raising the possibility of vasoconstriction affecting fetal blood flow.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Epinephrine/pharmacology , Receptors, Thromboxane/drug effects , Umbilical Veins/drug effects , Vasoconstrictor Agents/pharmacology , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic , Dose-Response Relationship, Drug , Drug Synergism , Endothelium, Vascular/metabolism , Fatty Acids, Unsaturated , Female , Humans , Hydrazines , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Protein Isoforms/metabolism , Receptors, Thromboxane/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins/metabolism , Vasoconstriction/drug effects
20.
Atherosclerosis ; 191(2): 250-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16797553

ABSTRACT

Sanguinarine is a plant alkaloid present in the root of Sanguinaria canadensis and Poppy fumaria species. Sanguinarine has been used as an antiseptic mouth rinse and a toothpaste additive to reduce dental plaque and gingival inflammation. In this study, we investigated the antiplatelet effects of sanguinarine, aiming to extend its potential pharmacological applications. Sanguinarine inhibited platelet aggregation induced by arachidonic acid (AA), collagen, U46619 and sub-threshold concentration of thrombin (0.05 U/ml) with IC(50) concentrations of 8.3, 7.7, 8.6 and 4.4 microM, respectively. Sanguinarine (5-10 microM) inhibited 10-31% of platelet TXB(2) production, but not platelet aggregation induced by higher concentration of thrombin (0.1 U/ml). SQ29548, a thromboxane receptor antagonist, inhibited the AA-induced platelet aggregation but not TXB(2) production. Sanguinarine suppressed cyclooxygenase-1 (COX-1) activity (IC(50)=28 microM), whereas its effect on COX-2 activity was minimal. Sanguinarine (8, 10 microM) further inhibited the AA-induced Ca(2+) mobilization by 27-62%. In addition, SQ22536, an adenylate cyclase inhibitor, attenuated the inhibitory effect of sanguinarine toward AA-induced platelet Ca(2+) mobilization and aggregation. These results suggest that sanguinarine is a potent antiplatelet agent, which activates adenylate cyclase, inhibits platelet Ca(2+) mobilization, TXB(2) production as well as suppresses COX-1 enzyme activity. Sanguinarine may have therapeutic potential for treatment of cardiovascular diseases related to platelet aggregation.


Subject(s)
Alkaloids/pharmacology , Benzophenanthridines/pharmacology , Blood Platelets/drug effects , Calcium/metabolism , Cyclic AMP/metabolism , Cyclooxygenase Inhibitors/pharmacology , Isoquinolines/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thromboxane B2/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenylyl Cyclases/metabolism , Animals , Arachidonic Acid/pharmacology , Blood Platelets/metabolism , Collagen/pharmacology , Cyclooxygenase 1/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , In Vitro Techniques , Rabbits , Receptors, Thromboxane/drug effects , Receptors, Thromboxane/metabolism , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL