Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 364
Filter
1.
J Pharmacol Sci ; 155(4): 148-151, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880549

ABSTRACT

We examined the inhibitory effects of α-linolenic acid (ALA) on the contractions of pig coronary arteries. ALA concentration-dependently inhibited the contractions elicited by U46619 and prostaglandin F2α without affecting those elicited by 80 mM KCl, histamine, acetylcholine, and serotonin. ALA rightward shifted the concentration-response curve of U46619, and Schild plot analysis revealed that ALA competitively antagonized U46619. Furthermore, ALA inhibited the increase in intracellular Ca2+ concentration caused by TP receptor stimulation but not that caused by FP receptor stimulation. These results suggest that ALA behaves as a selective antagonist of TP receptors in coronary arteries.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Calcium , Coronary Vessels , Receptors, Thromboxane , alpha-Linolenic Acid , Animals , Coronary Vessels/drug effects , alpha-Linolenic Acid/pharmacology , Swine , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Calcium/metabolism , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/metabolism , Dose-Response Relationship, Drug , Male , Dinoprost/pharmacology , Muscle Contraction/drug effects
2.
Int J Biochem Cell Biol ; 151: 106281, 2022 10.
Article in English | MEDLINE | ID: mdl-35995387

ABSTRACT

Excessive mitochondrial fission in podocytes serves as a central hub for the pathogenesis of diabetic nephropathy (DN), and the thromboxane/prostaglandin receptor (TP receptor) plays a potential role in DN. However, regulation of the TP receptor during mitochondrial dynamics disorder in podocytes remains unknown. Here, we firstly reported novel mechanistic details of TP receptor effects on mitochondrial dynamics in podocytes under diabetic conditions. Expression of the TP receptor was significantly upregulated in podocytes under diabetic conditions both in vivo and in vitro. S18886 attenuated podocyte mitochondrial fission, glomerular injury and renal dysfunction in diabetic mice. Furthermore, inhibition of the TP receptor by both genetic and pharmacological methods dramatically reduced mitochondrial fission and attenuated podocyte injury induced by high glucose through regulating dynamin-related protein 1 (Drp1) phosphorylation and its subsequent translocation to mitochondria. In contrast, TP receptor overexpression and application of TP receptor agonist U46619 in these podocytes showed the opposite effect on mitochondrial fission and podocyte injury. Furthermore, treatment with Y27632, an inhibitor of Rho-associated kinase1 (ROCK1), significantly blunted more fragmented mitochondria and reduced podocyte injuries in podocytes with TP receptor overexpression or after U46619 treatment. Finally, pharmacological inhibition of Drp1 alleviated excessive mitochondrial fragmentation and podocyte damage in TP receptor overexpressing podocytes. Our data suggests that increased expression of the TP receptor can occur in a human cultured podocyte cell line and in podocytes derived from streptozotocin (STZ)-induced diabetic mice, which contributes to mitochondrial excessive fission and podocyte injury via ROCK1-Drp1 signaling.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Mitochondrial Diseases , Podocytes , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/therapeutic use , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Dynamins/metabolism , Glucose/metabolism , Glucose/pharmacology , Humans , Mice , Mitochondrial Diseases/metabolism , Mitochondrial Dynamics , Prostaglandins/metabolism , Prostaglandins/pharmacology , Prostaglandins/therapeutic use , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin/therapeutic use , Receptors, Thromboxane/metabolism , Receptors, Thromboxane/therapeutic use , Streptozocin , Thromboxanes/metabolism , Thromboxanes/pharmacology , Thromboxanes/therapeutic use , rho-Associated Kinases/metabolism
3.
Am J Respir Crit Care Med ; 206(5): 596-607, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35728047

ABSTRACT

Rationale: Although persistent fibroblast activation is a hallmark of idiopathic pulmonary fibrosis (IPF), mechanisms regulating persistent fibroblast activation in the lungs have not been fully elucidated. Objectives: On the basis of our observation that lung fibroblasts express TBXA2R (thromboxane-prostanoid receptor) during fibrosis, we investigated the role of TBXA2R signaling in fibrotic remodeling. Methods: We identified TBXA2R expression in lungs of patients with IPF and mice and studied primary mouse and human lung fibroblasts to determine the impact of TBXA2R signaling on fibroblast activation. We used TBXA2R-deficient mice and small-molecule inhibitors to investigate TBXA2R signaling in preclinical lung fibrosis models. Measurements and Main Results: TBXA2R expression was upregulated in fibroblasts in the lungs of patients with IPF and in mouse lungs during experimental lung fibrosis. Genetic deletion of TBXA2R, but not inhibition of thromboxane synthase, protected mice from bleomycin-induced lung fibrosis, thereby suggesting that an alternative ligand activates profibrotic TBXA2R signaling. In contrast to thromboxane, F2-isoprostanes, which are nonenzymatic products of arachidonic acid induced by reactive oxygen species, were persistently elevated during fibrosis. F2-isoprostanes induced TBXA2R signaling in fibroblasts and mediated a myofibroblast activation profile due, at least in part, to potentiation of TGF-ß (transforming growth factor-ß) signaling. In vivo treatment with the TBXA2R antagonist ifetroban reduced profibrotic signaling in the lungs, protected mice from lung fibrosis in three preclinical models (bleomycin, Hermansky-Pudlak mice, and radiation-induced fibrosis), and markedly enhanced fibrotic resolution after bleomycin treatment. Conclusions: TBXA2R links oxidative stress to fibroblast activation during lung fibrosis. TBXA2R antagonists could have utility in treating pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Receptors, Thromboxane , Animals , Bleomycin/pharmacology , F2-Isoprostanes/metabolism , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Lung/metabolism , Mice , Mice, Inbred C57BL , Prostaglandins/metabolism , Receptors, Thromboxane/metabolism , Thromboxanes/metabolism , Transforming Growth Factor beta/metabolism
4.
Hypertension ; 79(7): 1361-1373, 2022 07.
Article in English | MEDLINE | ID: mdl-35477273

ABSTRACT

BACKGROUND: Abnormal accumulation of senescent cells in the vessel wall leads to a compromised vascular function contributing to vascular aging. Soluble DPP4 (dipeptidyl peptidase 4; sDPP4) secretion from visceral adipose tissue is enhanced in obesity, now considered a progeric condition. sDPP4 triggers vascular deleterious effects, albeit its contribution to vascular aging is unknown. We aimed to explore sDPP4 involvement in vascular aging, unraveling the molecular pathway by which sDPP4 acts on the endothelium. METHODS: Human endothelial cell senescence was assessed by senescence-associated ß-galactosidase assay, visualization of DNA damage, and expression of prosenescent markers, whereas vascular function was evaluated by myography over human dissected microvessels. In visceral adipose tissue biopsies from a cohort of obese patients, we explored several age-related parameters in vitro and ex vivo. RESULTS: By a common mechanism, sDPP4 triggers endothelial cell senescence and endothelial dysfunction in isolated human resistance arteries. sDPP4 activates the metabotropic receptor PAR2 (protease-activated receptor 2), COX-2 (cyclooxygenase 2) activity, and the production of TXA2 (thromboxane A2) acting over TP (thromboxane receptor) receptors (PAR2-COX-2-TP axis), leading to NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing 3) inflammasome activation. Obese patients exhibited impaired microarterial functionality in comparison to control nonobese counterparts. Importantly, endothelial dysfunction in obese patients positively correlated with greater expression of DPP4, prosenescent, and proinflammatory markers in visceral adipose tissue nearby the resistance arteries. Moreover, when DPP4 activity or sDPP4-induced prosenescent mechanism was blocked, endothelial dysfunction was restored back to levels of healthy subjects. CONCLUSIONS: These results reveal sDPP4 as a relevant mediator in early vascular aging and highlight its capacity activating main proinflammatory mediators in the endothelium that might be pharmacologically tackled.


Subject(s)
Cyclooxygenase 2 , Dipeptidyl Peptidase 4 , Inflammasomes , Biomarkers/metabolism , Cellular Senescence , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dipeptidyl Peptidase 4/metabolism , Endothelial Cells/metabolism , Humans , Inflammasomes/metabolism , Inflammasomes/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/metabolism , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Receptors, Thromboxane/genetics , Receptors, Thromboxane/metabolism
5.
Pharmacol Res Perspect ; 10(3): e00952, 2022 06.
Article in English | MEDLINE | ID: mdl-35466586

ABSTRACT

The inhibitory effects of docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and linoleic acid (LA) on the contractions induced by five prostanoids and U46619 (a TP receptor agonist) were examined in guinea pig gastric fundus smooth muscle (GFSM). Tension changes were isometrically measured, and the mRNA expression of prostanoid receptors was measured by RT-qPCR. DHA and EPA significantly inhibited contractions induced by the prostanoids and U46619, whereas LA inhibited those induced by prostaglandin D2 and U46619. The mRNA expression levels of the prostanoid receptors were TP ≈ EP3  >> FP > EP1 . The inhibition by DHA, EPA, and LA was positively correlated with that by SQ 29,548 (a TP receptor antagonist) but not with that by L-798,106 (an EP3 receptor antagonist). DHA and EPA suppressed high KCl-induced contractions by 35% and 25%, respectively, and the contractions induced by the prostanoids and U46619 were suppressed by verapamil, a voltage-dependent Ca2+ channel (VDCC) inhibitor, by 40%-85%. Although LA did not suppress high KCl-induced contractions, it suppressed U46619-induced contractions in the presence of verapamil. However, LA did not show significant inhibitory effects on U46619-induced Ca2+ increases in TP receptor-expressing cells. In contrast, LA inhibited U46619-induced contractions in the presence of verapamil, which was also suppressed by SKF-96365 (a store-operated Ca2+ channel [SOCC] inhibitor). These findings suggest that the TP receptor and VDCC are targets of DHA and EPA to inhibit prostanoid-induced contractions of guinea pig GFSM, and SOCCs play a significant role in LA-induced inhibition of U46619-induced contractions.


Subject(s)
Docosahexaenoic Acids , Eicosapentaenoic Acid , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Calcium Channels, L-Type/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Gastric Fundus/metabolism , Guinea Pigs , Muscle, Smooth , Prostaglandins/metabolism , Prostaglandins/pharmacology , RNA, Messenger/metabolism , Receptors, Thromboxane/metabolism , Verapamil/metabolism , Verapamil/pharmacology
6.
Arterioscler Thromb Vasc Biol ; 42(4): 444-461, 2022 04.
Article in English | MEDLINE | ID: mdl-35236104

ABSTRACT

BACKGROUND: TP (thromboxane A2 receptor) plays an eminent role in the pathophysiology of endothelial dysfunction and cardiovascular disease. Moreover, its expression is reported to increase in the intimal layer of blood vessels of cardiovascular high-risk individuals. Yet it is unknown, whether TP upregulation per se has the potential to affect the homeostasis of the vascular endothelium. METHODS: We combined global transcriptome analysis, lipid mediator profiling, functional cell analyses, and in vivo angiogenesis assays to study the effects of endothelial TP overexpression or knockdown/knockout on the angiogenic capacity of endothelial cells in vitro and in vivo. RESULTS: Here we report that endothelial TP expression induces COX-2 (cyclooxygenase-2) in a Gi/o- and Gq/11-dependent manner, thereby promoting its own activation via the auto/paracrine release of TP agonists, such as PGH2 (prostaglandin H2) or prostaglandin F2 but not TxA2 (thromboxane A2). TP overexpression induces endothelial cell tension and aberrant cell morphology, affects focal adhesion dynamics, and inhibits the angiogenic capacity of human endothelial cells in vitro and in vivo, whereas TP knockdown or endothelial-specific TP knockout exerts opposing effects. Consequently, this TP-dependent feedback loop is disrupted by pharmacological TP or COX-2 inhibition and by genetic reconstitution of PGH2-metabolizing prostacyclin synthase even in the absence of functional prostacyclin receptor expression. CONCLUSIONS: Our work uncovers a TP-driven COX-2-dependent feedback loop and important effector mechanisms that directly link TP upregulation to angiostatic TP signaling in endothelial cells. By these previously unrecognized mechanisms, pathological endothelial upregulation of the TP could directly foster endothelial dysfunction, microvascular rarefaction, and systemic hypertension even in the absence of exogenous sources of TP agonists.


Subject(s)
Endothelial Cells , Receptors, Thromboxane , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/pharmacology , Endothelial Cells/metabolism , Feedback , Homeostasis , Humans , Receptors, Thromboxane/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Thromboxane A2/metabolism , Thromboxanes/metabolism , Thromboxanes/pharmacology
7.
Environ Sci Pollut Res Int ; 29(22): 33095-33105, 2022 May.
Article in English | MEDLINE | ID: mdl-35025049

ABSTRACT

Airborne fine particulate matter (PM2.5) is a major cardiovascular disease environmental risk factor. However, the underlying mechanism of action is not fully understood. Thromboxane is widely known as an important vasoconstrictor substance that binds to G-protein-coupled receptors (GPCR) in arteries and is involved in various cardiovascular diseases. This study examined the effect of PM2.5 on thromboxane A2 receptor (TP) in the mesenteric arteries and the underlying intracellular signal mechanisms (by focusing on the mitogen-activated protein kinase (MAPK) pathway). Rat mesenteric artery segments were exposed to PM2.5 in the presence of MAPK pathway inhibitors. The contractile reactivity of mesenteric arteries was analyzed using wire myography. The mRNA and protein expression of TP receptor and MAPK pathway molecules were detected by real-time PCR and Western blot. Mesenteric artery receptor localization was assessed by immunohistochemistry. The results showed that TP receptor-mediated maximum contraction response was achieved after exposing arteries to 1.0 µg/mL PM2.5 for 16 h (Emax: 228 ± 16% of K+). Moreover, inhibitor U0126 (ERK1/2 inhibitor), SB203580 (p38 inhibitor), and SP600125 (JNK inhibitor) depressed the increased TP receptor-mediated contractile responses (reduced rage were 17.9 ~ 59.6%). These inhibitors also decreased the increased mRNA expression and protein of the TP receptor induced by PM2.5 (reduced by more than 50% and 46%, respectively). The immunoreactivity of increased TP receptor expression was primarily localized in the cytoplasm. In addition, phosphorylation quantitative analysis showed that in the presence of MAPK inhibitors, the PM2.5-induced phosphorylation of ERK1/2, p38, and JNK protein increased by more than 30.0 ~ 130.3%. These results suggest that PM2.5 upregulates the TP receptor of rat mesenteric arteries through activation of the ERK1/2, p38, and JNK MAPK pathways.


Subject(s)
Mitogen-Activated Protein Kinases , Receptors, Thromboxane , Animals , Mitogen-Activated Protein Kinases/metabolism , Particulate Matter/chemistry , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Thromboxane/metabolism , Thromboxanes , Up-Regulation
8.
Hypertension ; 79(1): 104-114, 2022 01.
Article in English | MEDLINE | ID: mdl-34784723

ABSTRACT

12/15-LO (12/15-lipoxygenase), encoded by Alox15 gene, metabolizes arachidonic acid to 12(S)-HETE (12-hydroxyeicosatetraenoic acid). Macrophages are the major source of 12/15-LO among immune cells, and 12/15-LO plays a crucial role in development of hypertension. Global Alox15- or macrophage-deficient mice are resistant to Ang II (angiotensin II)-induced hypertension. This study tests the hypothesis that macrophage 12(S)-HETE contributes to Ang II-mediated arterial constriction and thus to development of Ang II-induced hypertension. Ang II constricted isolated abdominal aortic and mesenteric arterial rings. 12(S)-HETE (100 nmol/L) alone was without effect; however, it significantly enhanced Ang II-induced constriction. The presence of wild-type macrophages also enhanced the Ang II-induced constriction, while Alox15-/- macrophages did not. Using this model, pretreatment of aortic rings with inhibitors, receptor agonists/antagonists, or removal of the endothelium, systematically uncovered an endothelium-mediated, Ang II receptor-2-mediated and superoxide-mediated enhancing effect of 12(S)-HETE on Ang II constrictions. The role of superoxide was confirmed using aortas from p47phox-/- mice where 12(S)-HETE failed to enhance constriction to Ang II. In cultured arterial endothelial cells, 12(S)-HETE increased the production of superoxide, and 12(S)-HETE or Ang II increased the production of an isothromboxane-like metabolite. A TP (thromboxane receptor) antagonist inhibited 12(S)-HETE enhancement of Ang II constriction. Both Ang II-induced hypertension and the enhancing effect of 12(S)-HETE on Ang II contractions were eliminated by a BLT2 (leukotriene B4 receptor-2) antagonist. These results outline a mechanism where the macrophage 12/15-LO pathway enhances the action of Ang II. 12(S)-HETE, acting on the BLT2, contributes to the hypertensive action of Ang II in part by promoting endothelial synthesis of a superoxide-derived TP agonist.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Angiotensin II/pharmacology , Aorta/drug effects , Mesenteric Arteries/drug effects , Receptors, Leukotriene B4/metabolism , Receptors, Thromboxane/metabolism , Animals , Aorta/metabolism , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Male , Mesenteric Arteries/metabolism , Mice , Mice, Knockout , Superoxides/metabolism
9.
Life Sci ; 287: 120130, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34767807

ABSTRACT

AIMS: We examined the potential stimulatory effects of U46619 (a prostanoid TP receptor agonist) and five prostanoids on the contractile activities of urinary bladder smooth muscle (UBSM), focusing on the role of the TP receptor and its associated Ca2+ influx routes to understand the roles of prostanoids in the regulation of UB contractile activity. MAIN METHODS: Changes in the basal tone and spontaneous contractile activity (amplitude and frequency) of isolated guinea pig UBSM were measured isotonically. The presence of TP receptors in UBSM was examined by RT-qPCR and immunofluorescence. KEY FINDINGS: U46619, prostaglandin (PG) E2, PGF2α, and PGA2 enhanced UBSM basal tone and spontaneous contractile activities, which were measured as amplitudes and frequencies. The enhancing effects of U46619 were completely suppressed by SQ 29,548 (a TP receptor antagonist), which also partially suppressed the stimulating effects of other prostanoids. The expression of TP receptors in UBSMs was verified at the mRNA and protein level. The enhancing effects of U46619 completely disappeared in Ca2+-free solution. U46619-enhanced basal tone was completely suppressed by verapamil, an inhibitor of voltage-dependent Ca2+ channels (VDCCs), and verapamil strongly decreased the spontaneous contraction frequency. The spontaneous contractions remaining in the presence of verapamil were strongly suppressed by SKF-96365 (an inhibitor of receptor-operated Ca2+ channels (ROCCs)/store-operated Ca2+ channels (SOCCs)), but not by LOE-908 (an inhibitor of ROCCs). SIGNIFICANCE: Prostanoids can enhance UBSM contractile activities and thus may be endogenous candidates for induction of detrusor overactivity. The TP receptor and TP-receptor-activated VDCCs/SOCCs are key molecules responsible for these effects.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Receptors, Thromboxane/metabolism , Urinary Bladder/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/therapeutic use , Animals , Guinea Pigs , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Organ Culture Techniques , Receptors, Thromboxane/agonists , Urinary Bladder/drug effects , Urinary Bladder Diseases/drug therapy , Urinary Bladder Diseases/metabolism , Vasoconstrictor Agents/pharmacology , Vasoconstrictor Agents/therapeutic use
10.
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
11.
FASEB J ; 35(10): e21941, 2021 10.
Article in English | MEDLINE | ID: mdl-34559928

ABSTRACT

Thromboxane receptor (TP) mediates nasal obstruction, a typical symptom of allergic rhinitis. Since it has been reported that several types of eicosanoids, such as non-enzymatic oxidation product of arachidonic acid isoprostane, act as a TP ligand, there is a possibility that some other eicosanoids contribute to the TP-mediated nasal obstruction. The aim of this study is to investigate the mechanisms of TP-mediated nasal obstruction. Intranasal challenges of ovalbumin (OVA) induced nasal obstruction in mice. Pharmacological blockade of TP receptor but not thromboxane A2 synthase inhibited OVA-induced nasal obstruction. Simultaneous analysis of eicosanoids in nasal lavage fluid and the responses in trans-endothelial resistance suggested that 8-iso-prostaglandin E2 (PGE2 ) can be a candidate for TP ligand. Intranasal challenge of 8-iso-PGE2 induced vascular hyperpermeability and nasal obstruction in TP receptor-dependent manner. Wholemount immunostaining of nasal septum mucosa revealed that 8-iso-PGE2 increased plasma leakage accompanied by distention of venous sinusoids. This study shows that 8-iso-PGE2 is a contributor in TP-mediated nasal obstruction in mice.


Subject(s)
Dinoprostone/analogs & derivatives , Disease Models, Animal , Isoprostanes/pharmacology , Nasal Obstruction/chemically induced , Nasal Obstruction/complications , Receptors, Thromboxane/metabolism , Rhinitis, Allergic/complications , Rhinitis, Allergic/metabolism , Administration, Intranasal , Animals , Capillary Permeability/drug effects , Dinoprostone/administration & dosage , Dinoprostone/pharmacology , Female , Isoprostanes/administration & dosage , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects
12.
FASEB J ; 35(9): e21877, 2021 09.
Article in English | MEDLINE | ID: mdl-34449098

ABSTRACT

Although commonly thought to produce prostacyclin (prostaglandin I2 ; PGI2 ) that evokes vasodilatation and protects vessels from the development of diseases, the endothelial cyclooxygenase (COX)-mediated metabolism has also been found to release substance(s) called endothelium-derived contracting factor(s) (EDCF) that causes endothelium-dependent contraction and implicates in endothelial dysfunction of disease conditions. Various mechanisms have been proposed for the process; however, the major endothelial COX metabolite PGI2 , which has been classically considered to activate the I prostanoid receptor (IP) that mediates vasodilatation and opposes the effects of thromboxane (Tx) A2 produced by COX in platelets, emerges as a major EDCF in health and disease conditions. Our recent studies from genetically altered mice further suggest that vasomotor reactions to PGI2 are collectively modulated by IP, the vasoconstrictor Tx-prostanoid receptor (TP; the prototype receptor of TxA2 ) and E prostanoid receptor-3 (EP3; a vasoconstrictor receptor of PGE2 ) although with differences in potency and efficacy; a contraction to PGI2 reflects activities of TP and/or EP3 outweighing that of the concurrently activated IP. Here, we discuss the history of endothelium-dependent contraction, evidences that support the above hypothesis, proposed mechanisms for the varied reactions to endothelial PGI2 synthesis as well as the relation of its dilator activity to the effect of another NO-independent vasodilator mechanism, the endothelium-derived hyperpolarizing factor. Also, we address the possible pathological and therapeutic implications as well as questions remaining to be resolved or limitations of our above findings obtained from genetically altered mouse models.


Subject(s)
Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Vasoconstriction/physiology , Animals , Endothelium, Vascular/drug effects , Humans , Mice , Prostaglandins/metabolism , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane/metabolism , Thromboxanes/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasomotor System/drug effects , Vasomotor System/metabolism
13.
Eur J Pharmacol ; 909: 174435, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34416239

ABSTRACT

Hypertension is one of the most common complications of chronic kidney disease (CKD). Some research has indicated that changes in large artery function especially caused by thromboxane A2 (TXA2) may be a novel factor acting to induce hypertension in CKD. We studied the 5/6 nephrectomy rat model and measured serum levels of creatinine (Cr), calcium (Ca), phosphorus (P), TXA2-stable metabolites (thromboxane B2, TXB2), and caudal artery pressure after nephrectomy. The tension variations in thoracic aortas were measured after stimulating by vasoconstrictor/vasodilator using the cumulative concentration administration method and then tested the expression of TXA2 receptors in the thoracic aortas through western blots. The CKD rats developed uremia, electrolyte imbalances,and hypertension. They also exhibited a significant increase in TXB2 concentration. The aortic rings of CKD rats showed an increased contraction response to U46619 (a TXA2 analogue) and the expression of TXA2 receptors also enhanced. In the meanwhile, the diastolic function decreased in the CKD group. Our results demonstrate that the impairment of artery contractile function caused by the increase of TXA2 receptors on the wall of aortic rings may be involved in hypertension in CKD rats.


Subject(s)
Hypertension/pathology , Receptors, Thromboxane/metabolism , Renal Insufficiency, Chronic/complications , Thromboxane A2/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Disease Models, Animal , Humans , Hypertension/etiology , Hypertension/physiopathology , Male , Rats , Receptors, Thromboxane/analysis , Thromboxane A2/analysis , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
14.
Curr Issues Mol Biol ; 43(1): 79-92, 2021 May 07.
Article in English | MEDLINE | ID: mdl-34066967

ABSTRACT

BACKGROUND: Vitamin D deficiency (VDD) may be considered an independent cardiovascular (CV) risk factor, and it is well known that CV risk is higher in males. Our goal was to investigate the pharmacological reactivity and receptor expression of intramural coronary artery segments of male rats in cases of different vitamin D supply. METHODS: Four-week-old male Wistar rats were divided into a control group (n = 11) with optimal vitamin D supply (300 IU/kgbw/day) and a VDD group (n = 11, <0.5 IU/kgbw/day). After 8 weeks of treatment, intramural coronary artery segments were microprepared, their pharmacological reactivity was examined by in vitro microangiometry, and their receptor expression was investigated by immunohistochemistry. RESULTS: Thromboxane A2 (TXA2)-agonist induced reduced vasoconstriction, testosterone (T) and 17-ß-estradiol (E2) relaxations were significantly decreased, a significant decrease in thromboxane receptor (TP) expression was shown, and the reduction in estrogen receptor-α (ERα) expression was on the border of significance in the VDD group. CONCLUSIONS: VD-deficient male coronary arteries showed deteriorated pharmacological reactivity to TXA2 and sexual steroids (E2, T). Insufficient vasoconstrictor capacity was accompanied by decreased TP receptor expression, and vasodilator impairments were mainly functional. The decrease in vasoconstrictor and vasodilator responses results in narrowed adaptational range of coronaries, causing inadequate coronary perfusion that might contribute to the increased CV risk in VDD.


Subject(s)
Arterioles/pathology , Coronary Artery Disease/pathology , Estradiol/pharmacology , Testosterone/pharmacology , Thromboxane A2/pharmacology , Vitamin D Deficiency/complications , Androgens/pharmacology , Animals , Arterioles/metabolism , Coronary Artery Disease/drug therapy , Coronary Artery Disease/etiology , Coronary Artery Disease/metabolism , Disease Models, Animal , Estrogens/pharmacology , Male , Rats , Rats, Wistar , Receptors, Thromboxane/metabolism , Vasoconstriction , Vitamin D Deficiency/metabolism , Vitamin D Deficiency/pathology
15.
Pharmacol Res ; 170: 105744, 2021 08.
Article in English | MEDLINE | ID: mdl-34182131

ABSTRACT

Enhanced platelet activation has been reported in patients with essential hypertension and heart failure. The possible contribution of platelet-derived thromboxane (TX)A2 in their pathophysiology remains unclear. We investigated the systemic TXA2 biosynthesis in vivo and gene expression of its receptor TP in 22 essential hypertension patients and a mouse model of salt-sensitive hypertension. The contribution of platelet TXA2 biosynthesis on enhanced blood pressure (BP) and overload-induced cardiac fibrosis was explored in mice by treating with low-dose Aspirin, resulting in selective inhibition of platelet cyclooxygenase (COX)-1-dependent TXA2 generation. In essential hypertensive patients, systemic biosynthesis of TXA2 [assessed by measuring its urinary metabolites (TXM) reflecting predominant platelet source] was enhanced together with higher gene expression of circulating leukocyte TP and TGF-ß, vs. normotensive controls. Similarly, in hypertensive mice with prostacyclin (PGI2) receptor (IP) deletion (IPKO) fed with a high-salt diet, enhanced urinary TXM, and left ventricular TP overexpression were detected vs. normotensive wildtype (WT) mice. Increased cardiac collagen deposition and profibrotic gene expression (including TGF-ß) was found. Low-dose Aspirin administration caused a selective inhibition of platelet TXA2 biosynthesis and mitigated enhanced blood pressure, cardiac fibrosis, and left ventricular profibrotic gene expression in IPKO but not WT mice. Moreover, the number of myofibroblasts and extravasated platelets in the heart was reduced. In cocultures of human platelets and myofibroblasts, platelet TXA2 induced profibrotic gene expression, including TGF-ß1. In conclusion, our results support tailoring low-dose Aspirin treatment in hypertensive patients with unconstrained TXA2/TP pathway to reduce blood pressure and prevent early cardiac fibrosis.


Subject(s)
Antifibrotic Agents/pharmacology , Antihypertensive Agents/pharmacology , Aspirin/pharmacology , Blood Platelets/drug effects , Blood Pressure/drug effects , Cardiomyopathies/prevention & control , Essential Hypertension/drug therapy , Myocytes, Cardiac/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thromboxane A2/blood , Adult , Animals , Biomarkers/blood , Blood Platelets/metabolism , Cardiomyopathies/blood , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Essential Hypertension/blood , Essential Hypertension/complications , Essential Hypertension/physiopathology , Female , Fibrosis , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/metabolism , Receptors, Thromboxane/metabolism
16.
Platelets ; 32(5): 618-625, 2021 Jul 04.
Article in English | MEDLINE | ID: mdl-32619120

ABSTRACT

The ADP receptor P2Y12, the thromboxane A2 receptor (TXA2R) and the C-type lectin-like receptor 2 (CLEC-2) mediate platelet activation by different mechanisms. Only little is known about the expression of the receptors in human megakaryopoiesis. Our study aimed to establish a flow cytometry (FC) method for the measurement of P2Y12, TXA2R, and CLEC-2 on platelets of healthy donors and to monitor receptor expression in ex vivo megakaryopoiesis. We determined mean fluorescence intensity (MFI) values of FITC, PE, or APC labeled antibodies binding to the receptors on platelets of 90 healthy donors. For cord blood-derived megakaryopoiesis (CBMK) differentiation of CD34+ cells was induced by IL-3, SCF, and TPO. At 6 time points between day 0 and day 21 of cell culture the MFI values for CD34, CD41, CD61, P2Y12, TXA2R, and CLEC-2 were measured. Quantitative PCR was used for relative quantification of the corresponding mRNA. Transcription factor (TF) binding sites were predicted by in silico analysis of the genes. Platelets showed expectable high MFI values for the platelet marker CD41 (13,716 median MFI). Lower MFI was found for P2Y12 (2,847 median MFI) and CLEC-2 (1,211 median MFI), whereas, binding of the TXA2R antibody revealed even higher values (21,458 median MFI) than CD41. In CBMK the CD34+ cells were negative for P2Y12, TXA2R, and CLEC-2 at day 0. A maximum of 21-fold and 6-fold increase of P2Y12 and TXA2R MFI values, respectively, was found on day 14 to 17. MFI for CLEC-2 increased by 58-fold within the first week and reached a maximum of 1,572-fold increase within the first two weeks of CBMK. Very similar results were obtained on the RNA level. The differential regulation of receptor expression in CBMK was further supported by significant differences in the numbers and types of TF binding sites. P2Y12 and TXA2R, both upregulated only to a low extent in CBMK, probably, are dispensable for megakaryopoiesis. Furthermore, we speculate that CLEC-2 strongly upregulated in early CMBK is important for megakaryopoiesis.


Subject(s)
Blood Platelets/metabolism , Fetal Blood/metabolism , Lectins, C-Type/metabolism , Megakaryocytes/metabolism , Platelet Activation/immunology , Receptors, Purinergic P2/metabolism , Receptors, Thromboxane/metabolism , Transcription Factors/metabolism , Fetal Blood/cytology , Humans
17.
Eur J Pharmacol ; 893: 173828, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33347824

ABSTRACT

This study was to determine how endothelium-dependent contractions (EDCs) change in iliac arteries of Wistar-Kyoto (WKYs) and spontaneously hypertensive rats (SHRs) during the transition from adolescence to adulthood and the underlying mechanism(s). We also aimed to elucidate effects of L-798106, an EP3 receptor antagonist, on EDCs and the blood pressure increase in adolescent SHRs. Blood vessels were isolated for functional and biochemical analyses. EDCs were comparable in adolescent iliac arteries of both strains, and contractions to ACh, prostacyclin (PGI2), the EP3 receptor agonist sulprostone and the TP receptor agonist U46619 in adult vessels were less prominent compared with those in the adolescents, while the attenuation of vasoconstrictions to ACh, PGI2 or U46619 with age was to a lesser extent in SHRs. PGI2 production was decreased to a similar level in adult arteries. TP and EP3 expressions were downregulated in adult vessels, whereas the extent of TP downregulation was less in SHRs. L-798106 partially suppressed the vasoconstrictions to U46619 and attenuated EDCs to a greater extent than SQ29548, and administration of L-798106 blunted the blood pressure increase with age in prehypertensive SHRs. These results demonstrate the comparable EDCs in iliac arteries of the adolescents are decreased in the adults, but relatively larger EDCs in adult SHRs can be a reflection of differential downregulation of TP and EP3 receptors during the transition from adolescence to adulthood. Also, our data suggest that blockade of both TP and EP3 receptors starting from the prehypertensive stage suppresses EDCs and the development of hypertension in SHRs.


Subject(s)
Blood Pressure , Endothelium, Vascular/metabolism , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Thromboxane/metabolism , Vasoconstriction , Age Factors , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Disease Models, Animal , Down-Regulation , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Hypertension/genetics , Hypertension/physiopathology , Hypertension/prevention & control , Iliac Artery/metabolism , Iliac Artery/physiopathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/genetics , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/genetics , Signal Transduction , Vasoconstriction/drug effects
18.
FASEB J ; 34(12): 16105-16116, 2020 12.
Article in English | MEDLINE | ID: mdl-33047360

ABSTRACT

Vasomotor reactions of prostacyclin (prostaglandin I2 ; PGI2 ) can be collectively modulated by thromboxane prostanoid receptor (TP), E-prostanoid receptor-3 (EP3), and the vasodilator I prostanoid receptor (IP). This study aimed to determine the direct effect of PGI2 on renal arteries and/or the whole renal vasculature and how each of these receptors is involved. Experiments were performed on vessels or perfused kidneys of wild-type mice and/or mice with deficiency in TP (TP-/- ) and/or EP3. Here we show that PGI2 did not evoke relaxation, but instead resulted in contraction of main renal arteries (from ~0.001-0.01 µM) or reduction of flow in perfused kidneys (from ~1 µM); either of them was reversed into a dilator response in TP-/- /EP3-/- counterparts. Also, we found that in renal arteries although it has a lesser effect than TP-/- on the maximal contraction to PGI2 (10 µM), EP3-/- but not TP-/- resulted in relaxation to the prostanoid at 0.01-1 µM. Meanwhile, TP-/- only significantly reduced the contractile activity evoked by PGI2 at ≥0.1 µM. These results demonstrate that PGI2 may evoke an overall vasoconstrictor response in the mouse renal vasculature, reflecting activities of TP and EP3 outweighing that of the vasodilator IP. Also, our results suggest that EP3, on which PGI2 can have a potency similar to that on IP, plays a major role in the vasoconstrictor effect of the prostanoid of low concentrations (≤1 µM), while TP, on which PGI2 has a lower potency but higher efficacy, accounts for a larger part of its maximal contractile activity.


Subject(s)
Epoprostenol/pharmacology , Kidney/drug effects , Prostaglandins/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Thromboxane/metabolism , Renal Artery/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Prostaglandins I/pharmacology , Renal Artery/metabolism , Vasoconstriction/drug effects
19.
Mol Cancer Ther ; 19(12): 2454-2464, 2020 12.
Article in English | MEDLINE | ID: mdl-33033174

ABSTRACT

Although new drug discoveries are revolutionizing cancer treatments, repurposing existing drugs would accelerate the timeline and lower the cost for bringing treatments to cancer patients. Our goal was to repurpose CPI211, a potent and selective antagonist of the thromboxane A2-prostanoid receptor (TPr), a G-protein-coupled receptor that regulates coagulation, blood pressure, and cardiovascular homeostasis. To identify potential new clinical indications for CPI211, we performed a phenome-wide association study (PheWAS) of the gene encoding TPr, TBXA2R, using robust deidentified health records and matched genomic data from more than 29,000 patients. Specifically, PheWAS was used to identify clinical manifestations correlating with a TBXA2R single-nucleotide polymorphism (rs200445019), which generates a T399A substitution within TPr that enhances TPr signaling. Previous studies have correlated 200445019 with chronic venous hypertension, which was recapitulated by this PheWAS analysis. Unexpectedly, PheWAS uncovered an rs200445019 correlation with cancer metastasis across several cancer types. When tested in several mouse models of metastasis, TPr inhibition using CPI211 potently blocked spontaneous metastasis from primary tumors, without affecting tumor cell proliferation, motility, or tumor growth. Further, metastasis following intravenous tumor cell delivery was blocked in mice treated with CPI211. Interestingly, TPr signaling in vascular endothelial cells induced VE-cadherin internalization, diminished endothelial barrier function, and enhanced transendothelial migration by tumor cells, phenotypes that were decreased by CPI211. These studies provide evidence that TPr signaling promotes cancer metastasis, supporting the study of TPr inhibitors as antimetastatic agents and highlighting the use of PheWAS as an approach to accelerate drug repurposing.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Repositioning , Genome-Wide Association Study/methods , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/genetics , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Metastasis , Phenotype , Polymorphism, Single Nucleotide , Receptors, Thromboxane/metabolism
20.
Exp Physiol ; 105(11): 1840-1854, 2020 11.
Article in English | MEDLINE | ID: mdl-32954541

ABSTRACT

NEW FINDINGS: What is the central question of this study? Do endoperoxide 4 and thromboxane A2 receptors, which are receptors for cyclooxygenase products of arachidonic metabolism, on thin fibre muscle afferents play a role in the chronic mechanoreflex sensitization present in rats with heart failure with reduced ejection fraction (HF-rEF)? What is the main finding and its importance? The data do not support a role for endoperoxide 4 receptors or thromboxane A2 receptors in the chronic mechanoreflex sensitization in HF-rEF rats. ABSTRACT: We investigated the role of cyclooxygenase metabolite-associated endoperoxide 4 receptors (EP4-R) and thromboxane A2 receptors (TxA2 -R) on thin fibre muscle afferents in the chronic mechanoreflex sensitization in rats with myocardial infarction-induced heart failure with reduced ejection fraction (HF-rEF). We hypothesized that injection of either the EP4-R antagonist L-161,982 (1 µg) or the TxA2 -R antagonist daltroban (80 µg) into the arterial supply of the hindlimb would reduce the increase in blood pressure and renal sympathetic nerve activity (RSNA) evoked in response to 30 s of static hindlimb skeletal muscle stretch (a model of isolated mechanoreflex activation) in decerebrate, unanaesthetized HF-rEF rats but not sham-operated control rats (SHAM). Ejection fraction was significantly reduced in HF-rEF (45 ± 11%) compared to SHAM (83 ± 6%; P < 0.01) rats. In SHAM and HF-rEF rats, we found that the EP4-R antagonist had no effect on the peak increase in mean arterial pressure (peak ΔMAP SHAM n = 6, pre: 15 ± 7, post: 15 ± 9, P = 0.99; HF-rEF n = 9, pre: 30 ± 11, post: 32 ± 15 mmHg, P = 0.84) or peak increase in RSNA (peak ΔRSNA SHAM pre: 33 ± 14, post: 47 ± 31%, P = 0.94; HF-rEF, pre: 109 ± 47, post: 139 ± 150%, P = 0.76) response to stretch. Similarly, in SHAM and HF-rEF rats, we found that the TxA2 -R antagonist had no effect on the peak ΔMAP (SHAM n = 7, pre: 13 ± 7, post: 19 ± 14, P = 0.15; HF-rEF n = 14, pre: 24 ± 13, post: 21 ± 13 mmHg, P = 0.47) or peak ΔRSNA (SHAM pre: 52 ± 43, post: 57 ± 67%, P = 0.94; HF-rEF, pre: 108 ± 93, post: 88 ± 72%, P = 0.30) response to stretch. The data do not support a role for EP4-Rs or TxA2 -Rs in the chronic mechanoreflex sensitization in HF-rEF.


Subject(s)
Heart Failure , Muscle Contraction , Animals , Blood Pressure , Heart Failure/drug therapy , Heart Failure/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Thromboxane/metabolism , Reflex , Thromboxanes/metabolism , Thromboxanes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL