Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Hypertension ; 65(5): 1055-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25733239

ABSTRACT

Cardiovascular disease is frequent in chronic kidney disease and has been related to angiotensin II, endothelin-1 (ET-1), thromboxane A2, and reactive oxygen species (ROS). Because activation of thromboxane prostanoid receptors (TP-Rs) can generate ROS, which can generate ET-1, we tested the hypothesis that chronic kidney disease induces cyclooxygenase-2 whose products activate TP-Rs to enhance ET-1 and ROS generation and contractions. Mesenteric resistance arterioles were isolated from C57/BL6 or TP-R+/+ and TP-R-/- mice 3 months after SHAM-operation (SHAM) or surgical reduced renal mass (RRM, n=6/group). Microvascular contractions were studied on a wire myograph. Cellular (ethidium: dihydroethidium) and mitochondrial (mitoSOX) ROS were measured by fluorescence microscopy. Mice with RRM had increased excretion of markers of oxidative stress, thromboxane, and microalbumin; increased plasma ET-1; and increased microvascular expression of p22(phox), cyclooxygenase-2, TP-Rs, preproendothelin and endothelin-A receptors, and increased arteriolar remodeling. They had increased contractions to U-46,619 (118 ± 3 versus 87 ± 6, P<0.05) and ET-1 (108 ± 5 versus 89 ± 4, P<0.05), which were dependent on cellular and mitochondrial ROS, cyclooxygenase-2, and TP-Rs. RRM doubled the ET-1-induced cellular and mitochondrial ROS generation (P<0.05). TP-R-/- mice with RRM lacked these abnormal structural and functional microvascular responses and lacked the increased systemic and the increased microvascular oxidative stress and circulating ET-1. In conclusion, RRM leads to microvascular remodeling and enhanced ET-1-induced cellular and mitochondrial ROS and contractions that are mediated by cyclooxygenase-2 products activating TP-Rs. Thus, TP-Rs can be upstream from enhanced ROS, ET-1, microvascular remodeling, and contractility and may thereby coordinate vascular dysfunction in chronic kidney disease.


Subject(s)
Arterioles/physiopathology , Endothelin-1/biosynthesis , Kidney/blood supply , Oxidative Stress , Receptors, Thromboxane/biosynthesis , Renal Insufficiency, Chronic/physiopathology , Vasoconstriction , Animals , Arterioles/metabolism , Disease Models, Animal , Endothelin-1/genetics , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , RNA/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Thromboxane/genetics , Renal Circulation , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Vascular Remodeling
2.
Prostaglandins Other Lipid Mediat ; 96(1-4): 41-4, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21983220

ABSTRACT

We have previously found increased expression of thromboxane synthase (TXAS) and thromboxane receptor (TP) beta isoform in the tissues of patients with bladder cancer. Studies in cell lines and mice have indicated a potential significant role of the thromboxane signaling pathway in the pathogenesis of human bladder cancer. This study was designed to determine if the changes observed in the tissues of patients with bladder cancer were mirrored by changes in the urine of these patients. We found increased levels of thromboxane B(2) (TXB(2)) the major metabolite of TXAS and increased levels of the TPß receptor. These results raised the possibility that patients with bladder cancer may be followed for progression or remission of their disease by quantitation of these substances in their urine.


Subject(s)
Biomarkers, Tumor/urine , Protein Isoforms/urine , Receptors, Thromboxane/biosynthesis , Signal Transduction/genetics , Thromboxane B2/urine , Thromboxane-A Synthase/urine , Urinary Bladder Neoplasms/urine , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Humans , Protein Isoforms/genetics , Real-Time Polymerase Chain Reaction , Receptors, Thromboxane/genetics , Thromboxane-A Synthase/genetics , United States , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/pathology
3.
Atherosclerosis ; 208(2): 376-81, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19735918

ABSTRACT

OBJECTIVES: Prostaglandins, such as thromboxane A(2) (TxA(2)) and prostacyclin (PGI(2)), are bioactive lipid mediators that are implicated in the pathogenesis of atherosclerosis. In the current study, we tested the hypothesis that thromboxane synthase (TXAS), prostacyclin synthase (PGIS) and thromboxane receptor (TP) are expressed within the atherosclerotic lesion. METHODS: Atherosclerotic aorta segments were obtained from low-density lipoprotein receptor deficient (LDL r-KO) mice on a high fat diet. Expression levels of TXAS, PGIS and TP were evaluated by real-time quantitative reverse transcription PCR, and immunohistochemistry; TxA(2) and PGI(2) biosynthesis was also assayed. RESULTS: After 8 weeks on the fat diet, aortic arches from LDL r-KO mice showed a significant increase in PGIS, TXAS, TP mRNA, TxA(2) and PGI(2) levels, when compared with controls. By contrast, after 16 weeks on the high fat diet PGIS and PGI(2) were significantly reduced, whereas TXAS and TP message and protein and TxA(2) levels were further and significantly increased in the atherosclerotic tissues when compared with the 8-week group. These changes correlated with the cellular composition of the atherosclerotic lesions. CONCLUSIONS: TXAS, PGIS and TP are all present within the atherosclerotic lesion areas, their levels change during progression of atherogenesis and contribute to TxA(2) and PGI(2) formation.


Subject(s)
Atherosclerosis/metabolism , Cytochrome P-450 Enzyme System/biosynthesis , Gene Expression Regulation, Enzymologic , Intramolecular Oxidoreductases/biosynthesis , Receptors, Thromboxane/biosynthesis , Thromboxane-A Synthase/biosynthesis , Animals , Atherosclerosis/enzymology , Atherosclerosis/pathology , Dietary Fats , Immunohistochemistry/methods , Lipids/chemistry , Male , Mice , Mice, Knockout , Prostaglandins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thromboxanes/metabolism
4.
J Thorac Cardiovasc Surg ; 139(4): 1033-40, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19660281

ABSTRACT

OBJECTIVE: We evaluated roles of serotonin 1B and 2A receptors, thromboxane synthase and receptor, and phospholipases A(2) and C in response to cardiopulmonary bypass. METHODS: Patients' atrial tissues were harvested before and after cardiopulmonary bypass with cardioplegia (n = 13). Coronary microvessels were assessed for vasoactive response to serotonin with and without inhibitors of serotonin 1B and 2A receptors and phospholipases A(2) and C. Expressions of serotonin receptor messenger RNA were determined with reverse transcriptase polymerase chain reaction. Expressions of serotonin receptors and thromboxane A(2) receptor and synthase proteins were determined with immunoblotting and immunohistochemistry. RESULTS: Microvessel exposure to serotonin elicited 7.3% +/- 2% relaxation before bypass, changing to contraction of -19.2% +/- 2% after bypass (P <.001). Additions of specific serotonin 1B receptor antagonist and inhibitor of phospholipase A(2) resulted in significantly decreased contraction, -8.6% +/- 1% (P < .001) and 2.8% +/- 3% (P = .001), respectively. Serotonin 1B receptor messenger RNA expression increased 1.82 +/- 0.34-fold after bypass (p = .044); serotonin 2A receptor messenger RNA expression did not change. Serotonin 1B but not 2A receptor protein expression increased after bypass by 1.35 +/- 0.7-fold (P = .0413). Thromboxane synthase and receptor expressions were unchanged after bypass. Serotonin 1B receptor increased mainly in arterial smooth muscle. There were no appreciable differences in arterial expressions of thromboxane synthase or receptor. CONCLUSIONS: Serotonin-induced vascular dysfunction after cardiopulmonary bypass with cardioplegic arrest may be mediated by increased expression of serotonin 1B receptor and subsequent phospholipase A(2) activation in myocardial coronary smooth muscle.


Subject(s)
Coronary Artery Bypass/adverse effects , Coronary Vessels/metabolism , Heart Atria/metabolism , Microcirculation/physiology , Receptors, Serotonin/biosynthesis , Vascular Diseases/physiopathology , Aged , Cardiopulmonary Bypass/adverse effects , Heart Arrest, Induced/adverse effects , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , Phospholipases A2/biosynthesis , Postoperative Period , Receptor, Serotonin, 5-HT1B/biosynthesis , Receptor, Serotonin, 5-HT2A/biosynthesis , Receptors, Thromboxane/biosynthesis , Thromboxane-A Synthase/biosynthesis , Type C Phospholipases/biosynthesis , Vascular Diseases/etiology , Vasoconstriction/physiology
5.
Am J Physiol Heart Circ Physiol ; 290(1): H295-303, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16199482

ABSTRACT

Previous reports showed that 17beta-estradiol implants attenuate in vivo coronary hyperreactivity (CH), characterized by long-duration vasoconstrictions (in coronary angiographic experiments), in menopausal rhesus monkeys. Prolonged Ca2+ contraction signals that correspond with CH in coronary vascular muscle cells (VMC) to the same dual-constrictor stimulus, serotonin + the thromboxane analog U-46619, in estrogen-deprived VMC were suppressed by >72 h in 17beta-estradiol. The purpose of this study was to test whether an endogenous estrogen metabolite with estrogen receptor-beta (ER-beta) binding activity, estriol (E3), suppresses in vivo and in vitro CH. E3 treatment in vivo for 4 wk significantly attenuated the angiographically evaluated vasoconstrictor response to intracoronary serotonin + U-46619 challenge. In vitro treatment of rhesus coronary VMC for >72 h with nanomolar E3 attenuated late Ca2+ signals. This reduction of late Ca2+ signals also appeared after >72 h of treatment with subnanomolar 5alpha-androstane-3beta,17beta-diol (3beta-Adiol), an endogenous dihydrotestosterone metabolite with ER-beta binding activity. R,R-tetrahydrochrysene, a selective ER-beta antagonist, significantly blocked the E3- and 3beta-Adiol-mediated attenuation of late Ca2+ signal increases. ER-beta and thromboxane-prostanoid receptor (TPR) were coexpressed in coronary arteries and aorta. In vivo E3 treatment attenuated aortic TPR expression. Furthermore, in vitro treatment with E3 or 3beta-Adiol downregulated TPR expression in VMC, which was blocked for both agonists by pretreatment with R,R-tetrahydrochrysene. E3- and 3beta-Adiol-mediated reduction in persistent Ca2+ signals is associated with ER-beta-mediated attenuation of TPR expression and may partly explain estrogen benefits in coronary vascular muscle.


Subject(s)
Coronary Vasospasm/drug therapy , Estriol/therapeutic use , Estrogen Receptor beta/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Administration, Cutaneous , Androstane-3,17-diol/pharmacology , Animals , Calcium Signaling/drug effects , Chrysenes/pharmacology , Coronary Vasospasm/chemically induced , Estriol/administration & dosage , Estriol/pharmacology , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Female , Gene Expression/drug effects , Genistein/pharmacology , Macaca mulatta , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nitriles/pharmacology , Propionates/pharmacology , Receptors, Thromboxane/biosynthesis , Serotonin/pharmacology , Vasoconstriction/drug effects
6.
J Immunol ; 171(12): 6389-95, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14662837

ABSTRACT

Metabolism of arachidonic acid by the cyclo-oxygenase (COX) pathway generates a family of prostanoid mediators. Nonsteroidal anti-inflammatory drugs (NSAIDs) act by inhibiting COX, thereby reducing prostanoid synthesis. The efficacy of these agents in reducing inflammation suggests a dominant proinflammatory role for the COX pathway. However, the actions of COX metabolites are complex, and certain prostanoids, such as PGE(2), in some circumstances actually inhibit immune and inflammatory responses. In these studies, we examine the hypothesis that anti-inflammatory actions of NSAIDs may be due, in part, to inhibition of thromboxane A(2) synthesis. To study the immunoregulatory actions of thromboxane A(2), we used mice with a targeted disruption of the gene encoding the thromboxane-prostanoid (TP) receptor. Both mitogen-induced responses and cellular responses to alloantigen were substantially reduced in TP(-/-) spleen cells. Similar attenuation was observed with pharmacological inhibition of TP signaling in wild-type splenocytes, suggesting that reduced responsiveness was not due to subtle developmental abnormalities in the TP-deficient mice. The absence of TP receptors reduced immune-mediated tissue injury following cardiac transplant rejection, an in vivo model of intense inflammation. Taken together, these findings show that thromboxane augments cellular immune responses and inflammatory tissue injury. Specific inhibition of the TP receptor may provide a more precise approach to limit inflammation without some of the untoward effects associated with NSAIDs.


Subject(s)
Adjuvants, Immunologic/physiology , Inflammation Mediators/physiology , Receptors, Thromboxane/physiology , Adjuvants, Immunologic/deficiency , Adjuvants, Immunologic/genetics , Animals , Calcium Signaling/genetics , Calcium Signaling/immunology , Cell Division/genetics , Cell Division/immunology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Heart Transplantation/immunology , Heart Transplantation/pathology , Immunity, Cellular/genetics , Inflammation Mediators/metabolism , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Thromboxane/biosynthesis , Receptors, Thromboxane/deficiency , Receptors, Thromboxane/genetics , Ribonucleases/metabolism , Spleen/cytology , Spleen/enzymology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thromboxane-A Synthase/antagonists & inhibitors
7.
Eur J Biochem ; 269(16): 4058-73, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12180983

ABSTRACT

In humans, thromboxane (TX) A2 signals through two TXA2 receptor (TP) isoforms, TPalpha and TPbeta, that diverge within their carboxyl terminal cytoplasmic (C) tail regions and arise by differential splicing. The human TP gene contains three exons E1-E3; while E1 exclusively encodes 5' untranslated region (UTR) sequence, E2 and E3 represent the main coding exons. An additional noncoding exon, E1b was identified within intron 1. Additionally, the TP gene contains two promoters P1 and P2 located 5' of E1 and E1b, respectively. Herein, we investigated the molecular basis of the differential expression of the TP isoforms by characterizing the 5' UTR of the TP transcripts. While E1 and E1b were found associated with TP transcript(s), their expression was mutually exclusive. 5' rapid amplification of cDNA ends (5' RACE) established that the major transcription initiation (TI) sites were clustered between -115 and -92 within E1 and at -99 within E1b. While E1 and E1b sequences were identified on TPalpha transcript(s), neither existed on TPbeta transcript(s). More specifically, TPalpha and TPbeta transcripts diverged within E2 and the major TI sites for TPbeta transcripts mapped to -12/-15 therein. Through genetic reporter assays, a previously unrecognized promoter, termed P3, was identified on the TP gene located immediately 5' of -12. The proximity of P3 to the TI site of TPbeta suggests a role for P3 in the control of TPbeta expression and implies that TPalpha and TPbeta, in addition to being products of differential splicing, are under the transcriptional control of distinct promoters.


Subject(s)
5' Untranslated Regions/genetics , Protein Isoforms/genetics , Receptors, Thromboxane/genetics , Base Sequence , Cell Line , Exons/genetics , Gene Expression Regulation/drug effects , Genes , Genes, Reporter , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Isoforms/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Thromboxane/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Transcription Initiation Site , Transcription, Genetic/drug effects
8.
Anesthesiology ; 96(5): 1147-52, 2002 May.
Article in English | MEDLINE | ID: mdl-11981155

ABSTRACT

BACKGROUND: Ketamine has been shown to suppress platelet aggregation, but its mechanisms of action have not been defined. The purpose of the current study is to clarify the effects of ketamine on human platelet aggregation and to elucidate the underlying mechanisms of its action. METHODS: Platelet aggregation was measured using an eight-channel aggregometer, and cytosolic free calcium concentration was measured in Fura-2/AM-loaded platelets using a fluorometer. Inositol 1,4,5-triphosphate (IP3) was measured with use of a commercially available IP3 assay kit. To estimate thromboxane A2 (TXA2) receptor binding affinity and expression, Scatchard analysis was performed using [3H]S145, a specific TXA2 receptor antagonist. TXA2 agonist binding assay was also performed. The membrane-bound guanosine 5'-triphosphatase activity was determined using [gamma-32P]guanosine triphosphate by liquid scintillation analyzer. RESULTS: Ketamine (500 microm) suppressed aggregation induced by adenosine diphosphate (0.5 microm), epinephrine (1 microm), (+)-9,11-epithia-11,12-methano-TXA2 (STA2) (0.5 microm), and thrombin (0.02 U/ml) to 39.1 +/- 30.9, 46.3 +/- 4.3, -2.0 +/- 16.8, and 86.6 +/- 1.4% of zero-control, respectively. Ketamine (250 microm-1 mm) also suppressed thrombin- and STA2-induced cytosolic free calcium concentration increase dose dependently. Although ketamine (2 mm) had no effect on TXA2 receptor expression and its binding affinity, it (1 mm) suppressed intracellular peak IP3 concentrations induced by thrombin and STA2 from 6.60 +/- 1.82 and 4.39 +/- 2.41 to 2.41 +/- 0.98 and 1.90 +/- 0.86 pmol/109 platelets, respectively, and it suppressed guanosine triphosphate hydrolysis induced by thrombin (0.02 units/ml) and STA2 (0.5 microm) to 50.3 +/- 3.2 and 67.5 +/- 5.5% versus zero-control, respectively. CONCLUSION: Ketamine inhibits human platelet aggregation possibly by suppressed IP3 formation and subsequent suppression of cytosolic free calcium concentration. The site of action of ketamine is neither TXA2 nor thrombin binding sites but possibly receptor-coupled mechanisms, including G-protein.


Subject(s)
Blood Platelets/metabolism , Calcium/metabolism , Cytosol/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Inosine Triphosphate/biosynthesis , Ketamine/pharmacology , Platelet Aggregation Inhibitors , Platelet Aggregation/drug effects , Adenosine Diphosphate/metabolism , Adult , Blood Platelets/drug effects , Epinephrine/metabolism , Fluorescent Dyes , Fura-2 , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Guanosine Triphosphate/metabolism , Humans , In Vitro Techniques , Receptors, Thromboxane/biosynthesis , Receptors, Thromboxane/drug effects , Receptors, Thromboxane/metabolism
9.
J Biol Chem ; 277(12): 9676-83, 2002 Mar 22.
Article in English | MEDLINE | ID: mdl-11777901

ABSTRACT

Thromboxane (TX) A(2) exerts contraction and proliferation of vascular smooth muscle cells (VSMCs) via its specific membrane TX receptor (TXR), possibly leading to the progression of atherosclerosis. A nuclear hormone receptor, peroxisome proliferator-activated receptor (PPAR)-gamma, has recently been reported to be expressed in VSMCs. Here we examined a role of PPAR-gamma in TXR gene expression in VSMCs. PPAR-gamma ligands 15-deoxy-Delta(12,14)-prostaglandin J(2) and troglitazone reduced TXR mRNA expression levels as well as cell growth as assessed by [(3)H]thymidine incorporation. Transcriptional activity of the TXR gene promoter was suppressed with PPAR-gamma ligands, and the suppression was augmented further by PPAR-gamma overexpression. By deletion and mutation analyses, the transcription suppression was shown to be the result of a -22/-7 GC box-related sequence (upstream of transcription start site). Electrophoretic mobility shift assays also showed that the sequence was bound by Sp1 but not by PPAR-gamma, and the formation of a Sp1 small middle dotDNA complex was inhibited either by coincubation with PPAR-gamma or PPAR-gamma ligand treatment of VSMCs. Moreover, glutathione S-transferase pull-down assays demonstrated a direct interaction between PPAR-gamma and Sp1. In conclusion, PPAR-gamma suppresses TXR gene transcription via an interaction with Sp1. PPAR-gamma may possibly have an antiatherosclerotic action by inhibiting TXR gene expression in VSMCs.


Subject(s)
Muscle, Smooth, Vascular/cytology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Thromboxane/biosynthesis , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Blotting, Northern , Cell Division , Cells, Cultured , DNA, Complementary/metabolism , Glutathione Transferase/metabolism , Ligands , Luciferases/metabolism , Plasmids/metabolism , Promoter Regions, Genetic , Protein Biosynthesis , RNA, Messenger/metabolism , Rats , Receptors, Thromboxane/genetics , Transfection , Up-Regulation
10.
Circ Res ; 87(9): 739-45, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-11055976

ABSTRACT

Thromboxane A(2) (TxA(2)) causes platelet aggregation, vasoconstriction, and inhibition of endothelial cell (EC) migration and prevents vascular tube formation via its specific receptors (TP), of which there are two isoforms (TPalpha and TPbeta), both expressed in human ECs. In this study, we demonstrate that the TxA(2) mimetic IBOP increases apoptosis of human ECs and inhibits the phosphorylation of Akt kinase, an intracellular mediator required for cell survival. Treatment with IBOP destroyed EC networks formed on a basement membrane matrix in vitro. To distinguish the role of the TP isoforms, each isoform was expressed in TP-null ECs to create TPalpha and TPbeta ECs. IBOP induced apoptosis and inhibited phosphorylation of Akt kinase in both TPalpha and TPbeta. IBOP increased cAMP levels in TPalpha but not in TPbeta. Apoptosis induced by IBOP in TPalpha was not affected by either the adenylyl cyclase activator forskolin or the protein kinase A inhibitor 14-22 amide or H-89, whereas that in TPbeta was suppressed by forskolin and enhanced by the protein kinase A inhibitor 14-22 amide or H-89, suggesting that the TP isoforms differ in their signal pathways in mediating apoptosis. In conclusion, apoptosis may be the mechanism by which TxA(2)-mediated destruction of vascular structures in ECs occurs; although both TP isoforms induce apoptosis, possibly via inhibiting Akt phosphorylation, the signaling differs in each isoform, in that activation of the adenylyl cyclase pathway prevents apoptosis caused by TPbeta, but not by TPalpha, stimulation.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis , Endothelium, Vascular/drug effects , Proto-Oncogene Proteins , Sulfonamides , Thromboxane A2/pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Bridged Bicyclo Compounds, Heterocyclic , Cell Survival , Clone Cells , Colforsin/pharmacology , Cyclic AMP/metabolism , Endothelium, Vascular/cytology , Enzyme Activation , Fatty Acids, Unsaturated , Humans , Hydrazines/pharmacology , Isoquinolines/pharmacology , Phosphorylation/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/biosynthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thromboxane A2/antagonists & inhibitors , Time Factors
11.
Biochim Biophys Acta ; 1496(2-3): 164-82, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10771086

ABSTRACT

We have investigated the functional coupling of alpha and beta isoforms of the human thromboxane A(2) receptor (TP) to Galpha(16) and Galpha(12) members of the G(q) and G(12) families of heterotrimeric G proteins in human embryonic kidney (HEK) 293 cell lines HEK.alpha10 or HEK.beta3, stably over-expressing TPalpha and TPbeta, respectively. Moreover, using HEK.TP(Delta328) cells which over-express a variant of TP truncated at the point of divergence of TPalpha and TPbeta, we investigated the requirement of the C-tail per se in mediating G protein coupling and effector activation. Both TPalpha and TPbeta couple similarly to Galpha(16) to affect increases in inositol 1,4,5-trisphosphate (IP(3)) and mobilisation of intracellular calcium ([Ca(2+)](i)) in response to the TP agonist U46619. Whilst both TP isoforms mediated [Ca(2+)](i) mobilisation in cells co-transfected with Galpha(12), neither receptor generated corresponding increases in IP(3), indicating that the Galpha(12)-mediated increases in [Ca(2+)](i) do not involve PLC activation. Verapamil, an inhibitor of voltage dependent Ca(2+) channels, reduced [Ca(2+)](i) mobilisation in TPalpha and TPbeta cells co-transfected with Galpha(12) to approximately 40% of that mobilised in its absence, whereas [8-(N,N-diethylamino)-octyl-3,4, 5-trimethoxybenzoate, hydrochloride] (TMB-8), an antagonist of intracellular Ca(2+) release, had no effect on [Ca(2+)](i) mobilisation by either receptor isoform co-transfected with Galpha(12). Despite the lack of differential coupling specificity by TPalpha and TPbeta, TP(Delta328) signalled more efficiently in the absence of a co-transfected G protein compared to the wild type receptors but, on the other hand, displayed an impaired ability to couple to co-transfected Galpha(11), Galpha(12) or Galpha(16) subunits. In studies investigating the role of the C-tail in influencing coupling to the effector adenylyl cyclase, similar to TPalpha but not TPbeta, TP(Delta328) coupled to Galpha(s), leading to increased adenosine 3',5'-cyclic monophosphate (cAMP), rather than to Galpha(i). Whereas TP(Delta328) signalled more efficiently in the absence of co-transfected G protein compared to the wild type TPalpha, co-transfection of Galpha(s) did not augment cAMP generation by TP(Delta328). Hence, from these studies involving the wild type TPalpha, TPbeta and TP(Delta328), we conclude that the C-tail sequences of TP are not a major determinant of G protein coupling specificity to Galpha(11) and Galpha(16) members of the G(q) family or to Galpha(12); it may play a role in determining G(s) versus G(i) coupling and may act as a determinant of coupling efficiency.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , Receptors, Thromboxane/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenylyl Cyclases/metabolism , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Enzyme Activation , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gq-G11 , GTP-Binding Protein alpha Subunits, Gs/pharmacology , GTP-Binding Proteins/pharmacology , Heterotrimeric GTP-Binding Proteins/pharmacology , Humans , Protein Isoforms/biosynthesis , Protein Isoforms/metabolism , Proto-Oncogene Proteins/pharmacology , Receptors, Thromboxane/agonists , Receptors, Thromboxane/biosynthesis , Signal Transduction , Transfection
12.
Br J Pharmacol ; 128(7): 1524-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10602332

ABSTRACT

1. Cumulative concentration-effect curves for the selective prostanoid TP receptor agonist, U46619, were constructed in strips of human non-pregnant myometrium grouped according to tissue excision site (top, lateral wall, lower uterine segment, sub-serosal or sub-endometrial), tissue orientation (strips cut either parallel or perpendicular to the serosa) and donor menstrual status (proliferative or secretory phase). 2. U46619 was excitatory in all tissues. There was no significant difference in either pEC50 or maximum response between groups (P<0.05). The range of pEC50 values was 6.8+/-0.1 (lateral wall, proliferative phase, n=5) to 7.1+/-0.3 (lateral wall, secretory phase, n=5). The range of maximum response values was 0.9+/-0.8 N cm-2 (lateral wall, proliferative phase, n=5) to 3.1+/-1.0 N cm-2 (lateral wall, secretory phase, n=5). 3. Saturation binding analyses were conducted using the radiolabelled TP receptor agonist, [125I]-BOP. Binding parameters were estimated for membranes prepared from human non-pregnant myometrium excised from the lateral wall and grouped according to donor menstrual status. 4. There were no significant differences in the mean pKd and [R]tot values for [125I]-BOP binding between the two groups (proliferative phase: pKd=8.3+/-0.3, [R]tot=412+/-319 fmol mg protein-1, n=5; secretory phase: pKd=8.5+/-0.4, [R]tot=369+/-192 fmol mg protein-1, n=6; P<0.05). 5. These data indicate that U46619-mediated responses in human non-pregnant myometrium are not influenced by tissue excision site, tissue orientation or donor menstrual status and that [125I]-BOP binding is not influenced by donor menstrual status. This suggests that the TP receptor population is homogeneous throughout the human non-pregnant myometrium, and not subject to hormonal regulation.


Subject(s)
Menstrual Cycle/physiology , Myometrium/metabolism , Receptors, Thromboxane/biosynthesis , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adult , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Female , Humans , In Vitro Techniques , Iodine Radioisotopes , Kinetics , Middle Aged , Myometrium/anatomy & histology , Myometrium/physiology , Myometrium/surgery , Pregnancy , Receptors, Thromboxane/agonists , Receptors, Thromboxane/metabolism , Uterine Contraction/drug effects , Vasoconstrictor Agents/pharmacology
13.
Biochim Biophys Acta ; 1425(3): 543-59, 1998 Nov 27.
Article in English | MEDLINE | ID: mdl-9838218

ABSTRACT

The human thromboxane A2 receptor (TP), a G protein-coupled receptor, exists as two isoforms, TPalpha and TPbeta, which arise by alternative mRNA splicing and differ exclusively in their carboxyl terminal cytoplasmic regions. In this study, a reverse transcriptase-polymerase chain reaction (RT-PCR)-based strategy was developed to examine the expression of the TPs in tissues of physiologic relevance to TXA2. Although most of the 17 different cell/tissue types examined expressed both TP isoforms, the liver hepatoblastoma HepG2 cell line was found to exclusively express TPalpha mRNA. In most cell types, TPalpha mRNA predominated over TPbeta mRNA. Moreover, although the levels of TPalpha mRNA expression were similar in most of the cell/tissue types examined, extensive differences in the levels of TPbeta mRNA were observed. Consequently, the relative expression of TPalpha: TPbeta mRNA varied considerably due to extensive differences in TPbeta mRNA expression. Most strikingly, primary HUVECs were found to express: (i) low levels of TPbeta and (ii) approximately 6-fold greater levels of TPalpha than TPbeta. These data were confirmed in the spontaneously transformed HUVEC derived ECV304 cell line. Expression of TP mRNAs in the various tissue/cells correlated with protein expression, as assessed by radioligand binding using the selective TP antagonist [3H]SQ29,548.


Subject(s)
Receptors, Thromboxane/biosynthesis , Blotting, Southern , Brain/metabolism , Bridged Bicyclo Compounds, Heterocyclic , Cell Line , Endothelium, Vascular/metabolism , Fatty Acids, Unsaturated , Humans , Hydrazines/pharmacology , Intestine, Small/metabolism , Protein Isoforms/biosynthesis , RNA, Messenger/biosynthesis , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Thromboxane A2/metabolism , Thymus Gland/metabolism , Trophoblasts/metabolism , Tumor Cells, Cultured
14.
Am J Physiol ; 275(2): F262-9, 1998 08.
Article in English | MEDLINE | ID: mdl-9691017

ABSTRACT

We examined the effects of thromboxane A2 (TxA2) on the activities of the plasminogen-plasmin system in glomerular mesangial cells. When mesangial cells are exposed to the TxA2 agonist U-46619, a substantial increase in production of plasminogen activator inhibitor-1 (PAI-1) protein is observed that is significantly greater than that induced by 10% serum alone. This increase in PAI-1 protein production is accompanied by an increase in steady-state levels of PAI-1 mRNA. This stimulation is specifically mediated by TxA2 (thromboxane prostanoid, TP) receptors, since U-46619 also stimulates PAI-1 expression in cells that are transfected with TP receptors, and this stimulation of PAI-1 production is completely blocked by the TxA2 receptor antagonist, SQ-29,548. Despite the increase in PAI-1 production, there was net stimulation of plasmin activity in the medium of mesangial cells that had been exposed to U-46619. Furthermore, U-46619 also caused an increase in tissue plasminogen activator (tPA) mRNA levels. Thus TxA2 stimulates the production of PAI-1 and plasminogen activators by mesangial cells through a receptor-dependent mechanism. In inflammatory renal diseases, the balance of these effects may modulate glomerular thrombosis and renal fibrosis.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Fibrinolysis/drug effects , Glomerular Mesangium/physiology , Plasminogen Activator Inhibitor 1/genetics , Receptors, Thromboxane/physiology , Thromboxane A2/physiology , Tissue Plasminogen Activator/genetics , Animals , Bridged Bicyclo Compounds, Heterocyclic , Cells, Cultured , Fatty Acids, Unsaturated , Fibrinolysin/metabolism , Glomerular Mesangium/drug effects , Hydrazines/pharmacology , Kinetics , Plasminogen Activator Inhibitor 1/biosynthesis , RNA, Messenger/biosynthesis , Rats , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/biosynthesis , Recombinant Proteins/biosynthesis , Thromboxane A2/agonists , Tissue Plasminogen Activator/biosynthesis , Transcription, Genetic , Transfection
15.
J Pharmacol Exp Ther ; 286(2): 1026-36, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9694965

ABSTRACT

The alpha and beta isoforms of the thromboxane A2 receptor (TP) mediate the actions of the prostanoid thromboxane A2 and its mimetics in humans. The amino terminal region of the TPs contains two consensus N-linked glycosylation sites at asparagine (N) residues N4 and N16. In this study, we explored the significance of N-linked glycosylation on the signaling and surface expression of the human TP isoforms. Inhibition of N-linked glycosylation reduced selective radioligand ([3H]SQ29,548) binding by either TP in both human erythroleukemia cells and in transfected human embryonic kidney 293 cells. Moreover, site-directed mutagenesis of the putative glycosylation sites of TPalpha revealed that radioligand binding also was reduced greatly for both the single (TPalphaN4-Q4, TPalphaN16-Q16) and double (TPalphaN4,N16-Q4,Q16) mutants, yielding levels of 8% binding relative to the wild-type TPalpha for the double mutants. Reductions in ligand binding were caused by decreased maximal binding and not by changes in affinity (Kd) or in specificity of the receptors for [3H]SQ29,548 or other ligands. Subcellular fractionation confirmed that, in relation to total TP expression, membrane expression was not altered in TPalphaN4-Q4 or TPalphaN16-Q16 but was reduced to levels of 55% of total expression in TPalphaN4,Q4-N16,Q16. Inhibition of glycosylation reduced, but did not abolish, agonist (U46619) mediated intracellular Ca++ mobilization by TPalpha or TPbeta and cAMP production by TPalpha. Thus, N-linked glycosylation of the human TP isoforms is important for ligand binding, efficient second messenger signaling and efficient membrane expression.


Subject(s)
Receptors, Thromboxane/biosynthesis , Signal Transduction/physiology , Thromboxane A2/metabolism , Binding, Competitive , Blotting, Northern , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Glycosylation , Hexosaminidases/metabolism , Humans , Isomerism , Mutagenesis, Site-Directed , Plasmids , Radioligand Assay , Receptors, Thromboxane/genetics , Signal Transduction/genetics , Transfection
16.
Naunyn Schmiedebergs Arch Pharmacol ; 357(1): 10-6, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9459567

ABSTRACT

This study describes the molecular cloning and functional characterization of the bovine thromboxane A2 (TP) receptor. Two partial nucleotide sequences coding for the bovine TP receptor were isolated from a bovine genomic and a bovine heart cDNA library. The deduced amino acid sequence suggests a heptahelical protein of 343 amino acids. The receptor protein is homologous with that of human placenta and endothelium at 84.0% and 81.4%, respectively. COS-7 cells were transfected with the bovine TP receptor cDNA, and binding affinities were assessed by radioligand binding studies. Specific displacement of [3H]SQ 29548 was demonstrated in COS-7 cell membranes with the unlabeled TP receptor antagonist SQ 29548 (Kd = 12.6+/-1.1 nM) and the TP receptor agonist U46619 (Kd = 192.1+/-58.9 nM), but not with other prostaglandins (PGD2, PGE1, PGF2alpha), or the PGI2 mimetic cicaprost. Agonist-induced stimulation of adenylyl cyclase in transfected COS-7 cells indicates a linkage to the cAMP signal transduction pathway via coupling to a stimulatory G-protein. Since bovine cells, e.g. vascular smooth muscle cells, are an established model to study the role of eicosanoids in cell signaling, this report on the molecular structure of the bovine TP receptor will allow further studies on receptor regulation.


Subject(s)
DNA, Complementary/genetics , Endothelium, Vascular/metabolism , Receptors, Thromboxane/biosynthesis , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , Adenylyl Cyclases/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive , Bridged Bicyclo Compounds, Heterocyclic , COS Cells , Cloning, Molecular , DNA, Complementary/chemistry , Fatty Acids, Unsaturated , Gene Library , Humans , Hydrazines/metabolism , Molecular Sequence Data , Prostaglandins/metabolism , Radioligand Assay , Receptors, Thromboxane/genetics , Sequence Homology , Transfection
17.
Zhonghua Jie He He Hu Xi Za Zhi ; 21(1): 19-22, 1998 Jan.
Article in Chinese | MEDLINE | ID: mdl-11263294

ABSTRACT

OBJECTIVE: To investigate the changes of thromboxane A2 receptors and the influence of their antagonist phenol red, and to study in molecular level the mechanism of action of thromboxane A2 in acute lung injury. METHODS: Acute lung injury model of rats was produced by oleic acid, and phenol red (5 mg/kg, 50 mg/kg) was injected via vein 5 minutes before oleic acid injection. Changes of thromboxane A2 receptors and their mRNA expression in the lung were determined at 6 hours after oleic acid injection. RESULTS: Down-regulation of thromboxane A2 receptors were observed, but their mRNA transcription increased significantly in the lung after oleic acid injection. The injection of phenol red before oleic acid could induce down-regulation of thromboxane A2 receptors and abolish the increase of their mRNA transcription after oleic acid injection. Phenol red could reduce LBI, RLW and improve PaO2. Histological examination of lung tissues demonstrated that the degree of lung injury had been reduced significantly. CONCLUSION: Thromboxane A2 plays an important role plays in pathological processes of acute lung injury. Phenol red in vivo could effectively reduce severity of acute lung injury by blocking thromboxane A2 receptors and regulating the expression of thromboxane A2 receptors.


Subject(s)
Lung/metabolism , Phenolsulfonphthalein/pharmacology , Receptors, Thromboxane/biosynthesis , Respiratory Distress Syndrome/metabolism , Animals , Female , Male , Oleic Acid , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Wistar , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/genetics , Respiratory Distress Syndrome/chemically induced
20.
Adv Exp Med Biol ; 433: 387-90, 1997.
Article in English | MEDLINE | ID: mdl-9561177

ABSTRACT

The activation of thrombin is the key event in clot formation after vascular injury. Thrombin itself, but also other clot-derived factors, such as thromboxane A2 (TXA2), are mitogenic for vascular smooth muscle cells. We have studied the possible interactions between thrombin and TXA2 in stimulation of coronary artery smooth muscle cell (SMC) proliferation. Thrombin (1 U/ml) caused a significant proliferatory response in SMC. U 46619, a stable TXA2 mimetic, had only a minor stimulating effect by its own but markedly potentiated the thrombin-induced mitogenesis. A possible mechanism for these potentiating effects is provided by the demonstration of a marked (6 fold) but transient (maximum after 20 min) increase in the expression of TXA2 receptor (TP receptor) mRNA in SMC by thrombin. Since a significant clot-related TXA2 generation was detected for at least 2 hours, the up-regulation of TP receptors by thrombin may represent a mechanism that is relevant for the in vivo situation of SMC proliferation after vessel injury.


Subject(s)
Coronary Vessels/drug effects , Muscle, Smooth, Vascular/drug effects , Receptors, Thromboxane/biosynthesis , Thrombin/pharmacology , Thromboxane A2/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Blood Coagulation , Blood Platelets/physiology , Cattle , Cell Division/drug effects , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/physiology , Drug Synergism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Thromboxane A2/biosynthesis , Thromboxane A2/blood , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...