Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Clin Transl Sci ; 15(10): 2378-2391, 2022 10.
Article in English | MEDLINE | ID: mdl-35791734

ABSTRACT

Postmenopausal women are at increased risk for a cardiovascular event due to platelet hyperactivity. There is evidence suggesting that ω-3 polyunsaturated fatty acids (PUFAs) and ω-6 PUFAs have cardioprotective effects in these women. However, a mechanistic understanding of how these fatty acids regulate platelet function is unknown. In this study, we supplemented postmenopausal women with fish oil (ω-3 fatty acids) or evening primrose oil (ω-6 fatty acids) and investigated the effects on their platelet activity. The effects of fatty acid supplementation on platelet aggregation, dense granule secretion, and activation of integrin αIIbß3 at basal levels and in response to agonist were tested in postmenopausal women following a supplementation and washout period. Supplementation with fish oil or primrose oil attenuated the thrombin receptor PAR4-induced platelet aggregation. Supplementation with ω-3 or ω-6 fatty acids decreased platelet dense granule secretion and attenuated basal levels of integrin αIIbß3 activation. Interestingly, after the washout period following supplementation with primrose oil, platelet aggregation was similarly attenuated. Additionally, for either treatment, the observed protective effects post-supplementation on platelet dense granule secretion and basal levels of integrin activation were sustained after the washout period, suggesting a long-term shift in platelet reactivity due to fatty acid supplementation. These findings begin to elucidate the underlying mechanistic effects of ω-3 and ω-6 fatty acids on platelet reactivity in postmenopausal women. Hence, this study supports the beneficial effects of fish oil or primrose oil supplementation as a therapeutic intervention to reduce the risk of thrombotic events in postmenopausal women. https://clinicaltrials.gov/ct2/show/NCT02629497.


Subject(s)
Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Fish Oils , Female , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/pharmacology , Fatty Acids, Omega-6/therapeutic use , Fish Oils/pharmacology , Fish Oils/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex , Postmenopause , Receptors, Thrombin , Humans
2.
Eur Heart J Cardiovasc Pharmacother ; 8(7): 728-737, 2022 Sep 29.
Article in English | MEDLINE | ID: mdl-35353154

ABSTRACT

AIMS: Dual pathway inhibition (DPI) by adding a vascular dose of rivaroxaban to a single antiplatelet agent has emerged as a promising antithrombotic strategy. However, in most studies the antiplatelet agent of choice used in adjunct to a vascular dose of rivaroxaban was aspirin, and data on a P2Y12 inhibitor and how this DPI regimen compares with standard dual antiplatelet therapy (DAPT) are limited. METHODS AND RESULTS: This investigation was a substudy analysis conducted in selected cohorts of patients with stable atherosclerotic disease enrolled from a larger prospective, open-label, parallel-group pharmacodynamic (PD) study. We analysed data from 40 patients treated with either clopidogrel- or ticagrelor-based DAPT first, and clopidogrel- or ticagrelor-based DPI thereafter. PD measures explored key pathways involved in thrombus formation and included markers of (1) P2Y12 reactivity, (2) platelet-mediated global thrombogenicity, (3) cyclooxygenase-1 activity, (4) thrombin receptor-activating peptide (TRAP)-induced platelet aggregation, (5) tissue factor (TF)-induced platelet aggregation, and (6) thrombin generation. Compared with DAPT, on a background of the same P2Y12 inhibitor (clopidogrel or ticagrelor), DPI was associated with reduced thrombin generation, increased markers of cyclooxygenase-1 activity and TRAP-induced platelet aggregation, and no differences in markers of P2Y12 signalling, platelet-mediated global thrombogenicity, and TF-induced platelet aggregation. In an analysis according to P2Y12 inhibitor type, ticagrelor reduced markers of platelet-mediated global thrombogenicity, P2Y12 signalling, and rates of high platelet reactivity compared with clopidogrel. CONCLUSION: Compared with DAPT with aspirin and a P2Y12 inhibitor, the use of a P2Y12 inhibitor in adjunct to a vascular dose of rivaroxaban as part of a DPI strategy is associated with similar effects on platelet-mediated global thrombogenicity but reduced thrombin generation. A DPI strategy with ticagrelor is associated with enhanced antithrombotic efficacy, the clinical implications of which warrant larger scale investigations. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03718429.


Subject(s)
Aspirin , Platelet Aggregation Inhibitors , Clopidogrel/adverse effects , Cyclooxygenase 1 , Fibrinolytic Agents/therapeutic use , Humans , Peptides , Prospective Studies , Receptors, Thrombin , Rivaroxaban/adverse effects , Thrombin , Thromboplastin , Ticagrelor/adverse effects
3.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502257

ABSTRACT

The catabolic and destructive activity of serine proteases in arthritic joints is well known; however, these enzymes can also signal pain and inflammation in joints. For example, thrombin, trypsin, tryptase, and neutrophil elastase cleave the extracellular N-terminus of a family of G protein-coupled receptors and the remaining tethered ligand sequence then binds to the same receptor to initiate a series of molecular signalling processes. These protease activated receptors (PARs) pervade multiple tissues and cells throughout joints where they have the potential to regulate joint homeostasis. Overall, joint PARs contribute to pain, inflammation, and structural integrity by altering vascular reactivity, nociceptor sensitivity, and tissue remodelling. This review highlights the therapeutic potential of targeting PARs to alleviate the pain and destructive nature of elevated proteases in various arthritic conditions.


Subject(s)
Arthritis/metabolism , Receptors, Proteinase-Activated/physiology , Animals , Humans , Receptor, PAR-1/physiology , Receptor, PAR-2/physiology , Receptors, Thrombin/physiology , Signal Transduction/physiology
4.
J Cell Biochem ; 122(8): 870-881, 2021 08.
Article in English | MEDLINE | ID: mdl-33748988

ABSTRACT

Hemostasis is a proteolytically regulated process that requires activation of platelets and the blood coagulation cascade upon vascular injury. Activated platelets create a thrombogenic environment and amplify the coagulation process. Plant latex proteases (PLPs) have been used as therapeutic components to treat various ailments by folk healers. One of the main applications of plant latices is to stop bleeding from minor injuries and to enhance wound healing activity. Although many studies have reported the pro-coagulant activities of PLPs, an in-depth investigation is required to understand the mechanism of action of PLPs on platelets. Here, the effect of PLPs on platelet aggregation was studied systematically to validate the observed pharmacological effect by folk healers. Among 29 latices from the Ficus genus tested, Ficus drupacea exhibited potent pro-coagulant and thrombin-like activity. Drupin, a thrombin-like cysteine protease responsible for platelet aggregation was purified from F. drupacea latex. Drupin exhibits pro-coagulant activity and reduces the bleeding time in mice tail. It induces platelet aggregation by activating mitogen-activated protein kinases and the nuclear factor-κB and PI3K/Akt signalling cascade, which, in turn, phosphorylats, cytosolic phospholipase A2  leading to the release of thromboxane A2 from the granules to activate the nearby platelets to aggregate. Furthermore, we investigated the involvement of protease-activated receptors in drupin-induced platelet aggregation using specific protease activated receptor 1 (PAR1) and PAR4 receptor antagonists. The results confirmed that the drupin-induced platelet aggregation was mediated by both PAR1 and PAR4, synergistically. Overall, drupin reduces the bleeding time by exerting pro-coagulant activity and induces platelet aggregation by activating the intracellular signalling cascade.


Subject(s)
Blood Platelets/metabolism , Ficus/enzymology , Peptide Hydrolases/pharmacology , Plant Proteins/pharmacology , Platelet Aggregation/drug effects , Receptors, Thrombin/metabolism , Animals , Male , Mice , Signal Transduction/drug effects
5.
Arterioscler Thromb Vasc Biol ; 39(4): 694-703, 2019 04.
Article in English | MEDLINE | ID: mdl-30727756

ABSTRACT

Objective- PAR4 (protease-activated receptor 4), one of the thrombin receptors in human platelets, has emerged as a promising target for the treatment of arterial thrombotic disease. Previous studies implied that thrombin exosite II, known as a binding site for heparin, may be involved in thrombin-induced PAR4 activation. In the present study, a heparin octasaccharide analog containing the thrombin exosite II-binding domain of heparin was chemically synthesized and investigated for anti-PAR4 effect. Approach and Results- PAR4-mediated platelet aggregation was examined using either thrombin in the presence of a PAR1 antagonist or γ-thrombin, which selectively activates PAR4. SCH-28 specifically inhibits PAR4-mediated platelet aggregation, as well as the signaling events downstream of PAR4 in response to thrombin. Moreover, SCH-28 prevents thrombin-induced ß-arrestin recruitment to PAR4 but not PAR1 in Chinese Hamster Ovary-K1 cells using a commercial enzymatic complementation assay. Compared with heparin, SCH-28 is more potent in inhibiting PAR4-mediated platelet aggregation but has no significant anticoagulant activity. In an in vitro thrombosis model, SCH-28 reduces thrombus formation under whole blood arterial flow conditions. Conclusions- SCH-28, a synthetic small-molecular and nonanticoagulant heparin analog, inhibits thrombin-induced PAR4 activation by interfering with thrombin exosite II, a mechanism of action distinct from other PAR4 inhibitors that target the receptor. The characteristics of SCH-28 provide a new strategy for targeting PAR4 with the potential for the treatment of arterial thrombosis.


Subject(s)
Antithrombins/pharmacology , Heparin/chemistry , Oligosaccharides/pharmacology , Platelet Aggregation/drug effects , Receptors, Thrombin/antagonists & inhibitors , Animals , Antithrombins/chemical synthesis , CHO Cells , Calcium Signaling/drug effects , Computer Simulation , Cricetulus , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Models, Molecular , Recombinant Proteins/drug effects , Thrombin/pharmacology , Thrombosis/prevention & control
6.
Cutan Ocul Toxicol ; 37(3): 207-209, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29171298

ABSTRACT

OBJECTIVE: The purpose of this experimental study was to investigate the role of vitamin supplements (Ocuvite, Vitalux Omega, and Nutrof Total) as possible inhibitors of the onset of age-related macular degeneration (AMD). MATERIALS AND METHODS: The anti-aggregating effect of each vitamin was determined against four accumulative factors namely, platelet activating factor (PAF), adenosine diphosphate (ADP), thrombin receptor-activating peptide (TRAP), and arachidonic acid (AA) in the platelet rich plasma (PRP) of healthy volunteers. RESULTS: Ocuvite, Vitalux Omega, and Nutrof Total were more potent inhibitors against PAF and ADP compared to TRAP and AA. Among the three vitamins, Nutrof Total displayed more potent inhibitions against TRAP and AA, while against PAF and ADP all the three vitamins revealed similar IC50 values. CONCLUSIONS: The vitamins Ocuvite, Vitalux Omega, and Nutrof Total have anti-aggregating effects and therefore can be used against AMD in healthy volunteers.


Subject(s)
Blood Platelets/physiology , Dietary Supplements , Macular Degeneration/prevention & control , Platelet Aggregation/drug effects , Vitamins/pharmacology , Adenosine Diphosphate/metabolism , Arachidonic Acid/antagonists & inhibitors , Arachidonic Acid/metabolism , Blood Platelets/drug effects , Healthy Volunteers , Humans , Inhibitory Concentration 50 , Platelet Activating Factor/antagonists & inhibitors , Platelet Activating Factor/metabolism , Receptors, Thrombin/antagonists & inhibitors , Receptors, Thrombin/metabolism , Vitamins/therapeutic use
7.
Article in English | WPRIM | ID: wpr-714640

ABSTRACT

BACKGROUND/AIMS: Although epigallocatechin-3-gallate (EGCG), which is found in high contents in the dried leaves of green tea, has been reported to have an anti-platelet effect, synergistic effects of EGCG in addition to current anti-platelet medications remains to be elucidated. METHODS: Blood samples were obtained from 40 participants who took aspirin (ASA, n = 10), clopidogrel (CPD, n = 10), ticagrelor (TCG, n = 10) and no anti-platelet medication (Control, n = 10). Ex vivo platelet aggregation and adhesion under various stimulators were analyzed by multiple electrode aggregometry (MEA) and Impact-R systems. PAC-1 and P-selectin expressions in human platelets were analyzed by flow cytometry. RESULTS: In MEA analysis, adenosine diphosphate (ADP) and thrombin receptor activating peptide (TRAP)-induced platelet aggregations were lower in the CPD and the TCG groups; arachidonic acid (AA)-induced platelet aggregation was lower in the ASA group, whereas collagen (COL)-induced platelet aggregations were comparable among four groups. EGCG significantly reduced ADP- and COL-induced platelet aggregation in dose-dependent manner (ADP, p = 0.04; COL, p < 0.01). There were no additional suppressions of platelet aggregation stimulated by AA in the ASA group, and by ADP in the CPD and TCG groups. Moreover, EGCG suppressed shear stress-induced platelet adhesion on Impact-R, and had no effect on P-selectin and PAC-1 expressions. CONCLUSIONS: Ex vivo treatment of EGCG inhibited platelet adhesion and aggregation without changes in P-selectin and PAC-1 expression. There was no additional suppressions in platelet aggregation stimulated by AA in the ASA group and ADP in the CPD and TCG groups.


Subject(s)
Humans , Adenosine Diphosphate , Arachidonic Acid , Aspirin , Blood Platelets , Catechin , Collagen , Electrodes , Flow Cytometry , P-Selectin , Platelet Aggregation , Platelet Aggregation Inhibitors , Receptors, Thrombin , Tea
8.
Br J Cancer ; 117(5): 695-703, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28697175

ABSTRACT

BACKGROUND: Selective platelet release of pro- or anti-angiogenic factors distinctly regulated angiogenesis. We hypothesised that selective release of platelet angiogenic factors could differently regulate tumour growth. METHODS: Breast cancer cell proliferation, cancer cell-induced endothelial tube formation in vitro, and tumour growth in vivo were studied in the presence of protease-activated receptor 1-stimulated platelet releasate (PAR1-PR; rich in pro-angiogenic factors) or PAR4-PR (rich in anti-angiogenic factors). RESULTS: The PAR1-PR and PAR4-PR supplementation (10%) similarly enhanced cell proliferation of MCF-7 and MDA-MB-231 breast cancer cells. The cancer cells triggered capillary-like tube formation of endothelial cells that was further enhanced by pro-angiogenic factor-rich PAR1-PR. The VEGF, but not SDF-1α, receptor blockade abolished PAR1-PR/PAR4-PR-enhanced cancer cell proliferation. Integrin blockade by RGDS had identical effects as VEGF inhibition. The Src and ERK inhibition diminished, whereas PI3K and PKC blockade abolished platelet releasate-enhanced cancer cell proliferation. Using a model of subcutaneous implantation of MDA-MB-231 cells in nude mice, PAR1-PR enhanced tumour growth more markedly than PAR4-PR, and seemed to achieve the exaggeration by promoting more profound tumour angiogenesis. CONCLUSIONS: Platelet releasate increases breast cancer cell proliferation through VEGF-integrin cooperative signalling. Pro-angiogenic factor-rich platelet releasate enhances cancer cell-induced angiogenesis more markedly, and thus exaggerates tumour growth in vivo.


Subject(s)
Blood Platelets/metabolism , Breast Neoplasms/metabolism , Integrins/metabolism , Neovascularization, Pathologic/metabolism , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Animals , Blood Platelets/drug effects , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Proliferation , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Human Umbilical Vein Endothelial Cells , Humans , Integrins/antagonists & inhibitors , MCF-7 Cells , Male , Mice , Mice, Nude , Middle Aged , Oligopeptides/pharmacology , Phenylurea Compounds/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Quinolines/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
9.
Thromb Res ; 154: 84-92, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28448853

ABSTRACT

Thrombin activates platelets via proteolytic cleavage of protease-activated receptors (PARs) 1 and 4. The two PARs have distinct but complementary roles. The mechanisms responsible for PAR1 activation by thrombin have been extensively studied. However, much less is known regarding thrombin activation of PAR4, especially the potential involvement of regions of PAR4 other than the N-terminal, which is bound to the catalytic site of thrombin. We have studied PAR4 in S. cerevisiae strain MMY12, an expression system in which the GPCR receptors are connected to a Lac Z reporter gene resulting in increased ß-galactosidase activity. This approach was used to assess PAR4 mutants to evaluate the contribution of different aspartic residues in facilitating PAR4 activation. Furthermore, peptides mimicking parts of the PAR4 N-terminal and the second extracellular loop (ECLII) were tested for their ability to inhibit platelet activation by thrombin. Binding of these peptides to γ-thrombin was studied by monitoring the decrease in tryptophan fluorescence intensity of thrombin. We conclude that not only the N-terminal but also the electronegative aspartic residues D224, D230 and D235 (located in ECLII) are be important for PAR4 binding to thrombin. We further suggest that they play a role for the tethered ligand binding to the receptor, as mutations also affected activation in response to a PAR4-activating peptide mimicking the new N-terminal formed after cleavage. This agrees with previous results on PAR1 and thrombin binding. We suggest that the ECLII of PAR4 could be a potential target for antithrombotic drug development.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Receptors, Thrombin/metabolism , Thrombin/metabolism , Amino Acid Sequence , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Binding Sites , Blood Platelets/cytology , Humans , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Thrombin/chemistry
10.
Thromb Res ; 138: 63-68, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26610745

ABSTRACT

BACKGROUND/OBJECTIVES: Novel (or non-vitamin K antagonist) oral anti-coagulants (NOACs) are antagonists of coagulation factors (F) Xa (rivaroxaban) or IIa (dabigatran), and their non-inferiority compared with vitamin K antagonists has been demonstrated in patients with non-valvular atrial fibrillation. However, it is still not fully understood if and how dabigatran and rivaroxaban impact platelet function. This observational study aimed to assess platelet function in patients receiving dabigatran or rivaroxaban. METHODS/RESULTS: This was a single centre, observational study quantifying platelet aggregation in 90 patients treated with NOACs by multiple electrode aggregometry. The thrombin receptor activating peptide (TRAP)-induced platelet aggregation was significantly higher in 35 patients receiving dabigatran (d) compared with control (c) patients (d 108±31 vs. c 85±30arbitrary units [AU]∗min, p<0.001). Patients receiving rivaroxaban (r) showed no differences compared with the control group (r 88±32 vs. c 85±30AU∗min, p=0.335). In intraindividual time courses of 16 patients, a significantly higher aggregation was found after the administration of dabigatran (before vs. after; 83±29 vs. 100±31AU∗min, p=0.009). CONCLUSION: In this observational study, the TRAP-induced platelet aggregation was enhanced in cardiovascular patients receiving dabigatran. This might be explained by a change in the expression profile of thrombin receptors on the surface of platelets. Rivaroxaban had no influence on platelet aggregation.


Subject(s)
Antithrombins/therapeutic use , Atrial Fibrillation/drug therapy , Blood Platelets/drug effects , Dabigatran/therapeutic use , Platelet Aggregation/drug effects , Receptors, Thrombin/metabolism , Rivaroxaban/therapeutic use , Aged , Aged, 80 and over , Antithrombins/pharmacology , Dabigatran/pharmacology , Female , Humans , Male , Middle Aged , Pulmonary Embolism/drug therapy , Rivaroxaban/pharmacology , Venous Thrombosis/drug therapy
11.
Chin J Integr Med ; 17(8): 625-30, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21826598

ABSTRACT

OBJECTIVE: To investigate the effects of Salvia Miltiorrhiza Liguspyragine Hydrochloride and Glucose Injection (, SLGI) on the expression of platelet membrane receptors proteinase-activated receptor-1 (PAR1) and proteinase-activated receptor-4 (PAR4) in end-stage renal disease (ESRD) patients on chronic haemodialysis (HD). METHODS: Eighty-six ESRD patients on HD (treated group) were treated with SLGI, 7 days as one therapeutic course, for two successive courses. The previous therapies were unchanged. Flow cytometry was used to assess the expression of platelet PAR1 and PAR4 in the patients, and turbidity method was used to determine the platelet maximum aggregation rate (MAR). Meanwhile, renal function was measured. The final data were compared with those before treatment and with those in the normal control group (54 healthy subjects). RESULTS: Compared with the normal control group, the expressions of PAR1 and PAR4 and platelet MAR in ESRD patients on HD was significantly higher before treatment (P=0.001, P=0.006, and P=0.008); after treatment with SLGI, the above indices in patients were remarkably decreased (P=0.036 and P=0.046), except PAR4 (P=0.067), but still higher than those in the normal control group, however, it was not statistically significant. CONCLUSIONS: (1) The overexpression of PAR1 and PAR4 might lead to increased platelet aggregation and this could be one of the reasons for the thrombotic events in ESRD patients on HD. (2) SLGI was able to down-regulate the expression of PAR1 in ESRD patients on HD, improve platelet function, and regulate platelet activation.


Subject(s)
Blood Platelets/metabolism , Drugs, Chinese Herbal/therapeutic use , Glucose/administration & dosage , Receptors, Thrombin/metabolism , Renal Dialysis , Salvia miltiorrhiza/chemistry , Blood Platelets/drug effects , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/pharmacology , Female , Glucose/pharmacology , Humans , Kidney Function Tests , Male , Middle Aged , Platelet Aggregation/drug effects
12.
J Surg Res ; 168(2): 197-205, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-19922946

ABSTRACT

BACKGROUND: Phytochemicals are an important source of emerging preventive and therapeutic agents for cancer. Triptolide/PG490, an extract of the Chinese herb Tripterygium wilfordii Hook F, is a potent anti-inflammatory agent that also possesses anticancer activity. While its antiproliferative effects are well-established, the potential antimigratory effects of triptolide have not been characterized. MATERIAL AND METHODS: Effects of triptolide on the proliferation and invasion of colon cancer cells and expression of cancer-related genes and proteins were assessed. RESULTS: Triptolide potently inhibited HT29 and HCT116 colon cancer cell growth and reduced basal and stimulated HCT116 migration through collagen by 65% to 80%. Triptolide inhibited mRNA expression of the positive cell cycle regulatory genes c-myc, and A, B, C, and D-type cyclins in multiple colon cancer cell lines. Additionally, we show that triptolide treatment decreased expression of VEGF and COX-2, which promote cancer progression and invasion, and inhibited the expression of multiple cytokine receptors potentially involved in cell migration and cancer metastasis, including the thrombin receptor, CXCR4, TNF receptors, and TGF-ß receptors. CONCLUSIONS: Triptolide is a potent inhibitor of colon cancer proliferation and migration in vitro. The down-regulation of multiple cytokine receptors, in combination with inhibition of COX-2 and VEGF and positive cell cycle regulators, may contribute to the antimetastatic action of this herbal extract.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cell Movement/drug effects , Cyclins/metabolism , Diterpenes/pharmacology , Phenanthrenes/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Cytokine/metabolism , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Drug Evaluation, Preclinical , Epidermal Growth Factor , Epoxy Compounds/pharmacology , HCT116 Cells , HT29 Cells , Humans , Neurotensin , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Receptors, CXCR4/metabolism , Receptors, Cytokine/antagonists & inhibitors , Receptors, Thrombin/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Tripterygium , Vascular Endothelial Growth Factor A/metabolism
13.
Article in English | WPRIM | ID: wpr-328445

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effects of Salvia Miltiorrhiza Liguspyragine Hydrochloride and Glucose Injection (, SLGI) on the expression of platelet membrane receptors proteinase-activated receptor-1 (PAR1) and proteinase-activated receptor-4 (PAR4) in end-stage renal disease (ESRD) patients on chronic haemodialysis (HD).</p><p><b>METHODS</b>Eighty-six ESRD patients on HD (treated group) were treated with SLGI, 7 days as one therapeutic course, for two successive courses. The previous therapies were unchanged. Flow cytometry was used to assess the expression of platelet PAR1 and PAR4 in the patients, and turbidity method was used to determine the platelet maximum aggregation rate (MAR). Meanwhile, renal function was measured. The final data were compared with those before treatment and with those in the normal control group (54 healthy subjects).</p><p><b>RESULTS</b>Compared with the normal control group, the expressions of PAR1 and PAR4 and platelet MAR in ESRD patients on HD was significantly higher before treatment (P=0.001, P=0.006, and P=0.008); after treatment with SLGI, the above indices in patients were remarkably decreased (P=0.036 and P=0.046), except PAR4 (P=0.067), but still higher than those in the normal control group, however, it was not statistically significant.</p><p><b>CONCLUSIONS</b>(1) The overexpression of PAR1 and PAR4 might lead to increased platelet aggregation and this could be one of the reasons for the thrombotic events in ESRD patients on HD. (2) SLGI was able to down-regulate the expression of PAR1 in ESRD patients on HD, improve platelet function, and regulate platelet activation.</p>


Subject(s)
Female , Humans , Male , Middle Aged , Blood Platelets , Metabolism , Drugs, Chinese Herbal , Pharmacology , Therapeutic Uses , Glucose , Pharmacology , Kidney Function Tests , Platelet Aggregation , Receptors, Thrombin , Metabolism , Renal Dialysis , Salvia miltiorrhiza , Chemistry
14.
Curr Pharm Des ; 15(19): 2262-9, 2009.
Article in English | MEDLINE | ID: mdl-19601827

ABSTRACT

The rising costs and time associated with bringing new medicines to the market have created a need for a new paradigm for reducing the attrition rates of drug candidates in both preclinical and clinical development stages. Early appraisal of drug metabolism and pharmacokinetic (DMPK) parameters is now possible due to several higher throughput in vitro and in vivo screens. This knowledge of DMPK properties should not only shorten the timelines for the selection of drug candidates but also enhance the probability of their success for development. The role of DMPK researchers in the drug research paradigm should not be limited to screening a large array of compounds during the lead optimization process but should include a strive for an understanding of the absorption, distribution, metabolism, excretion, and potential drug-related toxicities of a chemical series. As an example, in this article we present a specific DMPK research screening paradigm and describe a case study using the Thrombin Receptor Antagonist program. This screening paradigm followed by the extensive lead optimization process culminated in the selection of SCH 530348, a potent, selective and orally active thrombin receptor antagonist for the treatment of thrombosis.


Subject(s)
Drug Design , Drug Industry/methods , Pharmaceutical Preparations/metabolism , Animals , Drug Evaluation, Preclinical/methods , Drug Industry/economics , Drug-Related Side Effects and Adverse Reactions , Humans , Lactones/pharmacokinetics , Lactones/pharmacology , Lactones/therapeutic use , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, Thrombin/antagonists & inhibitors , Thrombosis/drug therapy , Thrombosis/physiopathology
15.
Neurogastroenterol Motil ; 21(11): 1189-e107, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19413681

ABSTRACT

Protease-activated receptor-4 (PAR(4)) belongs to the family of receptors activated by the proteolytic cleavage of their extracellular N-terminal domain and the subsequent binding of the newly released N-terminus. While largely expressed in the colon, the role of PAR(4) in gut functions has not been defined. We have investigated the effects of PAR(4) agonist on colonic sensations and sensory neuron signalling, and its role in visceral pain. We observed that a single administration of the PAR(4) agonist peptide (AYPGKF-NH(2)), but not the control peptide (YAPGKF-NH(2)) into the colon lumen of mice significantly reduced the visceromotor response to colorectal distension at different pressures of distension. Further, intracolonic administration of the PAR(4) agonist, but not the control peptide, was able to significantly inhibit PAR(2) agonist- and transcient receptor potential vanilloid-4 (TRPV4) agonist-induced allodynia and hyperalgesia in response to colorectal distension. Protease-activated receptor-4 was detected in sensory neurons projecting from the colon, and isolated from the dorsal root ganglia, where it co-expressed with PAR(2) and TRPV4. In total sensory neurons, PAR(4) agonist exposure inhibited free intracellular calcium mobilization induced by the pro-nociceptive agonists of PAR(2) and TRPV4. Finally, PAR(4)-deficient mice experienced increased pain behaviour in response to intracolonic administration of mustard oil, compared with wild-type littermates. These results show that PAR(4) agonists modulate colonic nociceptive response, inhibit colonic hypersensitivity and primary afferent responses to pro-nociceptive mediators. Endogenous activation of PAR(4) also plays a major role in controlling visceral pain. These results identify PAR(4) as a previously unknown modulator of visceral nociception.


Subject(s)
Hyperalgesia/physiopathology , Neurons, Afferent/metabolism , Pain/physiopathology , Receptors, Thrombin/metabolism , Visceral Afferents/metabolism , Animals , Behavior, Animal/drug effects , Catheterization , Ganglia, Spinal/cytology , Male , Mice , Mice, Inbred C57BL , Mustard Plant , Oligopeptides/pharmacology , Pain/metabolism , Plant Oils/pharmacology , Receptors, Thrombin/agonists , Sensory Receptor Cells/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism , Visceral Afferents/drug effects
16.
Oncol Rep ; 21(5): 1261-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19360302

ABSTRACT

Thrombin has been recently demonstrated to promote hepatocellular carcinoma (HCC) cell migration by activation of the proteinase-activated receptor (PAR) subtypes PAR1 and PAR4 suggesting a role of these proteinase-receptor systems in HCC progression. In this study, we investigated the effect of (-)-epigallocatechin-3-gallate (EGCG), the major polyphenolic compound of green tea on thrombin-PAR1/PAR4-mediated hepatocellular carcinoma cell invasion and p42/p44 MAPKinase activation. In this study we used the permanent liver carcinoma cell line HEP-3B and two primary cultures established from surgically resected HCCs. We found that stimulation of HCC cells with thrombin, the PAR1-selective activating peptide, TFLLRN-NH2, and the PAR4-selective activating peptide, AYPGKF-NH2, increased cell invasion across a Matrigel-coated membrane barrier and stimulated activation of p42/p44 MAPKinase phosphorylation. Both the effects on p42/p44 MAPKinases, and on cell invasiveness induced by thrombin and the PAR1/4 subtype-selective agonist peptides were effectively blocked by EGCG. The results clearly identify EGCG as a potent inhibitor of the thrombin-PAR1/PAR4-p42/p44 MAPKinase invasive signaling axis in hepatocellular carcinoma cells as a previously unrecognized mode of action for EGCG in cancer cells. Moreover, the results suggest that (-)-epigal-locatechin-3-gallate might have therapeutic potential for hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Catechin/analogs & derivatives , Liver Neoplasms/drug therapy , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Thrombin/antagonists & inhibitors , Anticarcinogenic Agents/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Catechin/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Oligopeptides/pharmacology , Phosphorylation , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/metabolism , Receptors, Thrombin/agonists , Receptors, Thrombin/antagonists & inhibitors , Receptors, Thrombin/metabolism , Tea/chemistry , Thrombin/pharmacology
17.
Biochem Pharmacol ; 77(5): 835-44, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19073150

ABSTRACT

Platelet secretion is an important physiological event in hemostasis. The protease-activated receptors, PAR 1 and PAR 4, and the thromboxane receptor activate the G(12/13) pathways, in addition to the G(q) pathways. Here, we investigated the contribution of G(12/13) pathways to platelet dense granule release. 2MeSADP, which does not activate G(12/13) pathways, does not cause dense granule release in aspirin-treated platelets. However, supplementing 2MeSADP with YFLLRNP (60muM), as selective activator of G(12/13) pathways, resulted in dense granule release. Similarly, supplementing PLC activation with G(12/13) stimulation also leads to dense granule release. These results demonstrate that supplemental signaling from G(12/13) is required for G(q)-mediated dense granule release and that ADP fails to cause dense granule release because the platelet P2Y receptors, although activate PLC, do not activate G(12/13) pathways. When RhoA, downstream signaling molecule in G(12/13) pathways, is blocked, PAR-mediated dense granule release is inhibited. Furthermore, ADP activated RhoA downstream of G(q) and upstream of PLC. Finally, RhoA regulated PKCdelta T505 phosphorylation, suggesting that RhoA pathways contribute to platelet secretion through PKCdelta activation. We conclude that G(12/13) pathways, through RhoA, regulate dense granule release and fibrinogen receptor activation in platelets.


Subject(s)
Blood Platelets/drug effects , Cytoplasmic Granules/physiology , GTP-Binding Proteins/physiology , Receptor, PAR-1/physiology , Receptors, Thrombin/physiology , rhoA GTP-Binding Protein/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Aspirin/pharmacology , Blood Platelets/metabolism , Blotting, Western , Enzyme Activation , Humans , In Vitro Techniques , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Protein Kinase C-delta/metabolism , Thionucleotides/pharmacology , Type C Phospholipases/metabolism
18.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 28(12): 1086-9, 2008 Dec.
Article in Chinese | MEDLINE | ID: mdl-19317163

ABSTRACT

OBJECTIVE: To observe the effect of Xiaoyu Zhixue Tablet (XYZXT) on expression of platelet membrane glycoproteins (GP) Ib/IX/V complex and its component GP Ibalpha in patients with hemorrhagic thrombopathy, so as to explore its possible mechanism. METHODS: Ninety-eight patients with hemorrhagic thrombopathy were randomly assigned to two groups, the TCM group (68 cases) treated with XYZXT and the Western medicine group (30 cases) treated with adrenosin, vitamins C, K and P, for 6 months totally. The hemostatic effect and the platelet aggregation recovery rate in the two groups were observed. And expressions of GPIb/IX/V complex and GP Ibalpha were analyzed by flow cytometry in both groups before and after treatment as well as in 34 healthy persons for control. RESULTS: The hemostatic effective rate was 89.7% in the TCM group and 46.7% in the Western medicine group (u= 5.68, P < 0.01); the platelet aggregation recovery rate in the two groups was 67.6% and 3.3% respectively (chi2 = 34. 49, P < 0.01). The fluorescence intensity of GPIb/IX/V complex and GPIbalpha were lower in both groups before treatment than those of the healthy control (P < 0.05), but after treatment the two markers elevated in the TCM group, approaching the control (P > 0.05), and significantly higher than those in the Western medicine group (P < 0.05). CONCLUSION: The partial mechanism of XYZXT in treating hemorrhagic thrombopathy might be its regulation on the expression of thrombin receptor at the receptor protein level.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Hemorrhage/drug therapy , Hemorrhage/genetics , Receptors, Thrombin/genetics , Adolescent , Adult , Aged , Female , Gene Expression/drug effects , Hemorrhage/metabolism , Humans , Male , Middle Aged , Receptors, Thrombin/metabolism , Tablets , Young Adult
19.
Article in Chinese | WPRIM | ID: wpr-337578

ABSTRACT

<p><b>OBJECTIVE</b>To observe the effect of Xiaoyu Zhixue Tablet (XYZXT) on expression of platelet membrane glycoproteins (GP) Ib/IX/V complex and its component GP Ibalpha in patients with hemorrhagic thrombopathy, so as to explore its possible mechanism.</p><p><b>METHODS</b>Ninety-eight patients with hemorrhagic thrombopathy were randomly assigned to two groups, the TCM group (68 cases) treated with XYZXT and the Western medicine group (30 cases) treated with adrenosin, vitamins C, K and P, for 6 months totally. The hemostatic effect and the platelet aggregation recovery rate in the two groups were observed. And expressions of GPIb/IX/V complex and GP Ibalpha were analyzed by flow cytometry in both groups before and after treatment as well as in 34 healthy persons for control.</p><p><b>RESULTS</b>The hemostatic effective rate was 89.7% in the TCM group and 46.7% in the Western medicine group (u= 5.68, P < 0.01); the platelet aggregation recovery rate in the two groups was 67.6% and 3.3% respectively (chi2 = 34. 49, P < 0.01). The fluorescence intensity of GPIb/IX/V complex and GPIbalpha were lower in both groups before treatment than those of the healthy control (P < 0.05), but after treatment the two markers elevated in the TCM group, approaching the control (P > 0.05), and significantly higher than those in the Western medicine group (P < 0.05).</p><p><b>CONCLUSION</b>The partial mechanism of XYZXT in treating hemorrhagic thrombopathy might be its regulation on the expression of thrombin receptor at the receptor protein level.</p>


Subject(s)
Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Drugs, Chinese Herbal , Gene Expression , Hemorrhage , Drug Therapy , Genetics , Metabolism , Receptors, Thrombin , Genetics , Metabolism , Tablets
20.
Alcohol Clin Exp Res ; 31(8): 1308-14, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17559545

ABSTRACT

BACKGROUND: Moderate and prolonged consumption of red wine is associated with decreased cardiovascular morbidity and mortality. Inhibition of platelet functions by ingredients in red wine is thought to be one of the causes. However, the molecular mechanism of this inhibition has remained unexplained. MATERIALS AND METHODS: We measured aggregation, changes in cytosolic Ca(2+) and tyrosine phosphorylation of the inhibitory receptor platelet endothelial cell adhesion molecule-1 (PECAM-1) in platelets stimulated with thrombin receptor (PAR-1) activating peptide (TRAP) and ADP and investigated the effects of alcohol-free polyphenolic grape extract (PGE), alcohol, and the polyphenols catechin, epi-catechin, resveratrol, trans-resveratrol, and gallic acid. RESULTS: Polyphenolic grape extract induced dose-dependent inhibition of TRAP-induced and ADP-induced platelet aggregation and Ca(2+) mobilization. Inhibition was accompanied by activation of PECAM-1. Apart from a slight inhibition by catechin, ethanol or other individual polyphenols failed to inhibit aggregation or activate PECAM-1. CONCLUSIONS: Red wine inhibits platelet functions through its PGE content, which stimulates the inhibitory receptor PECAM-1, thereby attenuating platelet activation.


Subject(s)
Cardiovascular Diseases/epidemiology , Flavonoids/pharmacology , Phenols/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Vitis/chemistry , Wine , Adenosine Diphosphate/pharmacology , Biotransformation/drug effects , Calcium/metabolism , Cytosol/metabolism , Flavonoids/isolation & purification , France/epidemiology , Fruit/chemistry , Humans , Indicators and Reagents , Phenols/isolation & purification , Phosphorylation , Plant Extracts/chemistry , Plant Extracts/pharmacology , Platelet Aggregation/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Polyphenols , Receptors, Thrombin/metabolism , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL