Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 43(8): 1494-1509, 2023 08.
Article in English | MEDLINE | ID: mdl-37381987

ABSTRACT

BACKGROUND: MAGT1 (magnesium transporter 1) is a subunit of the oligosaccharide protein complex with thiol-disulfide oxidoreductase activity, supporting the process of N-glycosylation. MAGT1 deficiency was detected in human patients with X-linked immunodeficiency with magnesium defect syndrome and congenital disorders of glycosylation, resulting in decreased cation responses in lymphocytes, thereby inhibiting the immune response against viral infections. Curative hematopoietic stem cell transplantation of patients with X-linked immunodeficiency with magnesium defect causes fatal bleeding and thrombotic complications. METHODS: We studied the role of MAGT1 deficiency in platelet function in relation to arterial thrombosis and hemostasis using several in vitro experimental settings and in vivo models of arterial thrombosis and transient middle cerebral artery occlusion model of ischemic stroke. RESULTS: MAGT1-deficient mice (Magt1-/y) displayed accelerated occlusive arterial thrombus formation in vivo, a shortened bleeding time, and profound brain damage upon focal cerebral ischemia. These defects resulted in increased calcium influx and enhanced second wave mediator release, which further reinforced platelet reactivity and aggregation responses. Supplementation of MgCl2 or pharmacological blockade of TRPC6 (transient receptor potential cation channel, subfamily C, member 6) channel, but not inhibition of store-operated calcium entry, normalized the aggregation responses of Magt1-/y platelets to the control level. GP (glycoprotein) VI activation of Magt1-/y platelets resulted in hyperphosphorylation of Syk (spleen tyrosine kinase), LAT (linker for activation of T cells), and PLC (phospholipase C) γ2, whereas the inhibitory loop regulated by PKC (protein kinase C) was impaired. A hyperaggregation response to the GPVI agonist was confirmed in human platelets isolated from a MAGT1-deficient (X-linked immunodeficiency with magnesium defect) patient. Haploinsufficiency of TRPC6 in Magt1-/y mice could normalize GPVI signaling, platelet aggregation, and thrombus formation in vivo. CONCLUSIONS: These results suggest that MAGT1 and TRPC6 are functionally linked. Therefore, deficiency or impaired functionality of MAGT1 could be a potential risk factor for arterial thrombosis and stroke.


Subject(s)
Cation Transport Proteins , Homeostasis , Infarction, Middle Cerebral Artery , Ischemic Stroke , Thrombosis , Animals , Humans , Mice , Blood Platelets/metabolism , Calcium/metabolism , Cations/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/complications , Ischemic Stroke/metabolism , Magnesium/metabolism , Platelet Activation , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Thrombosis/genetics , Thrombosis/metabolism , TRPC6 Cation Channel/metabolism , Cation Transport Proteins/deficiency
2.
Pharmacol Res ; 178: 106180, 2022 04.
Article in English | MEDLINE | ID: mdl-35288308

ABSTRACT

Metastasis remains a crucial obstacle to the clinical treatment of hepatocellular carcinoma (HCC). Investigating the potential anti-tumor compounds from medicinal herb against HCC metastasis is of particular interest. As a triterpenoid saponin, α-Hederin has been reported to exhibit cytotoxicity for diverse cancer cell lines by inducing mitochondrial related apoptosis or autophagic cell death. Nevertheless, little is known about the inhibitory effect of α-Hederin on the metastasis of HCC and its underlying mechanisms. Here, we integrated well-established target prediction webtool and molecular docking methods to predict the potential targets for α-Hederin, and finally focused on PTAFR, the receptor for platelet-activating factor (PAF). Activation of PAF/PTAFR pathways has been reported to be contribution to the initiation and progression of cancer. We showed for the first time that non-cytotoxic concentration of α-Hederin inhibited cell migration and invasion induced by PAF in HCC cells, as well as lung metastasis in vivo. Moreover, we demonstrated α-Hederin reduced the PAF-induced matrix metalloproteinase-2 expression through inhibiting the activation of STAT3 in PAF stimulated HCC cells. These findings suggest that α-Hederin functions as a prospective inhibitor of PTAFR and may be utilized as an optional candidate for treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Matrix Metalloproteinase 2 , Oleanolic Acid , Platelet Activating Factor , Saponins , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Molecular Docking Simulation , Neoplasm Metastasis , Oleanolic Acid/pharmacology , Platelet Activating Factor/antagonists & inhibitors , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , STAT3 Transcription Factor , Saponins/pharmacology , Signal Transduction/drug effects
3.
Drug Des Devel Ther ; 15: 3543-3560, 2021.
Article in English | MEDLINE | ID: mdl-34429584

ABSTRACT

PURPOSE: We aimed to investigate potential synergistic antiplatelet effects of Ginkgo biloba extract (GBE50) in combination with aspirin using in vitro models. METHODS: Arachidonic acid (AA), platelet activating factor (PAF), adenosine 5'-diphosphate (ADP) and collagen were used as inducers. The antiplatelet effects of GBE50, aspirin and 1:1 combination of GBE50 and aspirin were detected by microplate method using rabbit platelets. Synergy finder 2.0 was used to analyze the synergistic antiplatelet effect. The compounds in GBE50 were identified by UPLC-Q/TOF-MS analysis and the candidate compounds were screened by TCMSP database. The targets of candidate compounds and aspirin were obtained in TCMSP, CCGs, Swiss target prediction database and drugbank. Targets involving platelet aggregation were obtained from GenCLiP database. Compound-target network was constructed and GO and KEGG enrichment analyses were performed to identify the critical biological processes and signaling pathways. The levels of thromboxane B2 (TXB2), cyclic adenosine monophosphate (cAMP) and PAF receptor (PAFR) were detected by ELISA to determine the effects of GBE50, aspirin and their combination on these pathways. RESULTS: GBE50 combined with aspirin inhibited platelet aggregation more effectively. The combination displayed synergistic antiplatelet effects in AA-induced platelet aggregation, and additive antiplatelet effects occurred in PAF, ADP and collagen induced platelet aggregation. Seven compounds were identified as candidate compounds in GBE50. Enrichment analyses revealed that GBE50 could interfere with platelet aggregation via cAMP pathway, AA metabolism and calcium signaling pathway, and aspirin could regulate platelet aggregation through AA metabolism and platelet activation. ELISA experiments showed that GBE50 combined with aspirin could increase cAMP levels in resting platelets, and decreased the levels of TXB2 and PAFR. CONCLUSION: Our study indicated that GBE50 combined with aspirin could enhance the antiplatelet effects. They exerted both synergistic and additive effects in restraining platelet aggregation. The study highlighted the potential application of GBE50 as a supplementary therapy to treat thrombosis-related diseases.


Subject(s)
Aspirin/pharmacology , Plant Extracts/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Animals , Arachidonic Acid/metabolism , Aspirin/administration & dosage , Chromatography, High Pressure Liquid , Cyclic AMP/metabolism , Drug Synergism , Ginkgo biloba , Male , Mass Spectrometry , Plant Extracts/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Membrane Glycoproteins/metabolism , Rabbits , Receptors, G-Protein-Coupled/metabolism , Thromboxane B2/metabolism
4.
Thromb Haemost ; 121(10): 1337-1344, 2021 10.
Article in English | MEDLINE | ID: mdl-33690868

ABSTRACT

Platelet activation is characterized by shape change, granule secretion, activation of fibrinogen receptor (glycoprotein IIb/IIIa) sustaining platelet aggregation, and externalization of negatively charged aminophospholipids contributing to platelet procoagulant activity. Epinephrine (EPI) alone is a weak platelet activator. However, it is able to potentiate platelet activation initiated by other agonists. In this work, we investigated the role of EPI in the generation of procoagulant platelets. Human platelets were activated with convulxin (CVX), thrombin (THR) or protease-activated receptor (PAR) agonists, EPI, and combination thereof. Platelet aggregation was assessed by light transmission aggregometry or with PAC-1 binding by flow cytometry. Procoagulant collagen-and-THR (COAT) platelets, induced by combined activation with CVX-and-THR, were visualized by flow cytometry as Annexin-V-positive and PAC-1-negative platelets. Cytosolic calcium fluxes were monitored by flow cytometry using Fluo-3 indicator. EPI increased platelet aggregation induced by all agonist combinations tested. On the other hand, EPI dose-dependently reduced the formation of procoagulant COAT platelets generated by combined CVX-and-THR activation. We observed a decreased Annexin-V-positivity and increased binding of PAC-1 with the triple activation (CVX + THR + EPI) compared with CVX + THR. Calcium mobilization with triple activation was decreased with the higher EPI dose (1,000 µM) compared with CVX + THR calcium kinetics. In conclusion, when platelets are activated with CVX-and-THR, the addition of increasing concentrations of EPI (triple stimulation) modulates platelet response reducing cytosolic calcium mobilization, decreasing procoagulant activity, and enhancing platelet aggregation.


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/drug effects , Coagulants/pharmacology , Epinephrine/pharmacology , Platelet Aggregation/drug effects , Adolescent , Adult , Aged , Blood Platelets/metabolism , Calcium Signaling , Crotalid Venoms/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Kinetics , Lectins, C-Type , Male , Middle Aged , Platelet Membrane Glycoproteins/agonists , Platelet Membrane Glycoproteins/metabolism , Receptors, Proteinase-Activated/agonists , Receptors, Proteinase-Activated/metabolism , Thrombin/pharmacology , Young Adult
5.
Phytomedicine ; 80: 153363, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33070081

ABSTRACT

BACKGROUND: The ingestion of flavonoids has been reported to be associated with reduced cardiovascular disease risk. Quercitrin is a common flavonoid in nature, and it exhibits antioxidant properties. Although the process of thrombogenesis is intimately related to cardiovascular disease risk, it is unclear whether quercitrin plays a role in thrombogenesis. PURPOSE: The aim of this study was to examine the antiplatelet effect of quercitrin in platelet activation. METHODS: Platelet aggregation, granule secretion, calcium mobilization, and integrin activation were used to assess the antiplatelet activity of quercitrin. Antithrombotic effect was determined in mouse using ferric chloride (FeCl3)-induced arterial thrombus formation in vivo and thrombus formation on collagen-coated surfaces under arteriolar shear in vitro. Transection tail bleeding time was used to evaluate whether quercitrin inhibited primary hemostasis. RESULTS: Quercitrin significantly impaired collagen-related peptide-induced platelet aggregation, granule secretion, reactive oxygen species generation, and intracellular calcium mobilization. Outside-in signaling of αIIbß3 integrin was significantly inhibited by quercitrin in a concentration-dependent manner. The inhibitory effect of quercitrin resulted from inhibition of the glycoprotein VI-mediated platelet signal transduction during cell activation. Further, the antioxidant effect is derived from decreased phosphorylation of components of the TNF receptor-associated factor 4/p47phox/Hic5 axis signalosome. Oral administration of quercitrin efficiently blocked FeCl3-induced arterial thrombus formation in vivo and thrombus formation on collagen-coated surfaces under arteriolar shear in vitro, without prolonging bleeding time. Studies using a mouse model of ischemia/reperfusion-induced stroke indicated that treatment with quercitrin reduced the infarct volume in stroke. CONCLUSIONS: Our results demonstrated that quercitrin could be an effective therapeutic agent for the treatment of thrombotic diseases.


Subject(s)
Fibrinolytic Agents/pharmacology , Hemostasis/drug effects , Quercetin/analogs & derivatives , Thrombosis/drug therapy , Adenosine Triphosphate/metabolism , Animals , Arteries , Calcium/metabolism , Dose-Response Relationship, Drug , Humans , Male , Mice, Inbred C57BL , Phosphorylation/drug effects , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Quercetin/adverse effects , Quercetin/pharmacology , Reperfusion Injury/chemically induced , Thrombosis/chemically induced , Thrombosis/metabolism
6.
Molecules ; 25(21)2020 Nov 06.
Article in English | MEDLINE | ID: mdl-33172028

ABSTRACT

Saponins comprise a heterogenous group of chemical compounds containing a triterpene or steroid aglycone group and at least one sugar chain. They exist as secondary metabolites, occurring frequently in dicotyledonous plants and lower marine animals. Plant saponin extracts or single saponins have indicated antiplatelet and anticoagulant activity. Venous thromboembolism (VTE), including deep venous thrombosis and pulmonary embolism, is a multifactorial disease influenced by various patient characteristics such as age, immobility, previous thromboembolism and inherited thrombophilia. This mini-review (1) evaluates the current literature on saponins as modulators of the coagulation system, (2) discusses the impact of chemical structure on the modulation of the coagulation system, which may further provide a basis for drug or supplement design, (3) examines perspectives of their use in the prevention of VTE. It also describes the molecular mechanisms of action of the saponins involved in the prevention of VTE.


Subject(s)
Blood Coagulation/drug effects , Saponins/pharmacology , Venous Thrombosis/prevention & control , Animals , Arachidonic Acid/metabolism , Blood Coagulation/physiology , Humans , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/metabolism , Pulmonary Embolism/prevention & control , Saponins/adverse effects , Saponins/chemistry , Saponins/therapeutic use
7.
Thromb Haemost ; 119(8): 1212-1221, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31087308

ABSTRACT

Bruton's tyrosine kinase (Btk) is essential for B cell differentiation and proliferation, but also platelets express Btk. Patients with X-linked agammaglobulinemia due to hereditary Btk deficiency do not show bleeding, but a mild bleeding tendency is observed in high dose therapy of B-cell malignancies with ibrutinib and novel second-generation irreversible Btk inhibitors (acalabrutinib and ONO/GS-4059). This review discusses recent studies that may explain this apparent paradox and gives mechanistic insights that suggest a unique potential of low dose irreversible Btk inhibitors as atherothrombosis-focused antiplatelet drugs.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Platelet Aggregation Inhibitors/pharmacology , Thrombosis/drug therapy , Adenine/analogs & derivatives , Administration, Oral , Agammaglobulinaemia Tyrosine Kinase/deficiency , Agammaglobulinemia/drug therapy , Animals , Arteries/pathology , B-Lymphocytes/cytology , Benzamides/pharmacology , Blood Platelets/drug effects , Cell Differentiation , Genetic Diseases, X-Linked/drug therapy , Hemorrhage , Humans , Imidazoles/pharmacology , Mice , Piperidines , Platelet Membrane Glycoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction
8.
Platelets ; 30(5): 589-598, 2019.
Article in English | MEDLINE | ID: mdl-29870296

ABSTRACT

Panax ginseng (P. ginseng), one of the most valuable medicinal plants, is known for its healing and immunobooster properties and has been widely used in folk medicine against cardiovascular diseases, including stroke and heart attack. In this study, we explored the anti-platelet activity of gintonin (a recently discovered non-saponin fraction of ginseng) against agonist-induced platelet activation. In vitro effects of gintonin on agonist-induced human and rat platelet aggregation, granule secretion, integrin αIIbß3 activation, and intracellular calcium ion ([Ca2+]i) mobilization were examined. Western blot analysis and immunoprecipitation techniques were used to estimate the expression of mitogen-activated protein kinases (MAPKs) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) and interaction of glycoprotein VI (GPVI) signaling pathway molecules such as Src family kinases (SFK), tyrosine kinase Syk, and PLCγ2. In vivo effects were studied using acute pulmonary thromboembolism model in mice. Gintonin remarkably inhibited collagen-induced platelet aggregation and suppressed granule secretion, [Ca2+]i mobilization, and fibrinogen binding to integrin αIIbß3 in a dose-dependent manner and clot retraction. Gintonin attenuated the activation of MAPK molecules and PI3K/Akt pathway. It also inhibited SFK, Syk, and PLCγ2 activation and protected mice from thrombosis. Gintonin inhibited agonist-induced platelet activation and thrombus formation through impairment in GPVI signaling molecules, including activation of SFK, Syk, PLCγ2, MAPK, and PI3K/Akt; suggesting its therapeutic potential against platelet related CVD.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Plant Extracts/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/metabolism , Signal Transduction/drug effects , Thrombosis/metabolism , Animals , Biomarkers , Blood Coagulation/drug effects , Blood Platelets/ultrastructure , Disease Models, Animal , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Panax/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Thrombosis/drug therapy , Thrombosis/etiology
9.
J Nutr ; 147(10): 1917-1925, 2017 10.
Article in English | MEDLINE | ID: mdl-28855423

ABSTRACT

Background: Platelets play an important role in hemostasis, thrombosis, and atherosclerosis. Glycoprotein VI (GPVI) is a major platelet receptor that interacts with exposed collagen on injured vessel walls. Our previous studies have shown that anthocyanins (a type of natural plant pigment) attenuate platelet function; however, whether anthocyanins affect collagen-induced GPVI signaling remains unknown.Objective: The objective of this study was to explore the effects of cyanidin-3-glucoside (Cy-3-g, one of the major bioactive compounds in anthocyanins) on platelet activation and thrombosis and the GPVI signaling pathway.Methods: Platelets from healthy men and women were isolated and incubated with different concentrations (0, 0.5, 5, and 50 µM) of Cy-3-g. The expression of activated integrin αIIbß3, P-selectin, CD63, and CD40L, fibrinogen binding to platelets, and platelet aggregation were evaluated in vitro. Platelet adhesion and aggregation in whole blood under flow conditions were assessed in collagen-coated perfusion chambers. Thrombosis and hemostasis were assessed in 3-4-wk-old male C57BL/6J mice through FeCl3-induced intravital microscopy and tail bleeding time. The effect of Cy-3-g on collagen-induced human platelet GPVI signaling was explored with Western blot.Results: Cy-3-g attenuated platelet function in a dose-dependent manner. The 0.5-µM dose of Cy-3-g inhibited (P < 0.05) human platelet adhesion and aggregation to collagen at both venous (-54.02%) and arterial (-22.90%) shear stresses. The 5-µM dose inhibited (P < 0.05) collagen-induced human platelet activation (PAC-1: -48.21%, P-selectin: -50.63%), secretion (CD63: -73.89%, CD40L: -43.70%), fibrinogen binding (-56.79%), and aggregation (-17.81%). The 5-µM dose attenuated (P < 0.01) thrombus growth (-66.67%) without prolonging bleeding time in mice. The 50-µM dose downregulated (P < 0.05) collagen-induced GPVI signaling in human platelets and significantly decreased phosphorylation of Syk-linker for activation of T cells (LAT)-SLP76 (Syk: -39.08%, LAT: -32.25%, SLP76: -40.00%) and the expression of Lyn (-31.89%), Fyn (-36.27%), and phospholipase C-γ2 (-39.08%).Conclusions: Cy-3-g inhibits human platelet activation, aggregation, secretion, and thrombus formation, and downregulates the collagen-GPVI signaling pathway. Supplementation of Cy-3-g may have protective effects against atherothrombosis.


Subject(s)
Blood Platelets/metabolism , Phytotherapy , Plant Extracts/pharmacology , Plants, Edible/chemistry , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/metabolism , Thrombosis/prevention & control , Adaptor Proteins, Signal Transducing/blood , Adult , Aged , Animals , Anthocyanins/pharmacology , Anthocyanins/therapeutic use , Antigens, CD/blood , Atherosclerosis/blood , Atherosclerosis/diet therapy , Atherosclerosis/etiology , Collagen/blood , Female , Glucosides/pharmacology , Glucosides/therapeutic use , Hemostasis/drug effects , Humans , Male , Mice, Inbred C57BL , Middle Aged , P-Selectin/blood , Phosphoproteins/blood , Plant Extracts/therapeutic use , Platelet Activation/drug effects , Signal Transduction , Thrombosis/blood , Thrombosis/etiology
10.
Pharmacol Res ; 113(Pt A): 484-489, 2016 11.
Article in English | MEDLINE | ID: mdl-27693274

ABSTRACT

Anti Xa non-vitamin K oral anticoagulants (anti Xa NOACs) seem to possess antiplatelet effect in vitro, but it is unclear if this occurs also in vivo. Aim of the study was to compare the effect on platelet activation of two anti Xa NOACs, namely apixaban and rivaroxaban, to warfarin, and to investigate the potential underlying mechanism by evaluating soluble glycoprotein GPVI (sGPVI), a protein involved in platelet activation. We performed a cross-sectional including AF patients treated with warfarin (n=30), or apixaban 10mg/day (n=40), or rivaroxaban 20mg/day (n=40). Patients were balanced for sex, age and cardiovascular risk factors. Platelet activation by urinary excretion of 11-dehydro-thromboxane (Tx) B2 and soluble GPVI (sGPVI) were analysed at baseline and after 3 months of treatment. Baseline TxB2 value was 155.2±42.7ng/mg creatinine. The 3 months-variation of urinary excretion of TxB2 was -6.5% with warfarin (p=0.197), -29% with apixaban (p<0.001) and -31% with rivaroxaban (p<0.001). Use of anti Xa NOACs was independently associated to the variation of urinary TxB2 (B: -0.469, p<0.001), after adjustment for clinical characteristics; sGPVI was significantly lower in patients treated with NOACs at 3 months (p<0.001), while only a trend for the warfarin group (p=0.116) was observed. The variation of sGPVI was correlated with that of TxB2 in the NOACs group (Rs: 0.527, p<0.001). In 15 patients (5 per each group) platelet recruitment was significantly lowered at 3 months by NOACs (p<0.001), but not by warfarin. The study provides evidence that anti Xa NOACs significantly inhibit urinary TxB2 excretion compared to warfarin, suggesting that NOACs possess antiplatelet property.


Subject(s)
Anticoagulants/therapeutic use , Blood Platelets/drug effects , Factor Xa Inhibitors/therapeutic use , Platelet Activation/drug effects , Platelet Membrane Glycoproteins/metabolism , Aged , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Atrial Fibrillation/urine , Blood Platelets/metabolism , Cross-Sectional Studies , Female , Humans , Male , Platelet Aggregation Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyridones/therapeutic use , Risk Factors , Rivaroxaban/therapeutic use , Thromboxane B2/analogs & derivatives , Thromboxane B2/urine , Warfarin/therapeutic use
11.
Platelets ; 27(4): 322-32, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26559117

ABSTRACT

Anti-platelet agents play a central part in the treatment and prevention of acute thrombotic events. Discriminating animal models are needed for the development of novel agents. The chacma baboon has been extensively used as a model to evaluate anti-platelet agents. However, limited data exist to prove the translatability of this species to humans. We aimed to determine the suitability of the chacma baboon in preclinical human targeted GPIIb/IIIa, GPIbα and P2Y12 studies. Light-transmission platelet aggregometry (LTA), whole blood impedance aggregometry, receptor number quantification and genomic DNA sequencing were performed. Baboon ADP and arachidonic acid-induced LTA aggregation results differed significantly from human values, even at increased concentrations. LTA ristocetin-induced agglutination was comparable between species, but baboon platelets needed twice the concentration of ristocetin to elicit a similar response. Citrated baboon blood had significantly less aggregation than humans when evaluated with impedance aggregometry. However, hirudinised baboon whole blood gave similar aggregation as humans at the same agonist concentrations. GPIIb, GPIIIa and GPIbα numbers were significantly more on the baboon platelets. None of the amino acids deemed vital for receptor function, ligand binding or receptor inhibition, were radically different between the species. However, a conservative change in a calcium-binding region of GPIIb may render the baboon platelets more sensitive to calcium-binding agents. The chacma baboon may be used for the evaluation of human-targeted GPIIb/IIIa-, GPIbα- and P2Y12-inhibiting agents. However, the best anticoagulant, optimal agonist concentrations, increase in receptor number and sequence differences must be considered for any future studies.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Adenosine Diphosphate/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Arachidonic Acid/pharmacology , Drug Evaluation, Preclinical , Flow Cytometry , Humans , Male , Papio ursinus , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Ristocetin/pharmacology
12.
Platelets ; 26(6): 552-7, 2015.
Article in English | MEDLINE | ID: mdl-25165849

ABSTRACT

An effective and safe anti-platelet drug is central to the management of patients with acute coronary syndrome (ACS). Glycoprotein VI (GPVI) is currently regarded as a potential target for novel anti-platelet agents due to its collagen-binding potential. Development of anti-thrombotics is associated with testing in animals. We have previously successfully evaluated anti-platelet drugs in the Cape Chacma baboon (Papio ursinus). However, various anti-GPVI agents did not have an effect on baboons when evaluated in our arterial thrombosis model. To evaluate the suitability of baboons for GPVI studies, we performed collagen-induced platelet aggregation, GPVI quantification and DNA sequencing. Baboon platelets needed double the amount of collagen compared to human platelets to illicit proper aggregation. GPVI quantification was unsuccessful due to non-binding of monoclonal antibodies. Sequencing of the GPVI gene revealed 36 SNPs leading to 14 amino acid changes, as well as a 9 bp deletion causing a 3 amino acid deletion. Several of the amino acid changes were within the ligand binding region of GPVI, causing limited binding of humanized anti-GPVI antibodies to the baboon platelets. Therefore, the baboon was deemed not suitable to evaluate human targeted anti-GPVI agents.


Subject(s)
Antibodies, Monoclonal/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Amino Acid Substitution , Animals , Antibodies, Monoclonal/therapeutic use , Base Sequence , Collagen/metabolism , Collagen/pharmacology , Drug Evaluation, Preclinical , Humans , Models, Animal , Molecular Sequence Data , Papio ursinus , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Polymorphism, Single Nucleotide , Sequence Alignment
13.
Molecules ; 17(9): 10893-901, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22964504

ABSTRACT

The methanol extract of the leaves of Garcinia nervosa var. pubescens King, which showed strong inhibitory effects on platelet-activating factor (PAF) receptor binding, was subjected to bioassay-guided isolation to obtain a new biflavonoid, II-3,I-5, II-5,II-7,I-4',II-4'-hexahydroxy-(I-3,II-8)-flavonylflavanonol together with two known flavonoids, 6-methyl-4'-methoxyflavone and acacetin. The structures of the compounds were elucidated by spectroscopic methods. The compounds were evaluated for their ability to inhibit PAF receptor binding to rabbit platelets using ³H-PAF as a ligand. The biflavonoid and acacetin showed strong inhibition with IC50 values of 28.0 and 20.4 µM, respectively. The results suggest that these compounds could be responsible for the strong PAF antagonistic activity of the plant.


Subject(s)
Biflavonoids/pharmacology , Garcinia/chemistry , Platelet Activating Factor/antagonists & inhibitors , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Biflavonoids/chemistry , Biflavonoids/isolation & purification , Blood Platelets/drug effects , Blood Platelets/metabolism , Flavones/chemistry , Flavones/pharmacology , Inhibitory Concentration 50 , Plant Extracts/chemistry , Plant Leaves/chemistry , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/metabolism , Rabbits , Receptors, G-Protein-Coupled/metabolism
14.
Pharm Biol ; 50(3): 284-90, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22103812

ABSTRACT

CONTEXT: Enicosanthellum pulchrum (King) Heusden (Annonaceae) is a coniferous tree that is confined to mountain forests. The chemical constituents of this species have been studied previously; however, its biological activity has never been investigated before and is reported here for the first time. OBJECTIVE: The extracts, fractions and compounds from the roots of E. pulchrum were investigated for their inhibitory effects on platelet-activating factor (PAF) receptor binding to rabbit platelets using (3)H-PAF as a ligand. MATERIALS AND METHODS: The PAF receptor binding inhibitory effect using rabbit platelets was determined in vitro by measuring the difference between total amount of bound (3)H-PAF in the presence and the absence of excess unlabelled PAF. The compounds were isolated by bioassay-guided fractionation and their structures were elucidated by spectroscopic techniques. RESULTS AND DISCUSSION: Among the extracts tested, the ethyl acetate extract was the most active with 85.6% inhibition, while hexane and methanol extracts showed 40.2 and 42.5% inhibition, respectively. Fractionation of the ethyl acetate extract using vacuum liquid chromatography (VLC) yielded six fractions AEA(I--VI). Chromatography fraction AEA(VI) yielded a new compound, 1-(2',3',4'-trimethoxyphenyl)hexan-1-ol, while fraction AEA(III) afforded three compounds, namely liriodenine, cleistopholine and dehydroanonaine. 1-(2',3',4'-Trimethoxyphenyl)hexan-1-ol, cleistopholine and dehydroanonaine showed relatively strong inhibition with IC(50) values of 26.6, 50.2 and 45.4 µM, respectively. CONCLUSION: The results suggest that these compounds could be responsible for the PAF antagonistic activity of the ethyl acetate extract of this plant.


Subject(s)
Annonaceae/chemistry , Blood Platelets/metabolism , Plant Extracts/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Chromatography, Liquid , In Vitro Techniques , Inhibitory Concentration 50 , Plant Extracts/administration & dosage , Plant Roots , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein Binding , Rabbits , Receptors, G-Protein-Coupled/metabolism , Spectrophotometry, Ultraviolet
16.
Cardiology ; 118(3): 187-94, 2011.
Article in English | MEDLINE | ID: mdl-21701167

ABSTRACT

BACKGROUND: Prescription omega-3-acid ethyl esters (PO-3A) have been tested for outcome benefits in patients with coronary artery disease (CAD), arrhythmias and heart failure. Some evidence suggests that PO-3A may exert their benefit via inhibiting platelets. We tested the hypothesis that PO-3A may inhibit platelet activity in patients with documented stable CAD, beyond the antiplatelet properties of aspirin and statins. METHODS: Thirty patients with documented CAD and triglycerides over 250 mg/dl treated with aspirin (70-160 mg/daily) and statins (simvastatin equivalence dose: 5-40 mg/daily) were randomized 1:1:1 to Omacor™ 1 g/day (DHA/EPA ratio 1.25:1.0), Omacor 2 g/day, or a placebo for 2 weeks. Platelet tests including aggregometry and flow cytometry and cartridge analyzer readings were performed at baseline and at 1 and 2 weeks following PO-3A therapy. RESULTS: ADP-induced platelet aggregation (p = 0.037), GP IIb/IIIa antigen (p = 0.031) and activity (p = 0.024), and P-selectin (p = 0.041) were significantly reduced after PO-3A, while platelet/endothelial cell adhesion molecule (p = 0.09), vitronectin receptor (p = 0.16), formation of platelet-monocyte microparticles (p = 0.19) and the VerifyNow IIb/IIIa test (p = 0.27) only exhibited nonsignificant trends suggestive of reduced platelet activity. Finally, collagen- and arachidonic acid-induced aggregation, closure time with the PFA-100 device and expression of thrombospondin (CD36), GP Ib (CD42b), LAMP-3 (CD63), LAMP-1 (CD107a), CD40-ligand (CD154), GP37 (CD165), and PAR-1 receptor intact (SPAN 12) and cleaved (WEDE-15) epitopes were not affected by 2 weeks of PO-3A. CONCLUSION: Independently of the dose and already at 1 week, short-term therapy with PO-3A provided a modest reduction of platelet activity biomarkers, despite concomitant aspirin and statin therapy, when compared to a placebo. The effect of PO-3A is unique, differs from other known antiplatelet agents and suggests potential pleiotropism. These preliminary randomized data call for confirmation in prospective studies.


Subject(s)
Coronary Artery Disease/blood , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Hypertriglyceridemia/blood , Platelet Membrane Glycoproteins/metabolism , Aged , Biomarkers/blood , Coronary Artery Disease/complications , Coronary Artery Disease/drug therapy , Double-Blind Method , Drug Combinations , Female , Humans , Hypertriglyceridemia/complications , Hypertriglyceridemia/drug therapy , Male , Middle Aged , Platelet Activation/physiology , Platelet Function Tests
17.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 31(4): 483-6, 2011 Apr.
Article in Chinese | MEDLINE | ID: mdl-21608218

ABSTRACT

OBJECTIVE: To observe the effect of electro-needling at acupoints of the yangming meridian on the expression of platelet associated complement-1 (PAC-1) and lower limb functions in acute cerebral infarction (ACI) patients. METHODS: 58 ACI patients were randomly assigned to the treatment group and the control group. Conventional therapies were given to all patients. Additionally, the electro-needling at acupoints of the yangming meridian was given to patients in the treatment group. Changes of PAC-1 were detected using flow cytometry. Effect of lower limbs functions of ACI patients before and after electro-needling was assessed using Fugl-Meyer Index. Meanwhile, 20 healthy subjects were selected for reference value. RESULTS: In the acute stage, the PAC-1 level in ACI patients were significantly higher than that in healthy subjects (P<0.05). The PAC-1 level in the electro-needling group was obviously lowered after treatment (P<0.01). There was no significant difference in the control group between before and after treatment. Significant difference was found in Fugl-Meyer index in the same group between before and after two-week treatment (P<0.05). It was higher in the electro-needling group than in the control group, showing significant difference (P<0.01). CONCLUSIONS: Platelet activation exists in the acute stage of ACI. Electro-needling at acupoints of the yangming meridian showed obvious inhibition on PAC-1 levels, could improve lower limbs functions of ACI patients. It was inferred that electro-needling at acupoints of the yangming meridian promoted the recovery of paralyzed lower limbs at the early stage mainly by regulating PAC-1 levels, thus postponing the progress of ACI.


Subject(s)
Acupuncture Therapy , Cerebral Infarction/metabolism , Cerebral Infarction/therapy , Platelet Membrane Glycoproteins/metabolism , Acupuncture Points , Adult , Aged , Aged, 80 and over , Cerebral Infarction/rehabilitation , Electroacupuncture , Female , Humans , Lower Extremity , Male , Middle Aged , Recovery of Function , Stroke Rehabilitation , Treatment Outcome
18.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 31(3): 346-9, 2011 Mar.
Article in Chinese | MEDLINE | ID: mdl-21485076

ABSTRACT

OBJECTIVE: To study the correlation between prethrombotic biomarkers and traditional Uyghur medicinal syndromes of malignant tumor. METHODS: One hundred and fifty-nine malignant tumor inpatients were randomly selected and typed according to traditional Uyghur medicine theories. The expressions of peripheral platelet membrane glucoprotein (CD41 and CD62p), levels of serum endothelin (ET-1), plasma tissue plasminogen activator (t-PA), plasminogen activator inhibitor (PAI), plasma fibrinogen (FIB), prothrombin time (PT), and thrombin time (TT), and activated partial thromboplastin time (APTT) were detected by flow cytometry, radioimmunoassay, and enzyme-linked immunosorbent assay (ELISA) respectively. RESULTS: The patients were typed as 68 of abnormal Savda syndrome, 34 of abnormal Khan syndrome, 31 of the abnormal Sepra syndrome, and 26 of the abnormal Belghem syndrome. Compared with the control group, the levels of CD62p, PAI, and ET-1 increased, levels of FIB and t-PA decreased, PT prolongated and APTT shortened in the abnormal Savda group and the non-abnormal Savda group (including abnormal Khan, abnormal Sepra, and abnormal Belghem types) (all P < 0.05). No significant difference of CD41 or TT was shown in inter-group comparison (P > 0.05). Compared with abnormal Khan and Belghem groups, the ET-1 level increased in the abnormal Savda group (P < 0.05). Compared with the abnormal Sepra group, the CD62p positive percentage increased in abnormal Savda group (P < 0.05). No significant difference in CD41 positive percentage, t-PA or PAI-1 contents, PT, TT, or APTT was shown in patients of different traditional Uyghur medicine syndrome groups (P > 0.05). CONCLUSIONS: Prethrombotic changes existed in malignant tumor patients of different Uyghur abnormal syndrome types, manifested as injuries of vessel epithelial cells, platelet activation, increased blood viscosity, lowered fibrinolytic function. The prethrombotic changes were more obviously seen in the abnormal Savda group.


Subject(s)
Medicine, Chinese Traditional/methods , Neoplasms/blood , Neoplasms/diagnosis , Adult , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Minority Groups , Partial Thromboplastin Time , Plasminogen Activator Inhibitor 1/metabolism , Platelet Membrane Glycoproteins/metabolism , Tissue Plasminogen Activator/metabolism
19.
Am J Pathol ; 178(2): 699-708, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21281802

ABSTRACT

Platelet-activating factor (PAF), a potent biolipid mediator, is involved in a variety of cellular transduction pathways and plays a prominent role in inducing inflammation in different organs. We used K5.hTGF-ß1 transgenic mice, which exhibit an inflammatory skin disorder and molecular and cytokine abnormalities with strong similarities to human psoriasis, to study the pathogenic role of PAF. We found that injecting PAF into the skin of transgenic mice led to inflammation and accelerated manifestation of the psoriatic phenotype by a local effect. In contrast, injecting mice with PAF receptor antagonist PCA-4248 lowered the PAF level (most likely by depressing an autocrine loop) and neutrophil, CD68(+) cell (monocyte/macrophage), and CD3(+) T-cell accumulation in the skin and blocked progression of the psoriasis-like phenotype. This effect of PAF blockade was specific and similar to that of psoralen-UV-A and was paralleled by a decrease in abnormally elevated mRNA and/or protein levels of T-helper type 17 cell-related cytokines IL-17A, IL-17F, IL-23, IL-12A, and IL-6 and its transcription factor signal transducer and activator of transcription 3. In contrast, PCA-4248 treatment up-regulated mRNA levels of cyclooxygenase-2 and IL-10 in dorsal skin and release of IL-10 in serum and skin. Interfering with PAF may offer the opportunity to develop novel therapeutic strategies for inflammatory psoriasis and associated comorbidities, including metabolic syndrome and atherosclerosis, in which the IL-17 axis may be involved.


Subject(s)
Platelet Activating Factor/antagonists & inhibitors , Psoriasis/pathology , Signal Transduction , Th17 Cells/metabolism , Transforming Growth Factor beta1/genetics , Animals , Cytokines/genetics , Cytokines/metabolism , Dihydropyridines/administration & dosage , Dihydropyridines/pharmacology , Disease Progression , Down-Regulation/drug effects , Humans , Mice , Mice, Transgenic , PUVA Therapy , Phenotype , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/metabolism , Psoriasis/drug therapy , Psoriasis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism , Skin/pathology , Th17 Cells/drug effects , Th17 Cells/immunology , Transforming Growth Factor beta1/metabolism , Up-Regulation/drug effects
20.
Zhongguo Zhong Yao Za Zhi ; 35(24): 3382-5, 2010 Dec.
Article in Chinese | MEDLINE | ID: mdl-21438412

ABSTRACT

OBJECTIVE: To investigate the effects of Naoxinduotai capsule on the markers of prothrombotic state as plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor (vWF) and alpha-granular membrane protein (CD62p) in essential hypertension patients of yang hyperactivity and blood stagnation. METHOD: The 62 essential hypertension patients of yang hyperactivity and blood stagnation were divided into Naoxinduotai capsule + perindopril group (treatment group, 30 subjects) and Perindopril group (control group, 32 subjects). Clinical symptoms, blood pressure and the blood plasma PAI-1, vWF, CD62p of the patients were observed. The blood plasma PAI-1, vWF and CD62p were measured by enzyme-linked immunosorbant assay (ELISA). RESULT: After 8 weeks treatment, in treatment group the clinical symptoms became better, and there was a significant difference with control group (P < 0.05). Blood pressure was significantly degraded, but there was not a significant difference with control group. The blood plasma PAI-1, vWF, CD62p level was degraded ,and there were significant differences with control group in all the three makers (P < 0.05). CONCLUSION: Naoxinduotai capsule can treat the essential hypertension patients of Yang hyperactivity and blood stagnation, and it has conspicuous advantages in improving the clinical symptoms and the makers of prothrombotic state.


Subject(s)
Hypertension/drug therapy , Thrombosis/blood , Aged , Biomarkers/blood , Blood Pressure/drug effects , Capsules , Female , Humans , Hypertension/blood , Hypertension/physiopathology , Male , P-Selectin/blood , Plasminogen Activator Inhibitor 1/blood , Platelet Membrane Glycoproteins/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL