Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
Add more filters

Complementary Medicines
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.
Int Immunopharmacol ; 101(Pt A): 108181, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607229

ABSTRACT

Demyelinating diseases of the central nervous system are characterized by recurrent demyelination and progressive neurodegeneration, but there are no clinical drugs targeting myelin regeneration or improving functional disability in the treatment of multiple sclerosis. Total flavone of Epimedium (TFE) is the main active components of Epimedium, which exhibits the beneficial biological activities in the treatment of diseases, but there is no report in the treatment of demyelinating disorder. The purpose of this study was to explore the therapeutic potential and possible mechanism of TFE in the treatment of demyelination. The results showed that TFE efficiently improved the behavioural performance and histological demyelination in cuprizone (CPZ)-induced demyelinating model. In terms of action, TFE increased astrocytes enrichment in corpus callosum, striatum and cortex, and promoted astrocytes to express neurotrophic factors. Furthermore, the expression of platelet-activating factor receptor (PAFR) in astrocytes was induced by CPZ feeding and LPS stimulation, accompanied by the increase of inflammatory cytokines TNF-α,IL-6 and IL-1ß. TFE declined the expression of PAFR, and inhibited inflammatory response. At the same time, TFE also antagonized PAFR activation and inflammatory response triggered by PAF, which further confirmed that TFE, as a new PAFR antagonist, inhibited the astrocyte-derived inflammatory response by antagonizing PAFR-neuroinflammation axis, thus contributing to myelin protection and regeneration.


Subject(s)
Demyelinating Diseases/drug therapy , Epimedium/chemistry , Neuroinflammatory Diseases/drug therapy , Plant Extracts/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Cuprizone/administration & dosage , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Flavones/pharmacology , Flavones/therapeutic use , Humans , Male , Mice , Myelin Sheath/drug effects , Myelin Sheath/immunology , Myelin Sheath/pathology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Plant Extracts/therapeutic use
4.
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
5.
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
6.
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
7.
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
8.
Thromb Haemost ; 120(11): 1492-1504, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32772352

ABSTRACT

The pathogenesis of atherosclerotic vascular disease is driven by a multitude of risk factors intertwining metabolic and inflammatory pathways. Increasing knowledge about platelet biology sheds light on how platelets take part in these processes from early to later stages of plaque development. Recent insights from experimental studies and mouse models substantiate platelets as initiators and amplifiers in atherogenic leukocyte recruitment. These studies are complemented by results from genetics studies shedding light on novel molecular mechanisms which provide an interesting prospect as novel targets. For instance, experimental studies provide further details how platelet-decorated von Willebrand factor tethered to activated endothelial cells plays a role in atherogenic monocyte recruitment. Novel aspects of platelets as atherogenic inductors of neutrophil extracellular traps and particularities in signaling pathways such as cyclic guanosine monophosphate and the inhibitory adaptor molecule SHB23/LNK associating platelets with atherogenesis are shared. In summary, it was our intention to balance insights from recent experimental data that support a plausible role for platelets in atherogenesis against a paucity of clinical evidence needed to validate this concept in humans.


Subject(s)
Atherosclerosis/drug therapy , Blood Platelets/physiology , Animals , Blood Platelets/drug effects , Chemotaxis, Leukocyte , Coronary Disease/blood , Coronary Disease/genetics , Drug Evaluation, Preclinical , Endothelial Cells/pathology , Extracellular Traps/physiology , Genetic Predisposition to Disease , Lipids/blood , Lipids/physiology , Mice , Nitric Oxide/physiology , P-Selectin/physiology , Plaque, Atherosclerotic/metabolism , Platelet Adhesiveness , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/physiology , Risk , von Willebrand Factor/physiology
9.
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
10.
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
11.
Zhongguo Zhong Yao Za Zhi ; 43(7): 1392-1403, 2018 Apr.
Article in Chinese | MEDLINE | ID: mdl-29728028

ABSTRACT

Platelet activating factor(PAF), an endogenous synthesized phospholipid transmitter, has widely biological activities. It has "signal transmission" effect in various life processes, but abnormality of concentration in vivo will promote or aggravate the diseases, such as, cerebral ischemia, myocardial injury, multi-organ failure, asthma, injury of liver and kidney, severely affecting the normal life activities of body. In recent years, with the development of medical science and technology, more and more attention has been paid to the research of platelet activating factor receptor antagonist. Components of animals, plants, microbial fermentation, and synthetic composition all can reflect such activity. Ginkgolide B and cytopone are the most representative herbal compositions at present. This paper referred to the research status of platelet activating factor receptor antagonist in recent years, made a summary of the researches on biological effect of platelet activating factor and platelet receptor antagonist of different sources, so as to provide a reference for the exploration of effective and safe platelet activating factor receptor antagonists.


Subject(s)
Platelet Activating Factor , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Humans , Plants
12.
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
13.
Virus Res ; 233: 51-59, 2017 04 02.
Article in English | MEDLINE | ID: mdl-28279802

ABSTRACT

Viral infections may predispose the airways to secondary bacterial infections that can lead to unfavorable progression of principally self-limiting illnesses. Such complicated respiratory infections include pneumonia, bronchitis, sinusitis, acute otitis media, and sepsis, which cause high morbidity and lethality. Some of the pathogenic consequences of viral infections, like the expression of bacterial adhesion receptors and the disturbance of physical barrier integrity due to inflammation, may create permissive conditions for co-infections. Influenza virus A (H3N2) is a major pathogen that causes secondary bacterial infections and inflammation that lead to pneumonia. The herbal medicine Echinacea purpurea, on the other hand, has been widely used to prevent and treat viral respiratory infections, and recent clinical data suggest that it may prevent secondary infection complications as well. We investigated the role of standardized E. purpurea (Echinaforce® extract or EF) on H3N2-induced adhesion of live nontypeable Haemophilus influenzae (NTHi) and Staphylococcus aureus, along with the expression of bacterial receptors, intracellular adhesion molecule-1 (ICAM-1), fibronectin, and platelet activating factor receptor (PAFr), by BEAS-2B cells. Inflammatory processes were investigated by determining the cellular expression of IL-6 and IL-8 and the involvement of Toll-like receptor (TLR-4) and NFκB p65. We found that influenza virus A infection increased the adhesion of H. influenzae and S. aureus to bronchial epithelial cells via upregulated expression of the ICAM-1 receptor and, to some extent, of fibronectin and PAFr. Echinaforce (EF) significantly reduced the expression of ICAM-1, fibronectin, and PAFr and consequently the adhesion of both bacterial strains. EF also effectively prevented the super-expression of inflammatory cytokines by suppressing the expression of NFκB and possibly TLR-4. These results indicate that E. purpurea has the potential to reduce the risk of respiratory complications by preventing virus-induced bacterial adhesion and through the inhibition of inflammation super-stimulation (cytokine storms).


Subject(s)
Anti-Infective Agents/pharmacology , Echinacea/chemistry , Epithelial Cells/drug effects , Superinfection/prevention & control , Toll-Like Receptor 4/antagonists & inhibitors , Transcription Factor RelA/antagonists & inhibitors , Cell Line , Coinfection , Epithelial Cells/cytology , Epithelial Cells/microbiology , Epithelial Cells/virology , Fibronectins/genetics , Fibronectins/immunology , Gene Expression Regulation , Haemophilus influenzae/drug effects , Haemophilus influenzae/growth & development , Haemophilus influenzae/pathogenicity , Host-Pathogen Interactions , Humans , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/pathogenicity , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Lung/cytology , Lung/drug effects , Lung/microbiology , Lung/virology , Plant Extracts/pharmacology , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Signal Transduction , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , Superinfection/microbiology , Superinfection/virology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology
14.
Chin J Integr Med ; 23(12): 893-900, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28197937

ABSTRACT

OBJECTIVE: To explore the association of the platelet-activating factor receptor (PAFR) gene rs5938, rs313152 and rs76744145 polymorphisms with coronary heart disease (CHD) and blood stasis syndrome (BSS) of CHD in Chinese Han population. METHODS: A total of 570 CHD patients (299 with BSS and 271 with non-BSS) and 317 controls were enrolled. The PAFR gene rs5938, rs313152 and rs76744145 polymorphisms were genotyped using the multiplex SNaPshot technology. The statistical analysis was conducted using a multiple variable logistic regression model. RESULTS: Significant differences were detected in the genotypes frequency distributions of the rs5938 (P<0.01), but not the rs313152 (P>0.05), between the controls and CHD patients. Individuals with an rs5938 or rs313152 mutated allele had a low risk for CHD [adjusted odds ratio (aOR)=0.35, 95% confidence interval (CI): 0.23 to 0.56, P<0.01; aOR=0.65, 95% CI: 0.46 to 0.91, P<0.05, respectively]. After the CHD patients were stratified as BSS or non-BSS according to their Chinese medicine patterns, the rs5938 polymorphism mutated alleles had a significant association with a low risk for BSS of CHD (aOR=0.32, 95% CI: 0.18 to 0.57, P<0.01) and non-BSS of CHD (aOR=0.31, 95% CI: 0.17 to 0.55, P<0.01). The rs313152 polymorphism was associated with a low risk for BSS (aOR=0.51, 95% CI: 0.33 to 0.79, P<0.01), but not for non-BSS (aOR=1.22, 95% CI: 0.81 to 1.85, P<0.05). Furthermore, the interaction effect of the rs5938 and rs313152 polymorphisms for BSS of CHD was significantly based on an aOR value associated with the combination of the rs5938 GT genotype with the rs313152 TC genotype of 0.27 (95% CI: 0.1 to 0.7, P<0.01). CONCLUSION: The PAFR gene rs5938 or rs313152 polymorphisms might be a potential biomarker for susceptibility to CHD, especially to BSS of CHD in Chinese Han population.


Subject(s)
Asian People/genetics , Coronary Disease/genetics , Ethnicity/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Platelet Membrane Glycoproteins/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, G-Protein-Coupled/genetics , Demography , Epistasis, Genetic , Female , Humans , Male , Middle Aged , Odds Ratio , Syndrome
15.
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
16.
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
17.
Chin J Nat Med ; 13(1): 30-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25660286

ABSTRACT

The present study was designed to target fish for potential bioactive components contained in a Huang Lian Jie Du decoction (HLJDD) and identify the underlying mechanisms of action for the treatment of sepsis at the molecular level. he bioactive components database of HLJDD was constructed and the sepsis-associated targets were comprehensively investigated. The 3D structures of the PAFR and TXA2R proteins were established using the homology modelling (HM) method, and the molecular effects for sepsis treatment were analysed by comparing the bioactive components database and the sepsis targets using computational biology methods. The results of the screening were validated with biological testing against the human oral epidermal carcinoma cell line KB in vitro. We found that multiple bioactive compounds contained in the HLJDD interacted with multiple targets. We also predicted the promising compound leads for sepsis treatment, and the first 28 compounds were characterized. Several compounds, such as berberine, berberrubine and epiberberine, dose-dependently inhibited PGE2 production in human KB cells, and the effects were similar in the presence or absence of TPA. This study demonstrates a novel approach to identifying natural chemical compounds as new leads for the treatment of sepsis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Berberine/pharmacokinetics , Drugs, Chinese Herbal/pharmacokinetics , Sepsis/drug therapy , Berberine/analogs & derivatives , Dinoprostone/biosynthesis , Drugs, Chinese Herbal/chemistry , Humans , KB Cells , Platelet Membrane Glycoproteins/drug effects , Protein Transport , Receptors, G-Protein-Coupled/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/drug effects , Sepsis/metabolism , Tetradecanoylphorbol Acetate/pharmacokinetics
18.
Article in English | WPRIM | ID: wpr-812176

ABSTRACT

The present study was designed to target fish for potential bioactive components contained in a Huang Lian Jie Du decoction (HLJDD) and identify the underlying mechanisms of action for the treatment of sepsis at the molecular level. he bioactive components database of HLJDD was constructed and the sepsis-associated targets were comprehensively investigated. The 3D structures of the PAFR and TXA2R proteins were established using the homology modelling (HM) method, and the molecular effects for sepsis treatment were analysed by comparing the bioactive components database and the sepsis targets using computational biology methods. The results of the screening were validated with biological testing against the human oral epidermal carcinoma cell line KB in vitro. We found that multiple bioactive compounds contained in the HLJDD interacted with multiple targets. We also predicted the promising compound leads for sepsis treatment, and the first 28 compounds were characterized. Several compounds, such as berberine, berberrubine and epiberberine, dose-dependently inhibited PGE2 production in human KB cells, and the effects were similar in the presence or absence of TPA. This study demonstrates a novel approach to identifying natural chemical compounds as new leads for the treatment of sepsis.


Subject(s)
Humans , Anti-Inflammatory Agents, Non-Steroidal , Pharmacokinetics , Berberine , Pharmacokinetics , Dinoprostone , Drugs, Chinese Herbal , Chemistry , Pharmacokinetics , KB Cells , Platelet Membrane Glycoproteins , Protein Transport , Receptors, G-Protein-Coupled , Receptors, Thromboxane A2, Prostaglandin H2 , Sepsis , Drug Therapy , Metabolism , Tetradecanoylphorbol Acetate , Pharmacokinetics
19.
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
20.
Expert Opin Pharmacother ; 14(7): 917-27, 2013 May.
Article in English | MEDLINE | ID: mdl-23544968

ABSTRACT

INTRODUCTION: Recurrent ischemic events after an acute coronary syndrome (ACS) are frequent, despite current antiplatelet therapies and revascularization. Warfarin reduces recurrent ischemia but increases bleeding. Warfarin is difficult to administer with the proportion of time achieving therapeutic international normalized ratios varying within and among individuals. The newer oral anticoagulants have predictable dose-effect relationships and no monitoring is required. AREAS COVERED: The pharmacology of rivaroxaban , evidence for the use of rivaroxaban in ACS, and the management of bleeding complications are covered in this article. EXPERT OPINION: Arterial thrombosis is traditionally thought to be more platelet mediated than clotting factor mediated. Nonetheless, in the acute therapy for ACS, anticoagulation is a cornerstone treatment and there is persistent thrombin generation. Research has focused on finding a "sweet spot" of anticoagulation with high anti-ischemic and low bleeding effects. It follows that with chronic therapy post-ACS, there could also be a "sweet-spot" of anticoagulation. This may depend on the intrinsic vascular disease burden, the intervention delivered (stenting or bypass surgery), and background antiplatelet therapy. Rivaroxaban, a new oral factor Xa inhibitor, in a low-dose regimen-reduced ischemic events including mortality across a broad ACS population in the ATLAS-ACS trial with increased bleeding but without increasing fatal bleeding. Rivaroxaban is an attractive new treatment option for ACS.


Subject(s)
Acute Coronary Syndrome/drug therapy , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Morpholines/pharmacology , Morpholines/therapeutic use , Thiophenes/pharmacology , Thiophenes/therapeutic use , Acute Coronary Syndrome/physiopathology , Anticoagulants/adverse effects , Drug Approval , Drug Interactions , Europe , Humans , Morpholines/adverse effects , Platelet Membrane Glycoproteins/antagonists & inhibitors , Randomized Controlled Trials as Topic , Rivaroxaban , Thiophenes/adverse effects , United States , United States Food and Drug Administration
SELECTION OF CITATIONS
SEARCH DETAIL