Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.700
Filter
1.
Nutrients ; 16(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38999771

ABSTRACT

The study aimed to evaluate the antithrombotic action of Acrocomia aculeata pulp oil (AAPO) in natura, in an in vitro experimental model. AAPO was obtained by solvent extraction, and its chemical characterization was performed by gas chromatography coupled to a mass spectrometer (GC-MS). In vitro toxicity was evaluated with the Trypan Blue exclusion test and in vivo by the Galleria mellonella model. ADP/epinephrine-induced platelet aggregation after treatment with AAPO (50, 100, 200, 400, and 800 µg/mL) was evaluated by turbidimetry, and coagulation was determined by prothrombin activity time (PT) and activated partial thromboplastin time (aPTT). Platelet activation was measured by expression of P-selectin on the platelet surface by flow cytometry and intraplatelet content of reactive oxygen species (ROS) by fluorimetry. The results showed that AAPO has as major components such as oleic acid, palmitic acid, lauric acid, caprylic acid, and squalene. AAPO showed no toxicity in vitro or in vivo. Platelet aggregation decreased against agonists using treatment with different concentrations of AAPO. Oil did not interfere in PT and aPTT. Moreover, it expressively decreased ROS-induced platelet activation and P-selectin expression. Therefore, AAPO showed antiplatelet action since it decreased platelet activation verified by the decrease in P-selectin expression as well as in ROS production.


Subject(s)
Fibrinolytic Agents , P-Selectin , Plant Oils , Platelet Aggregation , Reactive Oxygen Species , Animals , Platelet Aggregation/drug effects , P-Selectin/metabolism , Humans , Plant Oils/pharmacology , Plant Oils/chemistry , Reactive Oxygen Species/metabolism , Fibrinolytic Agents/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Coagulation/drug effects , Platelet Activation/drug effects
2.
Nutrients ; 16(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999733

ABSTRACT

Natural and synthetic colorants present in food can modulate hemostasis, which includes the coagulation process and blood platelet activation. Some colorants have cardioprotective activity as well. However, the effect of genipin (a natural blue colorant) and synthetic blue colorants (including patent blue V and brilliant blue FCF) on hemostasis is not clear. In this study, we aimed to investigate the effects of three blue colorants-genipin, patent blue V, and brilliant blue FCF-on selected parameters of hemostasis in vitro. The anti- or pro-coagulant potential was assessed in human plasma by measuring the following coagulation times: thrombin time (TT), prothrombin time (PT), and activated partial thromboplastin time (APTT). Moreover, we used the Total Thrombus formation Analysis System (T-TAS, PL-chip) to evaluate the anti-platelet potential of the colorants in whole blood. We also measured their effect on the adhesion of washed blood platelets to fibrinogen and collagen. Lastly, the cytotoxicity of the colorants against blood platelets was assessed based on the activity of extracellular lactate dehydrogenase (LDH). We observed that genipin (at all concentrations (1-200 µM)) did not have a significant effect on the coagulation times (PT, APTT, and TT). However, genipin at the highest concentration (200 µM) and patent blue V at the concentrations of 1 and 10 µM significantly prolonged the time of occlusion measured using the T-TAS, which demonstrated their anti-platelet activity. We also observed that genipin decreased the adhesion of platelets to fibrinogen and collagen. Only patent blue V and brilliant blue FCF significantly shortened the APTT (at the concentration of 10 µM) and TT (at concentrations of 1 and 10 µM), demonstrating pro-coagulant activity. These synthetic blue colorants also modulated the process of human blood platelet adhesion, stimulating the adhesion to fibrinogen and inhibiting the adhesion to collagen. The results demonstrate that genipin is not toxic. In addition, because of its ability to reduce blood platelet activation, genipin holds promise as a novel and valuable agent that improves the health of the cardiovascular system and reduces the risk of cardiovascular diseases. However, the mechanism of its anti-platelet activity remains unclear and requires further studies. Its in vivo activity and interaction with various anti-coagulant and anti-thrombotic drugs, including aspirin and its derivatives, should be examined as well.


Subject(s)
Blood Coagulation , Blood Platelets , Food Coloring Agents , Iridoids , Humans , Iridoids/pharmacology , Blood Coagulation/drug effects , Food Coloring Agents/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Hemostasis/drug effects , Partial Thromboplastin Time , Platelet Adhesiveness/drug effects , Fibrinogen/metabolism , Benzenesulfonates/pharmacology , Prothrombin Time , Rosaniline Dyes/pharmacology , Hemostatics/pharmacology , Platelet Activation/drug effects , Thrombin Time
3.
Platelets ; 35(1): 2383642, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39072582

ABSTRACT

Hemolysis is associated with thrombosis and vascular dysfunction, which are the pathological components of many diseases. Hemolytic products, including hemoglobin and hemin, activate platelets (PLT). Despite its activation, the effect of hemolysis on platelet clearance remains unclear, It is critical to maintain a normal platelet count and ensure that circulating platelets are functionally viable. In this study, we used hemin, a degradation product of hemoglobin, as a potent agonist to treat platelets and simulate changes in vivo in mice. Hemin treatment induced activation and morphological changes in platelets, including an increase in intracellular Ca2+ levels, phosphatidylserine (PS) exposure, and cytoskeletal rearrangement. Fewer hemin-treated platelets were cleared by macrophages in the liver after transfusion than untreated platelets. Hemin bound to glycoprotein Ibα (GPIbα), the surface receptor in hemin-induced platelet activation and aggregation. Furthermore, hemin decreased GPIbα desialylation, as evidenced by reduced Ricinus communis agglutinin I (RCA- I) binding, which likely extended the lifetime of such platelets in vivo. These data provided new insight into the mechanisms of GPIbα-mediated platelet activation and clearance in hemolytic disease.


What is the context? Hemolysis is a primary hematological disease. Hemolysis is a pathological complication of several diseases.Hemin, a degradation product of cell-free hemoglobin, has been proven to be a more potent agonist than hemoglobin for directly activating platelets.Platelet membrane glycoproteins (GP), including GPIb-IX and GPIIb/IIIa complexes, play crucial roles in platelet hemostasis.Desialylation (loss of sialic acid residues) of GPIbα, is believed to regulate physiological platelet clearance through liver macrophages and hepatocytes.What is new? In this study, we evaluated the effects of hemolysis on platelet clearance. We first analyzed the influence of hemin at 0-50 µM on platelets in vitro before exploring the mechanism underlying hemin-induced platelet activation and its role in platelet clearance in vitro and in vivo.Our analyses suggest that: Hemin bound to GPIbα on the platelet surface with high affinity.Platelet clearance occurred slowly in the liver and spleen after hemin treatment.Platelets exhibited significant significantly reduced GPIbα surface expression and desialylation after hemin treatment.Platelets exhibited significant significantly reduced GPIbα surface expression and desialylation after hemin treatment.What is the impact? This study provides new insights into the role of hemin in the mechanisms of GPIbα-mediated platelets activation and clearance in diseases associated with hemolysis.


Subject(s)
Blood Platelets , Hemin , Platelet Glycoprotein GPIb-IX Complex , Mice , Animals , Blood Platelets/metabolism , Blood Platelets/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Hemin/pharmacology , Hemin/metabolism , Humans , Platelet Activation/drug effects , Hemolysis/drug effects , Protein Binding
4.
Platelets ; 35(1): 2322733, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38968449

ABSTRACT

Liver failure and cirrhosis are characterized by abnormal hemostasis with aberrant platelet activation. In particular, the consequences of cholestatic liver disease and molecular mechanisms, including the role of bile acids leading to impaired platelet responses, are not well understood. Here, we demonstrate that bile acids inhibit human and murine platelet activation, adhesion and spreading, leading to reduced thrombus formation under flow conditions. We identified the G-protein coupled receptor TGR5 in platelets and provide support for its role as mediator of bile acid-induced impairment of platelet activation. In the liver, TGR5 couples to Gαs proteins, activates the adenylate cyclase to induce a transient cAMP rise and stimulates the MAPK signaling pathway to regulate cholangiocyte proliferation, hepatocyte survival and inflammation. In this report, we demonstrate that the genetic deficiency of TGR5 in mice led to enhanced platelet activation and thrombus formation, suggesting that TGR5 plays an important role in hemostasis. Mechanistically, platelet inhibition is achieved by TGR5 mediated PKA activation and modulation of AKT and ERK1/2 phosphorylation. Thus, this report provides evidence for the ability of TGR5 ligands to reduce platelet activation and identifies TGR5 agonism as a new target for the prevention of cardiovascular diseases.


What is the context? Liver failure or cirrhosis are related to impaired hemostasis and a role of bile acids in impaired platelet responses is known but only less understood.Platelets express the bile acid receptor FXR. Ligand binding to the FXR on platelets causes a shift in platelet reactivity and is atheroprotective suggesting that the FXR is a potential target for the prevention of atherothrombotic diseases.What is new? Treatment of murine and human blood with bile acids in low molecular quantity led to reduced platelet activation, adhesion and thrombus formation.The bile acid receptor TGR5 was identified on human and murine platelets.TGR5 plays an important role in hemostasis because TGR5 deficient mice showed elevated platelet reactivity and enhanced thrombus formation.Loss of TGR5 led to enhanced PKA activation and modulated the phosphorylation of MAPK such as AKT and ERK1/2.What is the impact? Impairment of platelet activation by bile acids is mediated by TGR5 via the protein kinase A signaling pathway.Our findings provide evidence for the modulation of TGR5 activation as a potential new target of both, anti-platelet therapy in cardiovascular diseases and the restoration of hemostasis upon liver injury.


Subject(s)
Platelet Activation , Receptors, G-Protein-Coupled , Thrombosis , Receptors, G-Protein-Coupled/metabolism , Animals , Mice , Humans , Platelet Activation/drug effects , Thrombosis/metabolism , Blood Platelets/metabolism , Bile Acids and Salts/metabolism , Mice, Knockout , Signal Transduction
5.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062819

ABSTRACT

Platelets play a significant role in hemostasis, forming plugs at sites of vascular injury to limit blood loss. However, if platelet activation is not controlled, it can lead to thrombotic events, such as myocardial infarction and stroke. To prevent this, antiplatelet agents are used in clinical settings to limit platelet activation in patients at risk of arterial thrombotic events. However, their use can be associated with a significant risk of bleeding. An enhanced comprehension of platelet signaling mechanisms should facilitate the identification of safer targets for antiplatelet therapy. Over the past decade, our comprehension of the breadth and intricacy of signaling pathways that orchestrate platelet activation has expanded exponentially. Several recent studies have provided further insight into the regulation of platelet signaling events and identified novel targets against which to develop novel antiplatelet agents. Antiplatelet drugs are essential in managing atherothrombotic vascular disease. The current antiplatelet therapy in clinical practice is limited in terms of safety and efficacy. Novel compounds have been developed in response to patient variability and resistance to aspirin and/or clopidogrel. Recent studies based on randomized controlled trials and systematic reviews have definitively demonstrated the role of antiplatelet therapy in reducing the risk of cardiovascular events. Antiplatelet therapy is the recommended course of action for patients with established atherosclerosis. These studies compared monotherapy with a P2Y12 inhibitor versus aspirin for secondary prevention. However, in patients undergoing percutaneous coronary intervention, it is still unclear whether the efficacy of P2Y12 inhibitor monotherapy after a short course of dual antiplatelet therapy depends on the type of P2Y12 inhibitor. This paper focuses on the advanced-stage evaluation of several promising antiplatelet drugs.


Subject(s)
Platelet Aggregation Inhibitors , Purinergic P2Y Receptor Antagonists , Humans , Purinergic P2Y Receptor Antagonists/therapeutic use , Purinergic P2Y Receptor Antagonists/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/pharmacology , Receptors, Purinergic P2Y12/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Signal Transduction/drug effects , Platelet Activation/drug effects , Animals
6.
Biomed Pharmacother ; 177: 117154, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018868

ABSTRACT

This study investigates the antiplatelet properties of tomato pulp to combat cardiovascular diseases. Notably, it examines the formation of nitrated fatty acids (NO2-FA) in tomato pomace, renowned for its potential antiplatelet effects. Through diverse assays, including tandem mass spectrometry, microplate-based platelet aggregation, and flow cytometry, the research identifies NO2-OA, NO2-LA, and NO2-LnA as pivotal antiplatelet compounds. It demonstrates the concentration-dependent antiplatelet effects of nitrated tomato pomace against thrombin receptor activator peptide 6 (TRAP-6) and collagen-induced platelet activation, alongside the modulation of platelet activation markers. Additionally, synergistic effects were observed with nitrated tomato pomace extracts. The findings suggest therapeutic potential for NO2-FA derived from tomato pomace in preventing blood clot formation, with nitrated extracts exhibiting superior efficacy compared to non-nitrated ones. This research highlights the promising role of natural products, such as tomato pomace, in mitigating cardiovascular risks and proposes novel strategies for population health enhancement and cardiovascular disease management.


Subject(s)
Fatty Acids , Plant Extracts , Platelet Aggregation Inhibitors , Platelet Aggregation , Solanum lycopersicum , Solanum lycopersicum/chemistry , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation/drug effects , Humans , Plant Extracts/pharmacology , Plant Extracts/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Nitrates/pharmacology , Platelet Activation/drug effects
7.
PLoS One ; 19(6): e0304800, 2024.
Article in English | MEDLINE | ID: mdl-38924073

ABSTRACT

BACKGROUND: Despite Antiplatelet therapy (APT), cardiovascular patients undergoing revascularisation remain at high risk for thrombotic events. Individual response to APT varies substantially, resulting in insufficient protection from thrombotic events due to high on-treatment platelet reactivity (HTPR) in ≤40% of patients. Individual variation in platelet response impairs APT guidance on a single patient level. Unfortunately, little is known about individual platelet response to APT over time, timing for accurate residual platelet reactivity measurement, or the optimal test to monitor residual platelet reactivity. AIMS: To investigate residual platelet reactivity variability over time in individual patients undergoing carotid endarterectomy (CEA) treated with clopidogrel. METHODS: Platelet reactivity was determined in patients undergoing CEA in a prospective, single-centre, observational study using the VerifyNow (change in turbidity from ADP-induced binding to fibrinogen-coated beads), the VASP assay (quantification of phosphorylation of vasodilator-stimulated phosphoprotein), and a flow-cytometry-based assay (PACT) at four perioperative time points. Genotyping identified slow (CYP2C19*2 and CYP2C19*3) and fast (CYP2C19*17) metabolisers. RESULTS: Between December 2017 and November 2019, 50 patients undergoing CEA were included. Platelet reactivity measured with the VerifyNow (p = < .001) and VASP (p = .029) changed over time, while the PACT did not. The VerifyNow identified patients changing HTRP status after surgery. The VASP identified patients changing HTPR status after eight weeks (p = .018). CYP2C19 genotyping identified 13 slow metabolisers. CONCLUSION: In patients undergoing CEA, perioperative platelet reactivity measurements fluctuate over time with little agreement between platelet reactivity assays. Consequently, HTPR status of individual patients measured with the VerifyNow and VASP assay changed over time. Therefore, generally used perioperative platelet reactivity measurements seem unreliable for adjusting perioperative APT strategy.


Subject(s)
Blood Platelets , Clopidogrel , Endarterectomy, Carotid , Platelet Aggregation Inhibitors , Humans , Male , Female , Aged , Pilot Projects , Blood Platelets/metabolism , Prospective Studies , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/pharmacology , Clopidogrel/therapeutic use , Platelet Function Tests/methods , Middle Aged , Perioperative Period , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Vascular Surgical Procedures , Platelet Activation/drug effects , Aged, 80 and over , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/blood , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/blood
8.
Biochem Biophys Res Commun ; 724: 150140, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38852506

ABSTRACT

Sepsis is a severe inflammatory disease characterized by cytokine storm, often accompanied by disseminated intravascular coagulation (DIC). PANoptosis is a novel form of cell death triggered by cytokine storms, characterized by a cascade reaction of pyroptosis, apoptosis, and necroptosis. It exists in septic platelets and is closely associated with the onset and progression of DIC. However, there remains an unmet need for drugs targeting PANoptosis. The anti-PANoptosis effect of myricetin was predicted using network pharmacology and confirmed through molecular docking. In vitro platelet activation models demonstrated that myricetin significantly attenuated platelet particle release, integrin activation, adhesion, spreading, clot retraction, and aggregation. Moreover, in a sepsis model, myricetin reduced inflammatory infiltration in lung tissue and platelet activation while improving DIC. Additionally, whole blood sequencing samples from sepsis patients and healthy individuals were analyzed to elucidate the up-regulation of the PANoptosis targets. Our findings demonstrate the inhibitory effect of myricetin on septic platelet PANoptosis, indicating its potential as a novel anti-cellular PANoptosis candidate and therapeutic agent for septic DIC. Furthermore, our study establishes a foundation for utilizing network pharmacology in the discovery of new drugs to treat various diseases.


Subject(s)
Blood Platelets , Disseminated Intravascular Coagulation , Flavonoids , Sepsis , Flavonoids/pharmacology , Flavonoids/therapeutic use , Sepsis/drug therapy , Sepsis/blood , Humans , Blood Platelets/drug effects , Blood Platelets/metabolism , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/pathology , Disseminated Intravascular Coagulation/blood , Animals , Male , Molecular Docking Simulation , Platelet Activation/drug effects , Mice, Inbred C57BL , Mice , Pyroptosis/drug effects
9.
J Ethnopharmacol ; 333: 118485, 2024 Oct 28.
Article in English | MEDLINE | ID: mdl-38908490

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Xuefu Zhuyu Decoction (XZD), a renowned traditional Chinese medicine prescription, is widely employed for the management of conditions characterized by qi-stagnation and blood stasis. Although its anti-thrombotic effect on deep vein thrombosis (DVT) patients has been clinically observed, the underlying mechanism remains largely unexplored. AIM OF THE STUDY: Our aim was to investigate the mechanisms by which XZD exerted its effect on DVT. MATERIALS AND METHODS: The ultra performance liquid chromatography (UPLC) technique was employed to evaluate quality of XZD. To examine the effect of XZD on DVT, a DVT rat model with inferior vena cava (IVC) stenosis was established. The 4D-label-free proteomics approach was then utilized to uncover the possible mechanisms of XZD against DVT. Based on proteomics, citrullinated histone H3 (CitH3), along with serum levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1ß) were observed the inhibitory activity of XZD on neutrophil activation. Subsequently, the marker of platelet activation, specifically glycoprotein IIb (CD41) and glycoprotein IIIa (CD61), were assessed along with the secretion of von Willebrand factor (vWF) to investigate the inhibitory activity of XZD on platelet activation. Finally, we explored the impact of XZD on the sirtuin 1 (SIRT1)/nuclear factor kappa-B (NF-κB) pathway, which was associated with the activation of platelets and neutrophils. RESULTS: Eight distinct components were identified for the quality control of XZD. XZD effectively reduced thrombus weight and length in DVT rats, without affecting the coagulation function or hematological parameters in the systemic circulation. Proteomics analysis revealed that XZD alleviated DVT by inhibiting the activation of platelets and neutrophils. The protein expression of CitH3, along with serum levels of TNF-α and IL-1ß, were reduced in XZD-treated DVT rats. Similarly, protein expressions of CD41 and CD61, along with the release of vWF, were markedly down-regulated in XZD-treated DVT rats. Finally, treatment with XZD resulted in an up-regulation of SIRT1 protein expression and a down-regulation of both acetylated NF-κB/p65 and phosphorylated NF-κB/p65 protein expressions in endothelium. CONCLUSIONS: XZD alleviates DVT by inhibiting the activation of platelets and neutrophils at the injured endothelium via the regulation of SIRT1/NF-κB pathway.


Subject(s)
Blood Platelets , Drugs, Chinese Herbal , NF-kappa B , Neutrophils , Platelet Activation , Rats, Sprague-Dawley , Sirtuin 1 , Venous Thrombosis , Animals , Venous Thrombosis/drug therapy , Sirtuin 1/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , NF-kappa B/metabolism , Male , Neutrophils/drug effects , Neutrophils/metabolism , Platelet Activation/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Rats , Signal Transduction/drug effects , Disease Models, Animal , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Proteomics
11.
J Ethnopharmacol ; 333: 118457, 2024 Oct 28.
Article in English | MEDLINE | ID: mdl-38866117

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lian Qiao (LQ), the dried fruit of Forsythia suspensa (Thunb.) Vahl, is a well-documented traditional Chinese medicine known for its detoxifying and heat-clearing properties. Clinically, compounds containing LQ are widely used to treat thrombotic diseases, indicating that it may have antithrombotic effects. However, its exact mechanism of action remains unknown. AIM OF THE STUDY: This study aimed to verify the antithrombotic effect of LQ and further explore the material basis and target mechanism of its antithrombotic effect using various biological methods. MATERIALS AND METHODS: An epinephrine-collagen-thrombin-induced mouse model of acute pulmonary embolism (APE) was established to study the effects of LQ on thrombus development. A UPLC/Q/TOF-MS screening and identification system based on the inhibition of platelet aggregation and Ca2+ antagonism was established to determine the pharmacodynamic components of LQ that inhibit platelet activation. The inhibitory effect of active ingredients on platelet activation, and the determination of the target of their inhibitory effect on platelet activation have been studied using chemical proteomics. Furthermore, based on the structure and function of the target protein, a multidisciplinary approach was adopted to analyze the molecular mechanism of active ingredient binding to target proteins and to evaluate the effects of active ingredients on the downstream signaling pathways of target proteins. RESULTS: LQ showed significant anticoagulant effects in APE model mice. Phillyrin and phillygenin were the antiplatelet-activating components of LQ. PLCß3 was identified as a target for inhibiting platelet activation by phillyrin and its metabolites. The mechanism underlying the effect involves phillyrin and its metabolites inhibiting PLCß3 activity by blocking the binding of PLCß3 to Gαq through non-covalently targeting the ASN260 of PLCß3, thus inhibiting the downstream Gαq-PLCß3-Ca2+ signaling pathway, effectively hindering platelet activation and therefore playing an anticoagulant role. CONCLUSION: This study not only proposes and validates the antithrombotic effect of LQ for the first time but also finds that phillyrin and phillygenin are the main pharmacological substances through which LQ exerts antithrombotic activity and reveals a novel mechanism by which they exert antiplatelet activity by directly targeting and inhibiting PLCß3 activity. These findings significantly contribute to our understanding of the therapeutic potential of phillyrin and provide important clues for the discovery and development of new antiplatelet drugs.


Subject(s)
Platelet Activation , Pulmonary Embolism , Animals , Pulmonary Embolism/drug therapy , Mice , Platelet Activation/drug effects , Male , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Disease Models, Animal , Platelet Aggregation/drug effects , Glucosides/pharmacology , Glucosides/isolation & purification , Glucosides/therapeutic use , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use
12.
Am J Hematol ; 99(8): 1462-1474, 2024 08.
Article in English | MEDLINE | ID: mdl-38877813

ABSTRACT

Patients with essential thrombocythemia (ET) are treated with once-daily low-dose aspirin to prevent thrombosis, but their accelerated platelet turnover shortens the antiplatelet effect. The short-term Aspirin Regimens in EsSential Thrombocythemia trial showed that twice-daily aspirin dosing restores persistent platelet thromboxane (TX) inhibition. However, the long-term pharmacodynamic efficacy, safety and tolerability of twice-daily aspirin remain untested. We performed a multicenter, randomized, open-label, blinded-endpoint, phase-2 trial in which 242 patients with ET were randomized to 100 mg aspirin twice- or once-daily and followed for 20 months. The primary endpoint was the persistence of low serum TXB2, a surrogate biomarker of antithrombotic efficacy. Secondary endpoints were major and clinically relevant non-major bleedings, serious vascular events, symptom burden assessed by validated questionnaires, and in vivo platelet activation. Serum TXB2 was consistently lower in the twice-daily versus once-daily regimen on 10 study visits over 20 months: median 3.9 ng/mL versus 19.2 ng/mL, respectively; p < .001; 80% median reduction; 95% CI, 74%-85%. No major bleeding occurred. Clinically relevant non-major bleedings were non-significantly higher (6.6% vs. 1.7%), and major thromboses lower (0.8% vs. 2.5%) in the twice-daily versus once-daily group. Patients on the twice-daily regimen had significantly lower frequencies of disease-specific symptoms and severe hand and foot microvascular pain. Upper gastrointestinal pain was comparable in the two arms. In vivo platelet activation was significantly reduced by the twice-daily regimen. In patients with ET, twice-daily was persistently superior to once-daily low-dose aspirin in suppressing thromboxane biosynthesis and reducing symptom burden, with no detectable excess of bleeding and gastrointestinal discomfort.


Subject(s)
Aspirin , Drug Administration Schedule , Hemorrhage , Platelet Aggregation Inhibitors , Thrombocythemia, Essential , Humans , Aspirin/administration & dosage , Aspirin/therapeutic use , Thrombocythemia, Essential/drug therapy , Thrombocythemia, Essential/blood , Male , Middle Aged , Female , Aged , Adult , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/adverse effects , Hemorrhage/chemically induced , Thromboxane B2/blood , Platelet Activation/drug effects , Aged, 80 and over , Treatment Outcome
13.
Toxicon ; 247: 107798, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38871030

ABSTRACT

We investigated the hemotoxic effects of three North American pit vipers in healthy human donor blood. Using experiments focusing on platelet and red blood cell activity, we found differential effects of these venoms on these cellular components. Platelet aggregation was most induced by C. adamanteus. Platelet activation was highest with C. atrox. Red blood cells had calcium expression and erythrocyte formation most induced by C. adamanteus and A. piscivorus. These results demonstrate the complex interplay of individual cellular effects with clinical presentations seen in envenomings from these species.


Subject(s)
Blood Platelets , Erythrocytes , Humans , Erythrocytes/drug effects , Blood Platelets/drug effects , Animals , Crotalid Venoms/toxicity , Platelet Aggregation/drug effects , Calcium/metabolism , Calcium/blood , Platelet Activation/drug effects , Crotalinae
14.
Int J Biol Macromol ; 274(Pt 1): 133286, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908635

ABSTRACT

Thrombosis is associated with various fatal arteriovenous syndromes including ischemic stroke, myocardial infarction, and pulmonary embolism. However, current clinical thrombolytic treatment strategies still have many problems in targeting and safety to meet the thrombolytic therapy needs. Understanding the molecular mechanism that underlies thrombosis is critical in developing effective thrombolytic strategies. It is well known that platelets play a central role in thrombosis and the binding of fibrinogen to activated platelets is a common pathway in the process of clot formation. Based on this, a concept of biomimetic thrombus-targeted thrombolytic strategy inspired from fibrinogen binding to activated platelets in thrombosis was proposed, which could selectively bind to activated platelets at a thrombus site, thus enabling targeted delivery and local release of thrombolytic agents for effective thrombolysis. In this review, we first summarized the main characteristics of platelets and fibrinogen, and then introduced the classical molecular mechanisms of thrombosis, including platelet adhesion, platelet activation and platelet aggregation through the interactions of activated platelets with fibrinogen. In addition, we highlighted the recent advances in biomimetic thrombus-targeted thrombolytic strategies which inspired from fibrinogen binding to activated platelets in thrombosis. The possible future directions and perspectives in this emerging area are briefly discussed.


Subject(s)
Biomimetics , Blood Platelets , Fibrinogen , Platelet Activation , Thrombosis , Humans , Fibrinogen/metabolism , Fibrinogen/chemistry , Thrombosis/drug therapy , Thrombosis/metabolism , Blood Platelets/metabolism , Blood Platelets/drug effects , Platelet Activation/drug effects , Thrombolytic Therapy/methods , Protein Binding , Animals , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Fibrinolytic Agents/chemistry
15.
Biomed Khim ; 70(2): 99-108, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38711409

ABSTRACT

Platelet functional activity was assessed in healthy volunteers (HV, n=92), patients with stable angina pectoris (SA, n=42) and acute coronary syndrome (ACS, n=73), treated with acetylsalicylic acid (ASA) + clopidogrel and ASA + ticagrelor, respectively. In all HV and patients we have compared parameters of platelet aggregation (maximum light transmission and velocity, Tmax and Vmax) and parameters, characterizing exposure of platelet activation markers, evaluated by flow cytometry. HV platelets were activated by 10 µM, 1 µM TRAP, and 20 µM, 5 µM, 2.5 µM ADP; patient platelets were activated by 10 µM TRAP and by 20 µM and 5 µM ADP. Strong and significant correlations between the aggregation and flow cytometry parameters (the r correlation coefficient from 0.4 up to >0.6) most frequently were registered in HV platelet during activation by 1 µM TRAP and in SA patients during platelet activation by 20 µM and 5 µM ADP. However, in many other cases these correlations were rather weak (r < 0.3) and sometimes statistically insignificant. In HV the differences in PAC-1 binding parameters between platelets activated by 10 µM TRAP (the strongest agonist) and all ADP concentrations were negligible (≤ 10%), while CD62P binding (at all ADP concentrations) and LTA parameters for (5 µM and 2.5 µM ADP) were significantly lower (by 40-60%). Antiplatelet therapy in patients decreased all parameters as compared to HV, but to varying extents. For 10 µM TRAP the MFI index for PAC-1 binding (40-50% decrease) and for both ADP concentrations the Tmax values (60-85% decrease) appeared to be the most sensitive in comparison with the other parameters that decreased to a lesser extent. The data obtained indicate a possibility of inconsistency between different LTA and flow cytometry parameters in assessing platelet activity and efficacy of antiplatelet drugs.


Subject(s)
Acute Coronary Syndrome , Aspirin , Blood Platelets , Clopidogrel , Flow Cytometry , Platelet Aggregation Inhibitors , Platelet Aggregation , Humans , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Male , Aspirin/pharmacology , Aspirin/therapeutic use , Female , Blood Platelets/drug effects , Blood Platelets/metabolism , Middle Aged , Clopidogrel/pharmacology , Aged , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/blood , Adult , Ticagrelor/pharmacology , Ticagrelor/therapeutic use , Platelet Function Tests/methods , Platelet Activation/drug effects , Angina, Stable/drug therapy , Angina, Stable/blood , Adenosine Diphosphate/pharmacology
16.
Signal Transduct Target Ther ; 9(1): 110, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38724491

ABSTRACT

Previous studies have shown that low platelet count combined with high plasma total homocysteine (tHcy) increased stroke risk and can be lowered by 73% with folic acid. However, the combined role of other platelet activation parameters and the methylenetetrahydrofolate reductase (MTHFR) C677T genotypes on stroke risk and folic acid treatment benefit remain to be examined. This study aimed to investigate if platelet activation parameters and MTHFR genotypes jointly impact folic acid treatment efficacy in first stroke prevention. Data were derived from the China Stroke Primary Prevention Trial. This study includes a total of 11,185 adult hypertensive patients with relevant platelet activation parameters and MTHFR genotype data. When simultaneously considering both platelet activation parameters (plateletcrit, platelet count, mean platelet volume, platelet distribution width) and MTHFR genotypes, patients with both low plateletcrit (Q1) and the TT genotype had the highest stroke incidence rate (5.6%) in the enalapril group. This subgroup significantly benefited from folic acid treatment, with a 66% reduction in first stroke (HR: 0.34; 95% CI: 0.14-0.82; p = 0.016). Consistently, the subgroup with low plateletcrit (Q1) and the CC/CT genotype also benefited from folic acid treatment (HR: 0.40; 95% CI: 0.23-0.70; p = 0.001). In Chinese hypertensive adults, low plateletcrit can identify those who may greatly benefit from folic acid treatment, in particular, those with the TT genotype, a subpopulation known to have the highest stroke risk.


Subject(s)
Folic Acid , Genotype , Methylenetetrahydrofolate Reductase (NADPH2) , Stroke , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Folic Acid/administration & dosage , Folic Acid/genetics , Stroke/genetics , Stroke/prevention & control , Male , Female , Middle Aged , Aged , Hypertension/genetics , Platelet Activation/genetics , Platelet Activation/drug effects , China/epidemiology , Blood Platelets/metabolism , Blood Platelets/drug effects , Platelet Count , Adult
17.
J Thromb Haemost ; 22(6): 1749-1757, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811291

ABSTRACT

BACKGROUND: An iron overload status induces ferroptosis, an iron-dependent nonapoptotic cell death, in various pathological conditions. We previously reported that hemin (heme), protoporphyrin-IX with ferric iron, activates platelets via C-type lectin-like receptor-2 (CLEC-2) and glycoprotein VI/FcRγ, but protoporphyrin-IX alone blocks CLEC-2-dependent platelet activation. Therefore, we hypothesized that free iron has the ability to activate platelets. OBJECTIVES: This study aimed to elucidate platelet activation mechanisms of iron (ferric chloride), including the identification of signaling pathways and receptors, and to examine whether platelets regulate ferroptosis. METHODS: Platelet aggregometry, platelet activation marker expression, and protein phosphorylation were examined in ferric chloride-stimulated human and murine platelets. Inhibitors of platelet activation signaling pathways and receptor-deleted platelets were utilized to identify the responsible signaling pathway and receptor. The effect of platelets on ferroptosis of endothelial cells was investigated in vitro. RESULTS: Ferric chloride induced platelet activation dependent on Src family kinase pathways in humans and mice. Ferric chloride-induced platelet aggregation was almost lost in CLEC-2-depleted murine platelets and wild-type platelets preincubated with recombinant CLEC-2 proteins. Furthermore, coculture of wild-type platelets, but not CLEC-2-deficient platelets, attenuated ferroptosis of endothelial cells in vitro. CONCLUSION: Ferric chloride activates platelets via CLEC-2 and Src family kinase pathways, and platelets have a protective role in the ferroptosis of endothelial cells dependent on CLEC-2.


Subject(s)
Blood Platelets , Chlorides , Ferric Compounds , Ferroptosis , Lectins, C-Type , Mice, Inbred C57BL , Platelet Activation , Platelet Aggregation , Signal Transduction , Animals , Blood Platelets/metabolism , Blood Platelets/drug effects , Ferric Compounds/pharmacology , Humans , Platelet Activation/drug effects , Lectins, C-Type/metabolism , Chlorides/metabolism , Platelet Aggregation/drug effects , Ferroptosis/drug effects , src-Family Kinases/metabolism , Phosphorylation , Mice, Knockout , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mice , Human Umbilical Vein Endothelial Cells/metabolism
18.
Exp Clin Transplant ; 22(4): 284-293, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38742319

ABSTRACT

OBJECTIVES: Splenectomy during liver transplant can affect platelet function. In this study, our primary aim was to assess the perioperative platelet function by rotational thromboelastometry and the effects of splenectomy on platelet function. MATERIALS AND METHODS: We studied 40 consecutive liver transplant recipients with end-stage liver disease (50% as a result of hepatitis C). Patients with splenectomy were compared with patients without splenectomy (n = 20/group). Three platelet function parameters by rotational thromboelastometry were studied: platelet activation with arachidonic acid, platelet activation with adenosine diphosphate, and platelet activation with thrombin receptor-activating peptide 6. Patients were monitored perioperatively and until postoperative day 21. Heparin was infused for 2 days postoperatively (60-180 U/kg/day), followed by administration of subcutaneous low-molecular-weight heparin (40 mg/24 h) on postoperative days 2 and 3 and oral acetylsalicylic acid when platelet count was >50 × 103/µL. RESULTS: Liver disease contributed to low perioperative platelet count and function. Patients showed significant improvement by postoperative day 14 and day 21, particularly after splenectomy. Platelet count was significantly correlated with the 3 platelet function parameters by rotational thromboelastometry (P < .001). Acetyl salicylic acid was required earlier (postoperative day 3) for patients with splenectomy (8/20) but only affected the platelet function represented by platelet activation with arachidonic acid, whereas other platelet activation pathways were less affected. Patients received no transfusions of platelet units. CONCLUSIONS: End-stage liver disease significantly contributed to low platelet function and counts before transplant. Two weeks were required for recovery of patients posttransplant, with further enhancement by splenectomy. Some recipients showed recovery that exceeded the normal reference range, which warranted monitoring. Acetyl salicylic acid only affected 1 platelet activation receptor.


Subject(s)
Blood Coagulation , Blood Platelets , End Stage Liver Disease , Liver Transplantation , Predictive Value of Tests , Splenectomy , Thrombelastography , Humans , Liver Transplantation/adverse effects , Male , Female , Middle Aged , Splenectomy/adverse effects , Treatment Outcome , Blood Coagulation/drug effects , Adult , End Stage Liver Disease/surgery , End Stage Liver Disease/diagnosis , End Stage Liver Disease/blood , Time Factors , Blood Platelets/drug effects , Platelet Activation/drug effects , Platelet Function Tests , Platelet Aggregation Inhibitors/administration & dosage , Anticoagulants/administration & dosage , Platelet Count , Blood Coagulation Tests , Aspirin/administration & dosage , Prospective Studies
19.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732081

ABSTRACT

Flavonoid aglycones are secondary plant metabolites that exhibit a broad spectrum of pharmacological activities, including anti-inflammatory, antioxidant, anticancer, and antiplatelet effects. However, the precise molecular mechanisms underlying their inhibitory effect on platelet activation remain poorly understood. In this study, we applied flow cytometry to analyze the effects of six flavonoid aglycones (luteolin, myricetin, quercetin, eriodictyol, kaempferol, and apigenin) on platelet activation, phosphatidylserine externalization, formation of reactive oxygen species, and intracellular esterase activity. We found that these compounds significantly inhibit thrombin-induced platelet activation and decrease formation of reactive oxygen species in activated platelets. The tested aglycones did not affect platelet viability, apoptosis induction, or procoagulant platelet formation. Notably, luteolin, myricetin, quercetin, and apigenin increased thrombin-induced thromboxane synthase activity, which was analyzed by a spectrofluorimetric method. Our results obtained from Western blot analysis and liquid chromatography-tandem mass spectrometry demonstrated that the antiplatelet properties of the studied phytochemicals are mediated by activation of cyclic nucleotide-dependent signaling pathways. Specifically, we established by using Förster resonance energy transfer that the molecular mechanisms are, at least partly, associated with the inhibition of phosphodiesterases 2 and/or 5. These findings underscore the therapeutic potential of flavonoid aglycones for clinical application as antiplatelet agents.


Subject(s)
Blood Platelets , Flavonoids , Platelet Activation , Platelet Aggregation Inhibitors , Reactive Oxygen Species , Flavonoids/pharmacology , Humans , Platelet Aggregation Inhibitors/pharmacology , Platelet Activation/drug effects , Blood Platelets/metabolism , Blood Platelets/drug effects , Reactive Oxygen Species/metabolism , Apigenin/pharmacology , Quercetin/pharmacology , Luteolin/pharmacology , Signal Transduction/drug effects , Kaempferols/pharmacology , Thrombin/metabolism , Flavanones
20.
Life Sci ; 350: 122746, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38810792

ABSTRACT

AIMS: Dysregulated platelet aggregation is a fatal condition in many bacterial- and virus-induced diseases. However, classical antithrombotics cannot completely prevent immunothrombosis, due to the unaddressed mechanisms towards inflammation. Thus, targeting platelet hyperactivation together with inflammation might provide new treatment options in diseases, characterized by immunothrombosis, such as COVID-19 and sepsis. The aim of this study was to investigate the antiaggregatory effect and mode of action of 1.8-cineole, a monoterpene derived from the essential oil of eucalyptus leaves, known for its anti-inflammatory proprieties. MAIN METHODS: Platelet activity was monitored by measuring the expression and release of platelet activation markers, i.e., P-selectin, CD63 and CCL5, as well as platelet aggregation, upon treatment with 1.8-cineole and stimulation with several classical stimuli and bacteria. A kinase activity assay was used to elucidate the mode of action, followed by a detailed analysis of the involvement of the adenylyl-cyclase (AC)-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) pathway by Western blot and ELISA. KEY FINDINGS: 1.8-cineole prevented the expression and release of platelet activation markers, as well as platelet aggregation, upon induction of aggregation with classical stimuli and immunological agonists. Mechanistically, 1.8- cineole influences the activation of the AC-cAMP-PKA pathway, leading to higher cAMP levels and vasodilator-stimulated phosphoprotein (VASP) phosphorylation. Finally, blocking the adenosine A2A receptor reversed the antithrombotic effect of 1.8-cineole. SIGNIFICANCE: Given the recognized anti-inflammatory attributes of 1.8-cineole, coupled with our findings, 1.8-cineole might emerge as a promising candidate for treating conditions marked by platelet activation and abnormal inflammation.


Subject(s)
Cyclic AMP , Eucalyptol , Platelet Activation , Platelet Aggregation , Receptor, Adenosine A2A , Eucalyptol/pharmacology , Receptor, Adenosine A2A/metabolism , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Humans , Cyclic AMP/metabolism , Blood Platelets/metabolism , Blood Platelets/drug effects , Signal Transduction/drug effects , P-Selectin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Platelet Aggregation Inhibitors/pharmacology , Anti-Inflammatory Agents/pharmacology , COVID-19/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL