Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.871
Filtrer
1.
Life Sci ; 351: 122848, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38885879

RÉSUMÉ

AIMS: This research aimed to study the changes in platelet function and their underlying mechanisms in iron deficiency anemia. MAIN METHODS: Initially, we evaluated platelet function in an IDA mice model. Due to the inability to accurately reduce intracellular Fe2+ concentrations, we investigated the impact of Fe2+ on platelet function by introducing varying concentrations of Fe2+. To probe the underlying mechanism, we simultaneously examined the dynamics of calcium in the cytosol, and integrin αIIbß3 activation in Fe2+-treated platelets. Ferroptosis inhibitors Lip-1 and Fer-1 were applied to determine whether ferroptosis was involved in this process. KEY FINDINGS: Our study revealed that platelet function was suppressed in IDA mice. Fe2+ concentration-dependently facilitated platelet activation and function in vitro. Mechanistically, Fe2+ promoted calcium mobilization, integrin αIIbß3 activation, and its downstream outside-in signaling. Additionally, we also demonstrated that ferroptosis might play a role in this process. SIGNIFICANCE: Our data suggest an association between iron and platelet activation, with iron deficiency resulting in impaired platelet function, while high concentrations of Fe2+ contribute to platelet activation and function by promoting calcium mobilization, αIIbß3 activation, and ferroptosis.


Sujet(s)
Anémie par carence en fer , Plaquettes , Calcium , Ferroptose , Souris de lignée C57BL , Activation plaquettaire , Animaux , Souris , Plaquettes/métabolisme , Anémie par carence en fer/métabolisme , Anémie par carence en fer/sang , Ferroptose/physiologie , Calcium/métabolisme , Activation plaquettaire/physiologie , Mâle , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Fer/métabolisme , Modèles animaux de maladie humaine
2.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38928286

RÉSUMÉ

Integrin αIIbß3 mediates platelet aggregation by binding the Arginyl-Glycyl-Aspartic acid (RGD) sequence of fibrinogen. RGD binding occurs at a site topographically proximal to the αIIb and ß3 subunits, promoting the conformational activation of the receptor from bent to extended states. While several experimental approaches have characterized RGD binding to αIIbß3 integrin, applying computational methods has been significantly more challenging due to limited sampling and the need for a priori information regarding the interactions between the RGD peptide and integrin. In this study, we employed all-atom simulations using funnel metadynamics (FM) to evaluate the interactions of an RGD peptide with the αIIb and ß3 subunits of integrin. FM incorporates an external history-dependent potential on selected degrees of freedom while applying a funnel-shaped restraint potential to limit RGD exploration of the unbound state. Furthermore, it does not require a priori information about the interactions, enhancing the sampling at a low computational cost. Our FM simulations reveal significant molecular changes in the ß3 subunit of integrin upon RGD binding and provide a free-energy landscape with a low-energy binding mode surrounded by higher-energy prebinding states. The strong agreement between previous experimental and computational data and our results highlights the reliability of FM as a method for studying dynamic interactions of complex systems such as integrin.


Sujet(s)
Simulation de dynamique moléculaire , Oligopeptides , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Liaison aux protéines , Oligopeptides/composition chimique , Oligopeptides/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/composition chimique , Humains , Plaquettes/métabolisme , Sites de fixation , Intégrine bêta3/métabolisme , Intégrine bêta3/composition chimique
3.
Clin Transl Sci ; 17(5): e13823, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38771157

RÉSUMÉ

This study aims to investigate the mechanism of platelet activation-induced thrombosis in patients with acute non-ST segment elevation myocardial infarction (NSTEMI) by detecting the expression of autophagy-associated proteins in platelets of patients with NSTEMI. A prospective study was conducted on 121 patients with NSTEMI who underwent emergency coronary angiography and optical coherence tomography. The participants were divided into two groups: the ST segment un-offset group (n = 64) and the ST segment depression group (n = 57). We selected a control group of 60 patients without AMI during the same period. The levels of autophagy-associated proteins and the expression of autophagy-associated proteins in platelets were measured using immunofluorescence staining and Western blot. In NSTEMI, the prevalence of red thrombus was higher in the ST segment un-offset myocardial infarction (STUMI) group, whereas white thrombus was more common in the ST segment depression myocardial infarction (STDMI) group. Furthermore, the platelet aggregation rate was significantly higher in the white thrombus group compared with the red thrombus group. Compared with the control group, the autophagy-related protein expression decreased, and the expression of αIIbß3 increased in NSTEMI. The overexpression of Beclin1 could activate platelet autophagy and inhibit the expression of αIIbß3. The results suggested that the increase in platelet aggregation rate in patients with NSTEMI may be potentially related to the change in autophagy. And the overexpression of Beclin1 could reduce the platelet aggregation rate by activating platelet autophagy. Our findings demonstrated that Beclin1 could be a potential therapeutic target for inhibiting platelet aggregation in NSTEMI.


Sujet(s)
Autophagie , Bécline-1 , Plaquettes , Infarctus du myocarde sans sus-décalage du segment ST , Activation plaquettaire , Thrombose , Humains , Bécline-1/métabolisme , Mâle , Femelle , Infarctus du myocarde sans sus-décalage du segment ST/sang , Adulte d'âge moyen , Sujet âgé , Études prospectives , Plaquettes/métabolisme , Thrombose/sang , Thrombose/métabolisme , Coronarographie , Agrégation plaquettaire , Études cas-témoins , Tomographie par cohérence optique , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme
4.
Cell Biochem Funct ; 42(4): e4039, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38751189

RÉSUMÉ

Platelet hyperreactivity contributes to the pathogenesis of COVID-19, which is associated with a hypercoagulability state and thrombosis disorder. It has been demonstrated that Vitamin D deficiency is associated with the severity of COVID-19 infection. Vitamin D supplement is widely used as a dietary supplement due to its safety and health benefits. In this study, we investigated the direct effects and underlying mechanisms of 1,25(OH)2D3 on platelet hyperreactivity induced by SRAS-CoV-2 spike protein via Western blot and platelet functional studies in vitro. Firstly, we found that 1,25(OH)2D3 attenuated platelet aggregation and Src-mediated signaling. We further observed that 1,25(OH)2D3 attenuated spike protein-potentiated platelet aggregation in vitro. Mechanistically, 1,25(OH)2D3 attenuated spike protein upregulated-integrin αIIbß3 outside-in signaling such as platelet spreading and the phosphorylation of ß3, c-Src and Syk. Moreover, using PP2, the Src family kinase inhibitor to abolish spike protein-stimulated platelet aggregation and integrin αIIbß3 outside-in signaling, the combination of PP2 and 1,25(OH)2D3 did not show additive inhibitory effects on spike protein-potentiated platelet aggregation and the phosphorylation of ß3, c-Src and Syk. Thus, our data suggest that 1,25(OH)2D3 attenuates platelet aggregation potentiated by spike protein via downregulating integrin αIIbß3 outside-in signaling.


Sujet(s)
Agrégation plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Transduction du signal , Glycoprotéine de spicule des coronavirus , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/antagonistes et inhibiteurs , Glycoprotéine de spicule des coronavirus/métabolisme , Humains , Transduction du signal/effets des médicaments et des substances chimiques , SARS-CoV-2/effets des médicaments et des substances chimiques , COVID-19/métabolisme , Plaquettes/métabolisme , Plaquettes/effets des médicaments et des substances chimiques , Calcitriol/pharmacologie , src-Family kinases/métabolisme , src-Family kinases/antagonistes et inhibiteurs , Syk kinase/métabolisme , Syk kinase/antagonistes et inhibiteurs , Phosphorylation/effets des médicaments et des substances chimiques , Traitements médicamenteux de la COVID-19
5.
Protein J ; 43(3): 603-612, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38734856

RÉSUMÉ

Disintegrins, a family of snake venom protein, which are capable of modulating the activity of integrins that play a fundamental role in the regulation of many physiological and pathological processes. The main purpose of this study is to obtain the recombinant disintegrin (r-DI) and evaluate its biological activity. In this study, we explored a high-level expression prokaryotic system and purification strategy for r-DI. Then, r-DI was treated to assay effects on cell growth, migration, and invasion. The affinity for the interactions of r-DI with integrin was determined using Surface plasmon resonance (SPR) analyses. The r-DI can be expressed in Escherichia coli and purified by one-step chromatography. The r-DI can inhibit B16F10 cells proliferation, migration, and invasion. Also, we found that r-DI could interact with the integrin αIIbß3 (GPIIb/IIIa). The r-DI can be expressed, purified, characterized through functional assays, and can also maintain strong biological activities. Thus, this study showed potential therapeutic effects of r-DI for further functional and structural studies.


Sujet(s)
Désintégrines , Escherichia coli , Protéines recombinantes , Escherichia coli/génétique , Escherichia coli/métabolisme , Animaux , Désintégrines/composition chimique , Désintégrines/génétique , Désintégrines/isolement et purification , Désintégrines/pharmacologie , Protéines recombinantes/génétique , Protéines recombinantes/composition chimique , Protéines recombinantes/isolement et purification , Protéines recombinantes/biosynthèse , Protéines recombinantes/métabolisme , Souris , Viperidae/génétique , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/génétique , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/composition chimique , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Lignée cellulaire tumorale , Expression des gènes , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Venins de crotalidé/composition chimique , Venins de crotalidé/génétique , Crotalinae ,
6.
Blood Adv ; 8(13): 3388-3401, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38701351

RÉSUMÉ

ABSTRACT: Glycoprotein Ibα (GPIbα), the ligand-binding subunit of platelet GPIb-IX complex, interacts with von Willebrand factor (VWF) exposed at the injured vessel wall, initiating platelet adhesion, activation, hemostasis, and thrombus formation. The cytoplasmic tail of GPIbα interacts with 14-3-3ζ, regulating the VWF-GPIbα-elicited signal transduction and VWF binding function of GPIbα. However, we unexpectedly found that the GPIbα-14-3-3ζ association, beyond VWF-dependent function, is essential for general platelet activation. We found that the myristoylated peptide of GPIbα C-terminus MPαC, a potential GPIbα inhibitor, by itself induced platelet aggregation, integrin αIIbß3 activation, granule secretion, and phosphatidylserine (PS) exposure. Conversely, the deletion of the cytoplasmic tail of GPIbα in mouse platelets (10aa-/-) decreased platelet aggregation, integrin αIIbß3 activation, granule secretion, and PS exposure induced by various physiological agonists. Phosphoproteome-based kinase activity profiling revealed significantly upregulated protein kinase C (PKC) activity in MPαC-treated platelets. MPαC-induced platelet activation was abolished by the pan-PKC inhibitor and PKCα deletion. Decreased PKC activity was observed in both resting and agonist-stimulated 10aa-/- platelets. GPIbα regulates PKCα activity by sequestering 14-3-3ζ from PKCα. In vivo, the deletion of the GPIbα cytoplasmic tail impaired mouse hemostasis and thrombus formation and protected against platelet-dependent pulmonary thromboembolism. Therefore, our findings demonstrate an essential role for the GPIbα cytoplasmic tail in regulating platelet general activation and thrombus formation beyond the VWF-GPIbα axis.


Sujet(s)
Plaquettes , Activation plaquettaire , Complexe glycoprotéique GPIb-IX plaquettaire , Complexe glycoprotéique GPIb-IX plaquettaire/métabolisme , Animaux , Souris , Humains , Plaquettes/métabolisme , Protéines 14-3-3/métabolisme , Facteur de von Willebrand/métabolisme , Thrombose/métabolisme , Transduction du signal , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Souris knockout , Agrégation plaquettaire
7.
Chembiochem ; 25(13): e202400188, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38743506

RÉSUMÉ

Plastics, omnipresent in the environment, have become a global concern due to their durability and limited biodegradability, especially in the form of microparticles and nanoparticles. Polystyrene (PS), a key plastic type, is susceptible to fragmentation and surface alterations induced by environmental factors or industrial processes. With widespread human exposure through pollution and diverse industrial applications, understanding the physiological impact of PS, particularly in nanoparticle form (PS-NPs), is crucial. This study focuses on the interaction of PS-NPs with model blood proteins, emphasising the formation of a protein corona, and explores the subsequent contact with platelet membrane mimetics using experimental and theoretical approaches. The investigation involves αIIbß3-expressing cells and biomimetic membranes, enabling real-time and label-free nanoscale precision. By employing quartz-crystal microbalance with dissipation monitoring studies, the concentration-dependent cytotoxic effects of differently functionalised ~210 nm PS-NPs on HEK293 cells overexpressing αIIbß3 are evaluated in detail. The study unveils insights into the molecular details of PS-NP interaction with supported lipid bilayers, demonstrating that a protein corona formed in the presence of exemplary blood proteins offers protection against membrane damage, mitigating PS-NP cytotoxicity.


Sujet(s)
Nanoparticules , Polystyrènes , Couronne de protéines , Humains , Polystyrènes/composition chimique , Couronne de protéines/composition chimique , Couronne de protéines/métabolisme , Nanoparticules/composition chimique , Cellules HEK293 , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/métabolisme , Double couche lipidique/composition chimique , Double couche lipidique/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/composition chimique
8.
Structure ; 32(7): 899-906.e3, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38579706

RÉSUMÉ

Integrin αIIbß3 is the key receptor regulating platelet retraction and accumulation and a proven drug-target for antithrombotic therapies. Here we resolve the cryo-EM structures of the full-length αIIbß3, which covers three distinct states along the activation pathway. Firstly, we obtain the αIIbß3 structure at 3 Å resolution in the inactive state, revealing the overall topology of the heterodimer with the transmembrane (TM) helices and the ligand-binding domain tucked in a specific angle proximity to the TM region. After the addition of a Mn2+ agonist, we resolve two coexisting structures representing two new states between inactive and active state. Our structures show conformational changes of the αIIbß3 activating trajectory and a unique twisting of the integrin legs, which is required for platelets accumulation. Our structure provides direct structural evidence for how the lower legs are involved in full-length integrin activation mechanisms and offers a new strategy to target the αIIbß3 lower leg.


Sujet(s)
Cryomicroscopie électronique , Modèles moléculaires , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/composition chimique , Humains , Liaison aux protéines , Multimérisation de protéines , Sites de fixation , Manganèse/métabolisme , Manganèse/composition chimique , Conformation des protéines
9.
Int J Mol Sci ; 25(5)2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38474244

RÉSUMÉ

Adrenaline has recently been found to trigger phosphatidylserine (PS) exposure on blood platelets, resulting in amplification of the coagulation process, but the mechanism is only fragmentarily established. Using a panel of platelet receptors' antagonists and modulators of signaling pathways, we evaluated the importance of these in adrenaline-evoked PS exposure by flow cytometry. Calcium and sodium ion influx into platelet cytosol, after adrenaline treatment, was examined by fluorimetric measurements. We found a strong reduction in PS exposure after blocking of sodium and calcium ion influx via Na+/H+ exchanger (NHE) and Na+/Ca2+ exchanger (NCX), respectively. ADP receptor antagonists produced a moderate inhibitory effect. Substantial limitation of PS exposure was observed in the presence of GPIIb/IIIa antagonist, phosphoinositide-3 kinase (PI3-K) inhibitors, or prostaglandin E1, a cyclic adenosine monophosphate (cAMP)-elevating agent. We demonstrated that adrenaline may develop a procoagulant response in human platelets with the substantial role of ion exchangers (NHE and NCX), secreted ADP, GPIIb/IIIa-dependent outside-in signaling, and PI3-K. Inhibition of the above mechanisms and increasing cytosolic cAMP seem to be the most efficient procedures to control adrenaline-evoked PS exposure in human platelets.


Sujet(s)
Plaquettes , Activation plaquettaire , Humains , Plaquettes/métabolisme , Calcium/métabolisme , Épinéphrine/métabolisme , Antiagrégants plaquettaires/pharmacologie , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Sodium/métabolisme , Thrombine/métabolisme
10.
J Thromb Haemost ; 22(6): 1715-1726, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38508397

RÉSUMÉ

BACKGROUND: Protease-activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated that a single nucleotide polymorphism in extracellular loop 3 and PAR4-P310L (rs2227376) leads to a hyporeactive receptor. OBJECTIVES: The goal of this study was to determine how the hyporeactive PAR4 variant in extracellular loop 3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L). METHODS: A point mutation was introduced into the PAR4 gene F2rl3 via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbß3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model. RESULTS: PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbß3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis. CONCLUSION: PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts.


Sujet(s)
Plaquettes , Souris de lignée C57BL , Agrégation plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Récepteurs à la thrombine , Thrombine , Animaux , Plaquettes/métabolisme , Récepteurs à la thrombine/génétique , Récepteurs à la thrombine/métabolisme , Thrombine/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/génétique , Modèles animaux de maladie humaine , Venins de crotalidé/pharmacologie , Venins de crotalidé/toxicité , ADP/métabolisme , ADP/pharmacologie , Sélectine P/métabolisme , Sélectine P/génétique , Mutation ponctuelle , Techniques de knock-in de gènes , Transduction du signal , Thrombose/génétique , Thrombose/sang , Mâle , Chlorures , Souris , Activation plaquettaire , Systèmes CRISPR-Cas , Humains , Phénotype , Composés du fer III , Oligopeptides , Lectines de type C , Récepteurs activés par la protéinase
11.
J Thromb Haemost ; 22(7): 1806-1818, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38518897

RÉSUMÉ

There have been substantial advances in vascular protein disulfide isomerases (PDIs) in platelet function and thrombosis in recent years. There are 4 known prothrombotic thiol isomerases; PDI, endoplasmic reticulum protein (ERp)57, ERp72, and ERp46, and 1 antithrombotic PDI; transmembrane protein 1. A sixth PDI, ERp5, may exhibit either prothrombotic or antithrombotic properties in platelets. Studies on ERp46 in platelet function and thrombosis provide insight into the mechanisms by which these enzymes function. ERp46-catalyzed disulfide cleavage in the αIIbß3 platelet integrin occurs prior to PDI-catalyzed events to maximally support platelet aggregation. The transmembrane PDI transmembrane protein 1 counterbalances the effect of ERp46 by inhibiting activation of αIIbß3. Recent work on the prototypic PDI found that oxidized PDI supports platelet aggregation. The a' domain of PDI is constitutively oxidized, possibly by endoplasmic reticulum oxidoreductase-1α. However, the a domain is normally reduced but becomes oxidized under conditions of oxidative stress. In contrast to the role of oxidized PDI in platelet function, reduced PDI downregulates activation of the neutrophil integrin αMß2. Intracellular platelet PDI cooperates with Nox1 and contributes to thromboxane A2 production to support platelet function. Finally, αIIb and von Willebrand factor contain free thiols, which alter the functions of these proteins, although the extent to which the PDIs regulate these functions is unclear. We are beginning to understand the substrates and functions of vascular thiol isomerases and the redox network they form that supports hemostasis and thrombosis. Moreover, the disulfide bonds these enzymes target are being defined. The clinical implications of the knowledge gained are wide-ranging.


Sujet(s)
Plaquettes , Oxydoréduction , Protein Disulfide-Isomerases , Thrombose , Humains , Protein Disulfide-Isomerases/métabolisme , Plaquettes/métabolisme , Plaquettes/enzymologie , Thrombose/sang , Thrombose/enzymologie , Animaux , Agrégation plaquettaire , Activation plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Protéines membranaires/métabolisme , Stress oxydatif , Thiols/métabolisme , Glycoprotéines membranaires/métabolisme
12.
Curr Eye Res ; 49(7): 731-741, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38482878

RÉSUMÉ

PURPOSE: To explore the role of coagulation and fibrinolytic factors, and the potential mechanism of platelet aggregation in the pathogenesis of retinal vein occlusion. METHODS: Coagulation and fibrinolytic parameters in patients with retinal vein occlusion were determined using hemagglutinin and HISCL-5000. Relationships between these elevated parameters and factors representing typical clinical manifestations of retinal vein occlusion were examined, and these parameters were analyzed using a STRING database to indicate the potential role of platelet aggregation. Platelet glycoprotein IIb/IIIa (GPIIb/IIIa) levels were evaluated by flow cytometry after antiplatelet treatment in patients and mouse models. Furthermore, the GPIIb/IIIa ligand fibrinogen in peripheral blood and retina of mouse models was assessed by the turbidimetric method and real-time PCR, respectively. RESULTS: In patients, significant increases in peripheral blood fibrinogen and GPIIb/IIIa levels were observed (p = 0.0040, p < 0.0001, respectively). A positive correlation was observed between macular thickness (MT) and both fibrinogen and GPIIb/IIIa (r = 0.4528, p = 0.0063; r = 0.3789, p = 0.0427, respectively). After intravitreal injections of anti-vascular endothelial growth factor drugs, a significant reduction in fibrinogen levels was observed (p = 0.0072). In addition, the use of antiplatelet drugs resulted in a significant decrease in GPIIb/IIIa (p < 0.0001). In a mouse model, antiplatelet therapy significantly reduced both peripheral blood and retina fibrinogen levels and the overall rate of vein occlusion 3 days after occlusion (p < 0.0005). In addition, the reduction in GPIIb/IIIa levels after antiplatelet therapy was remarkable. CONCLUSION: Fibrinogen and GPIIb/IIIa may be involved in retinal vein occlusion and blocking platelet aggregation may be a new therapeutic approach for retinal vein occlusion.


Sujet(s)
Modèles animaux de maladie humaine , Fibrinogène , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Occlusion veineuse rétinienne , Sujet âgé , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Plaquettes/métabolisme , Fibrinogène/métabolisme , Cytométrie en flux , Souris de lignée C57BL , Agrégation plaquettaire/physiologie , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Antiagrégants plaquettaires/usage thérapeutique , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/antagonistes et inhibiteurs , Réaction de polymérisation en chaine en temps réel , Occlusion veineuse rétinienne/traitement médicamenteux , Occlusion veineuse rétinienne/métabolisme , Intégrine alpha2/métabolisme
13.
J Agric Food Chem ; 72(13): 7043-7054, 2024 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-38509000

RÉSUMÉ

14-3-3ζ protein, the key target in the regulation and control of integrin ß3 outside-in signaling, is an attractive new strategy to inhibit thrombosis without affecting hemostasis. In this study, 4'-O-methylbavachalconeB (4-O-MB) in Psoraleae Fructus was identified as a 14-3-3ζ ligand with antithrombosis activity by target fishing combined with ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) analysis. The competitive inhibition analysis showed that 4-O-MB targeted 14-3-3ζ and blocked the 14-3-3ζ/integrin ß3 interaction with inhibition constant (Ki) values of 9.98 ± 0.22 µM. Molecular docking and amino acid mutation experiments confirmed that 4-O-MB specifically bound to 14-3-3ζ through LSY9 and SER28 to regulate the 14-3-3ζ/integrin ß3 interaction. Besides, 4-O-MB affected the integrin ß3 early outside-in signal by inhibiting AKT and c-Src phosphorylation. Meanwhile, 4-O-MB could inhibit ADP-, collagen-, or thrombin-induced platelet aggregation function but had no effect on platelet adhesion to collagen-coated surfaces in vivo. Administration of 4-O-MB could significantly inhibit thrombosis formation without disturbing hemostasis in mice. These findings provide new prospects for the antithrombotic effects of Psoraleae Fructus and the potential application of 4-O-MB as lead compounds in the therapy of thrombosis by targeting 14-3-3ζ.


Sujet(s)
Agrégation plaquettaire , Thrombose , Souris , Animaux , Intégrine bêta3/génétique , Intégrine bêta3/composition chimique , Intégrine bêta3/métabolisme , Protéines 14-3-3/génétique , Protéines 14-3-3/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/génétique , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/pharmacologie , Simulation de docking moléculaire , Thrombose/traitement médicamenteux , Thrombose/génétique , Thrombose/métabolisme , Collagène/métabolisme , Plaquettes/métabolisme
14.
Phytomedicine ; 125: 155276, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38295661

RÉSUMÉ

BACKGROUND: Coronary microembolism (CME) is commonly seen in the peri-procedural period of Percutaneous Coronary Intervention (PCI), where local platelet activation and endothelial cell inflammation crosstalk may lead to micro thrombus erosion and rupture, with serious consequences. Qihuang Zhuyu Formula (QHZYF) is a Chinese herbal compound with high efficacy against coronary artery disease, but its antiplatelet mechanism is unclear. HYPOTHESIS/PURPOSE: This study aimed to elucidate the effects and mechanisms of QHZYF on sodium laurate-induced CME using network pharmacology and in vitro and in vivo experiments. METHODS: We employed high-performance liquid chromatography mass spectrometry to identify the main components of QHZYF. Network pharmacology analysis, molecular docking and surface plasmon resonance (SPR) were utilized to predict the primary active components, potential therapeutic targets, and intervention pathways mediating the effects of QHZYF on platelet activation. Next, we pretreated a sodium laurate-induced minimally invasive CME rat model with QHZYF. In vivo experiments were performed to examine cardiac function in rats, to locate coronary arteries on heart sections to observe internal microthrombi, to extract rat Platelet-rich plasma (PRP) for adhesion assays and CD62p and PAC-1 (ITGB3/ITGA2B) flow assays, and to measure platelet-associated protein expression in PRP. In vitro clot retraction and Co-culture of HUVECs with PRP were performed and the gene pathway was validated through flow cytometry and immunofluorescence. RESULTS: Combining UPLC-Q-TOF/MS technology and database mining, 78 compounds were finally screened as the putative and representative compounds of QHZYF, with 75 crossover genes associated with CME. QHZYF prevents CME mainly by regulating key pathways of the inflammation and platelets, including Lipid and atherosclerosis, Fluid shear stress, platelet activation, and PI3K-Akt signaling pathways. Five molecules including Calyson, Oroxin A, Protosappanin A,Kaempferol and Geniposide were screened and subjected to molecular docking and SPR validation in combination with Lipinski rules (Rule of 5, Ro5). In vivo experiments showed that QHZYF not only improved myocardial injury but also inhibited formation of coronary microthrombi. QHZYF inhibited platelet activation by downregulating expression of CD62p receptor and platelet membrane protein αIIbß3 and reduced the release of von Willebrand Factor (vWF), Ca2+ particles and inflammatory factor IL-6. Further analysis revealed that QHZYF inhibited the activation of integrin αIIbß3, via modulating the PI3K/Akt pathways. In in vitro experiments, QHZYF independently inhibited platelet clot retraction. Upon LPS induction, the activation of platelet membrane protein ITGB3 was inhibited via the PI3K/Akt pathway, revealing an important mechanism for attenuating coronary microthrombosis. We performed mechanistic validation using PI3K inhibitor LY294002 and Akt inhibitor MK-2206 to show that QHZYF inhibited platelet membrane protein activation and inflammation to improved coronary microvessel embolism by regulating PI3K/Akt/αIIbß3 pathways, mainly by inhibiting PI3K and Akt phosphorylation. CONCLUSION: QHZYF interferes with coronary microthrombosis through inhibition of platelet adhesion, activation and inflammatory crosstalk, thus has potential in clinical anti-platelet applications. Calyson, Oroxin A, Protosappanin A, Kaempferol and Geniposide may be the major active ingredient groups of QHZYF that alleviate coronary microthrombosis.


Sujet(s)
Médicaments issus de plantes chinoises , Iridoïdes , Intervention coronarienne percutanée , Phénols , Thrombose , Rats , Animaux , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Kaempférols/pharmacologie , Agrégation plaquettaire , Simulation de docking moléculaire , Activation plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Thrombose/traitement médicamenteux , Inflammation , Médicaments issus de plantes chinoises/pharmacologie
15.
J Thromb Haemost ; 22(7): 2009-2017, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38266679

RÉSUMÉ

BACKGROUND: Kindlin-3 in platelets plays an essential role in supporting integrin αIIbß3 activation, platelet spreading, aggregation, and clot retraction by binding to the integrin ß3 cytoplasmic tail. However, the mechanism by which kindlin-3 mediates the crosstalk between integrin αIIbß3 and myosin in platelets remains unknown. OBJECTIVES: To examine the role of myosin light chain 6 (Myl6) in supporting integrin αIIbß3 activation in platelets. METHODS: Myl6fl/flPF4-Cre mice with a deficiency of Myl6 in the megakaryocyte lineage were generated, and integrin αIIbß3 activation in Myl6-deficient platelets was analyzed. RESULTS: We identified a novel kindlin-3 binding protein, Myl6, an essential light chain of myosin in platelets. Myl6fl/flPF4-Cre mice exhibited significant macrothrombocytopenia resulting from defective proplatelet formation. In the absence of Myl6, integrin αIIbß3 activation in platelets was significantly suppressed, and platelet aggregation was substantially impaired. Interestingly, the deficiency of Myl6 in platelets preferentially affected the binding of a multivalent ligand compared to a monovalent ligand to integrin αIIbß3 upon activation, indicating that Myl6 may contribute to the avidity modulation of integrin αIIbß3 by binding to kindlin-3. Furthermore, blood coagulation ability was impaired in Myl6fl/flPF4-Cre mice, and consistently, these mice exhibited defects in both hemostatic and thrombotic functions. CONCLUSION: In summary, these results suggest that Myl6, as a novel kindlin-3 binding partner, is required to support integrin αIIbß3 activation in platelets, which plays an important role in both hemostasis and thrombosis.


Sujet(s)
Plaquettes , Chaînes légères de myosine , Agrégation plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Liaison aux protéines , Animaux , Plaquettes/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Chaînes légères de myosine/métabolisme , Thrombopénie/sang , Activation plaquettaire , Souris knockout , Mégacaryocytes/métabolisme , Humains , Souris de lignée C57BL , Souris , Transduction du signal , Thrombose/métabolisme , Thrombose/sang , Thrombose/génétique , Protéines du cytosquelette
16.
J Thromb Haemost ; 22(2): 558-564, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37866516

RÉSUMÉ

BACKGROUND: Leukocyte adhesion deficiency III (LAD-III) is a rare autosomal recessive syndrome characterized by functional deficiencies of platelets and leukocytes that occurs due to mutations in the FERMT3 gene encoding kindlin-3. Kindlin-3 is a FERM domain-containing adaptor protein that is essential in integrin activation. We have previously demonstrated that the FERM domain of kindlin-3 is structurally compact and plays an important role in supporting integrin activation in a mouse model. The impact of destabilizing the compact FERM domain in kindlin-3 on the development of LAD-III in humans remains uncertain. OBJECTIVES: To use primary cells from a patient with LAD-III to validate the role of the compact FERM domain in kindlin-3 function in platelets and leukocytes. METHODS: The patient is a 4-year-old girl who since infancy has displayed clinical features of LAD-III. Patient platelets and leukocytes were functionally analyzed, and structural analysis of the kindlin-3 variant was conducted. RESULTS: We identified a novel homozygous missense mutation in the FERMT3 (c.412G>A, p.E138K) FERM domain. Substantially reduced levels of kindlin-3 were detected in the proband's platelets and leukocytes. Functional evaluation verified that integrin αIIbß3-mediated platelet activation, spreading, and aggregation and ß2-integrin-mediated neutrophil adhesion and spreading were significantly compromised. Structural analysis revealed that this newly identified E138K substitution in kindlin-3 destabilizes the compacted FERM domain, resulting in poor expression of kindlin-3 in blood cells and subsequent LAD-III. CONCLUSION: We have identified a novel missense mutation and verified the functional significance of the compact kindlin-3 FERM domain in supporting integrin functions in platelets and leukocytes.


Sujet(s)
Domaines FERM , Déficit d'adhérence leucocytaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Animaux , Enfant d'âge préscolaire , Femelle , Humains , Souris , Adhérence cellulaire/génétique , Protéines du cytosquelette/métabolisme , Déficit d'adhérence leucocytaire/génétique , Leucocytes/métabolisme , Mutation , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme
17.
Artif Organs ; 48(1): 28-36, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37792630

RÉSUMÉ

BACKGROUND: When nonphysiological stenosis occurs, the transient high shear stress formed in vessels increases the risk of thrombosis and is a potential factor for cardiovascular diseases. But the platelet adhesion and aggregation behavior at nonphysiological post-stenosis and its affecting factors are not fully understood yet. METHODS: In this experiment, platelet aggregation on collagen and fibrinogen at different shear stresses and different hematocrits were observed by microfluidic technology. Platelet activation (P-selectin, glycoprotein IIb/IIIa) and monocyte-platelet aggregate (MPA) levels under different shear stresses were analyzed by flow cytometry. RESULTS: On fibrinogen, platelets aggregate more at higher shear stress conditions. While on collagen, it becomes more difficult for platelets to form stable aggregation at higher shear stress conditions. If platelets adhere initially at low shear stress, stable platelet aggregation can be formed at subsequent high shear stress. Moreover, when the shear stress increases, platelet activity markers (P-selectin, glycoprotein IIb/IIIa and MPAs) increase significantly. Hematocrit affects the degree of platelet aggregation, and the influence of hematocrit is obvious at high shear stress. CONCLUSION: Transient high shear stress (46 ms) can effectively activate platelets. Platelet aggregation behavior was different for coated fibrinogen and collagen protein. Stable platelet adhesion at post-stenosis is more dependent on fibrinogen and platelet aggregation is stable on both fibrinogen and collagen. Hematocrit can significantly affect the formation of platelet aggregation.


Sujet(s)
Microfluidique , Sélectine P , Humains , Sténose pathologique/métabolisme , Activation plaquettaire/physiologie , Agrégation plaquettaire/physiologie , Plaquettes/métabolisme , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Fibrinogène/métabolisme , Collagène/métabolisme
18.
Thromb Res ; 233: 119-126, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-38039724

RÉSUMÉ

BACKGROUND: Microvesicles (MVs) produced by platelets upon activation possess high procoagulant activity and represent a possible thrombotic risk marker. However, direct experimental evaluation of the adhesive properties of MVs and their potential role in thrombus growth is lacking. OBJECTIVES: We investigated integrin αIIbß3 status and adhesive properties of plasma-circulating and platelet-derived MVs from healthy individuals. METHODS: MVs were isolated from whole blood or produced from activated platelets. Flow cytometry was used for quantification of fluorescently labeled PAC-1 and fibrinogen binding to MVs. Confocal microscopy was used for evaluation of MVs adhesion to fibrinogen and for estimation of their involvement in whole blood thrombus formation in a parallel-plate flow chambers under arterial shear conditions. RESULTS AND CONCLUSIONS: Neither circulating plasma MVs, nor platelet-activation-produced MVs bound PAC-1. However, both types of MVs specifically and weakly bound fibrinogen (about 400 molecules of bound fibrinogen per MV versus >100,000 per non-procoagulant activated platelet). Still, the MVs did not adhere stably to the immobilized fibrinogen. Both types of MVs were weakly incorporated into a thrombus and did not affect thrombus formation: average thrombus height in the recalcified whole blood in the presence of platelet-activation-produced MVs was 4.19 ± 1.38 µm versus 4.87 ± 1.72 µm (n = 6, p > 0.05) in the control experiments. This suggests that MVs present in plasma of healthy individuals are not likely to be directly involved in thrombus formation under arterial flow conditions.


Sujet(s)
Plaquettes , Thrombose , Humains , Plaquettes/métabolisme , Activation plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Fibrinogène/métabolisme
19.
Artif Organs ; 48(5): 514-524, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38112069

RÉSUMÉ

BACKGROUND: von Willebrand factor (vWF) plays a crucial role in physiological hemostasis through platelet and subendothelial collagen adhesion. However, its role in shear-induced platelet activation and functional alteration under non-physiological conditions common to blood-contacting medical devices (BCMDs) is not well investigated. METHODS: Fresh healthy human blood was treated with an anti-vWF antibody to block vWF-GPIbα interaction. Untreated blood was used as a control. They were exposed to three levels of non-physiological shear stress (NPSS) (75, 125, and 175 Pa) through a shearing device with an exposure time of 0.5 s to mimic typical shear conditions in BCMDs. Flow cytometric assays were used to measure the expression levels of PAC-1 and P-Selectin and platelet aggregates for platelet activation and the expression levels of GPIbα, GPIIb/IIIa, and GPVI for receptor shedding. Collagen/ristocetin-induced platelet aggregation capacity was characterized by aggregometry. RESULTS: The levels of platelet activation and aggregates increased with increasing NPSS in the untreated blood. More receptors were lost with increasing NPSS, resulting in a decreased capacity of collagen/ristocetin-induced platelet aggregation. In contrast, the increase in platelet activation and aggregates after exposure to NPSS, even at the highest level of NPSS, was significantly lower in treated blood. Nevertheless, there was no notable difference in receptor shedding, especially for GPIIb/IIIa and GPVI, between the two blood groups at the same level of NPSS. The block of vWF exacerbated the decreased capacity of collagen/ristocetin-induced platelet aggregation. CONCLUSIONS: High NPSS activates platelets mainly by enhancing the vWF-GPIbα interaction. Platelet activation and receptor shedding induced by high NPSS likely occur through different pathways.


Sujet(s)
Ristocétine , Facteur de von Willebrand , Humains , Facteur de von Willebrand/métabolisme , Ristocétine/métabolisme , Activation plaquettaire , Plaquettes/métabolisme , Agrégation plaquettaire , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Collagène/métabolisme , Contrainte mécanique
20.
Cells ; 12(20)2023 10 10.
Article de Anglais | MEDLINE | ID: mdl-37887268

RÉSUMÉ

Platelet activation during hemostasis and thrombosis is facilitated by agonist-induced inside-out and integrin αIIbß3-initiated outside-in signaling via protein kinases and phosphatases. Pharmacological inhibitor studies suggest that the serine/threonine protein phosphatase 1 (PP1) promotes platelet activation. However, since phosphatase inhibitors block all the isoforms of the catalytic subunit of PP1 (PP1c), the role of specific PP1c isoform in platelet signaling remains unclear. Here, we employed a platelet-specific PP1cα-/- mice to explore the contribution of a major PP1 isoform in platelet functions. Loss of PP1cα moderately decreased activation of integrin αIIbß3, binding of soluble fibrinogen, and aggregation to low-dose thrombin, ADP, and collagen. In contrast, PP1cα-/- platelets displayed increased adhesion to immobilized fibrinogen, fibrin clot retraction, and thrombus formation on immobilized collagen. Mechanistically, post-fibrinogen engagement potentiated p38 mitogen-activated protein kinase (MAPK) activation in PP1cα-/- platelets and the p38 inhibitor blocked the increased integrin-mediated outside-in signaling function. Tail bleeding time and light-dye injury-induced microvascular thrombosis in the cremaster venules and arterioles were not altered in PP1cα-/- mice. Thus, PP1cα displays pleiotropic signaling in platelets as it amplifies agonist-induced signaling and attenuates integrin-mediated signaling with no impact on hemostasis and thrombosis.


Sujet(s)
Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire , Thrombose , Souris , Animaux , Protein Phosphatase 1/métabolisme , Domaine catalytique , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Isoformes de protéines/métabolisme , Collagène , Fibrinogène/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...