Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Thromb Res ; 238: 41-51, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669962

ABSTRACT

BACKGROUND AND PURPOSE: Hypolipidemia and platelet activation play key roles in atherosclerotic diseases. Pirinixic acid (WY-14643) was originally developed as a lipid-lowering drug. Here we focused on its antiplatelet and antithrombotic abilities and the underlying mechanism. EXPERIMENTAL APPROACH: The effects of WY-14643 on platelet aggregation was measured using a lumi-aggregometer. Clot retraction and spreading on fibrinogen were also assayed. PPARα-/- platelets were used to identify the target of WY-14643. The interaction between WY-14643 and glycoprotein Ibα (GPIbα) was detected using cellular thermal shift assay (CETSA), surface plasmon resonance (SPR) spectroscopy and molecular docking. GPIbα downstream signaling was examined by Western blot. The antithrombotic effect was investigated using mouse mesenteric arteriole thrombosis model. Mouse tail bleeding model was used to study its effect on bleeding side effects. KEY RESULTS: WY-14643 concentration-dependently inhibits human washed platelet aggregation, clot retraction, and spreading. Significantly, WY-14643 inhibits thrombin-induced activation of human washed platelets with an IC50 of 7.026 µM. The antiplatelet effect of WY-14643 is mainly dependent of GPIbα. CESTA, SPR and molecular docking results indicate that WY-14643 directly interacts with GPIbα and acts as a GPIbα antagonist. WY-14643 also inhibits phosphorylation of PLCγ2, Akt, p38, and Erk1/2 induced by thrombin. Noteworthily, 20 mg/kg oral administration of WY-14643 inhibits FeCl3-induced thrombosis of mesenteric arteries in mice similarly to clopidogrel without increasing bleeding. CONCLUSION AND IMPLICATIONS: WY-14643 is not only a PPARα agonist with lipid-lowering effect, but also an antiplatelet agent as a GPIbα antagonist. It may have more significant therapeutic advantages than current antiplatelet agents for the treatment of atherosclerotic thrombosis, which have lipid-lowering effects without bleeding side effects.


Subject(s)
Fibrinolytic Agents , Platelet Aggregation Inhibitors , Platelet Aggregation , Platelet Glycoprotein GPIb-IX Complex , Pyrimidines , Animals , Mice , Platelet Glycoprotein GPIb-IX Complex/metabolism , Humans , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Platelet Aggregation/drug effects , Thrombosis/drug therapy , Blood Platelets/metabolism , Blood Platelets/drug effects , Male , Molecular Docking Simulation , Mice, Inbred C57BL
2.
Arterioscler Thromb Vasc Biol ; 44(1): 254-270, 2024 01.
Article in English | MEDLINE | ID: mdl-37916416

ABSTRACT

BACKGROUND: Hyperglycemia-a symptom that characterizes diabetes-is highly associated with atherothrombotic complications. However, the underlying mechanism by which hyperglycemia fuels platelet activation and arterial thrombus formation is still not fully understood. METHODS: The profiles of polyunsaturated fatty acid metabolites in the plasma of patients with diabetes and healthy controls were determined with targeted metabolomics. FeCl3-induced carotid injury model was used to assess arterial thrombus formation in mice with endothelial cell (EC)-specific YAP (yes-associated protein) deletion or overexpression. Flow cytometry and clot retraction assay were used to evaluate platelet activation. RNA sequencing and multiple biochemical analyses were conducted to unravel the underlying mechanism. RESULTS: The plasma PGE2 (prostaglandin E2) concentration was elevated in patients with diabetes with thrombotic complications and positively correlated with platelet activation. The PGE2 synthetases COX-2 (cyclooxygenase-2) and mPGES-1 (microsomal prostaglandin E synthase-1) were found to be highly expressed in ECs but not in other type of vessel cells in arteries from both patients with diabetes and hyperglycemic mice, compared with nondiabetic individuals and control mice, respectively. A combination of RNA sequencing and ingenuity pathway analyses indicated the involvement of YAP signaling. EC-specific deletion of YAP limited platelet activation and arterial thrombosis in hyperglycemic mice, whereas EC-specific overexpression of YAP in mice mimicked the prothrombotic state of diabetes, without affecting hemostasis. Mechanistically, we found that hyperglycemia/high glucose-induced endothelial YAP nuclear translocation and subsequently transcriptional expression of COX-2 and mPGES-1 contributed to the elevation of PGE2 and platelet activation. Blockade of EP3 (prostaglandin E receptor 3) activation by oral administration of DG-041 reversed the hyperactivity of platelets and delayed thrombus formation in both EC-specific YAP-overexpressing and hyperglycemic mice. CONCLUSIONS: Collectively, our data suggest that hyperglycemia-induced endothelial YAP activation aggravates platelet activation and arterial thrombus formation via PGE2/EP3 signaling. Targeting EP3 with DG-041 might be therapeutic for diabetes-related thrombosis.


Subject(s)
Diabetes Mellitus , Hyperglycemia , Thrombosis , Animals , Humans , Mice , Blood Platelets/metabolism , Cyclooxygenase 2/metabolism , Diabetes Mellitus/metabolism , Dinoprostone/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Mice, Obese , Thrombosis/genetics , Thrombosis/metabolism
3.
Sci Total Environ ; 912: 169235, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38097078

ABSTRACT

BACKGROUND: Accumulating epidemiological evidence suggests the association between low ambient temperature exposure and the risk of ischemic stroke, but the underlying mechanisms remain unclear. OBJECTIVE: Given the crucial role of platelet activation and thrombosis in ischemic stroke, this study aims to investigate the effect of ambient temperature on platelet activation through multi-center clinical data in Tianjin as well as animal experiments. METHODS: From 2018 to 2020, nearly 3000 ischemic stroke patients from three stroke centers in Tianjin were included in the analysis, among them the ADP induced platelet aggregation rate was available. Meteorological data from the same period had also been collected. After controlling for confounding factors, the generalized additive mixed model (GAMM) was used to evaluate the correlation between environmental temperature and platelet aggregation rate. In further animal experiments, platelet function assessments were conducted on mice from the cold exposure group and the normal temperature group, including platelet aggregation, spreading, and clot retraction. Additionally, tail bleeding and mesentery thrombosis were also tested to monitor hemostasis and thrombosis in vivo. RESULT: A nonlinear "S" shaped relationship between outdoor temperature and platelet aggregation was found. Each 1 °C decrease of mean temperature was associated with an increase of 7.77 % (95 % CI: 2.06 % - 13.48 %) in platelet aggregation. The ambient temperature is not related to other platelet parameters. Subgroup analysis found that males, people aged ≥65 years, and hypertensive individuals are more susceptible to temperature changes. Furthermore, animal experiments demonstrated that the increased CIRBP levels and subsequent activation of p-AKT/p-ERK may be one of the reasons for cold exposure induced platelets activation. CONCLUSION: Both clinical data and basic research support that low ambient temperature exposure has the potential to increase platelet activation. These results provide a basis for understanding the potential mechanism of temperature variations on the pathogenesis of cerebrovascular diseases.


Subject(s)
Ischemic Stroke , Stroke , Thrombosis , Male , Humans , Mice , Animals , Temperature , Platelet Activation/physiology , Platelet Aggregation , Stroke/epidemiology , RNA-Binding Proteins
4.
Exp Ther Med ; 23(5): 344, 2022 May.
Article in English | MEDLINE | ID: mdl-35401796

ABSTRACT

Arctium lappa L., also known as burdock, is a popular medicinal plant in traditional Chinese medicine due to its potential therapeutic properties. Saccharides from Arctium lappa L. root (ALR-S) have been extensively studied for their anti-inflammatory and anti-diabetes effects. Platelets play a pivotal role in thrombosis. The present study describes the effects of ALR-S on platelet activation and thrombosis using a laser injury thrombosis in vivo model. The study also measured the effects of ALR-S on platelet activation by analysing aggregation, ATP release, platelet spreading, adhesion and clot retraction in vitro. Specifically, the effects were ALR-S concentration-dependent inhibition of platelet aggregation and ATP release. Activated platelets pretreated with ALR-S showed diminished CD62P expression levels and fibrinogen binding, as measured by flow cytometry. ALR-S inhibited platelet spreading on fibrinogen and adhesion on collagen under shear. ALR-S attenuated platelet activation by decreasing oxidative stress and thrombus formation. These results demonstrated the antiplatelet effects of ALR-S, suggesting the antithrombotic and cardiovascular protective activities of ALR-S as a functional food.

5.
Eur Heart J ; 43(37): 3556-3574, 2022 10 07.
Article in English | MEDLINE | ID: mdl-35165707

ABSTRACT

AIMS: Galectin-3, a ß-galactoside-binding lectin, is abnormally increased in cardiovascular disease. Plasma Galectin-3 receives a Class II recommendation for heart failure management and has been extensively studied for multiple cellular functions. The direct effects of Galectin-3 on platelet activation remain unclear. This study explores the direct effects of Galectin-3 on platelet activation and thrombosis. METHODS AND RESULTS: A strong positive correlation between plasma Galectin-3 concentration and platelet aggregation or whole blood thrombus formation was observed in patients with coronary artery disease (CAD). Multiple platelet function studies demonstrated that Galectin-3 directly potentiated platelet activation and in vivo thrombosis. Mechanistic studies using the Dectin-1 inhibitor, laminarin, and Dectin-1-/- mice revealed that Galectin-3 bound to and activated Dectin-1, a receptor not previously reported in platelets, to phosphorylate spleen tyrosine kinase and thus increased Ca2+ influx, protein kinase C activation, and reactive oxygen species production to regulate platelet hyperreactivity. TD139, a Galectin-3 inhibitor in a Phase II clinical trial, concentration dependently suppressed Galectin-3-potentiated platelet activation and inhibited occlusive thrombosis without exacerbating haemorrhage in ApoE-/- mice, which spontaneously developed increased plasma Galectin-3 levels. TD139 also suppressed microvascular thrombosis to protect the heart from myocardial ischaemia-reperfusion injury in ApoE-/- mice. CONCLUSION: Galectin-3 is a novel positive regulator of platelet hyperreactivity and thrombus formation in CAD. As TD139 has potent antithrombotic effects without bleeding risk, Galectin-3 inhibitors may have therapeutic advantages as potential antiplatelet drugs for patients with high plasma Galectin-3 levels.


Subject(s)
Platelet Aggregation , Thrombosis , Animals , Apolipoproteins E/metabolism , Blood Platelets , Calcium/metabolism , Fibrinolytic Agents/pharmacology , Galectin 3/metabolism , Galectin 3/pharmacology , Lectins, C-Type , Mice , Mice, Knockout, ApoE , Platelet Activation , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Protein Kinase C , Reactive Oxygen Species/metabolism , Syk Kinase/metabolism , Syk Kinase/pharmacology , Thrombosis/metabolism
6.
Stem Cell Res ; 58: 102622, 2022 01.
Article in English | MEDLINE | ID: mdl-34929444

ABSTRACT

A 25-years-old hypertrophic cardiomyopathy male patient donated his peripheral blood mononuclear cells (PBMCs) with heterozygote mutation in theTNNT2 gene. We generated induced pluripotent stem cell (iPSC) with normal karyotypic and expressing NANOG, Lin28, GDF3 and DNMT3. The iPSC line has demonstrated pluripotency by differentiating into three germ layers in vitro. The ZZUNEUi021-A would serve as an in vitro model for loss of TNNT2 function.


Subject(s)
Cardiomyopathy, Hypertrophic , Induced Pluripotent Stem Cells , Adult , Cardiomyopathy, Hypertrophic/genetics , Cell Differentiation , Genes, Homeobox , Humans , Leukocytes, Mononuclear , Male , Mutation/genetics , Troponin T/genetics
7.
Biochem Pharmacol ; 194: 114822, 2021 12.
Article in English | MEDLINE | ID: mdl-34748820

ABSTRACT

BACKGROUND: Platelets from septic patients exhibit increased reactivity. However, the underlying mechanism of sepsis-induced platelet hyperactivity is still not completely understood. OBJECTIVE: P2Y12 is a central receptor for platelet activation. In this study, we investigated the role of platelet P2Y12 in platelet hyperactivity during sepsis. METHODS: We measured platelet P2Y12 expression and aggregation in response to ADP in septic patients and cecal ligation and puncture (CLP)-treated mice. We also detected the downstream signaling of P2Y12 in resting platelets from patients and mice with sepsis. The role of nucleotide-binding oligomerization domain 2 (NOD2)/RIP2/NF-κB/P65 pathway in sepsis-induced platelet P2Y12 high expression was also investigated. Finally, we compared the antiplatelet and antithrombotic effects of clopidogrel, prasugrel, and ticagrelor in experimental sepsis in mice and rats. RESULTS: Compared to healthy subjects, platelets from septic patients exhibit P2Y12 hyperactivity and higher P2Y12 expression. pAkt is enhanced and pVASP is impaired in resting platelets from the patients, indicating the constitutive activation of platelet P2Y12 receptor. Mouse sepsis model recapitulates the findings in septic patients. NOD2 deficiency attenuates sepsis-induced platelet P2Y12 high expression, hyperactivity, and thrombosis. Prasugrel and ticagrelor are potent P2Y12 inverse agonists, and exhibit superior antiplatelet and antithrombotic efficacy over clopidogrel in mice and rats with sepsis. CONCLUSIONS: NOD2 activation upregulates platelet P2Y12 expression, which is constitutively activated and contributes to platelet hyperactivity in septic status. Compared to clopidogrel, prasugrel and ticagrelor are potent P2Y12 inverse agonists with superior antiplatelet and antithrombotic efficacy in experimental sepsis.


Subject(s)
Nod2 Signaling Adaptor Protein/biosynthesis , Platelet Activation/physiology , Receptors, Purinergic P2Y12/biosynthesis , Sepsis/metabolism , Thrombosis/metabolism , Up-Regulation/physiology , Animals , Cell Line , Female , Humans , Male , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Prasugrel Hydrochloride/pharmacology , Prasugrel Hydrochloride/therapeutic use , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
8.
Cell Death Dis ; 12(11): 955, 2021 10 16.
Article in English | MEDLINE | ID: mdl-34657146

ABSTRACT

Platelets are generated from the cytoplasm of megakaryocytes (MKs) via actin cytoskeleton reorganization. Zyxin is a focal adhesion protein and wildly expressed in eukaryotes to regulate actin remodeling. Zyxin is upregulated during megakaryocytic differentiation; however, the role of zyxin in thrombopoiesis is unknown. Here we show that zyxin ablation results in profound macrothrombocytopenia. Platelet lifespan and thrombopoietin level were comparable between wild-type and zyxin-deficient mice, but MK maturation, demarcation membrane system formation, and proplatelet generation were obviously impaired in the absence of zyxin. Differential proteomic analysis of proteins associated with macrothrombocytopenia revealed that glycoprotein (GP) Ib-IX was significantly reduced in zyxin-deficient platelets. Moreover, GPIb-IX surface level was decreased in zyxin-deficient MKs. Knockdown of zyxin in a human megakaryocytic cell line resulted in GPIbα degradation by lysosomes leading to the reduction of GPIb-IX surface level. We further found that zyxin was colocalized with vasodilator-stimulated phosphoprotein (VASP), and loss of zyxin caused diffuse distribution of VASP and actin cytoskeleton disorganization in both platelets and MKs. Reconstitution of zyxin with VASP binding site in zyxin-deficient hematopoietic progenitor cell-derived MKs restored GPIb-IX surface expression and proplatelet generation. Taken together, our findings identify zyxin as a regulator of platelet biogenesis and GPIb-IX surface expression through VASP-mediated cytoskeleton reorganization, suggesting possible pathogenesis of macrothrombocytopenia.


Subject(s)
Blood Platelets/metabolism , Cell Membrane/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Zyxin/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Blood Platelets/ultrastructure , Bone Marrow/ultrastructure , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Line , Female , Fibrinogen/pharmacology , Humans , Lysosomes/metabolism , Male , Megakaryocytes/metabolism , Megakaryocytes/ultrastructure , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Mutant Proteins/metabolism , Phosphoproteins/metabolism , Platelet Count , Protein Binding/drug effects , Proteolysis , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thrombin/pharmacology , Thrombocytopenia , Zyxin/deficiency
9.
Stem Cell Res ; 51: 102191, 2021 03.
Article in English | MEDLINE | ID: mdl-33529980

ABSTRACT

Urine cells (or renal tubular cells) can be isolated from human urine samples efficiently. This noninvasive and cost-effective method to collect biological sample provide us favorable access to donor cells from human. In the present study, we generate ZZUNEUi022-A, a urine cells-derived induced pluripotent stem cell (iPSC) line, from a 29-year-old healthy male via Sendai virus delivery system. ZZUNEUi022-A showed stable karyotype, and could differentiate into three germ layers (ectoderm, mesoderm, and endoderm) readily in an embryoid body formation model.


Subject(s)
Induced Pluripotent Stem Cells , Adult , Cell Differentiation , Embryoid Bodies , Endoderm , Germ Layers , Humans , Male , Sendai virus
10.
Biochem Pharmacol ; 183: 114315, 2021 01.
Article in English | MEDLINE | ID: mdl-33152345

ABSTRACT

BACKGROUND AND PURPOSE: Previously, we discovered that the activation of nucleotide-binding oligomerization domain 2 (NOD2) enhances platelet activation. We here investigated the antiplatelet and antithrombotic potential of GSK669, a NOD2 antagonist. EXPERIMENTAL APPROACH: Effects of GSK669 on platelet functions, reactive oxygen species (ROS) and proinflammatory cytokine generation were detected. NOD2-/- platelets were used to confirm GSK669 target. The interaction between GSK669 and glycoprotein VI (GPVI) was detected using surface plasmon resonance (SPR) spectroscopy. GPVI downstream signaling was examined by Western blot. The antithrombotic and antioxidative effects were investigated using mouse mesenteric arteriole thrombosis model and pulmonary embolism model. KEY RESULTS: GSK669 significantly inhibits platelet proinflammatory cytokine release induced by muramyl dipeptide, platelet aggregation, ATP release, and ROS generation induced by collagen and collagen related peptide (CRP). Platelet spreading and clot retraction are also inhibited. GSK669 also decreases collagen-induced phosphorylation of Src, Syk, PLCγ2, and Akt. The antiplatelet effect of GSK669 is NOD2-independent and mediated by GPVI antagonism. Consistent with its antiplatelet activity as a GPVI antagonist, GSK669 inhibits platelet adhesion on collagen in flow condition. Notably, GSK669 inhibits mouse mesenteric arteriole thrombosis similarly to aspirin without bleeding. The antithrombotic effect of GSK669 is further confirmed in the pulmonary embolism model; decreased malonaldehyde (MDA) and increased superoxide dismutase (SOD) levels in mouse plasma reveal a significant antioxidant effect of GSK669. CONCLUSION AND IMPLICATIONS: Beyond its anti-inflammatory effect as a NOD2 antagonist, GSK669 is also an efficient and safe antiplatelet agent combined with antioxidant effect by targeting GPVI. An antiplatelet agent bearing antioxidative and anti-inflammatory activities without bleeding risk may have therapeutic advantage over current antiplatelet drugs for atherothrombosis.


Subject(s)
Blood Platelets/drug effects , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Oxidative Stress/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Membrane Glycoproteins/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/metabolism , Blood Platelets/metabolism , Drug Delivery Systems/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/metabolism , Oxidative Stress/physiology , Platelet Aggregation Inhibitors/metabolism , Platelet Membrane Glycoproteins/metabolism , Thrombosis/metabolism
11.
Circulation ; 143(1): 45-61, 2021 01 05.
Article in English | MEDLINE | ID: mdl-32988222

ABSTRACT

BACKGROUND: PCSK9 (proprotein convertase subtilisin/kexin 9), mainly secreted by the liver and released into the blood, elevates plasma low-density lipoprotein cholesterol by degrading low-density lipoprotein receptor. Pleiotropic effects of PCSK9 beyond lipid metabolism have been shown. However, the direct effects of PCSK9 on platelet activation and thrombosis, and the underlying mechanisms, as well, still remain unclear. METHODS: We detected the direct effects of PCSK9 on agonist-induced platelet aggregation, dense granule ATP release, integrin αIIbß3 activation, α-granule release, spreading, and clot retraction. These studies were complemented by in vivo analysis of FeCl3-injured mouse mesenteric arteriole thrombosis. We also investigated the underlying mechanisms. Using the myocardial infarction (MI) model, we explored the effects of PCSK9 on microvascular obstruction and infarct expansion post-MI. RESULTS: PCSK9 directly enhances agonist-induced platelet aggregation, dense granule ATP release, integrin αIIbß3 activation, P-selectin release from α-granules, spreading, and clot retraction. In line, PCSK9 enhances in vivo thrombosis in a FeCl3-injured mesenteric arteriole thrombosis mouse model, whereas PCSK9 inhibitor evolocumab ameliorates its enhancing effects. Mechanism studies revealed that PCSK9 binds to platelet CD36 and thus activates Src kinase and MAPK (mitogen-activated protein kinase)-extracellular signal-regulated kinase 5 and c-Jun N-terminal kinase, increases the generation of reactive oxygen species, and activates the p38MAPK/cytosolic phospholipase A2/cyclooxygenase-1/thromboxane A2 signaling pathways downstream of CD36 to enhance platelet activation, as well. Using CD36 knockout mice, we showed that the enhancing effects of PCSK9 on platelet activation are CD36 dependent. It is important to note that aspirin consistently abolishes the enhancing effects of PCSK9 on platelet activation and in vivo thrombosis. Last, we showed that PCSK9 activating platelet CD36 aggravates microvascular obstruction and promotes MI expansion post-MI. CONCLUSIONS: PCSK9 in plasma directly enhances platelet activation and in vivo thrombosis, and MI expansion post-MI, as well, by binding to platelet CD36 and thus activating the downstream signaling pathways. PCSK9 inhibitors or aspirin abolish the enhancing effects of PCSK9, supporting the use of aspirin in patients with high plasma PCSK9 levels in addition to PCSK9 inhibitors to prevent thrombotic complications.


Subject(s)
Blood Platelets/metabolism , CD36 Antigens/metabolism , Myocardial Infarction/metabolism , Platelet Activation/physiology , Proprotein Convertase 9/metabolism , Thrombosis/metabolism , Animals , Aspirin/pharmacology , Aspirin/therapeutic use , Blood Platelets/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/drug therapy , PCSK9 Inhibitors , Platelet Activation/drug effects , Platelet Aggregation/physiology , Thrombosis/drug therapy
12.
Stem Cell Res ; 50: 102139, 2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33383407

ABSTRACT

In this study, we report a human induced pluripotent stem cell (iPSC) line from a healthy 27-year-old female individual using non-integrative Sendai viral reprogramming technology. The cell line expresses stemness markers, exhibits a normal female karyotype, and can differentiate into three germ layers in vivo. This iPSC line from a healthy individual provides a control group for studying disease mechanisms, drug screening, and toxicity testing.

13.
J Hematol Oncol ; 13(1): 120, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887634

ABSTRACT

BACKGROUND: Critically ill patients diagnosed with COVID-19 may develop a pro-thrombotic state that places them at a dramatically increased lethal risk. Although platelet activation is critical for thrombosis and is responsible for the thrombotic events and cardiovascular complications, the role of platelets in the pathogenesis of COVID-19 remains unclear. METHODS: Using platelets from healthy volunteers, non-COVID-19 and COVID-19 patients, as well as wild-type and hACE2 transgenic mice, we evaluated the changes in platelet and coagulation parameters in COVID-19 patients. We investigated ACE2 expression and direct effect of SARS-CoV-2 virus on platelets by RT-PCR, flow cytometry, Western blot, immunofluorescence, and platelet functional studies in vitro, FeCl3-induced thrombus formation in vivo, and thrombus formation under flow conditions ex vivo. RESULTS: We demonstrated that COVID-19 patients present with increased mean platelet volume (MPV) and platelet hyperactivity, which correlated with a decrease in overall platelet count. Detectable SARS-CoV-2 RNA in the blood stream was associated with platelet hyperactivity in critically ill patients. Platelets expressed ACE2, a host cell receptor for SARS-CoV-2, and TMPRSS2, a serine protease for Spike protein priming. SARS-CoV-2 and its Spike protein directly enhanced platelet activation such as platelet aggregation, PAC-1 binding, CD62P expression, α granule secretion, dense granule release, platelet spreading, and clot retraction in vitro, and thereby Spike protein enhanced thrombosis formation in wild-type mice transfused with hACE2 transgenic platelets, but this was not observed in animals transfused with wild-type platelets in vivo. Further, we provided evidence suggesting that the MAPK pathway, downstream of ACE2, mediates the potentiating role of SARS-CoV-2 on platelet activation, and that platelet ACE2 expression decreases following SARS-COV-2 stimulation. SARS-CoV-2 and its Spike protein directly stimulated platelets to facilitate the release of coagulation factors, the secretion of inflammatory factors, and the formation of leukocyte-platelet aggregates. Recombinant human ACE2 protein and anti-Spike monoclonal antibody could inhibit SARS-CoV-2 Spike protein-induced platelet activation. CONCLUSIONS: Our findings uncovered a novel function of SARS-CoV-2 on platelet activation via binding of Spike to ACE2. SARS-CoV-2-induced platelet activation may participate in thrombus formation and inflammatory responses in COVID-19 patients.


Subject(s)
Betacoronavirus/metabolism , Blood Platelets/metabolism , Coronavirus Infections/metabolism , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/metabolism , Thrombosis/metabolism , Adult , Aged , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/genetics , COVID-19 , Caco-2 Cells , Coronavirus Infections/virology , Female , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , PC-3 Cells , Pandemics , Peptidyl-Dipeptidase A/genetics , Platelet Aggregation/immunology , Platelet Count , Pneumonia, Viral/virology , RNA, Viral/blood , SARS-CoV-2 , Serine Endopeptidases/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Thrombosis/virology
14.
Thromb Haemost ; 120(11): 1536-1547, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32854120

ABSTRACT

Platelet activation plays a pivotal role in physiological hemostasis and pathological thrombosis causing heart attack and stroke. Previous studies conclude that simultaneous activation of Gi and G12/13 signaling pathways is sufficient to cause platelet aggregation. However, using Gq knockout mice and Gq-specific inhibitors, we here demonstrated that platelet aggregation downstream of coactivation of Gi and G12/13 depends on agonist concentrations; coactivation of Gi and G12/13 pathways only induces platelet aggregation under higher agonist concentrations. We confirmed Gi and G12/13 pathway activation by showing cAMP (cyclic adenosine monophosphate) decrease and RhoA activation in platelets stimulated at both low and high agonist concentrations. Interestingly, we found that though Akt and PAK (p21-activated kinase) translocate to the platelet membrane upon both low and high agonist stimulation, membrane-translocated Akt and PAK only phosphorylate at high agonist concentrations, correlating well with platelet aggregation downstream of concomitant Gi and G12/13 pathway activation. PAK inhibitor abolishes Akt phosphorylation, inhibits platelet aggregation in vitro and arterial thrombus formation in vivo. We propose that the PAK-PI3K/Akt pathway mediates platelet aggregation downstream of Gi and G12/13, and PAK may represent a potential antiplatelet and antithrombotic target.


Subject(s)
Platelet Aggregation , Signal Transduction/physiology , p21-Activated Kinases/physiology , Adenosine Diphosphate/pharmacology , Animals , Cell Shape , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, G12-G13/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , Mice , Mice, Knockout , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Platelet Aggregation/drug effects , Protein Transport , Proto-Oncogene Proteins c-akt/physiology , Rats , Thromboxane A2/pharmacology , rhoA GTP-Binding Protein/metabolism
15.
Stem Cell Res ; 47: 101882, 2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32592953

ABSTRACT

FLNA gene encodes an actin-binding protein filamin A and mutations in FLNA can causes X-Linked cardiac valvular dysplasia. In this study, we report the generation of ZZUNEUi008-A, a human induced pluripotent stem cell line from a 10-year-old male patient with c. 84G â†’ A in FLNA gene using non-integrative Sendai viral reprogramming technology. The ZZUNEUi008-A iPSC line expresses pluripotency markers, exhibits a normal male karyotype (46, XY) and can differentiate into three germ layers in vivo.

16.
Stem Cell Res ; 50: 102138, 2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33385793

ABSTRACT

An induced pluripotent stem cell (iPSC) line was generated from peripheral blood mononuclear cells (PBMCs) of a 41-year-old male patient with hypertrophic cardiomyopathy who carries a G3755A heterozygote mutation in the MYH6 gene. The generated iPSC line expressed pluripotency markers, exhibited a normal karyotype, presented the specific mutation, and demonstrated differentiation potential into three germ layers in vitro.

17.
J Med Virol ; 92(12): 3319-3326, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31769518

ABSTRACT

Recurrent hepatitis activity during chronic hepatitis B virus infection results in fibrosis and even hepatocellular carcinoma. It is still unclear what causes acute exacerbation. As platelets have recently been identified as a significant role in inflammation, we here investigated the role of platelets in mediating liver damage in patients with chronic hepatitis B virus infection. Platelet aggregation testing and flow cytometry were carried out to evaluate platelet activation status in 121 patients chronically infected with hepatitis B across different phases of the condition. The correlation between platelet aggregation rate and liver inflammation or liver fibrosis index was evaluated. To investigate the genesis of platelet activation, several serum cytokines were also assessed by MILLIPLEX microsphere-based multiplex cytokine assay. Active hepatitis patients showed a higher aggregation rate than others. Levels of CD62p, a marker of platelet activation, were also increased in this group of patients. Positive correlations between platelet aggregation rate and liver inflammation or liver fibrosis were also noted, indicating a significant role of platelet in the progression of liver disease. The level of tumor necrosis factor-alpha, which is known to trigger platelet activation, was markedly higher in the active hepatitis group (P < .005). Based on the findings in our study, platelet activation plays a vital role in the progression of chronic hepatitis B virus infection. Antiplatelet therapy may provide a new means of hepatitis B infection treatment.

19.
Circulation ; 136(9): 817-833, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28637879

ABSTRACT

BACKGROUND: Platelets from patients with diabetes mellitus are hyperactive. Hyperactivated platelets may contribute to cardiovascular complications and inadequate responses to antiplatelet agents in the setting of diabetes mellitus. However, the underlying mechanism of hyperactivated platelets is not completely understood. METHODS: We measured P2Y12 expression on platelets from patients with type 2 diabetes mellitus and on platelets from rats with diabetes mellitus. We also assayed platelet P2Y12 activation by measuring cAMP and VASP phosphorylation. The antiplatelet and antithrombotic effects of AR-C78511 and cangrelor were compared in rats. Finally, we explored the role of the nuclear factor-κB pathway in regulating P2Y12 receptor expression in megakaryocytes. RESULTS: Platelet P2Y12 levels are 4-fold higher in patients with type 2 diabetes mellitus compared with healthy subjects. P2Y12 expression correlates with ADP-induced platelet aggregation (r=0.89, P<0.01). P2Y12 in platelets from patients with diabetes mellitus is constitutively activated. Although both AR-C78511, a potent P2Y12 inverse agonist, and cangrelor have similar antiplatelet efficacy on platelets from healthy subjects, AR-C78511 exhibits more powerful antiplatelet effects on diabetic platelets than cangrelor (aggregation ratio 36±3% versus 49±5%, respectively, P<0.05). Using a FeCl3-injury mesenteric arteriole thrombosis model in rats and an arteriovenous shunt thrombosis model in rats, we found that the inverse agonist AR-C78511 has greater antithrombotic effects on GK rats with diabetes mellitus than cangrelor (thrombus weight 4.9±0.3 mg versus 8.3±0.4 mg, respectively, P<0.01). We also found that a pathway involving high glucose-reactive oxygen species-nuclear factor-κB increases platelet P2Y12 receptor expression in diabetes mellitus. CONCLUSIONS: Platelet P2Y12 receptor expression is significantly increased and the receptor is constitutively activated in patients with type 2 diabetes mellitus, which contributes to platelet hyperactivity and limits antiplatelet drug efficacy in type 2 diabetes mellitus.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Type 2/pathology , Receptors, Purinergic P2Y12/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Chlorides/toxicity , Cyclic AMP/analysis , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Drug Inverse Agonism , Ferric Compounds/toxicity , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Humans , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NF-kappa B/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Thrombosis/chemically induced , Thrombosis/drug therapy , Thrombosis/pathology
20.
Adv Exp Med Biol ; 906: 307-324, 2017.
Article in English | MEDLINE | ID: mdl-27628007

ABSTRACT

P2Y12 receptor is a 342 amino acid Gi-coupled receptor predominantly expressed on platelets. P2Y12 receptor is physiologically activated by ADP and inhibits adenyl cyclase (AC) to decrease cyclic AMP (cAMP) level, resulting in platelet aggregation. It also activates PI3 kinase (PI3K) pathway leading to fibrinogen receptor activation, and may protect platelets from apoptosis. Abnormalities of P2Y12 receptor include congenital deficiencies or high activity in diseases like diabetes mellitus (DM) and chronic kidney disease (CKD), exposing such patients to a prothrombotic condition. A series of clinical antiplatelet drugs, such as clopidogrel and ticagrelor, are designed as indirect or direct antagonists of P2Y12 receptor to reduce incidence of thrombosis mainly for patients of acute coronary syndrome (ACS) who are at high risk of thrombotic events. Studies on novel dual-/multi-target antiplatelet agents consider P2Y12 receptor as a promising part in combined targets. However, the clinical practical phenomena, such as "clopidogrel resistance" due to gene variations of cytochrome P450 or P2Y12 receptor constitutive activation, call for better antiplatelet agents. Researches also showed inverse agonist of P2Y12 receptor could play a better role over neutral antagonists. Personalized antiplatelet therapy is the most ideal destination for antiplatelet therapy in ACS patients with or without other underlying diseases like DM or CKD, however, there is still a long way to go.


Subject(s)
Adenosine Diphosphate/blood , Adenylyl Cyclases/blood , Blood Platelets/metabolism , Cyclic AMP/blood , Receptors, Purinergic P2Y12/blood , Thrombosis/blood , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/complications , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/pathology , Adenosine/analogs & derivatives , Adenosine/therapeutic use , Adenylyl Cyclases/genetics , Blood Platelets/drug effects , Blood Platelets/pathology , Clopidogrel , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Gene Expression Regulation , Humans , Phosphatidylinositol 3-Kinases/blood , Phosphatidylinositol 3-Kinases/genetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Receptors, Fibrinogen/blood , Receptors, Fibrinogen/genetics , Receptors, Purinergic P2Y12/genetics , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Thrombosis/complications , Thrombosis/drug therapy , Thrombosis/pathology , Ticagrelor , Ticlopidine/analogs & derivatives , Ticlopidine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...