Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Mol Sci ; 22(7)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807403

ABSTRACT

Platelets play a crucial role in the physiology of primary hemostasis and pathological processes such as arterial thrombosis; thus, developing a therapeutic target that prevents platelet activation can reduce arterial thrombosis. Pterostilbene (PTE) has remarkable pharmacological activities, including anticancer and neuroprotection. Few studies have reported the effects of pterostilbene on platelet activation. Thus, we examined the inhibitory mechanisms of pterostilbene in human platelets and its role in vascular thrombosis prevention in mice. At low concentrations (2-8 µM), pterostilbene strongly inhibited collagen-induced platelet aggregation. Furthermore, pterostilbene markedly diminished Lyn, Fyn, and Syk phosphorylation and hydroxyl radical formation stimulated by collagen. Moreover, PTE directly hindered integrin αIIbß3 activation through interfering with PAC-1 binding stimulated by collagen. In addition, pterostilbene affected integrin αIIbß3-mediated outside-in signaling, such as integrin ß3, Src, and FAK phosphorylation, and reduced the number of adherent platelets and the single platelet spreading area on immobilized fibrinogen as well as thrombin-stimulated fibrin clot retraction. Furthermore, pterostilbene substantially prolonged the occlusion time of thrombotic platelet plug formation in mice. This study demonstrated that pterostilbene exhibits a strong activity against platelet activation through the inhibition of integrin αIIbß3-mediated inside-out and outside-in signaling, suggesting that pterostilbene can serve as a therapeutic agent for thromboembolic disorders.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Stilbenes/metabolism , Animals , Blood Coagulation/drug effects , Blood Platelets/drug effects , Clot Retraction/drug effects , Collagen , Fibrinogen/metabolism , Hemostasis/drug effects , Humans , Integrin alpha2/drug effects , Integrin alpha2/metabolism , Integrin beta3/drug effects , Integrin beta3/metabolism , Integrins/drug effects , Integrins/metabolism , Mice , P-Selectin/metabolism , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Thrombosis/metabolism
2.
Biochem Pharmacol ; 188: 114519, 2021 06.
Article in English | MEDLINE | ID: mdl-33737052

ABSTRACT

Mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) is implicated in type 2 diabetes mellitus, insulin tolerance, inflammation, cancer, and atherosclerosis. We found that GNE 495 and PF 06260933 (both potent and selective MAP4K4 inhibitors) regulated human platelet activation. Immunoblotting revealed human platelets express MAP4K4, and that GNE 495 and PF 06260933 inhibited collagen-, ADP-, and thrombin-induced platelet aggregation and eventually suppressed granule release, TXA2 generation, integrin αIIbß3 activation, and clot retraction. In addition, both inhibitors elevated intracellular levels of cAMP, and coincubation with GNE 495 and aspirin or dipyridamole (a phosphodiesterase inhibitor) synergistically inhibited collagen-induced platelet aggregation and TXA2 generation. Moreover, both inhibitors phosphorylated VASP (ser157), IP3 receptor, and PKA and attenuated MAPK and PI3K/Akt/GSK3ß signaling pathways. This study is the first to demonstrate that MAP4K4 inhibitors reduce thrombus formation by inhibiting platelet activation. These findings also suggest MAP4K4 be considered an emerging target protein for the treatment of thrombosis.


Subject(s)
Aminopyridines/pharmacology , Clot Retraction/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Platelet Aggregation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adolescent , Adult , Clot Retraction/physiology , Dose-Response Relationship, Drug , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Platelet Aggregation/physiology , Protein Serine-Threonine Kinases/metabolism , Young Adult
3.
Toxins (Basel) ; 12(12)2020 11 27.
Article in English | MEDLINE | ID: mdl-33260875

ABSTRACT

Envenomation by viperid snakes may lead to severe bleeding, consumption coagulopathy, and thrombotic microangiopathy symptoms. The exact etiology or toxins responsible for thrombotic microangiopathy symptoms after snake envenomation remain obscure. Snake C-type lectin-like proteins (snaclecs) are one of the main non-enzymatic protein constituents in viper venoms, of which a majority are considered as modulators of thrombosis and hemostasis. In this study, we demonstrated that two snaclecs (mucetin and stejnulxin), isolated and identified from Protobothrops mucrosquamatus and Trimeresurus stejnegeri venoms, directly induced platelet degranulation and clot-retraction in vitro, and microvascular thrombosis has been confirmed in various organs in vivo. These snaclecs reduced cerebral blood flow and impaired motor balance and spatial memories in mice, which partially represent the thrombotic microangiopathy symptoms in some snakebite patients. The functional blocking of these snaclecs with antibodies alleviated the viper venom induced platelet activation and thrombotic microangiopathy-like symptoms. Understanding the pathophysiology of thrombotic microangiopathy associated with snake envenoming may lead to emerging therapeutic strategies.


Subject(s)
Antivenins/pharmacology , Brain Ischemia/etiology , Lectins, C-Type/physiology , Snake Bites/complications , Thrombotic Microangiopathies/etiology , Animals , Cell Degranulation/drug effects , Clot Retraction/drug effects , Female , Humans , Lectins, C-Type/isolation & purification , Male , Mice , Mice, Inbred BALB C , Platelet Activation/drug effects , Thrombotic Microangiopathies/pathology , Viper Venoms/pharmacology , Viperidae
4.
Biomed Pharmacother ; 130: 110535, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32707441

ABSTRACT

Pathophysiological reaction of platelets in the blood vessel is an indispensable part of thrombosis and cardiovascular disease, which is the most common cause of death in the world. In this study, we performed in vitro assays to evaluate antiplatelet activity of arctigenin in human platelets and attempted to identify the mechanism responsible for thromboxane A2 (TXA2) generation, integrin αIIbß3 activation and cAMP pathway. Arctigenin exhibited obvious inhibitory effects on collagen-, thrombin-, and ADP-induced human platelet aggregation, granule secretion, TXA2 generation, integrin αIIbß3 activation, and clot retraction. Additionally, we found that arctigenin attenuated PI3K/Akt/mTOR/GSK-3ß and MAPK signaling pathways, and increased cAMP level. Accordingly, the findings support that arctigenin attenuates thrombotic events through the inhibition of platelet activation and platelet plug formation. Therefore, we suggest that arctigenin may have therapeutic potential as an antiplatelet and antithrombotic agent.


Subject(s)
Clot Retraction/drug effects , Cyclic AMP , Fibrinolytic Agents/pharmacology , Furans/pharmacology , Lignans/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction/drug effects , Thromboxane A2/biosynthesis , Blood Platelets/drug effects , Glycogen Synthase Kinase 3 beta/drug effects , Humans , In Vitro Techniques , Mitogen-Activated Protein Kinases , Oncogene Protein v-akt/drug effects , Phosphatidylinositol 3-Kinases/drug effects , TOR Serine-Threonine Kinases/drug effects
5.
Clin Respir J ; 14(6): 549-556, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32052554

ABSTRACT

BACKGROUND: This study assessed predictors of pulmonary thromboembolism (PE) resolution and their implications for clinical outcome. METHOD: A total of 150 patients with acute PE diagnosed by computed tomography pulmonary angiography (CTPA) were included. All patients received anticoagulant therapy for 3-6 months and were followed-up for at least 2 years. d-dimer levels in plasma were assayed at the first admission and during follow-up. RESULTS: The rate of CTPA-confirmed PE resolution was 48.67% at 6 months, 68% at 12 months, and 78.67% at 24 months. Thirty-nine patients had recurrent thrombosis after anticoagulation therapy was stopped, whereas 93 patients had complete resolution. The initial d-dimer level positively correlated with the pulmonary artery obstruction index (PAOI) (r = 0.21; P = 0.015), but did not significantly differ between patients experiencing resolution or recurrence. In contrast, the follow-up mean d-dimer level was significantly higher in the recurrent group (P < 0.001), and this level was an independent risk factor for recurrent PE after the termination of anticoagulation treatment (OR 1.003, 95%CI 1.002 to 1.004; P < 0.001). Higher initial thromboembolic burden measured by PAOI was associated with residual thromboemboli (P = 0.004) and recurrence (P = 0.03), but was not an independent risk factor for either. CONCLUSIONS: Elevated d-dimer is an independent risk factor for PE recurrence. A higher initial thromboembolic burden may be associated with unresolved thromboemboli or recurrence.


Subject(s)
Anticoagulants/pharmacology , Clot Retraction/drug effects , Fibrin Fibrinogen Degradation Products/analysis , Pulmonary Embolism/blood , Aged , Anticoagulants/therapeutic use , Computed Tomography Angiography/methods , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Pulmonary Artery/diagnostic imaging , Pulmonary Artery/pathology , Pulmonary Embolism/diagnosis , Pulmonary Embolism/drug therapy , Recurrence , Retrospective Studies , Risk Factors , Thrombosis/metabolism , Tomography, X-Ray Computed/methods , Venous Thrombosis/blood , Venous Thrombosis/complications , Venous Thrombosis/diagnosis
6.
Int J Mol Sci ; 20(21)2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31683623

ABSTRACT

Tyrosine kinase inhibitors (TKI) such as the BCR-ABL inhibitor dasatinib and nilotinib are highly effective therapies for chronic myeloid leukemia (CML). However, several lines of evidence suggest that dasatinib can induce bleeding which may be due to impaired collagen-induced platelet adhesion, aggregation, and secretion. Sarcoma family kinases (SFK) play central role in the GPVI-induced signaling pathway. We aimed to investigate whether and how dasatinib can modulate SFK-mediated platelet procoagulant activity in a purified system and in dasatinib/nilotinib treated CML patients. In platelet rich plasmas of healthy volunteers, dasatinib dose-dependently reduced convulxin-induced phosphatidylserine exposure and attenuated thrombin formation. Similarly to these changes, integrin activation and clot retraction were also significantly inhibited by 100 nM dasatinib. Platelets isolated from dasatinib treated patients showed a significantly lower phosphatidylserine expression upon convulxin activation compared to premedication levels. In these samples, thrombin generation was significantly slower, and the quantity of formed thrombin was less compared to the trough sample. Western blot analyses showed decreased phosphorylation levels of the C-terminal tail and the activation loop of SFKs upon dasatinib administration. Taken together, these results suggest that dasatinib inhibits the formation of procoagulant platelets via the GPVI receptor by inhibiting phosphorylation of SFKs.


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/drug effects , Clot Retraction/drug effects , Dasatinib/pharmacology , Platelet Activation/drug effects , Blood Platelets/metabolism , Crotalid Venoms/pharmacology , Humans , Lectins, C-Type , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Phosphatidylserines/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thrombin/metabolism , src-Family Kinases/metabolism
7.
Eur J Pharmacol ; 865: 172734, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31614139

ABSTRACT

Morin hydrate is an active constituent of Morus alba L, Prunus dulcis, and Cudrania tricuspidata and has been reported to inhibit platelet activation in vivo and in vitro, but no reports have been issued on its regulation of αIIbß3, a platelet-specific integrin and thromboxane A2 (TXA2), positive feedback molecule. In this study, we investigated the anti-platelet activity of morin hydrate in collagen- and thrombin-induced human platelets and attempted to identify the mechanism responsible for integrin αIIbß3 activation and TXA2 generation. Our results demonstrated that morin hydrate (25-100 µM) inhibited collagen- and thrombin-induced platelet aggregation, granule secretion (P-selectin expression, ATP, and serotonin release), calcium mobilization, TXA2 production, integrin αIIbß3 activation, and clot retraction. Additionally, morin hydrate attenuated the phosphorylations of phospholipase Cγ2 (PLCγ2), cytosolic phospholipase A2 (cPLA2), phosphoinositide 3-kinase (PI3K), Akt, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK), and enhanced the phosphorylations of inositol trisphosphate receptor (IP3 receptor) and cyclic adenosine monophosphate (cAMP) generation. However, it had no effect on the coagulation pathway. Taken together, these observations indicate morin hydrate inhibits platelet-mediated thrombosis by down-regulating TXA2 production and integrin αIIbß3 activation, and by upregulating cAMP generation, and thus, inhibits clot retraction. These results suggest morin hydrate may have therapeutic potential as a treatment for platelet-activation-related diseases.


Subject(s)
Clot Retraction/drug effects , Cyclic AMP/metabolism , Flavonoids/pharmacology , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thromboxane A2/metabolism , Blood Coagulation/drug effects , Collagen/pharmacology , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Thrombin/pharmacology
8.
Thromb Haemost ; 119(10): 1655-1664, 2019 10.
Article in English | MEDLINE | ID: mdl-31370073

ABSTRACT

All-trans retinoic acid (ATRA) is widely used for induction of complete remission in patients with acute promyelocytic leukemia (APL). ATRA also regulates protein kinase C (PKC) activity. Therapeutic use of ATRA reportedly interferes with hemostatic function in APL patients, including effects on coagulation or other vascular cells, although effects of ATRA on platelets remain unclear. This study aims to investigate the effect of therapeutic-relevant doses of ATRA on platelet function. Human platelets were preincubated with ATRA (0-20 µM) for 1 hour at 37°C, followed by analysis of aggregation, granule secretion, receptor expression by flow cytometry, platelet spreading, or clot retraction. Additionally, ATRA (10 mg/kg) was injected intraperitoneally into mice and tail bleeding time and arterial thrombus formation were evaluated. ATRA inhibited platelet aggregation and adenosine triphosphate release induced by collagen (5 µg/mL) or thrombin (0.05 U/mL) in a dose-dependent manner without affecting P-selectin expression or surface levels of glycoprotein (GP) Ibα, GPVI, or αIIbß3. ATRA-treated platelets demonstrated reduced spreading on immobilized fibrinogen or collagen and reduced thrombin-induced clot retraction together with reduced phosphorylation of Syk and PLCγ2. In addition, ATRA-treated mice displayed significantly impaired hemostasis and arterial thrombus formation in vivo. Further, in platelets stimulated with either collagen-related peptide or thrombin, ATRA selectively inhibited phosphorylation of PKCßI (Ser661) and PKCδ (Thr505), but not PKCα or PKCßII phosphorylation (Thr638/641). In conclusion, ATRA inhibits platelet function and thrombus formation, possibly involving direct or indirect inhibition of PKCßI/δ, indicating that ATRA might be beneficial for the treatment of thrombotic or cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Protein Kinase C beta/metabolism , Protein Kinase C-delta/metabolism , Thrombosis/drug therapy , Tretinoin/pharmacology , Animals , Blood Coagulation/drug effects , Blood Platelets/metabolism , Clot Retraction/drug effects , Collagen/chemistry , Cytoskeleton/metabolism , Hemostasis/drug effects , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Function Tests , Signal Transduction/drug effects
9.
Free Radic Biol Med ; 141: 426-437, 2019 09.
Article in English | MEDLINE | ID: mdl-31279970

ABSTRACT

Hypochlorite (HOCl), a strong oxidant and antimicrobial agent, has been proposed to be associated with hemostatic abnormalities during inflammatory response. However, its complex impact on hemostasis is not completely understood. In this report we studied the effect of clinically relevant (micromolar) HOCl concentrations on thrombus formation under flow, kinetics of platelet-fibrin clot formation, its architecture, retraction, and lysis. We found that HOCl (up to 500 µM) did not affect kinetics of coagulation measured in whole blood. HOCl (500-1000 µM) markedly diminished thrombus formation under flow. Clot retraction rate was reduced by HOCl dose-dependently (50-500 µM). HOCl (125-500 µM) inhibited fibrinolysis in whole blood and in platelet-depleted plasma, dose-dependently. Activity of plasmin was reduced by HOCl at concentrations started from 500 µM. HOCl (up to 500 µM) did not reduce plasminogen binding to fibrin under flow. HOCl (125-500 µM) modulated architecture of fibrin- and platelet-fibrin clots towards structures made of thin and densely packed fibers. Exposure of pure fibrinogen to HOCl (10-1000 µM) resulted in formation of dityrosine and was associated with altered fibrin structure derived from such modified fibrinogen. HOCl-altered fibrin net structure was not related with modulation of platelet procoagulant response, thrombin generation, and factor XIII activity. We conclude that, in human blood, clinically relevant HOCl concentrations may inhibit thrombus formation under flow, clot retraction and fibrinolysis. Fibrinolysis and clot retraction seem to be the most sensitive to HOCl-evoked inhibition. HOCl-modified fibrinogen and altered clot structure associated with it are likely to be primary sources of attenuated fibrinolysis.


Subject(s)
Clot Retraction/drug effects , Hypochlorous Acid/pharmacology , Inflammation/drug therapy , Thrombosis/drug therapy , Blood Coagulation/drug effects , Blood Platelets/metabolism , Factor XIII/drug effects , Fibrin/metabolism , Fibrinogen/metabolism , Fibrinolysis/drug effects , Hemostasis/drug effects , Humans , Hypochlorous Acid/metabolism , Inflammation/blood , Inflammation/pathology , Peroxidase/metabolism , Thrombin/metabolism , Thrombosis/blood , Thrombosis/pathology
10.
J Transl Med ; 16(1): 311, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442147

ABSTRACT

BACKGROUND: Platycodin D (PD) is one of the major bioactive components of the roots of Platycodon grandiflorum and possesses multiple biological and pharmacological properties, such as antiviral, anti-inflammatory, and anti-cancer activities. However, whether it affects platelet function remains unclear. This study aims to evaluate the role of PD in platelet function and thrombus formation. METHODS: Platelets were treated with PD followed by measuring platelet aggregation, activation, spreading, clot retraction, expression of glycoprotein receptors. Moreover, mice platelets were treated with PD and infused into wild-type mice for analysis of in vivo hemostasis and arterial thrombosis. RESULTS: Platycodin D treatment significantly inhibited platelet aggregation in response to collagen, ADP, arachidonic acid and epinephrine, reduced platelet P-selectin expression, integrin αIIbß3 activation, spreading on fibrinogen as well as clot retraction, accompanied with decreased phosphorylation of Syk and PLCγ2 in collagen-related peptide or thrombin-stimulated platelets. Moreover, PD-treated mice platelets presented significantly impaired in vivo hemostasis and arterial thrombus formation. Interestingly, PD induced internalization of glycoprotein receptors αIIbß3, GPIbα and GPVI. However, GM6001, cytochalasin D, BAPTA-AM and wortmannin did not prevent PD-induced internalization of receptors. CONCLUSIONS: Our study demonstrates that PD inhibits platelet aggregation, activation and impairs hemostasis and arterial thrombosis, suggesting it might be a potent anti-thrombotic drug.


Subject(s)
Blood Platelets/metabolism , Blood Platelets/pathology , Endocytosis/drug effects , Platelet Membrane Glycoproteins/metabolism , Saponins/pharmacology , Thrombosis/pathology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Arteries/drug effects , Arteries/pathology , Blood Platelets/drug effects , Clot Retraction/drug effects , Hemostasis/drug effects , Humans , Mice, Inbred C57BL , P-Selectin/metabolism , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Signal Transduction/drug effects , Syk Kinase/metabolism
11.
J Biochem Mol Toxicol ; 32(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-29077251

ABSTRACT

We discovered recently in vitro and in vivo antithrombotic and cytotoxicity effects of ferulic acid. The cytotoxicity assays showed that ferulic acid (∼300 µg/mL) did not cause any significant toxicity on three cell lines, platelets, leukocytes, and erythrocytes. In vitro assays showed inhibitory effects of ferulic acid on thrombin (THR)- or collagen/epinephrine-stimulated platelet activation by inhibiting platelet aggregation, and decreasing clot retraction activity. The in vitro effect of ferulic acid on THR-stimulated platelet activation was proved by the decrease in the secretion of serotonin from the platelets. The anticoagulant effects of ferulic acid were confirmed by the prolongation of the intrinsic or/and extrinsic pathways and the delay of recalcification time in plasma coagulation. Ferulic acid had antithrombotic effect in acute thromboembolism model in vivo, and decreased the expression of αIIb ß3 /FIB and phosphorylation of AKT in THR-stimulated platelet activation in vivo, and their antithrombotic efficacies hold promise for therapeutic targeting in our ongoing studies.


Subject(s)
Anticoagulants/therapeutic use , Coumaric Acids/therapeutic use , Disease Models, Animal , Platelet Aggregation Inhibitors/therapeutic use , Thromboembolism/prevention & control , 3T3-L1 Cells , Animals , Anticoagulants/adverse effects , Anticoagulants/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Survival/drug effects , Clot Retraction/drug effects , Coumaric Acids/adverse effects , Coumaric Acids/pharmacology , Erythrocytes/cytology , Erythrocytes/drug effects , Hemostasis/drug effects , Hep G2 Cells , Humans , Leukocytes/cytology , Leukocytes/drug effects , Male , Mice , Mice, Inbred ICR , NIH 3T3 Cells , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacology , Serotonin/chemistry , Serotonin/metabolism , Serotonin Antagonists/adverse effects , Serotonin Antagonists/pharmacology , Serotonin Antagonists/therapeutic use
12.
Acta Pharmacol Sin ; 38(5): 651-659, 2017 May.
Article in English | MEDLINE | ID: mdl-28260800

ABSTRACT

The signal transducer and activator of transcription 3 (STAT3) plays a critical role in platelet functions. This study sought to understand the effects of the STAT3 inhibitor SC99 on platelet activation and aggregation. Immunoblotting assays were applied to measure the effects of SC99 on the STAT3 signaling pathway. A ChronoLog aggregometer was used to evaluate platelet aggregation. A flow cytometer was used to evaluate P-selectin expression in the presence of SC99. AlamarBlue and Annexin-V staining were used to evaluate platelet viability and apoptosis, respectively. A fluorescence microscope was applied to analyze platelet spreading. SC99 inhibited the phosphorylation of JAK2 and STAT3 in human platelets but had no effects on the phosphorylation of AKT, p65 or Src, all of which are involved in platelet activation. Further studies revealed that SC99 inhibited human platelet aggregation induced by collagen and thrombin in a dose-dependent manner. SC99 inhibited thrombin-induced P-selectin expression and fibrinogen binding to single platelets. Moreover, SC99 inhibited platelet spreading on fibrinogen and clot retraction mediated by outside-in signaling. SC99 inhibited platelet aggregation in mice but it did not significantly prolong the bleeding time. Taken together, the present study revealed that SC99 inhibited platelet activation and aggregation as a STAT3 inhibitor. This agent can be developed as a promising treatment for thrombotic disorders.


Subject(s)
Hydrazones/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Bleeding Time , Clot Retraction/drug effects , Humans , Hydrazones/toxicity , Mice, Inbred C57BL , Platelet Aggregation Inhibitors/toxicity , Signal Transduction
13.
J Asthma ; 53(6): 589-98, 2016 08.
Article in English | MEDLINE | ID: mdl-27145190

ABSTRACT

OBJECTIVE: Asthma enhances the risk of pulmonary embolism. The mechanism of this phenomenon is unclear. METHODS: We evaluated the kinetics of clot formation, clot retraction rate (CRR), clot volume at 40 min, the rate of lactate production (a marker of aerobic glycolysis in platelets in contracting clots), blood eosinophil count (EOS), nitric oxide in exhaled breath (FENO), and spirometry (FEV1) in 50 healthy controls and in 81 allergic asthmatics (41 subjects with steroid-naïve asthma and 40 with steroid-treated asthma). RESULTS: Thromboelastometry revealed that only steroid-treated asthmatics had slightly activated coagulation. Compared with healthy controls, whole asthmatics demonstrated (p < 0.05) reduced CRR, higher clot volume at 40 minutes, higher FENO, decreased FEV1, elevated EOS, and augmented lactate production in retracting clots. Reduced CRR was observed also in the absence of native plasma. In whole study population (asthmatics and healthy controls), CRR positively correlated with spirometry (rS = 0.668, p = <0.001) and negatively with FENO (rS = -0.543; p < 0.001), EOS (rS = -0.367, p < 0.002), and lactate production (rS = -0.791; p < 0.001). However, in steroid-treated asthmatics, the CRR did not correlate with FENO and EOS. In all study patients lactate production negatively correlated with FEV1 and positively with FENO. CONCLUSION: Collectively, this data is consistent with the hypothesis that, in asthmatics, reactive nitrogen species produced in the lungs may reduce platelet contractility (and CRR) through the diminution of platelet energy production. CRR inhibition would predispose asthmatics to pulmonary embolism.


Subject(s)
Asthma/blood , Blood Platelets/metabolism , Clot Retraction/physiology , Thrombosis/metabolism , Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Asthma/drug therapy , Case-Control Studies , Clot Retraction/drug effects , Eosinophils/metabolism , Female , Humans , Lactic Acid/metabolism , Male , Middle Aged , Nitric Oxide/metabolism , Respiratory Function Tests , Thrombelastography , Young Adult
14.
Thromb Haemost ; 114(5): 1004-19, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26245294

ABSTRACT

P2Y12 receptor is required for sustained activation of integrin αIIbß3, irreversible platelet aggregation and thrombus stabilisation. Tetraspanin superfamily member CD151 associates with integrin αIIbß3 and plays critical roles in regulation of thrombus growth and stability in vivo. The possible functional relationship between P2Y12 and CD151 in a molecular cluster in platelets may affect thrombus formation. Hence our aim was to investigate the physical and functional requirements for this association in platelets. Our investigations reveal a specific and constitutive association between CD151 and P2Y12 receptor in human platelets shown by immunoprecipitation/western blot studies and by flow cytometry. Specifically, the prominent association involves CD151 with P2Y12 oligomers, and to a lesser extent P2Y12 monomers. This association is not altered by platelet aggregation induced by different agonists. There is also a distinct complex of tetraspanin CD151 with ADP purinergic receptor P2Y12 but not P2Y1. P2Y12 oligomer interaction with CD151 is selective as compared to other tetraspanins. To investigate the functional relationship between these receptors in platelets we used wild-type or CD151 knockout (KO) mice treated with either PBS or 50 mg/kg clopidogrel. CD151 KO mice treated with clopidogrel exhibited synergy in delayed kinetics of clot retraction, in PAR-4 and collagen-mediated platelet aggregation, platelet spreading on fibrinogen and without restricting cAMP inhibition. Clopidogrel treated CD151 KO arterioles showed smaller and less stable thrombi with increased tendency to embolise ex vivo and in vivo. These studies demonstrate a complementary role between CD151 and P2Y12 receptor in platelets in regulating thrombus growth and stability.


Subject(s)
Blood Platelets/physiology , Receptors, Purinergic P2Y12/metabolism , Tetraspanin 24/metabolism , Tetraspanins/metabolism , Thrombosis/physiopathology , Animals , Blood Platelets/drug effects , Cells, Cultured , Clopidogrel , Clot Retraction/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Platelet Aggregation Inhibitors/administration & dosage , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding/drug effects , Protein Binding/genetics , Tetraspanin 24/genetics , Tetraspanins/genetics , Ticlopidine/administration & dosage , Ticlopidine/analogs & derivatives
15.
Free Radic Res ; 48(12): 1443-53, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25236568

ABSTRACT

Using porcine blood, we examined the impact of hypochlorite, product of activated inflammatory cells, on clot retraction (CR), an important step of hemostasis. We found that, in vitro, HOCl is able to reduce CR rate and enlarge final clot size in whole blood (t.c. 100 µM), platelet-rich plasma (PRP) threshold concentration (t.c. 50 µM), and an artificial system (washed platelets and fibrinogen) (t.c. 25 nM). Combination of low HOCl and peroxynitrite concentrations resulted in synergistic inhibition of CR by these stressors. Concentrations of HOCl completely inhibiting CR failed to affect the kinetics of coagulation measured in PRP and in platelet-free plasma. Concentrations of HOCl reducing CR rate in PRP augmented production of lactate, inhibited consumption of oxygen by platelets, and decreased total adenosine triphosphate (ATP) content in PRP-derived clots. In an artificial system, concentrations of HOCl resulting in inhibition of CR (25-100 nM) reduced mitochondrial transmembrane potential and did not affect actin polymerization in thrombin-stimulated platelets. These concentrations of HOCl failed to affect the adhesion of washed platelets to fibrinogen and to evoke sustained calcium signal, thus excluding stressor action on glycoprotein IIb/IIIa receptors. Exogenously added Mg-ATP almost completely recovered HOCl-mediated retardation of CR. Concentrations of HOCl higher than those affecting CR reduced thromboelastometric variables (maximum clot firmness and α angle). We conclude that low clinically relevant HOCl concentrations may evoke the inhibition of CR via the reduction of platelet contractility resulted from malfunction of platelet mitochondria. At the inflammatory conditions, CR may be the predominant HOCl target.


Subject(s)
Blood Platelets/drug effects , Clot Retraction/drug effects , Energy Metabolism/drug effects , Hypochlorous Acid/pharmacology , Mitochondria/drug effects , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Dose-Response Relationship, Drug , Mitochondria/metabolism , Structure-Activity Relationship , Swine , Time Factors
16.
Thromb Haemost ; 111(3): 508-17, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24352480

ABSTRACT

3-phosphoinositide-dependent protein kinase 1 (PDK1), a member of the protein A,G and C (AGC) family of proteins, is a Ser/Thr protein kinase that can phosphorylate and activate other protein kinases from the AGC family, including Akt at Thr308, all of which play important roles in mediating cellular responses. The functional role of PDK1 or the importance of phosphorylation of Akt on Thr308 for its activity has not been investigated in human platelets. In this study, we tested two pharmacological inhibitors of PDK1, BX795 and BX912, to assess the role of Thr308 phosphorylation on Akt. PAR4-induced phosphorylation of Akt on Thr308 was inhibited by BX795 without affecting phosphorylation of Akt on Ser473. The lack of Thr308 phosphorylation on Akt also led to the inhibition of PAR4-induced phosphorylation of two downstream substrates of Akt, viz. GSK3ß and PRAS40. In vitro kinase activity of Akt was completely abolished if Thr308 on Akt was not phosphorylated. BX795 caused inhibition of 2-MeSADP-induced or collagen-induced aggregation, ATP secretion and thromboxane generation. Primary aggregation induced by 2-MeSADP was also inhibited in the presence of BX795. PDK1 inhibition also resulted in reduced clot retraction indicating its role in outside-in signalling. These results demonstrate that PDK1 selectively phosphorylates Thr308 on Akt thereby regulating its activity and plays a positive regulatory role in platelet physiological responses.


Subject(s)
Blood Platelets/physiology , Oncogene Protein v-akt/metabolism , Threonine/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphate/metabolism , Blood Platelets/drug effects , Cells, Cultured , Clot Retraction/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Phosphorylation/drug effects , Platelet Activation/drug effects , Pyrimidines/pharmacology , Receptors, Thrombin/metabolism , Serine/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Thromboxanes/metabolism
17.
Life Sci ; 93(20): 707-13, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24096143

ABSTRACT

AIMS: We studied the role of serine/threonine phosphatases (PSTPs) on αIIbß3 signaling and the potential selectivity of the level of PSTP inhibition with okadaic acid (OA) on αIIbß3 signaling for regulation of platelet aggregation and clot retraction. MAIN METHODS: We used washed platelets from normal donors and OA as inhibitor of PSTPs. Clot retraction was induced by 1U/mL of thrombin. Reorganized cytoskeleton was isolated from Triton X-100 lysed platelets. The presence of proteins incorporated to the cytoskeleton was assayed by immunoblotting with specific antibodies. KEY FINDINGS: We found that both 100 and 500 nM OA blocked platelet mediated clot retraction. In contrast, only 500 nM OA inhibited thrombin-induced inside-out αIIbß3 activation, platelet aggregation, and cytoskeletal reorganization. Among markers of αIIbß3 outside-in signaling, 500 nM OA inhibited the incorporation to the cytoskeleton of syk, src, and FAK (Focal Adhesion Kinase) tyrosine kinases and the incorporation and phosphorylation at Tyr(759) of the ß3 chain of αIIbß3, while 100 nM OA only inhibited the FAK translocation and its tyrosine phosphorylation. SIGNIFICANCE: The level of inhibition of PSTPs by low or high OA concentration (33% and 73% inhibition, respectively) in intact whole cells differentially regulates platelet aggregation and integrin signaling, but have a common effect in blocking clot retraction. The latter may be associated with the presence of phosphorylated FAK in the cytoskeleton. This study reveals a novel target for anti-platelet treatment to block clot retraction without affecting the platelet hemostatic function by a partial inhibition of PSTPs.


Subject(s)
Blood Platelets/drug effects , Clot Retraction/drug effects , Okadaic Acid/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Blood Platelets/cytology , Blood Platelets/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Platelet Aggregation/drug effects , Signal Transduction/drug effects
18.
Blood ; 122(19): 3340-8, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24002447

ABSTRACT

Membrane rafts are spatially and functionally heterogenous in the cell membrane. We observed that lysenin-positive sphingomyelin (SM)-rich rafts are identified histochemically in the central region of adhered platelets where fibrin and myosin are colocalized on activation by thrombin. The clot retraction of SM-depleted platelets from SM synthase knockout mouse was delayed significantly, suggesting that platelet SM-rich rafts are involved in clot retraction. We found that fibrin converted by thrombin translocated immediately in platelet detergent-resistant membrane (DRM) rafts but that from Glanzmann's thrombasthenic platelets failed. The fibrinogen γ-chain C-terminal (residues 144-411) fusion protein translocated to platelet DRM rafts on thrombin activation, but its mutant that was replaced by A398A399 at factor XIII crosslinking sites (Q398Q399) was inhibited. Furthermore, fibrin translocation to DRM rafts was impaired in factor XIII A subunit-deficient mouse platelets, which show impaired clot retraction. In the cytoplasm, myosin translocated concomitantly with fibrin translocation into the DRM raft of thrombin-stimulated platelets. Furthermore, the disruption of SM-rich rafts by methyl-ß-cyclodextrin impaired myosin activation and clot retraction. Thus, we propose that clot retraction takes place in SM-rich rafts where a fibrin-αIIbß3-myosin complex is formed as a primary axis to promote platelet contraction.


Subject(s)
Blood Platelets/metabolism , Clot Retraction/genetics , Factor XIII/metabolism , Fibrin/metabolism , Myosins/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Sphingomyelins/metabolism , Animals , Blood Coagulation/drug effects , Blood Coagulation/genetics , Blood Platelets/cytology , Blood Platelets/drug effects , Clot Retraction/drug effects , Factor XIII/genetics , Fibrin/genetics , Gene Expression , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Myosins/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Transport , Signal Transduction , Thrombin/pharmacology , Transferases (Other Substituted Phosphate Groups)/deficiency , Transferases (Other Substituted Phosphate Groups)/genetics
19.
Acad Emerg Med ; 20(5): 449-55, 2013 May.
Article in English | MEDLINE | ID: mdl-23672358

ABSTRACT

OBJECTIVES: Recombinant tissue plasminogen activator (rt-PA) is a lytic medication widely used in the emergency department to treat acute thrombotic disorders such as ischemic stroke and myocardial infarction. It is known in the clinical use of this drug that it can be less effective in approximately 25% of individuals receiving such treatment. However, there are no data on the variation of lytic efficacy of rt-PA in decreasing individuals' clot size over time. In this study, in vitro lytic efficacy was determined by measuring the decrease in clot diameter after 30 minutes of drug exposure. The authors sought to explore whether there are individuals who do not respond to this lytic therapy and to estimate the rate of nonresponse. METHODS: Human whole blood clots were made from blood drawn from 22 adult volunteers. The only exclusion criterion was the use of aspirin within 72 hours of the blood draw. Blood clots were allowed to spontaneously form at room temperature and were then incubated at 37°C for 3 hours to ensure complete clot retraction. Sample clots from the same individuals were then exposed to human fresh-frozen plasma (hFFP) control or rt-PA in hFFP (rt-PA) at a concentration of 3.15 µg/mL. All clots were exposed at 37°C for 30 minutes, and clot diameter was measured as a function of time, using a microscopic imaging technique. The fractional clot loss (FCL), which is the percentage decrease in clot diameter at 30 minutes, was used as a measure of lytic efficacy. RESULTS: Means with standard deviation (SD) FCL values were 8.6% (±3.0%) for control and 20.6% (±9.3%) for rt-PA-treated clots. The mean (±SD) difference in FCL values was 12.0% (±8.8%) and was significant (p < 0.05, paired t-test). Five of the 22 subjects (23%) were "rt-PA nonresponders," in that their FCL (rt-PA) values fell within that of the FCL control values. CONCLUSIONS: Overall, rt-PA does not produce clot lysis in vitro in clots from a substantial minority of the population, likely due to individual variations in clot composition and structure.


Subject(s)
Blood Coagulation/drug effects , Clot Retraction/drug effects , Fibrinolytic Agents/pharmacology , Tissue Plasminogen Activator/pharmacology , Adult , Female , Humans , Male , Middle Aged
20.
Blood ; 121(18): 3718-26, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23444402

ABSTRACT

The effects of phosphoinositide-dependent protein kinase 1 (PDK1), a master kinase in the phosphoinositide 3-kinase/Akt pathway, on platelet activation are unknown. Accordingly, platelet-specific PDK1-deficient mice were characterized to elucidate the platelet-related function(s) of PDK1. We found that PDK1 deficiency caused mild thrombocytopenia. The aggregation of PDK1(-/-) platelets was diminished in response to low levels of thrombin, U46619, and adenosine 5'-diphosphate. Further results demonstrated that PDK1 regulates thrombin-induced platelet activation by affecting αIIbß3-mediated outside-in signaling. This result provided an explanation for the diminished spreading of PDK1(-/-) platelets on immobilized fibrinogen (Fg) and the decreased rate of clot retraction in platelet-rich plasma (PRP) containing PDK1(-/-) platelets. PDK1 deficiency diminished agonist-induced Akt Ser473 phosphorylation and thoroughly abolished Akt Thr308 and Gsk3ß Ser9 phosphorylation in response to agonist treatment and platelet spreading, respectively. A Gsk3ß inhibitor fully restored the aggregation of PDK1(-/-) platelets in response to low levels of thrombin, normal spreading of PDK1(-/-) platelets on Fg, and normal clot retraction in PRP containing PDK1(-/-) platelets. Those results indicated that Gsk3ß is one of the major downstream effectors of PDK1 in thrombin-induced platelet activation and αIIbß3-mediated outside-in signaling. In addition, in vivo data demonstrated that PDK1 is an important regulator in arterial thrombosis formation.


Subject(s)
Platelet Activation/genetics , Protein Serine-Threonine Kinases/physiology , Thrombosis/genetics , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Arteries/drug effects , Arteries/metabolism , Arteries/pathology , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Blood Platelets/physiology , Clot Retraction/drug effects , Clot Retraction/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Thrombin/pharmacology , Thrombosis/pathology , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...