Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Clin Biochem ; 113: 64-69, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36610469

ABSTRACT

BACKGROUND: α-thalassemia is an inherited blood disorder caused by variants in the α-globin gene cluster. Identification of the pathogenic α-globin gene variants is important for the diagnosis and management of thalassemia. METHODS: Two suspected families from Xiantao, Hubei Province were recruited in this study. The family members underwent hemoglobin testing. Polymerase Chain Reaction based reverse dot blot (PCR-RDB) was employed to identify the known variants. Next-generation sequencing (NGS) and third-generation sequencing (TGS) were performed to screen the potential disease-causing variants, which were validated by Sanger sequencing and multiplex ligation-dependent probe amplification (MLPA). RESULTS: Hematological analysis suggested that proband A had α-thalassemia traits, and proband B had HbH disease traits. However, only a -α3.7 mutation had been detected by PCR-RDB and NGS in the proband of family B. Subsequent TGS identified a novel 10.3 kb deletion (NC_000016.10:g.172342-182690del) covering the HBA1, HBQ1 and HBA2 genes in the α-globin gene cluster in both family A and B, which was confirmed by Sanger sequencing and MLPA. These results indicated that the novel deletion is likely responsible for α-thalassemia. CONCLUSION: A novel α-thalassemia deletion was identified for the two families by TGS. Our work broadened the molecular spectrum of α-thalassemia, and was beneficial for the diagnosis, genetic counseling and management of α-thalassemia.


Subject(s)
alpha-Thalassemia , Humans , alpha-Thalassemia/diagnosis , alpha-Thalassemia/genetics , Pedigree , Mutation , Multiplex Polymerase Chain Reaction , alpha-Globins/genetics
2.
Chin J Integr Med ; 23(1): 18-24, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27299461

ABSTRACT

OBJECTIVE: To compare the effect between nebulized and intravenous administration of Shenmai Injection () on pulmonary gas exchange function of patients following tourniquet-induced lower limb ischemia-reperfusion. METHODS: Thirty-eight patients scheduled for lower extremity surgery were randomized into three groups using the closed envelop method: Shenmai Injection was administered 30 min before tourniquet inflflation by nebulization [0.6 mL/kg in 10 mL normal saline (NS)] in the nebulization group or by intravenous drip (0.6 mL/kg dissolved in 250 mL of 10% glucose) in the intravenous drip group, and equal volume of NS was given intravenously in the NS group; 15 in each group. Arterial blood gases were analyzed, serum levels of malonaldehyde (MDA) and interleukine-6 (IL-6) and interleukine-8 (IL-8) were determined using the method of thiobarbituric acid reaction and enzyme-linked immuno sorbent assay respectively just before tourniquet inflflation (T0), and at 0.5 h (T1), 2 h (T2), 6 h (T3) after tourniquet deflflation. RESULTS: Compared with baselines at T0, MDA levels signifificantly increased at T2, T3 in the NS group and at T3 in the nebulization group, and IL-6 and IL-8 levels were signifificantly increased at T2, T3 in NS, the intravenous drip and the nebulization groups (P <0.05). Arterial pressure of oxygen (PaO2) at T3 was decreased, while alveolararterial oxygen tension showed difference (PA-aDO2) at T3 in the NS group; RI at T3 in both intravenous drip and the nebulization groups were enhanced (P <0.05). Compared with the NS group, MDA and IL-8 levels at T2, T3, IL-6 at T3 in the intravenous drip group, and IL-8 at T3 in the nebulization group were all remarkably increased (P <0.05). Additionally, MDA level at T3 in the nebulization group was higher than that in the intravenous drip group (P <0.05). CONCLUSIONS: Intravenous administration of Shenmai Injection provided a better protective effect than nebulization in mitigating pulmonary gas exchange dysfunction in patients following tourniquet-induced limb ischemia-reperfusion.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/therapeutic use , Pulmonary Gas Exchange , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Tourniquets/adverse effects , Adult , Blood Gas Analysis , Drug Administration Routes , Drug Combinations , Drugs, Chinese Herbal/pharmacology , Female , Humans , Injections , Interleukin-6/blood , Interleukin-8/blood , Male , Malondialdehyde/blood , Pulmonary Gas Exchange/drug effects , Reperfusion Injury/blood
3.
Thromb Haemost ; 116(5): 931-940, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27465150

ABSTRACT

Phospholipase C (PLC)-ß2 (gene PLCB2) is a critical regulator of platelet responses upon activation. Mechanisms regulating of PLC-ß2 expression in platelets/MKs are unknown. Our studies in a patient with platelet PLC-ß2 deficiency revealed the PLCB2 coding sequence to be normal and decreased platelet PLC-ß2 mRNA, suggesting a defect in transcriptional regulation. PLCB2 5'- upstream region of the patient revealed a heterozygous 13 bp deletion (-1645/-1633 bp) encompassing a consensus sequence for nuclear factor-κB (NF-κB). This was subsequently detected in three of 50 healthy subjects. To understand the mechanisms regulating PLC-ß2, we studied the effect of this variation in the PLCB2. Gel-shift studies using nuclear extracts from human erythroleukaemia (HEL) cells or recombinant p65 showed NF-κB binding to oligonucleotide with NF-κB site; in luciferase reporter studies its deletion reduced PLCB2 promoter activity. PLCB2 expression was decreased by siRNA knockdown of NF-κB p65 subunit and increased by p65 overexpression. By immunoblotting platelet PLC-ß2 in 17 healthy subjects correlated with p65 (r=0.76, p=0.0005). These studies provide the first evidence that NF-κB regulates MK/platelet PLC-ß2 expression. This interaction is important because of the major role of PLC-ß2 in platelet activation and of NF-κB in processes, including inflammation and atherosclerosis, where both are intimately involved.


Subject(s)
Blood Platelets/enzymology , NF-kappa B/metabolism , Phospholipase C beta/metabolism , Binding Sites , Consensus Sequence , Female , Gene Expression Regulation, Enzymologic , Humans , Male , Megakaryocytes/enzymology , NF-kappa B/genetics , Phospholipase C beta/genetics , Promoter Regions, Genetic , Sequence Deletion
4.
Thromb Haemost ; 114(3): 558-68, 2015 Aug 31.
Article in English | MEDLINE | ID: mdl-25947062

ABSTRACT

ADP-induced thromboxane generation depends on Src family kinases (SFKs) and is enhanced with pan-protein kinase C (PKC) inhibitors, but it is not clear how these two events are linked. The aim of the current study is to investigate the role of Y311 phosphorylated PKCδ in regulating ADP-induced platelet activation. In the current study, we employed various inhibitors and murine platelets from mice deficient in specific molecules to evaluate the role of PKCδ in ADP-induced platelet responses. We show that, upon stimulation of platelets with 2MeSADP, Y311 on PKCδ is phosphorylated in a P2Y1/Gq and Lyn-dependent manner. By using PKCδ and Lyn knockout murine platelets, we also show that tyrosine phosphorylated PKCδ plays a functional role in mediating 2MeSADP-induced thromboxane generation. 2MeSADP-induced PKCδ Y311 phosphorylation and thromboxane generation were potentiated in human platelets pre-treated with either a pan-PKC inhibitor, GF109203X or a PKC α/ß inhibitor and in PKC α or ß knockout murine platelets compared to controls. Furthermore, we show that PKC α/ß inhibition potentiates the activity of SFK, which further hyper-phosphorylates PKCδ and potentiates thromboxane generation. These results show for the first time that tyrosine phosphorylated PKCδ regulates ADP-induced thromboxane generation independent of its catalytic activity and that classical PKC isoforms α/ß regulate the tyrosine phosphorylation on PKCδ and subsequent thromboxane generation through tyrosine kinase, Lyn, in platelets.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/drug effects , Platelet Activation/drug effects , Protein Kinase C/blood , Purinergic P2Y Receptor Agonists/pharmacology , Thromboxane A2/blood , Animals , Blood Platelets/enzymology , Humans , Isoenzymes , Mice, Knockout , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/metabolism , Signal Transduction/drug effects , src-Family Kinases/blood , src-Family Kinases/genetics
5.
J Biol Chem ; 288(25): 18194-203, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23640884

ABSTRACT

Proline-rich tyrosine kinase 2 (Pyk2) is activated by various agonists in platelets. We evaluated the signaling mechanism and the functional role of Pyk2 in platelets by using pharmacological inhibitors and Pyk2-deficient platelets. We found that platelet aggregation and secretion in response to 2-methylthio-ADP (2-MeSADP) and AYPGKF were diminished in the presence of Pyk2 inhibitors or in Pyk2-deficient platelets, suggesting that Pyk2 plays a positive regulatory role in platelet functional responses. It has been shown that ADP-, but not thrombin-induced thromboxane (TxA2) generation depends on integrin signaling. Unlike ADP, thrombin activates G12/13 pathways, and G12/13 pathways can substitute for integrin signaling for TxA2 generation. We found that Pyk2 was activated downstream of both G12/13 and integrin-mediated pathways, and both 2-MeSADP- and AYPGKF-induced TxA2 generation was significantly diminished in Pyk2-deficient platelets. In addition, TxA2 generation induced by co-stimulation of Gi and Gz pathways, which is dependent on integrin signaling, was inhibited by blocking Pyk2. Furthermore, inhibition of 2-MeSADP-induced TxA2 generation by fibrinogen receptor antagonist was not rescued by co-stimulation of G12/13 pathways in the presence of Pyk2 inhibitor. We conclude that Pyk2 is a common signaling effector downstream of both G12/13 and integrin αIIbß3 signaling, which contributes to thromboxane generation.


Subject(s)
Blood Platelets/metabolism , Focal Adhesion Kinase 2/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thromboxane A2/biosynthesis , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/genetics , Humans , Mice , Mice, Knockout , Oligopeptides/pharmacology , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Signal Transduction/drug effects , Thionucleotides/pharmacology , Time Factors , Tyrphostins/pharmacology
6.
Circ Res ; 112(3): 498-509, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23104882

ABSTRACT

RATIONALE: In the failing heart, persistent ß-adrenergic receptor activation is thought to induce myocyte death by protein kinase A (PKA)-dependent and PKA-independent activation of calcium/calmodulin-dependent kinase II. ß-adrenergic signaling pathways also are capable of activating cardioprotective mechanisms. OBJECTIVE: This study used a novel PKA inhibitor peptide to inhibit PKA activity to test the hypothesis that ß-adrenergic receptor signaling causes cell death through PKA-dependent pathways and cardioprotection through PKA-independent pathways. METHODS AND RESULTS: In PKA inhibitor peptide transgenic mice, chronic isoproterenol failed to induce cardiac hypertrophy, fibrosis, and myocyte apoptosis, and decreased cardiac function. In cultured adult feline ventricular myocytes, PKA inhibition protected myocytes from death induced by ß1-adrenergic receptor agonists by preventing cytosolic and sarcoplasmic reticulum Ca(2+) overload and calcium/calmodulin-dependent kinase II activation. PKA inhibition revealed a cardioprotective role of ß-adrenergic signaling via cAMP/exchange protein directly activated by cAMP/Rap1/Rac/extracellular signal-regulated kinase pathway. Selective PKA inhibition causes protection in the heart after myocardial infarction that was superior to ß-blocker therapy. CONCLUSIONS: These results suggest that selective block of PKA could be a novel heart failure therapy.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Failure/enzymology , Heart Failure/prevention & control , Intracellular Signaling Peptides and Proteins/metabolism , Myocytes, Cardiac/enzymology , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Apoptosis , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cats , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cytosol/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Failure/chemically induced , Heart Failure/pathology , Heart Failure/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , Isoproterenol , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Receptors, Adrenergic, beta/drug effects , Recombinant Fusion Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Signal Transduction , Stroke Volume , Time Factors , Transfection , rac GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism
7.
Arterioscler Thromb Vasc Biol ; 32(5): 1211-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22362759

ABSTRACT

OBJECTIVE: Members of the protein kinase C (PKC) family are shown to positively and negatively regulate platelet activation. Although positive regulatory roles are extensively studied, negative regulatory roles of PKCs are poorly understood. We investigated the mechanism and specific isoforms involved in PKC-mediated negative regulation of ADP-induced functional responses. METHODS AND RESULTS: A pan-PKC inhibitor, GF109203X, potentiated ADP-induced cPLA(2) phosphorylation and thromboxane generation as well as ERK activation and intracellular calcium (Ca(2+)(i)) mobilization, 2 signaling molecules, upstream of cPLA(2) activation. Thus, PKCs inhibit cPLA(2) activation by inhibiting ERK and Ca(2+)(i) mobilization. Because the inhibitor of classic PKC isoforms, GO-6976, did not affect ADP-mediated thromboxane generation, we investigated the role of novel class of PKC isoforms. ADP-induced thromboxane generation, calcium mobilization, and ERK phosphorylation were potentiated in PKCε null murine platelets compared with platelets from wild-type littermates. Interestingly, when thromboxane release is blocked, ADP-induced aggregation in PKCε knockout and wild-type was similar, suggesting that PKCε does not affect ADP-induced aggregation directly. PKCε knockout mice exhibited shorter times to occlusion in an FeCl(3)-induced arterial injury model and shorter bleeding times in tail-bleeding experiments. CONCLUSIONS: We conclude that PKCε negatively regulates ADP-induced thromboxane generation in platelets and offers protection against thrombosis.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/metabolism , Calcium/metabolism , Platelet Aggregation/physiology , Protein Kinase C-epsilon/metabolism , Thromboxanes/metabolism , Animals , Disease Models, Animal , Humans , Intracellular Fluid/metabolism , Mice , Platelet Aggregation/drug effects , Platelet Count , Signal Transduction , Thrombosis/metabolism , Thrombosis/pathology
8.
Zhongguo Zhong Yao Za Zhi ; 36(15): 2153-6, 2011 Aug.
Article in Chinese | MEDLINE | ID: mdl-22066459

ABSTRACT

OBJECTIVE: To investigate the effect of Shenmai injection on vascular endothelial active facters nitric oxide (NO) and endothelin-1 (ET-1), and pulmonary gas exchange induced by tourniquet deflation in patients undergoing lower extremity surgery. METHOD: Twenty-six patients scheduled for unilateral lower extremity surgery were randomly divided into 2 groups: control group (group C, n = 14) and Shenmai injection group (group SM, n = 12). All the patients agreed to a combined spinal-epidural anesthesia at the L2-L3 interspace and a radial artery catheter was placed for sampling. Patients in group SM were injected Shenmai injection 0.6 mL x kg(-1) and physiological saline 100 mL, while patients in group C were injected equal volume of normal saline instead 15 min before tourniquet inflation. Blood samples which were used for blood gas analysis and measurement of nitric oxide (NO) and endothelin-1 (ET-1) were taken before tourniquet inflation (T0, baseline) and 30 min (T1), 2 h (T2), 6 h (T3), 24 h (T4) after tourniquet deflation. RESULT: Compared with the baseline values at T0, in group C at T3 P(a) O2 and the levels of NO were significantly decreased, while P(A-a) DO2 and the levels of ET-1 at T3 were significantly increased (P < 0.05 or P < 0.01), in group SM, the levels of NO at T3 were significantly decreased (P < 0.05). Compared with group C, the changes of P(a)O2, P(A-a) DO2, NO and ET-1 were significantly mitigated in group SM. CONCLUSION: The concentrations of NO and ET-1 is connected with the pulmonary gas exchange impairment induced by tourniquet application. Shenmai injection can improve the pulmonary gas exchange based on rising the level of NO, reducing the level of ET-1.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Endothelin-1/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Pulmonary Gas Exchange , Tourniquets/adverse effects , Adult , Drug Combinations , Drugs, Chinese Herbal/administration & dosage , Endothelin-1/blood , Endothelium, Vascular/physiopathology , Female , Humans , Injections, Intravenous , Male , Middle Aged , Nitric Oxide/blood , Pulmonary Gas Exchange/drug effects
9.
PLoS One ; 6(2): e16586, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347357

ABSTRACT

BACKGROUND: We have previously shown that ADP-induced TXA(2) generation requires signaling from αIIbß3 integrin in platelets. Here we observed that, unlike ADP, protease-activated receptor (PAR)-mediated TXA(2) generation occurs independently of αIIbß3. PAR agonists, but not ADP, activate G(12/13) signaling pathways. Hence, we evaluated the role of these pathways in TXA(2) generation. PRINCIPAL FINDINGS: Inhibition of ADP-induced thromboxane generation by fibrinogen receptor antagonist SC57101 was rescued by co-stimulation of G(12/13) pathways with YFLLRNP. This observation suggested an existence of a common signaling effector downstream of integrins and G(12/13) pathways. Hence, we evaluated role of three potential tyrosine kinases; c-Src, Syk and FAK (Focal Adhesion Kinase) that are known to be activated by integrins. c-Src and Syk kinase did not play a role in ADP-induced functional responses in platelets. Selective activation of G(12/13) pathways resulted in the activation of FAK, in the absence of integrin signaling. Interestingly, αIIbß3-mediated FAK activation occurred in a Src family kinase (SFK)-independent manner whereas G(12/13) pathway caused FAK activation in a SFK and RhoA-dependent manner. A FAK selective inhibitor TAE-226, blocked TXA(2) generation. However, in comparison to WT mice, Pf4-Cre/Fak-Floxed mice did not show any difference in platelet TXA(2) generation. CONCLUSIONS: Therefore, we conclude that differential activation of FAK occurs downstream of Integrins and G(12/13) pathways. However, the common effector molecule, possibly a tyrosine kinase downstream of integrins and G(12/13) pathways contributing to TXA(2) generation in platelets remains elusive.


Subject(s)
Blood Platelets/cytology , Blood Platelets/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction , Thromboxane A2/biosynthesis , Adenosine Diphosphate/metabolism , Animals , Blood Platelets/enzymology , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Syk Kinase , rho-Associated Kinases/metabolism
10.
Platelets ; 21(8): 604-9, 2010.
Article in English | MEDLINE | ID: mdl-21067313

ABSTRACT

Antiplatelet therapy for the management of patients with cardiovascular risks often includes a combination therapy of aspirin and clopidogrel, acting through inhibition of thromboxane generation and blockade of G(i)-coupled P2Y12 receptor, respectively. We hypothesized that ADP acting through P2Y12 regulates physiological thromboxane levels. The serum thromboxane levels in mice (n = 3) dosed with clopidogrel and prasugrel were decreased by 83.1 ± 5.3% and 94.26 ± 1.75% respectively compared to untreated mice. Pre-treatment of human blood (n = 3) ex vivo with active metabolites of clopidogrel or prasugrel led to a reduction in thromboxane levels to 16.3 ± 3.2% and 4.9 ± 0.8% respectively, compared to untreated human serum. We also evaluated serum thromboxane levels in P2Y receptor null mice (n = 4). Whereas serum thromboxane levels in P2Y1 null mice were similar to those in wild type littermates, those in the P2Y12 null mice were inhibited by 83.15 ± 3.8%. Finally, in a pilot study, serum thromboxane levels were reduced by 76.05 ± 8.41% in healthy human volunteers (n = 6) upon dosing with clopidogrel, compared to the levels before dosing. In conclusion, P2Y12 antagonism alone can decrease physiological thromboxane levels. Thus, this study could pave way the for newer/modified treatment regimens for the management of patients with thrombotic complications who are allergic or non-responsive to aspirin.


Subject(s)
Blood Platelets , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/metabolism , Thromboxanes/blood , Adenosine Diphosphate/metabolism , Adolescent , Adult , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Clopidogrel , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Pilot Projects , Piperazines , Platelet Aggregation Inhibitors/pharmacology , Prasugrel Hydrochloride , Receptors, Purinergic P2Y12/genetics , Thiophenes , Ticlopidine/analogs & derivatives , Young Adult
11.
Thromb Haemost ; 104(4): 845-57, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20806121

ABSTRACT

Though antiplatelet drugs are proven beneficial to patients with coronary heart disease and stroke, more effective and safer antiplatelet drugs are still needed. In this study we report the antiplatelet effects and mechanism of BF0801, a novel adenine derivative. BF0801 dramatically inhibited platelet aggregation and ATP release induced by ADP, 2MeSADP, AYPGKF, SFLLRN or convulxin without affecting shape change in vitro . It also potentiated the inhibitory effects of adenosine-based P2Y12 antagonist AR-C69931MX or phosphodiesterase (PDE) inhibitor IBMX on platelet aggregation. The cAMP levels in both resting and forskolin-stimulated platelets were increased by BF0801 suggesting its PDE inhibitor activity, which is further confirmed by the concentration-dependent suppression of BF0801 on the native and recombinant PDE. Similar to AR-C69931MX, BF0801 drastically inhibited 2MeSADP- induced adenylyl cyclase inhibition in platelets indicating its P2Y12 antagonism activity, which is substantiated by the inhibition of BF0801 on the interaction between ADP and P2Y12 receptor expressed in CHO-K1 cells measured by atomic force microscopy. Moreover, we confirmed the antiplatelet effects of BF0801 using platelets from rats intravenously given BF0801. In summary, for the first time we developed a novel adenine derivative bearing dual activities of PDE inhibition and P2Y12 antagonism, which may have therapeutic advantage as a potential antithrombotic drug.


Subject(s)
Adenine/pharmacology , Blood Platelets/drug effects , Coronary Artery Disease/drug therapy , Phosphoric Diester Hydrolases/metabolism , Receptors, Purinergic P2Y12/metabolism , Adenine/administration & dosage , Adenine/analogs & derivatives , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/genetics , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Mice, Knockout , Phosphoric Diester Hydrolases/genetics , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y12/genetics
12.
Eur J Pharmacol ; 644(1-3): 10-6, 2010 Oct 10.
Article in English | MEDLINE | ID: mdl-20599922

ABSTRACT

The P2Y(12) receptor, a Gi protein-coupled receptor, plays a central role in platelet activation. In this study, we did a mutational analysis of residues possibly involved in the ligand interactions with the human P2Y(12) receptor. Mutant receptors were stably expressed in CHO-K1 cells with an HA-tag at the N-terminus. Expression of wild-type and mutant receptors was confirmed by detecting the HA-tag on the cell membrane. Residues in transmembrane helical domains (TMs) 3, 5, 6, and 7, which are homologous to residues important for P2Y(1) receptor activation and ligand recognition, were replaced by site-directed mutagenesis. ADP-induced inhibition of forskolin-stimulated cAMP levels in the presence or absence of antagonist AR-C69931MX were investigated for each of the mutant receptors. F104S and S288P significantly increased agonist-induced receptor function without affecting the antagonism by AR-C69931MX. Arg256 in TM6 and Arg 265 in extracellular loop 3 (EL3) are more important for antagonist recognition than effect on agonist-mediated receptor function. Compared to wild-type P2Y(12) receptor, mutations in Arg 256 or/and Arg 265 significantly increased the sensitivity to antagonist AR-C69931MX. Our study shows that the cytosolic side of TM3 and the exofacial side of TM5 are critical for P2Y(12) receptor function, which is different from P2Y(1). Arg 256 in TM6 and Arg265 in EL3 appear to play a role in antagonist recognition rather than effects on agonist-induced receptor function.


Subject(s)
Adenosine Diphosphate/pharmacology , Cyclic AMP/metabolism , Receptors, Purinergic P2Y12/metabolism , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , DNA Mutational Analysis/methods , Humans , Ligands , Mutagenesis, Site-Directed , Protein Binding , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/genetics
13.
J Biol Chem ; 285(23): 17282-91, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20400514

ABSTRACT

Cbl-b, a member of the Cbl family of E3 ubiquitin ligases, plays an important role in the activation of lymphocytes. However, its function in platelets remains unknown. We show that Cbl-b is expressed in human platelets along with c-Cbl, but in contrast to c-Cbl, it is not tyrosine-phosphorylated upon glycoprotein VI (GPVI) stimulation. Cbl-b, unlike c-Cbl, is not required for Syk ubiquitylation downstream of GPVI activation. Phospholipase Cgamma2 (PLCgamma2) and Bruton's tyrosine kinase (BTK) are constituently associated with Cbl-b. Cbl-b-deficient (Cbl-b(-/-)) platelets display an inhibition in the concentration-response curve for GPVI-specific agonist-induced aggregation, secretion, and Ca(2+) mobilization. A parallel inhibition is found for activation of PLCgamma2 and BTK. However, Syk activation is not affected by the absence of Cbl-b, indicating that Cbl-b acts downstream of Syk but upstream of BTK and PLCgamma2. When Cbl-b(-/-) mice were tested in the ferric chloride thrombosis model, occlusion time was increased and clot stability was reduced compared with wild type controls. These data indicate that Cbl-b plays a positive modulatory role in GPVI-dependent platelet signaling, which translates to an important regulatory role in hemostasis and thrombosis in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Adaptor Proteins, Signal Transducing/genetics , Agammaglobulinaemia Tyrosine Kinase , Animals , Blood Platelets/metabolism , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phospholipase C gamma/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-cbl/genetics
14.
Platelets ; 20(3): 191-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19437337

ABSTRACT

Plasmin, a major extracellular protease, activates platelets through PAR4 receptors. Plasmin-induced full aggregation is achieved at lower concentrations (0.1 U/mL) in murine platelets as compared to human platelets (1 U/mL). In COS7 cells expressing the murine PAR4 (mPAR4) receptor, 1 U/mL plasmin caused a higher intracellular calcium mobilization than in cells expressing the human PAR4 (hPAR4) receptor. This difference was reversed when the tethered ligand sequences of mPAR4 and hPAR4 were interchanged through site-directed mutagenesis. We further investigated whether PAR3 expressed in murine platelets serves as a co-receptor for PAR4 activation by plasmin. In COS7 cells, co-expressing mPAR3 and mPAR4, plamsin produced a smaller intracellular calcium mobilization compared to cells expressing mPAR4 alone, suggesting that PAR3 might inhibit plasmin-induced PAR4 stimulation. Consistent with these results, PAR3 null murine platelets also showed a greater plasmin-induced calcium mobilization and aggregation compared to wild-type murine platelets. In conclusion, murine platelets are more sensitive to activation by plasmin than human platelets due to differences in the primary sequence of PAR4. In contrast to thrombin-dependent activation of platelets, wherein PAR3 acts as a co-receptor, mPAR3 inhibits plasmin-induced PAR4 activation.


Subject(s)
Blood Platelets/metabolism , Fibrinolysin/pharmacology , Platelet Aggregation/physiology , Receptors, Thrombin/blood , Animals , Blood Platelets/drug effects , COS Cells , Calcium/blood , Chlorocebus aethiops , Fibrinolysin/physiology , Humans , Mice , Platelet Aggregation/drug effects , Receptors, Thrombin/physiology
15.
Biochem J ; 417(1): 113-20, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18652571

ABSTRACT

Phosphorylation of activation loop threonine (Thr(505)) and regulatory domain tyrosine (Tyr(311)) residues are key regulators of PKC (protein kinase C) delta function in platelets. In the present study, we show that G(q) and G(12/13) pathways regulate the Thr(505) and Tyr(311) phosphorylation on PKCdelta in an interdependent manner. DiC8 (1,2-dioctanoylglycerol), a synthetic analogue of DAG (diacylglycerol), caused Thr(505), but not Tyr(311), phosphorylation on PKCdelta, whereas selective activation of G(12/13) pathways by the YFLLRNP peptide failed to cause phosphorylation of either residue. However, simultaneous activation by DiC8 and YFLLRNP resulted in Thr(505) and Tyr(311) phosphorylation on PKCdelta. In addition, we found that the activation of SFKs (Src family tyrosine kinases) is essential for G(12/13)-mediated Tyr(311) phosphorylation of PKCdelta. These results were confirmed using G(q)-deficient mouse platelets. Finally, we investigated whether Thr(505) phosphorylation is required for Tyr(311) phosphorylation. A T505A PKCdelta mutant failed to be phosphorylated at Tyr(311), even upon stimulation of both G(q) and G(12/13) pathways. We conclude that (i) PKCdelta binding to DAG, downstream of G(q) pathways, and its translocation results in Thr(505) phosphorylation, (ii) G(12/13) pathways activate SFKs required for the phosphorylation of Tyr(311) on Thr(505)-phosphorylated PKCdelta, and (iii) Thr(505) phosphorylation is a prerequisite for Tyr(311) phosphorylation on PKCdelta.


Subject(s)
Blood Platelets/metabolism , GTP-Binding Proteins/metabolism , Protein Kinase C-delta/metabolism , Threonine/metabolism , Tyrosine/metabolism , Animals , Blood Platelets/drug effects , Blotting, Western , COS Cells , Chlorocebus aethiops , Diglycerides/metabolism , Diglycerides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , Humans , Mice , Mice, Knockout , Phosphorylation/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
16.
Biochem Pharmacol ; 77(5): 835-44, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19073150

ABSTRACT

Platelet secretion is an important physiological event in hemostasis. The protease-activated receptors, PAR 1 and PAR 4, and the thromboxane receptor activate the G(12/13) pathways, in addition to the G(q) pathways. Here, we investigated the contribution of G(12/13) pathways to platelet dense granule release. 2MeSADP, which does not activate G(12/13) pathways, does not cause dense granule release in aspirin-treated platelets. However, supplementing 2MeSADP with YFLLRNP (60muM), as selective activator of G(12/13) pathways, resulted in dense granule release. Similarly, supplementing PLC activation with G(12/13) stimulation also leads to dense granule release. These results demonstrate that supplemental signaling from G(12/13) is required for G(q)-mediated dense granule release and that ADP fails to cause dense granule release because the platelet P2Y receptors, although activate PLC, do not activate G(12/13) pathways. When RhoA, downstream signaling molecule in G(12/13) pathways, is blocked, PAR-mediated dense granule release is inhibited. Furthermore, ADP activated RhoA downstream of G(q) and upstream of PLC. Finally, RhoA regulated PKCdelta T505 phosphorylation, suggesting that RhoA pathways contribute to platelet secretion through PKCdelta activation. We conclude that G(12/13) pathways, through RhoA, regulate dense granule release and fibrinogen receptor activation in platelets.


Subject(s)
Blood Platelets/drug effects , Cytoplasmic Granules/physiology , GTP-Binding Proteins/physiology , Receptor, PAR-1/physiology , Receptors, Thrombin/physiology , rhoA GTP-Binding Protein/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Aspirin/pharmacology , Blood Platelets/metabolism , Blotting, Western , Enzyme Activation , Humans , In Vitro Techniques , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Protein Kinase C-delta/metabolism , Thionucleotides/pharmacology , Type C Phospholipases/metabolism
17.
J Invasive Cardiol ; 20(7): 349-53, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18599893

ABSTRACT

Several contrast agents have been approved in the United States for radiographic imaging purposes. Most of the older ionic, high-osmolar contrast agents are no longer used because of their side effect profile. Therefore, newer nonionic, low or iso-osmolar contrast agents have been widely accepted as an alternative due to their improved tolerability and safety. We investigated the thrombogenicity of the 6 different nonionic radiocontrast media in terms of their platelet reactivity and noted some minor differences among them. In the 50% contrast concentration group, all of the nonionic contrast agents inhibited aggregation, whereas in the 10% contrast concentration group, all agents showed similar aggregation curves in comparison to the normal control. At 50% contrast concentration, the inhibitory effect of aggregation appeared to be related to the inhibition of calcium mobilization, which may be one of the mechanistic effects.


Subject(s)
Cardiac Catheterization/methods , Contrast Media/adverse effects , Coronary Thrombosis/chemically induced , Coronary Thrombosis/epidemiology , Blood Platelets/drug effects , Contrast Media/pharmacology , Coronary Thrombosis/physiopathology , Dose-Response Relationship, Drug , Humans , Iohexol/adverse effects , Iohexol/analogs & derivatives , Iohexol/pharmacology , Iopamidol/adverse effects , Iopamidol/pharmacology , Platelet Aggregation/drug effects , Risk Factors , Triiodobenzoic Acids/adverse effects , Triiodobenzoic Acids/pharmacology
18.
Biochem Pharmacol ; 75(2): 438-47, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17950254

ABSTRACT

A new peptide (TFRRRLSRATR), derived from the c-terminal of human platelet P2Y(1) receptor, was synthesized and its biological function was evaluated. This peptide activated platelets in a concentration-dependent manner, causing shape change, aggregation, secretion and calcium mobilization. Of the several receptor antagonists tested, only BMS200261, a protease activated receptor 1 (PAR-1) specific antagonist, totally abolished the peptide-induced platelet aggregation, secretion and calcium mobilization. The TFRRR-peptide-pretreated washed platelets failed to aggregate in response to SFLLRN (10 microM) but not to AYPGKF (500 microM). In addition, in mouse platelets, peptide concentrations up to 600 microM failed to cause platelet activation, indicating that the TFRRR-peptide activated platelets through the PAR-1 receptor, rather than through the PAR-4 receptor. The shape change induced by 10 microM peptide was totally abolished by Y-27632, an inhibitor of p160(ROCK) which is a downstream mediator of G12/13 pathways. The TFRRR-peptide, YFLLRNP, and the physiological agonist thrombin selectively activated G12/13 pathways at low concentrations and began to activate both Gq and G12/13 pathways with increasing concentrations. Similar to SFLLRN, the TFRRR-peptide caused phosphorylation of Akt and Erk in a P2Y(12) receptor-dependent manner, and p-38 MAP kinase activation in a P2Y(12)-independent manner. The effects of this peptide are elicited by the first six amino acids (TFRRRL) whereas the remaining peptide (LSRATR), TFERRN, or TFEERN had no effects on platelets. We conclude that TFRRRL activates human platelets through PAR-1 receptors.


Subject(s)
Peptide Fragments/pharmacology , Receptor, PAR-1/agonists , Receptors, Purinergic P2/physiology , Adenosine Diphosphate/physiology , Animals , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , Mice , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Receptor, PAR-1/physiology , Receptors, Purinergic P2Y1 , Structure-Activity Relationship , Thrombin/pharmacology
19.
Blood ; 107(3): 947-54, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16223779

ABSTRACT

Protease-activated receptors (PARs) activate Gq and G(12/13) pathways, as well as Akt (protein kinase B [PKB/Akt]) in platelets. However, the relative contribution of different G-protein pathways to Akt phosphorylation has not been elucidated. We investigated the contribution of Gq and G(12/13) to Gi/Gz-mediated Akt phosphorylation downstream of PAR activation. Selective G(12/13) activation failed to cause Akt phosphorylation in human and Galpha q-deficient mouse platelets. However, supplementing Gi/Gz signaling to G(12/13) caused significant increase in Akt phosphorylation, confirming that G(12/13) potentiates Akt phosphorylation. Inhibition of PAR-mediated Akt phosphorylation in the presence of the Gq-selective inhibitor YM-254890 was restored to the normal extent achieved by PAR agonists if supplemented with Gi signaling, indicating that Gq does not have any direct effect on Akt phosphorylation. Selective G(12/13) activation resulted in Src kinase activation, and Akt phosphorylation induced by costimulation of G(12/13) and Gi/Gz was inhibited by a Src kinase inhibitor but not by a Rho kinase inhibitor. These data demonstrate that G(12/13), but not Gq, is essential for thrombin-induced Akt phosphorylation in platelets, whereas Gq indirectly contributes to Akt phosphorylation through Gi stimulation by secreted ADP. G(12/13) activation might mediate its potentiating effect through Src activation, and Src kinases play an important role in thrombin-mediated Akt phosphorylation.


Subject(s)
Blood Platelets/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction/physiology , Adenosine Diphosphate/metabolism , Animals , Blood Platelets/cytology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Humans , Mice , Mice, Knockout , Peptides, Cyclic/pharmacology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Signal Transduction/drug effects , Thrombin/metabolism , src-Family Kinases/metabolism
20.
Blood ; 106(2): 550-7, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15811957

ABSTRACT

Thrombin has been known to cause tyrosine phosphorylation of protein kinase C delta (PKCdelta) in platelets, but the molecular mechanisms and function of this tyrosine phosphorylation is not known. In this study, we investigated the signaling pathways used by protease-activated receptors (PARs) to cause tyrosine phosphorylation of PKCdelta and the role of this event in platelet function. PKCdelta was tyrosine phosphorylated by either PAR1 or PAR4 in a concentration- and time-dependent manner in human platelets. In particular, the tyrosine 311 residue was phosphorylated downstream of PAR receptors. Also the tyrosine phosphorylation of PKCdelta did not occur in Galpha(q)-deficient mouse platelets and was inhibited in the presence of a phospholipase C (PLC) inhibitor U73122 and calcium chelator BAPTA (5,5'-dimethyl-bis(o-aminophenoxy)ethane-N, N, N ', N '-tetraacetic acid), suggesting a role for Galpha(q) pathways and calcium in this event. Both PAR1 and PAR4 caused a time-dependent activation of Src (pp60c-src) tyrosine kinase and Src tyrosine kinase inhibitors completely blocked the tyrosine phosphorylation of PKCdelta. Inhibition of tyrosine phosphorylation or the kinase activity of PKCdelta dramatically blocked PAR-mediated thromboxane A2 generation. We conclude that thrombin causes tyrosine phosphorylation of PKCdelta in a calcium- and Src-family kinase-dependent manner in platelets, with functional implications in thromboxane A2 generation.


Subject(s)
Blood Platelets/metabolism , Egtazic Acid/analogs & derivatives , Protein Kinase C/blood , Thrombin/metabolism , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Humans , In Vitro Techniques , Mice , Models, Biological , Phosphorylation , Protein Kinase C/chemistry , Protein Kinase C-delta , Pyrrolidinones/pharmacology , Receptor, PAR-1/blood , Receptors, Thrombin/blood , Signal Transduction , Threonine/chemistry , Type C Phospholipases/antagonists & inhibitors , Tyrosine/chemistry , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...