Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Clin Neurol Neurosurg ; 160: 54-58, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28668524

ABSTRACT

OBJECTIVE: To assess the effects of dexmedetomidine (Dex) on CD42a+/CD14+,HLADR+/CD14+ and inflammatory cytokine levels in patients undergoing multilevel spinal fusion. Patients and methods Forty ASA I-II patients undergoing multilevel spinal fusion were randomly divided into Dex and control groups (n=20). A continuous intravenous infusion of Dex (0.5µg/kg/h) or normal saline was started 10min prior to induction and was stopped 15min before operation completion. Serum levels of CD42a+/CD14+, HLADR+/CD14+, WBC, PLT, CRP, IL-6, IL-10, and TNF-α were measured before induction (T1), 30min (T2) after operation initiation, and 60min (T3), 1d (T4), 3d (T5), and 5d (T6) post-operation. VAS values were obtained at T3, T4, T5 and T6, as well as hospital days. RESULTS: Treatment with Dex significantly decreased CD42a+/CD14+ at T2, T3, and T4, and markedly increased HLADR+/CD14+ at T4 and T5 when compared with controls. CRP and WBC were markedly decreased at T2, T3, T4 and T5 (P<0.01 or P<0.05). Serum IL-6 and TNF-α level in Dex group was significantly increased at T3 and T4 (P<0.05), and IL-6 and TNF-α level in control group was significantly increased at T2, T3, T4 and T5 (P<0.05) when compared with their respective preoperative levels (T1). IL-6 and TNF-α levels at T2, T3, T4 and T5 in Dex group were significantly lower than those in control group (P<0.05). There were no significant differences in operation time, hospital days or VAS values between the two groups (P>0.05). CONCLUSION: Dex can inhibit the inflammatory response and reduce immunosuppression in patients undergoing multilevel spinal fusion.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , HLA-DR Antigens/drug effects , Interleukin-10 , Interleukin-6 , Lipopolysaccharide Receptors/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects , Spinal Fusion/methods , Tumor Necrosis Factor-alpha/drug effects , Adult , Female , HLA-DR Antigens/blood , Humans , Interleukin-10/blood , Interleukin-6/blood , Lipopolysaccharide Receptors/blood , Male , Middle Aged , Platelet Glycoprotein GPIb-IX Complex/analysis , Tumor Necrosis Factor-alpha/blood
2.
Arq Bras Cardiol ; 90(1): 54-63, 2008 Jan.
Article in English, Portuguese | MEDLINE | ID: mdl-18317641

ABSTRACT

OBJECTIVE: To assess the importance of the interaction between leukocyte integrin Mac-1 (a Mb 2) and platelet glycoprotein (GP) Ib-a for leukocyte recruitment after vascular injury and the effect of the neutralization of the Mac-1-GPIba interaction on cell proliferation and the neointimal hyperplasia triggered by the vascular injury. METHODS: A peptide called M2 or anti-M2 antibody was developed to block the Mac-1-GPIba interaction. This peptide was injected and compared to a control-peptide in C57B1/6J mice submitted to vascular injury of the femoral artery with a guide wire. One, five or 28 days after the vascular injury, the femoral arteries were removed for morphometric and immunohistochemical analyses. RESULTS: The blocking of the Mac-1-GPIba interaction promoted a statistically significant reduction in the number of leukocytes in the neointimal layer on the first day after the vascular injury (control: 7.9+/-5.0% of the cell total versus anti-M2: 2.0+/-1.6%, p=0.021), as well as determined a statistically significant decrease in leukocyte accumulation in the neointimal layer on days 5 and 28 (control: 42.3+/-12.9% versus anti-M2: 24.6+/-10.8%, p=0.047 and control: 7.9+/-3.0% versus anti-M2: 3.3+/-1.3%, p=0.012; respectively). Cell proliferation in the neointimal layer of the vessel five days post-injury was reduced with the blocking of the Mac-1-GPIba interaction (control: 5.0+/-2.9% of the cell total versus anti-M2: 1.8+/-0.5%; p=0.043), along with a significant decrease in cell proliferation in the vessel neointimal layer 28 days post-injury (control: 3.8+/-1.7% versus anti-M2: 2.0+/-1.2%; p=0.047). The blocking of the Mac-1-GPIba interaction also determined a statistically significant decrease of the intimal thickening 28 days post-injury (control: 10,395+/-3,549 microm(2) versus anti-M2: 4,561+/-4,915 microm(2); p=0.012). CONCLUSION: Leukocyte recruitment after a vascular injury depends on the Mac-1-GPIba interaction and the neutralization of this interaction inhibits cell proliferation and neointimal formation.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Femoral Artery/injuries , Leukocytes/physiology , Macrophage-1 Antigen/physiology , Peptides/administration & dosage , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/physiology , Animals , Antibodies, Monoclonal/immunology , Blood Platelets/metabolism , Cell Proliferation , Femoral Artery/metabolism , Immunoglobulin G/administration & dosage , Inflammation/metabolism , Macrophage-1 Antigen/analysis , Male , Mice , Mice, Inbred C57BL , Models, Animal , Peptides/immunology , Platelet Adhesiveness/physiology , Rabbits , Statistics, Nonparametric , Tunica Intima/immunology , Tunica Intima/pathology
3.
Arq. bras. cardiol ; 90(1): 54-63, jan. 2008. ilus, graf, tab
Article in English, Portuguese | LILACS | ID: lil-476046

ABSTRACT

OBJETIVO: Avaliar a importância da interação entre a integrina Mac-1 dos leucócitos (a Mb 2) e a glicoproteína (GP) Iba das plaquetas para o recrutamento de leucócitos após a lesão vascular e o efeito da neutralização da interação Mac-1-GPIba sobre a proliferação celular e a hiperplasia neointimal desencadeadas por lesão vascular. MÉTODOS: Um peptídeo denominado M2 ou anticorpo anti-M2 foi desenvolvido para bloquear a interação Mac-1-GPIba . Esse peptídeo foi injetado e comparado com anticorpo-controle em camundongos C57B1/6J submetidos a lesão vascular da artéria femoral com corda-guia. Um, cinco ou 28 dias após a lesão vascular, as artérias femorais foram retiradas para a realização de morfometria e imuno-histoquímica. RESULTADOS: O bloqueio da interação Mac-1-GPIba promoveu uma redução estatisticamente significativa do número de leucócitos na camada média no primeiro dia após a lesão vascular (controle: 7,9±5,0 por cento do total de células versus anti-M2: 2,0±1,6 por cento, p=0,021), bem como determinou uma diminuição estatisticamente significativa do acúmulo de leucócitos na neoíntima em cinco e 28 dias (controle: 42,3±12,9 por cento versus anti-M2: 24,6±10,8 por cento, p=0,047 e controle: 7,9±3,0 por cento versus anti-M2: 3,3±1,3 por cento, p=0,012; respectivamente). A proliferação celular na camada média do vaso em cinco dias pós-lesão foi reduzida com o bloqueio da interação Mac-1-GPIba (controle: 5,0±2,9 por cento do total de células versus anti-M2: 1,8±0,5 por cento; p=0,043), assim como houve diminuição significativa da proliferação celular na camada íntima do vaso em 28 dias (controle: 3,8±1,7 por cento versus anti-M2: 2,0±1,2 por cento; p=0,047). O bloqueio da interação Mac-1-GPIba também determinou uma redução estatisticamente significativa do espessamento intimal em 28 dias pós-lesão (controle: 10.395±3.549 µm² versus anti-M2: 4.561±4.915 ...


OBJECTIVE: To assess the importance of the interaction between leukocyte integrin Mac-1 (a Mb 2) and platelet glycoprotein (GP) Ib-a for leukocyte recruitment after vascular injury and the effect of the neutralization of the Mac-1-GPIba interaction on cell proliferation and the neointimal hyperplasia triggered by the vascular injury. METHODS: A peptide called M2 or anti-M2 antibody was developed to block the Mac-1-GPIba interaction. This peptide was injected and compared to a control-peptide in C57B1/6J mice submitted to vascular injury of the femoral artery with a guide wire. One, five or 28 days after the vascular injury, the femoral arteries were removed for morphometric and immunohistochemical analyses. RESULTS: The blocking of the Mac-1-GPIba interaction promoted a statistically significant reduction in the number of leukocytes in the neointimal layer on the first day after the vascular injury (control: 7.9±5.0 percent of the cell total versus anti-M2: 2.0±1.6 percent, p=0.021), as well as determined a statistically significant decrease in leukocyte accumulation in the neointimal layer on days 5 and 28 (control: 42.3±12.9 percent versus anti-M2: 24.6±10.8 percent, p=0.047 and control: 7.9±3.0 percent versus anti-M2: 3.3±1.3 percent, p=0.012; respectively). Cell proliferation in the neointimal layer of the vessel five days post-injury was reduced with the blocking of the Mac-1-GPIba interaction (control: 5.0±2.9 percent of the cell total versus anti-M2: 1.8±0.5 percent; p=0.043), along with a significant decrease in cell proliferation in the vessel neointimal layer 28 days post-injury (control: 3.8±1.7 percent versus anti-M2: 2.0±1.2 percent; p=0.047). The blocking of the Mac-1-GPIba interaction also determined a statistically significant decrease of the intimal thickening 28 days post-injury (control: 10,395±3,549 µm² versus anti-M2: 4,561±4,915 µm²; ...


Subject(s)
Animals , Male , Mice , Rabbits , Antibodies, Monoclonal/administration & dosage , Femoral Artery/injuries , Leukocytes/physiology , Macrophage-1 Antigen/physiology , Peptides/administration & dosage , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/physiology , Antibodies, Monoclonal/immunology , Blood Platelets/metabolism , Cell Proliferation , Femoral Artery/metabolism , Immunoglobulin G/administration & dosage , Inflammation/metabolism , Models, Animal , Macrophage-1 Antigen/analysis , Peptides/immunology , Platelet Adhesiveness/physiology , Statistics, Nonparametric , Tunica Intima/immunology , Tunica Intima/pathology
4.
Arch Inst Pasteur Tunis ; 85(1-4): 69-80, 2008.
Article in French | MEDLINE | ID: mdl-19469418

ABSTRACT

Snake venoms are a rich natural source of bioactive molecules, such as peptides, proteins and enzymes, more and more used in biomedical research in diagnostic or therapeutic purposes. The protein components of snake venoms belong to diverse families such as serine proteases, phospholipases, disintegrins, metalloproteinases and C-type lectins. Due to their effects on various receptors such as GPIb, GPVI, alpha2beta1..., the C-type lectins were considered, in first time, as modulators of the platelet aggregation. Recently, some of them have been described for their anti-tumoral potential effect due to their capacity to inhibit adhesion, migration, proliferation and invasion of different cancer cell lines. Also, the C-type lectins have a powerful antiangiogenic effect in vivo and in vitro by interacting with integrins of endothelial cells.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Lectins, C-Type/therapeutic use , Neovascularization, Pathologic/prevention & control , Platelet Aggregation/drug effects , Snake Venoms/chemistry , Animals , Cell Line, Tumor/drug effects , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , Integrin alpha2beta1/drug effects , Integrin alpha5beta1/drug effects , Lectins, C-Type/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects
5.
Mol Pharmacol ; 72(4): 984-92, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17609416

ABSTRACT

Binding of von Willebrand factor (vWF) to platelet glycoprotein (GP) Ib-IX-V mediates platelet activation in the early stage of thrombus formation. Kistomin, a snake venom metalloproteinase (SVMP) purified from venom of Calloselasma rhodostoma, has been shown to inhibit vWF-induced platelet aggregation. However, its action mechanism, structure-function relationship, and in vivo antithrombotic effects are still largely unknown. In the present study, cDNA encoding kistomin precursor was cloned and revealed that kistomin is a P-I class SVMP with only a proteinase domain. Further analysis indicated that kistomin specifically inhibited vWF-induced platelet aggregation through binding and cleavage of platelet GPIbalpha and vWF. Cleavage of platelet GPIbalpha by kistomin resulted in release of 45- and 130-kDa soluble fragments, indicating that kistomin cleaves GPIbalpha at two distinct sites. In parallel, cleavage of vWF by kistomin also resulted in the formation of low-molecular-mass multimers of vWF. In ex vivo and in vivo studies, kistomin cleaved platelet GPIbalpha in whole blood. Moreover, GPIbalpha agonist-induced platelet aggregation ex vivo was inhibited, and tail-bleeding time was prolonged in mice administered kistomin intravenously. Kistomin's in vivo antithrombotic effect was also evidenced by prolonging the occlusion time in mesenteric microvessels of mice. In conclusion, kistomin, a P-I class metalloproteinase, has a relative specificity for GPIbalpha and vWF and its proteolytic activity on GPIbalpha-vWF is responsible for its antithrombotic activity both in vitro and in vivo. Kistomin can be useful as a tool for studying metalloproteinase-substrate interactions and has a potential being developed as an antithrombotic agent.


Subject(s)
Antithrombins/pharmacology , Crotalid Venoms/enzymology , Metalloproteases/pharmacology , Platelet Glycoprotein GPIb-IX Complex/drug effects , von Willebrand Factor/drug effects , Amino Acid Sequence , Animals , Base Sequence , Crotalid Venoms/chemistry , Crotalid Venoms/pharmacology , DNA Primers , DNA, Complementary , Humans , Male , Metalloproteases/chemistry , Mice , Mice, Inbred ICR , Molecular Sequence Data , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/metabolism
6.
Eur J Pharmacol ; 546(1-3): 142-7, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16890935

ABSTRACT

Thrombin activates human platelets through proteolytic activation of two protease-activated receptors (PARs), PAR1 and PAR4. In the present study, we show that, RWJ-56110, a potent synthetic PAR1 antagonist, inhibited platelet aggregation caused by a low concentration (0.05 U/ml) of thrombin, but lost its effectiveness when higher concentrations of thrombin were used as stimulators. YD-3, a non-peptide PAR4 antagonist, alone had little or no effect on thrombin-induced platelet aggregation, significantly enhanced the anti-aggregatory activity of PAR1 antagonist. In addition, we demonstrate for the first time that P-selectin expression in thrombin-stimulated platelets can be synergistically prevented by combined treatment of PAR1 antagonist and PAR4 antagonist. These results indicate that thrombin-induced platelet activation cannot be effectively inhibited by just blocking either single thrombin receptor pathway, and suggest a rationale for potential combination therapy in arterial thrombosis.


Subject(s)
Blood Platelets/drug effects , Platelet Activation/drug effects , Receptor, PAR-1/antagonists & inhibitors , Receptors, Thrombin/antagonists & inhibitors , Thrombin/metabolism , Blood Platelets/metabolism , Crotalid Venoms/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Humans , In Vitro Techniques , Indazoles/pharmacology , Oligopeptides/pharmacology , P-Selectin/metabolism , Peptide Fragments/pharmacology , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Urea/analogs & derivatives , Urea/pharmacology
7.
Transfusion ; 45(6): 911-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15934989

ABSTRACT

BACKGROUND: Several strategies are being developed to reduce the risk of pathogen transmission associated with platelet (PLT) transfusion. STUDY DESIGN AND METHODS: The impact of a new technology for pathogen reduction based on riboflavin plus illumination (Mirasol PRT, Navigant Biotechnologies, Inc.) at 6.2 and 12.3 J per mL on functional and biochemical characteristics of PLTs was evaluated. PLT concentrates (PCs) obtained by apheresis were treated with Mirasol PRT and stored at 22 degrees C. Modifications in major PLT glycoproteins (GPIbalpha, GPIV, and GPIIb-IIIa), adhesive ligands (von Willebrand factor [VWF], fibrinogen [Fg], and fibronectin), activation antigens (P-selectin and LIMP), and apoptotic markers (annexin V binding and factor [F]Va) were analyzed by flow cytometry. Adhesive and cohesive PLT functions were evaluated with well-established perfusion models. Studies were performed on the preparation day (Day 0) and during PCs storage (Days 3 and 5). RESULTS: Levels of glycoproteins remained stable during storage in PCs treated with 6.2 J per mL pathogen reduction technology (PRT) and similar to those observed in nontreated PCs. When 12.3 J per mL PRT was applied, however, levels of GPIbalpha moderately decreased on Days 3 and 5. VWF, Fg, and FVa were not modified in their expression levels, either by treatment or by storage period. Fibronectin appeared more elevated in all PRT samples. A progressive increase in P-selectin and LIMP expression and in annexin V binding was observed during storage of PRT-treated PCs. Functional studies indicated that 6.2 J per mL Mirasol PRT-treated PLTs preserved adhesive and cohesive functions to levels compatible with those observed in the respective control PCs. CONCLUSION: PLT function was well preserved in PCs treated with 6.2 J per mL Mirasol PRT and stored for 5 days.


Subject(s)
Blood Platelets , Blood Platelets/drug effects , Blood Platelets/radiation effects , Blood Preservation , Riboflavin/pharmacology , Ultraviolet Rays , Annexin A5/analysis , Annexin A5/drug effects , Annexin A5/radiation effects , Antigens, CD/analysis , Antigens, CD/drug effects , Antigens, CD/radiation effects , Blood Platelets/chemistry , Blood Platelets/cytology , Blood Platelets/physiology , Fibrinogen/analysis , Fibrinogen/drug effects , Fibrinogen/radiation effects , Fibronectins/analysis , Fibronectins/drug effects , Fibronectins/radiation effects , Flow Cytometry , Humans , Lysosomal Membrane Proteins , P-Selectin/analysis , P-Selectin/drug effects , P-Selectin/radiation effects , Platelet Activation/drug effects , Platelet Activation/radiation effects , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/radiation effects , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/analysis , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/radiation effects , Platelet Glycoprotein GPIb-IX Complex/analysis , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/radiation effects , Platelet Membrane Glycoprotein IIb/analysis , Platelet Membrane Glycoprotein IIb/drug effects , Platelet Membrane Glycoprotein IIb/radiation effects , Platelet Transfusion , Plateletpheresis , Temperature , Time Factors , von Willebrand Factor/analysis , von Willebrand Factor/drug effects , von Willebrand Factor/radiation effects
8.
Hepatogastroenterology ; 51(59): 1417-21, 2004.
Article in English | MEDLINE | ID: mdl-15362767

ABSTRACT

BACKGROUND/AIMS: Interferon alpha used in treatment of chronic hepatitis C significantly influences the blood platelets. The role of platelets in initiating and developing pathological processes in hepatic diseases is still barely known. We studied the effects of interferon alpha 2b (IFN alpha2b) on blood platelets in chronic hepatitis C. METHODOLOGY: The studies were conducted in 16 patients who underwent IFN alpha2b treatment 3 times a week at 6MU. The examination was carried out before and on the 14th day of the treatment of IFN alpha2b. Morphological parameters of blood platelets were determined by hematological methods and flow cytometry. Expression of receptors on blood platelet surfaces (CD41, CD42a, CD62P) and thrombopoietin, platelet-derived growth factor, soluble form sP-selectin, IL-6, and tumor necrosis factor alpha were also determined. RESULTS: The use of IFN alpha2b in patients with chronic hepatitis C significantly effects blood platelets morphology by causing the decrease in their number, the change in population size, and the increase in large platelet count. Interferon decreases P-selectin expression on platelets, sP-selectin and platelet-derived growth factor concentration in plasma. During interferon therapy we noted increase concentration of thrombopoietin, tumor necrosis factor alpha, IL-6 in chronic hepatitis C. CONCLUSIONS: IFN alpha2b stabilizes activated platelets and probably decreases their participation in inflammatory and fibrotic processes in the liver.


Subject(s)
Antiviral Agents/therapeutic use , Blood Platelets/drug effects , Hepatitis C, Chronic/drug therapy , Interferon-alpha/therapeutic use , Platelet Activation/drug effects , Adult , Blood Platelets/pathology , Female , Hepatitis C, Chronic/blood , Humans , Interferon alpha-2 , Liver Function Tests , Male , Middle Aged , P-Selectin/blood , P-Selectin/drug effects , Platelet Count , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoprotein IIb/blood , Platelet Membrane Glycoprotein IIb/drug effects , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/metabolism , Recombinant Proteins , Thrombopoiesis/drug effects
9.
Exp Cell Res ; 300(1): 84-93, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15383317

ABSTRACT

The expression of the small GTPase Rap1 in human megakaryocytes (MKs) differentiated from cord blood (CB)-derived progenitors was investigated. High levels of Rap1 were detected in the majority of mature megakaryocytes independently of days of culture, while a very low percentage of immature megakaryocytes was found to express a small amount of the protein. Rap1 was predominantly detected on internal alpha-granule but not on the plasma membrane. By contrast, CD41 was clearly present on the peripheral plasma membrane, although it also displayed an intracellular localization similar to that of Rap1. Upon thrombin stimulation, both Rap1 and CD41 translocated to the periphery of the cell. At the opposite, RhoA GTPase and glycoprotein Ibalpha were predominantly located at the plasma membrane and did not undergo relocation upon thrombin stimulation. Thrombin induced a dose- and time-dependent activation of Rap1 in mature megakaryocytes. By using a confocal microscopy approach with a specific probe, active Rap1 was detected exclusively at the peripheral plasma membrane. These results demonstrate that expression of Rap1 occurs during maturation rather than differentiation of megakaryocytes from cord blood progenitor cells. Moreover, we demonstrate that thrombin-activated Rap1 is exclusively localized at the peripheral plasma membrane.


Subject(s)
Cell Membrane/metabolism , Fetal Blood/metabolism , Megakaryocytes/metabolism , rap1 GTP-Binding Proteins/metabolism , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Membrane/drug effects , Cells, Cultured , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Dose-Response Relationship, Drug , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/enzymology , Humans , Infant, Newborn , Megakaryocytes/cytology , Megakaryocytes/drug effects , Megakaryocytes/enzymology , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoprotein IIb/drug effects , Platelet Membrane Glycoprotein IIb/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Thrombin/metabolism , Thrombin/pharmacology , rap1 GTP-Binding Proteins/drug effects , rhoA GTP-Binding Protein/drug effects , rhoA GTP-Binding Protein/metabolism
10.
J Trauma ; 57(1 Suppl): S13-21, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15280745

ABSTRACT

BACKGROUND: Investigations were performed to determine whether poly-N-acetyl glucosamine (p-GlcNAc) induces hemostasis by the activation of platelets. METHODS: Platelets were isolated from human blood, fixed in the presence poly-N-acetyl glucosamine fibers, and visualized with scanning electron microscopy. Platelet activation surface markers were measured by fluorescence multiphoton microscopy. Platelet aggregation in the presence of p-GlcNAc fibers and integrin receptor blockers was measured. RESULTS: Scanning electron microscopy indicated that contact of platelets with poly-N-acetyl glucosamine fibers resulted in platelet activation. Fluorescent microscopy showed that contact of platelets with the marine polymer increased intracellular levels of free calcium and resulted in surface exposure of platelet phosphatidylserine, P selectin, and the alphaIIbbeta3 integrin. Antibody inhibitors of the platelet alphaIIbbeta3 integrin inhibited p-GlcNAc to stimulate fibrin polymerization. CONCLUSION: Poly-N-acetyl glucosamine fiber material promotes hemostasis by the activation of platelets.


Subject(s)
Acetylglucosamine/pharmacology , Blood Platelets/drug effects , Chitin/analogs & derivatives , Hemostatics/pharmacology , Platelet Activation/drug effects , Acetylglucosamine/chemistry , Blood Platelets/ultrastructure , Calcium/physiology , Chitin/chemistry , Chitin/pharmacology , Chitosan , Drug Evaluation, Preclinical , Fibrin/drug effects , Hemostasis, Surgical/methods , Hemostatics/chemistry , Humans , Integrin alpha2 , Integrin alpha5/drug effects , Integrin alpha6/drug effects , Integrin beta3/drug effects , Intracellular Fluid/drug effects , Membrane Glycoproteins/drug effects , Microscopy, Electron, Scanning , Microscopy, Fluorescence, Multiphoton , P-Selectin/drug effects , Phosphatidylserines/physiology , Platelet Activation/physiology , Platelet Adhesiveness/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Membrane Glycoprotein IIb/drug effects , Spectrophotometry , Time Factors
11.
J Trauma ; 57(1 Suppl): S22-5; discussion S25, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15280746

ABSTRACT

BACKGROUND: This study was performed to assess the effect of poly-N-acetyl glucosamine fiber slurry on plasma clotting proteins, platelets, and red blood cells in the clotting of the blood. METHODS: Citrate phosphate dextrose whole blood was stored at 22degreesC for 48 hours to prepare platelet-poor plasma, platelet-rich plasma (PRP), and PRP plus red blood cells with hematocrit values of 20%, 35%, and 45% with and without an equal volume of poly-N-acetyl glucosamine fibers (1 mg/mL 0.9% NaCl). RESULTS: Thromboelastogram data show that poly-N-acetyl glucosamine fibers (p-GlcNAc) significantly reduced the R time in platelet-poor plasma, PRP, and PRP supplemented with red blood cells. Poly-N-acetyl glucosamine fibers increased, but not significantly, Annexin V and factor X binding to platelets, platelet microparticles, and red blood cell Annexin V binding. Poly-N-acetyl glucosamine fibers increased the production of thromboxane B2 by PRP. CONCLUSION: Poly-N-acetyl glucosamine slurry activates platelets.


Subject(s)
Acetylglucosamine/pharmacology , Blood Platelets/drug effects , Erythrocytes/drug effects , Hemostatics/pharmacology , Platelet Activation/drug effects , Acetylglucosamine/chemistry , Annexin A5/analysis , Annexin A5/blood , Annexin A5/drug effects , Blood Coagulation Factors/drug effects , Blood Platelets/chemistry , Drug Evaluation, Preclinical , Erythrocytes/chemistry , Factor X/analysis , Factor X/drug effects , Factor X/metabolism , Fibrin Fibrinogen Degradation Products/drug effects , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/analysis , Fibrinogen/drug effects , Fibrinogen/metabolism , Hematocrit , Hemostatics/chemistry , Humans , P-Selectin/analysis , P-Selectin/blood , P-Selectin/drug effects , Peptide Fragments/blood , Peptide Fragments/drug effects , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/analysis , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/analysis , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Prothrombin/drug effects , Thrombelastography , Thromboxane B2/blood , Time Factors
12.
J Thromb Haemost ; 2(7): 1118-26, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15219195

ABSTRACT

Circulating procoagulant microparticles (MP) were measured as markers of vascular damage and prothrombotic risk in patients undergoing ST-segment myocardial infarction (STEMI) treated by primary percutaneous transluminal coronary angioplasty (PTCA) and additional GPIIb-IIIa antagonists. Cells possibly more responsive to GPIIb-IIIa (alpha(IIb)beta(3)) antagonists were evidenced through MP phenotypes by comparison with healthy volunteers (HV) and STEMI patients treated by PTCA without GPIIb-IIIa antagonist (CP). In 50 STEMI patients, blood samples were collected at day 1 and day 6. Circulating procoagulant MP were captured on annexin V and quantified by prothrombinase assay as nanomolar phosphatidylserine equivalents (nm PhtdSer). Platelet activation by thrombin was confirmed through independent measurement of soluble GPV (sGPV). With respect to HV, procoagulant MP levels were high in patients with STEMI or unstable angina, platelet-derived MP and elevated sGPV testifying to significant platelet activation. A substantial release of endothelial-derived MP was evidenced simultaneously. In abciximab-treated patients, procoagulant MP, mainly of platelet origin, decreased precociously at day 1 (4.2 +/- 0.6 vs. CP 15.5 +/- 2.1 nm PhtdSer; P = 0.001) together with sGPV (36 +/- 3 vs. CP 58 +/- 8 ng mL(-1); P = 0.02). Leukocyte-derived MP decreased at day 6 (0.12 +/- 0.04 vs. CP 0.56 +/- 0.12 nm PhtdSer; P = 0.01) suggesting a possible effect on underlying inflammatory status. In patients presenting cardiovascular events at 6-month follow-up, procoagulant MP levels at day 1 could be indicative of a worsened outcome. MP could constitute a relevant parameter for the follow-up of STEMI patients treated by GPIIb-IIIa antagonists.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Myocardial Infarction/blood , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/analysis , Thrombophilia/drug therapy , Abciximab , Adrenergic beta-Antagonists/therapeutic use , Adult , Aged , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Aspirin/therapeutic use , Biomarkers/blood , Female , Follow-Up Studies , Humans , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin Fab Fragments/therapeutic use , Male , Middle Aged , Myocardial Infarction/therapy , Particle Size , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/drug effects , Predictive Value of Tests , Prognosis , Solubility , Thrombophilia/diagnosis , Thrombophilia/etiology
13.
J Thorac Cardiovasc Surg ; 126(2): 428-35, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12928640

ABSTRACT

OBJECTIVE: Contact between blood and the synthetic surfaces of a cardiopulmonary bypass circuit leads to platelet activation, and resultant platelet dysfunction contributes to postoperative bleeding. We compared the effects of various platelet inhibitors on preservation of platelet function during simulated cardiopulmonary bypass circulation. METHODS: Fresh human blood was recirculated in an in vitro cardiopulmonary bypass model circuit. We measured various platelet activation markers including expressions of PAC-1 and P-selectin, annexin V binding, and microparticle formations by means of whole-blood flow cytometry. RESULTS: Two types of glycoprotein IIb/IIIa complex antagonists, peptide-mimetic FK633 and abciximab and prostaglandin E(1), significantly prevented platelet loss and the increase in binding of PAC-1, an antibody specific for fibrinogen receptor on activated platelets, during extracorporeal circulation of heparinized blood. These antagonists significantly suppressed but did not abolish P-selectin expression, annexin V binding, and microparticle formation. Anti-von Willebrand factor monoclonal antibody and aurin tricarboxylic acid (an inhibitor of glycoprotein Ib) had no effect on platelet activation during simulated cardiopulmonary bypass circulation. These data suggest that inhibition of fibrinogen binding glycoprotein IIb/IIIa complex is partly effective in attenuating platelet activation in a heparinized cardiopulmonary bypass model circuit. The direct thrombin inhibitor argatroban prevented platelet loss and expression of P-selectin significantly more than did heparin. A combination of FK633 with argatroban as a substitute for heparin further prevented platelet loss and platelet secretion during simulated cardiopulmonary bypass circulation, although the inhibition of microparticle formation was less. CONCLUSION: The inhibition of both platelet adhesion and thrombin may be effective to preserve platelet number and function during cardiopulmonary bypass circulation.


Subject(s)
Anesthesia , Blood Platelets/drug effects , Blood Platelets/physiology , Extracorporeal Circulation , Pipecolic Acids/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Abciximab , Alprostadil/blood , Alprostadil/pharmacology , Annexin A5/drug effects , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacology , Anticoagulants/pharmacology , Arginine/analogs & derivatives , Dipeptides/blood , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Dual Specificity Phosphatase 2 , Heparin/pharmacology , Humans , Immunoglobulin Fab Fragments/blood , Immunoglobulin Fab Fragments/pharmacology , P-Selectin/drug effects , Pipecolic Acids/blood , Platelet Activation/drug effects , Platelet Activation/physiology , Platelet Aggregation Inhibitors/blood , Platelet Count , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Membrane Glycoprotein IIb/drug effects , Protein Phosphatase 2 , Protein Tyrosine Phosphatases/drug effects , Sulfonamides , Treatment Outcome , von Willebrand Factor/drug effects
14.
Article in English | MEDLINE | ID: mdl-12769639

ABSTRACT

Platelet adhesion to a damaged blood vessel is the initial trigger for arterial hemostasis and thrombosis. Platelets adhere to the subendothelium through an interaction with von Willebrand factor (VWF), which forms a bridge between collagen within the damaged vessel wall and the platelet receptor glycoprotein Ib/V/IX (GPIb), an interaction especially important under high shear conditions[1]. This reversible adhesion allows platelets to roll over the damaged area, which is then followed by a firm adhesion mediated by the collagen receptors (alpha(2)beta(1), GPVI, ) in addition[2] resulting in platelet activation. This leads to the conformational activation of the platelet alpha(IIb)beta3 receptor, fibrinogen binding and finally to platelet aggregation. Over the past decades, modulation of platelet function has been a strategy for the control of cardiovascular disease. Lately, drugs have been developed that target the fibrinogen receptor alphaIIbbeta3 or the ADP receptor and many of these promising compounds have been tested in clinical trials. However the development of products that interfere with the first step of hemostasis, i.e. the platelet adhesion, has lagged behind. In this review we want to discuss (i) the in vivo studies that were performed with compounds that target proteins involved in different adhesion steps i.e. the VWF-GPIb-axis, the collagen-VWF axis and the collagen-collagen receptor axis and (ii) the possible advantages these putative new drugs could have over the current antiplatelet agents.


Subject(s)
Collagen/physiology , Fibrinolytic Agents/pharmacology , Platelet Adhesiveness/drug effects , Animals , Humans , Integrin alpha2beta1/drug effects , Integrin alpha2beta1/metabolism , Integrin alpha2beta1/physiology , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/physiology , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/physiology , von Willebrand Factor/drug effects , von Willebrand Factor/metabolism , von Willebrand Factor/physiology
15.
Blood Coagul Fibrinolysis ; 14(2): 159-67, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12632026

ABSTRACT

Thrombin-induced platelet activation involves cleavage of protease-activated receptors (PARs) 1 and 4, and interaction, via glycoprotein (Gp)Ibalpha, with the platelet GpIb/IX/V complex. This study investigated inhibition of platelet activation by thrombin inhibitors with different modes of action: two reversible direct thrombin inhibitors, melagatran and inogatran; hirudin, a tightly binding direct thrombin inhibitor; and two indirect thrombin inhibitors, heparin and dalteparin. Up-regulation of P-selectin (CD62P) and PAR-1 cleavage was measured in human whole blood, by flow cytometry. The thrombin concentration that induced 50% of maximum (EC50 ) PAR-1 cleavage was 0.028 nmol/l, while that of platelet activation (CD62P) was over two-fold higher (0.64 nmol/l). The EC50 of a PAR-1-independent component, defined as a further activating effect of thrombin on top of the maximum PAR-1-activating peptide (AP) effect, was 3.2 nmol/l. All anticoagulants were concentration-dependent inhibitors of thrombin-induced platelet activation and PAR-1 cleavage, but none inhibited PAR-1-AP or PAR-4-AP induced activation. Melagatran and inogatran were more potent inhibitors of CD62P up-regulation than of PAR-1 cleavage; conversely, hirudin, heparin and dalteparin were more potent inhibitors of PAR-1 cleavage.Thus, reversible direct thrombin inhibitors, such as melagatran, are potent inhibitors of thrombin-induced platelet activation, acting mainly by inhibition of a PAR-1-independent component.


Subject(s)
Anticoagulants/pharmacology , Glycine/analogs & derivatives , Platelet Activation/drug effects , Thrombin/pharmacology , Adult , Azetidines , Benzylamines , Dalteparin/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Glycine/pharmacology , Heparin/pharmacology , Hirudins/pharmacology , Humans , Male , P-Selectin/blood , P-Selectin/drug effects , P-Selectin/metabolism , Piperidines/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptor, PAR-1 , Receptors, Thrombin/blood , Receptors, Thrombin/drug effects , Receptors, Thrombin/metabolism , Statistics as Topic , Thrombin/antagonists & inhibitors , Thrombin/metabolism
16.
Thromb Haemost ; 87(4): 692-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12008953

ABSTRACT

Alboluxin, a potent platelet activator, was purified from Trimeresurus albolabris venom with a mass of 120 kDa non-reduced and, after reduction, subunits of 17 and 24 kDa. Alboluxin induced a tyrosine phosphorylation profile in platelets that resembles those produced by collagen and convulxin, involving the time dependent tyrosine phosphorylation of Fc receptor gamma chain (Fc gamma), phospholipase Cgamma2 (PLCgamma2), LAT and p72SYK. Antibodies against both GPIb and GPVI inhibited platelet aggregation induced by alboluxin, whereas antibodies against alpha2beta1 had no effect. Inhibition of alphaIIb beta3 reduced the aggregation response to alboluxin, as well as tyrosine phosphorylation of platelet proteins, showing that activation of alphaIIb beta3 and binding of fibrinogen are involved in alboluxin-induced platelet aggregation and it is not simply agglutination. N-terminal sequence data from the beta-subunit of alboluxin indicates that it belongs to the snake C-type lectin family. The C-type lectin subunits are larger than usual possibly due to post-translational modifications such as glycosylation. Alboluxin is a hexameric (alphabeta)3 snake C-type lectin which activates platelets via both GPIb and GPVI.


Subject(s)
Crotalid Venoms/chemistry , Crotalid Venoms/isolation & purification , Lectins/isolation & purification , Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Membrane Glycoproteins/drug effects , Alprostadil/pharmacology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Biotinylation , Carrier Proteins , Chromatography, Ion Exchange , Crotalid Venoms/antagonists & inhibitors , Crotalid Venoms/pharmacology , Edetic Acid/pharmacology , Egtazic Acid/pharmacology , Glycosylation , Humans , Lectins/pharmacology , Molecular Sequence Data , Molecular Weight , Phosphorylation , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/immunology , Platelet Glycoprotein GPIb-IX Complex/physiology , Platelet Membrane Glycoproteins/immunology , Platelet Membrane Glycoproteins/physiology , Protein Processing, Post-Translational/drug effects , Protein Subunits , Proteins/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid , Stimulation, Chemical , Viper Venoms
17.
Blood ; 99(11): 3971-7, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12010796

ABSTRACT

Ultralarge von Willebrand factor (ULVWF) multimers have been implicated in the pathogenesis of the catastrophic microangiopathic disorder, thrombotic thrombocytopenic purpura. Spontaneous ULVWF binding to platelets has been ascribed to increased avidity due to the greatly increased number of binding sites for platelets (the A1 domain) per molecule. To address the mechanism of enhanced ULVWF binding to platelets, we used optical tweezers to study the unbinding forces from the glycoprotein Ib-IX (GP Ib-IX) complex of plasma VWF, ULVWF, and isolated A1 domain. The unbinding force was defined as the minimum force required to pull ligand-coated beads away from their attachment with GP Ib-IX-expressing cells. Beads coated with plasma VWF did not bind to the cells spontaneously, requiring the modulators ristocetin or botrocetin. The force required to break the ristocetin- and botrocetin-induced plasma VWF-GP Ib-IX bonds occurred in integer multiples of 6.5 pN and 8.8 pN, respectively, depending on the number of bonds formed. In contrast, beads coated with either ULVWF or A1 domain bound the cells in the absence of modulators, with bond strengths in integer multiples of approximately 11.4 pN for both. Thus, in the absence of shear stress, ULVWF multimers form spontaneous high-strength bonds with GP Ib-IX, while plasma VWF requires exogenous modulators. The strength of individual bonds formed with GP Ib-IX was similar for both ULVWF and the isolated A1 domain and greater than those of plasma VWF induced by either modulator. Therefore, we suggest that the conformational state of ULVWF multimers is more critical than their size for interaction with platelets.


Subject(s)
Platelet Glycoprotein GPIb-IX Complex/chemistry , von Willebrand Factor/chemistry , Animals , Antibodies, Monoclonal , Blood Platelets/physiology , Cricetinae , Humans , Kinetics , Macromolecular Substances , Platelet Glycoprotein GPIb-IX Complex/drug effects , Protein Binding , Recombinant Proteins/chemistry , Ristocetin/pharmacology , Transfection , von Willebrand Factor/drug effects
18.
Br J Pharmacol ; 134(5): 1013-20, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11682449

ABSTRACT

1. The pharmacological characteristics of solid-phase von Willebrand factor (svWF), a novel platelet agonist, were studied. 2. Washed platelet suspensions were obtained from human blood and the effects of svWF on platelets were measured using aggregometry, phase-contrast microscopy, flow cytometry and zymography. 3. Incubation of platelets with svWF (0.2 - 1.2 microg ml(-1)) resulted in their adhesion to the ligand, while co-incubations of svWF with subthreshold concentrations of ADP, collagen and thrombin resulted in aggregation. 4. 6B4 inhibitory anti-glycoprotein (GP)Ib antibodies abolished platelet adhesion stimulated by svWF, while aggregation was reduced in the presence of 6B4 and N-Acetyl-Pen-Arg-Gly-Asp-Cys, an antagonist of GPIIb/IIIa. 5. Platelet adhesion stimulated with svWF was associated with a concentration-dependent increase in expression of GPIb, but not of GPIIb/IIIa. 6. In contrast, collagen (0.5 - 10.0 microg ml(-1)) caused down-regulation of GPIb and up-regulation of GPIIb/IIIa in platelets. 7. Solid-phase vWF (1.2 microg ml(-1)) resulted in the release of MMP-2 from platelets. 8. Inhibition of MMP-2 with phenanthroline (10 microM), but not with aspirin or apyrase, inhibited platelet adhesion stimulated with svWF. 9. In contrast, human recombinant MMP-2 potentiated both the effects of svWF on adhesion and up-regulation of GPIb. 10. Platelet adhesion and aggregation stimulated with svWF were reduced by S-nitroso-n-acetyl-penicillamine, an NO donor, and prostacyclin. 11. Thus, stimulation of human platelets with svWF leads to adhesion and aggregation that are mediated via activation of GPIb and GPIIb/IIIa, respectively. 12. Mechanisms of activation of GPIb by svWF involve the release of MMP-2, and are regulated by NO and prostacyclin.


Subject(s)
Blood Platelets/drug effects , Penicillamine/analogs & derivatives , von Willebrand Factor/pharmacology , Biological Transport/drug effects , Blood Platelets/metabolism , Collagen/pharmacology , Dose-Response Relationship, Drug , Epoprostenol/pharmacology , Humans , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 2/metabolism , Nitric Oxide Donors/pharmacology , Penicillamine/pharmacology , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism
19.
Br J Pharmacol ; 134(5): 1104-12, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11682459

ABSTRACT

1. We have studied the effects of a novel agonist, solid-phase von Willebrand Factor (sVWF), on tumour cell-induced platelet aggregation (TCIPA). 2. Washed platelet suspensions were obtained from human blood and the effects of HT-1080 human fibrosarcoma cells and sVWF on platelets were studied using aggregometry, phase-contrast microscopy, and flow cytometry. 3. Incubation of platelets with sVWF (1.2 microg ml(-1)) and HT-1080 cells (5 x 10(3) ml(-1)) resulted in a two-phased reaction characterized first by the adhesion of platelets to sVWF, then by aggregation. 4. TCIPA in the presence of sVWF was inhibited by S-nitroso-glutathione (GSNO, 100 microM) and prostacyclin (PGI(2), 30 nM). 5. Platelet activation in the presence of tumour cells and sVWF resulted in the decreased surface expression of platelet glycoprotein (GP)Ib and up-regulation of GPIIb/IIIa receptors. 6. Pre-incubation of platelets with PGI(2) (30 nM) resulted in inhibition of sVWF-tumour cell-stimulated platelet surface expression of GPIIb/IIIa as measured by flow cytometry using antibodies directed against both non-activated and activated receptor. In contrast, GSNO (100 microM) did not affect sVWF-tumour cell-stimulated platelet surface expression of GPIIb/IIIa. 7. Flow cytometry performed with PAC-1 antibodies that bind only to the activated GPIIb/IIIa revealed that GSNO (100 microM) caused inhibition of activation of GPIIb/IIIa. 8. The inhibitors exerted no significant effects on TCIPA-mediated changes in GPIb. 9. Thus, sVWF potentiates the platelet-aggregatory activity of HT-1080 cells and these effects appear to be mediated via up-regulation of platelet GPIIb/IIIa. 10. Prostacyclin and NO inhibit TCIPA-sVWF-mediated platelet aggregation. The mechanisms of inhibition of this aggregation by PGI(2) differ from those of NO.


Subject(s)
Platelet Aggregation/drug effects , von Willebrand Factor/pharmacology , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Coculture Techniques , Epoprostenol/pharmacology , Humans , Nitric Oxide/physiology , Nitric Oxide Donors/pharmacology , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/biosynthesis , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/biosynthesis , Platelet Glycoprotein GPIb-IX Complex/drug effects , Protein Binding , S-Nitrosoglutathione/pharmacology , Time Factors , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , von Willebrand Factor/metabolism
20.
Environ Health Perspect ; 109(3): 225-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11333182

ABSTRACT

We studied the effect of nickel ions on platelet function because hypernickelemia has been found in patients with acute myocardial infarction. We previously demonstrated that nickel can activate an intracellular pathway leading to cytoskeleton reorganization consequent to tyrosine phosphorylation of p60(src) in human platelets independently of integrin alpha-IIb-beta(3). Moreover, in von Willebrand factor-stimulated platelets, the tyrosine phosphorylation of pp60(c-src) is closely associated with the activation of phosphatidylinositol 3-kinase (PIK), and two adhesion receptors, glycoprotein (Gp)Ib and GpIIb/IIIa(alpha-IIb-beta(3)), are involved. In our study, 1 and 5 mM nickel in the presence of fibrinogen induced platelet aggregation (independently of protein kinase C activation) and secretion. The pretreatment with a PIK inhibitor, wortmannin, strongly decreased nickel-induced platelet aggregation. Platelet treatment with mocarhagin, a cobra venom metalloproteinase that cleaves GpIba, significantly reduced aggregation induced by 5 mM without affecting the response to other agonists such as adenosine diphosphate (ADP). Moreover, nickel caused PIK translocation to the cytoskeleton. Taken together, these observations suggest a partial involvement of both integrins alpha-IIb-beta(3) and GpIb-V-IX complex in Ni(2+)-induced platelet activation.


Subject(s)
Environmental Pollutants/pharmacology , Nickel/pharmacology , Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Membrane Glycoproteins , Cytoskeleton/drug effects , Cytoskeleton/physiology , Humans , Immunoblotting , Myocardial Infarction/etiology , Occupational Exposure/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...