Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
2.
Curr Opin Hematol ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38529832

ABSTRACT

PURPOSE OF REVIEW: Loss of surface sialic acid by neuraminidases is known as 'desialylation'. Platelets are desialylated in bacterial or viral infections, during storage, senescence, various mutations, platelet auto antibodies, hemostasis and shear stress. In this review the recent literature on the different sialic acid capped glycan structures will be covered as well as platelet desialylation in inherited glycan disorders and induced by external neuraminidases. RECENT FINDINGS: Neuraminidases are released from platelet intracellular stores and translocated to the platelet surface. Apart from clearance, loss of surface sialic acid by neuraminidases ('desialylation') affects platelet signaling including ligand binding and their procoagulant function. Platelets are also desialylated in infections, various mutations, presence of platelet auto antibodies. SUMMARY: Since platelet desialylation occurs in various healthy and pathological conditions, measuring desialylation might be a new diagnostic tool.

4.
Transfusion ; 62(6): 1289-1301, 2022 06.
Article in English | MEDLINE | ID: mdl-35467785

ABSTRACT

BACKGROUND: Platelets shed platelet microparticles (PMP) when activated or stored. As the removal of sialic acid (desialylation) promotes platelet uptake and clearance from the circulation, similar mechanisms for PMP uptake were hypothesized. The aim of the study was to investigate the role of surface glycans in the in vitro uptake of PMP from stored platelet components. STUDY DESIGN AND METHODS: Apheresis platelet components were stored in 40% plasma/60% SSP+ and sampled on day 1, 5, and 7 post-collection. PMP were characterized by staining with annexin-V (AnV) for phosphatidylserine (PS)-exposure, CD41 antibody, and fluorescently labeled glycan-binding lectins using flow cytometry. The procoagulant function of PMP following desialylation by neuraminidase treatment was assessed by AnV binding and a procoagulant phospholipid assay. PMP were isolated and stained with Deep Red, and phagocytosis by HepG2 cells was measured. Isolated PMP were deglycosylated with neuraminidase and galactosidase to assess the involvement of glycans in mediating phagocytosis. RESULTS: While the overall platelet surface glycan profile was unchanged during storage, PS+ platelets were sialylated, indicating different glycoproteins were changed. In contrast, sialic acid was removed from PS+ and CD41+ PMP, which specifically lost α-2,3-linked sialic acid during platelet storage. PMP were phagocytized by HepG2 cells, and PMP from platelets stored for 7 days were phagocytized to a lesser extent than on day 1. Desialylation by neuraminidase induced PS-exposure on PMP, decreased PPL clotting time, and increased PMP phagocytosis. CONCLUSION: PMP glycans change during platelet storage. Desialylation influences the procoagulant function of PMP and phagocytosis by HepG2 cells.


Subject(s)
Blood Component Removal , Blood Platelets , Annexin A5/metabolism , Blood Platelets/metabolism , Flow Cytometry , Humans , N-Acetylneuraminic Acid/analysis , N-Acetylneuraminic Acid/metabolism , Neuraminidase/analysis , Neuraminidase/metabolism , Neuraminidase/pharmacology , Phagocytosis , Phosphatidylserines/metabolism , Polysaccharides/analysis , Polysaccharides/metabolism
5.
Transfusion ; 62(2): 273-278, 2022 02.
Article in English | MEDLINE | ID: mdl-34761380

ABSTRACT

BACKGROUND: Platelet collection and processing methods, as well as donor attributes, can influence platelet function and quality during ex vivo storage. In this study, activation and procoagulant responses in platelets collected from donors experiencing a citrate reaction (CR) were investigated. STUDY DESIGN AND METHODS: Apheresis platelet components (n = 54) were stored in 100% autologous plasma and tested on days 1 and 5 post-collection. Platelet components were categorized into two groups according to whether the donor had experienced a CR during donation (n = 10; non-CR group, n = 44). Platelet aggregation was initiated with collagen and thrombin. Platelet phenotype was characterized by flow cytometry. Fibrinogen binding was assessed following collagen + thrombin stimulation (COATed platelets), and procoagulant activity was assessed using a procoagulant phospholipid assay (PPL). Platelet microparticle (PMP) subsets were enumerated by flow cytometry. RESULTS: Basal von Willebrand factor (VWF) binding was higher in the CR donations when compared with the non-CR group. Collagen aggregation was significantly higher in platelets from CR donations, in contrast to aggregation induced by thrombin. The proportion of phosphatidylserine (PS) positive PMP and PPL clotting time were higher in the CR group, in contrast to the number of basal PS+ platelets and COATed platelets following stimulation. CONCLUSION: Platelets donated by donors who experienced a CR during donation had higher platelet activation response and possibly a more procoagulant PMP phenotype, suggesting that this donor reaction might lead to increased platelet activation.


Subject(s)
Blood Component Removal , Blood Platelets , Blood Platelets/metabolism , Citrates , Citric Acid/metabolism , Citric Acid/pharmacology , Collagen/metabolism , Collagen/pharmacology , Humans , Phenotype , Phosphatidylserines/metabolism , Platelet Activation , Thrombin/analysis
6.
Transfusion ; 61(11): 3081-3086, 2021 11.
Article in English | MEDLINE | ID: mdl-34554562

ABSTRACT

BACKGROUND: Irradiation of selected blood components is standard practice for the prevention of transfusion-associated graft-versus-host disease (TA-GvHD). Currently, gamma-irradiation is the most widely used form of irradiation, but there is an increasing interest in X-irradiation, which is considered to be functionally equivalent and safer. However, there is a paucity of contemporary data regarding the ability of X-irradiation to inactivate lymphocytes in blood components. Therefore, the effect of gamma- and X-irradiation on lymphocyte viability and function in blood components was compared. STUDY DESIGN AND METHODS: Lymphocytes were isolated from venous blood by density gradient centrifugation, spiked into plasma/SSP+ to simulate a blood component, and either gamma- or X-irradiated. The phenotype of the isolated lymphocytes was confirmed. Lymphocyte viability was measured using a LIVE/DEAD assay, and function was assessed using mixed lymphocyte culture and CD69 expression post-phorbol-12 myristate 13-acetate (PMA) stimulation. RESULTS: Lymphocyte viability and CD69 expression following PMA stimulation were significantly reduced by both gamma-irradiation and X-irradiation in simulated blood components. Allorecognition and allostimulation were also significantly reduced by both gamma-irradiation and X-irradiation. CONCLUSION: Lymphocyte viability and function are reduced to a similar extent by gamma- and X-irradiation in simulated blood components. As such, X-irradiation is suitable for the irradiation of blood components and, in terms of lymphocyte inactivation, could be used instead of gamma-irradiation.


Subject(s)
Graft vs Host Disease , Transfusion Reaction , Blood Component Transfusion , Gamma Rays , Graft vs Host Disease/prevention & control , Humans , Lymphocytes/radiation effects
7.
Res Pract Thromb Haemost ; 5(2): 253-260, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33733023

ABSTRACT

As a result of the coronavirus disease 2019 pandemic, the International Society on Thrombosis and Haemostasis (ISTH), like many societies around the world, canceled their in-person hematology congress planned for Milan, Italy, in July 2020. As a result, the first virtual ISTH congress in the organisation's 51-year history was delivered, inviting free registration from across the globe. As part of the social media support, marketing, and scientific dissemination efforts for the virtual congress, the ISTH assembled a group of official Twitter Ambassadors, which represented the broad and diverse ISTH community. Ambassadors were tasked to tweet daily throughout the congress and to share their commentary on the hematology research being presented with the "#ISTH2020" hashtag. Ambassadors were also supported by Twitter activities from the two official ISTH-affiliated journals: the Journal of Thrombosis and Haemostasis (JTH) and Research and Practice in Thrombosis and Haemostasis (RPTH). In this forum and through the Twitter ambassadors' lens, we present the Twitter Ambassadors' experience, reflect on the impact of social media on the ISTH 2020 congress, and share this experience with the wider scientific community. Specifically, we report on the role of Twitter communication for virtual meetings, discuss the pros and cons of the virtual congress, and offer Twitter-related recommendations for future virtual or blended congresses. We conclude that the ISTH Twitter Ambassador program broadened social media engagement and offers a novel route to improve social connectivity in the virtual research congress setting.

8.
Res Pract Thromb Haemost ; 4(1): 36-45, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31989083

ABSTRACT

Platelets are key mediators of hemostasis and thrombosis and can be inhibited by nonsteroidal anti-inflammatory drugs (NSAIDs). As a result, platelet donors are temporarily deferred from donating if they have recently taken NSAIDs such as aspirin or ibuprofen. Despite these measures, a proportion of platelet donations show exposure to these drugs; however, little is known about the effect of NSAIDs and their metabolites on platelet quality in vivo and during storage. In this review, the effect of NSAIDs on platelet function is summarized, with a focus on the widely consumed over-the-counter (OTC) medications aspirin, ibuprofen, and the non-NSAID paracetamol. Aspirin and ibuprofen have well-defined antiplatelet effects. In comparison, studies regarding the effect of paracetamol on platelets report variable findings. The timing and order of NSAID intake is important, as concurrent NSAID use can inhibit or potentiate platelet activation depending on the drug taken. NSAID deferral periods and maximum platelet shelf-life is set by each country and are revised regularly. Reduced donor deferral periods and longer platelet storage times may affect the quality of platelet products, and it is therefore important to identify the possible impact of NSAID intake on platelet quality before and after storage.

9.
Haematologica ; 105(4): 1081-1094, 2020 04.
Article in English | MEDLINE | ID: mdl-31273092

ABSTRACT

Upon vascular injury, platelets adhere to von Willebrand Factor (VWF) via glycoprotein Ibα (GPIbα). GPIbα contains many glycans, capped by sialic acid. Sialic acid cleavage (desialylation) triggers clearance of platelets. Neuraminidases (NEU) are responsible for desialylation and so far, NEU1-4 have been identified. However, the role of NEU in healthy platelets is currently unknown. Aim of the study was to study the role of NEU1 and NEU2 in platelet signalling. Membrane association of platelet attached glycans, NEU1 and NEU2 was measured following activation with agonists using flow cytometry. Adhesion on fibrinogen, aggregation and fibrinogen-binding were assessed with/without the NEU-inhibitor, 2-deoxy-2-3-dide-hydro-N-acetylneuraminic acid. Cellular localisation of NEU1 and NEU2 was examined by fluorescence microscopy. Desialylation occurred following GPIbα-clustering by VWF. Basal levels of membrane NEU1 were low; glycoprotein Ibα-clustering induced a four-fold increase (n=3, P<0.05). Inhibition of αIIbß3-integrin prevented the increase in NEU1 membrane-association by ~60%. Membrane associated NEU2 increased two-fold (n=3, P<0.05) upon VWF-binding, while inhibition/removal of GPIbα reduced the majority of membrane associated NEU1 and NEU2 (n=3, P<0.05). High shear and addition of fibrinogen increased membrane NEU1 and NEU2. NEU-inhibitior prevented VWF-induced αIIbß3-integrin activation by 50% (n=3, P<0.05), however, promoted VWF-mediated agglutination, indicating a negative feedback mechanism for NEU activity. NEU1 or NEU2 were partially co-localised with mitochondria and α-granules respectively. Neither NEU1 nor NEU2 co-localised with lysosomal-associated membrane protein 1. These findings demonstrate a previously unrecognised role for NEU1 and NEU2 in GPIbα-mediated and αIIbß3-integrin signalling.


Subject(s)
Neuraminidase , Platelet Glycoprotein GPIIb-IIIa Complex , Blood Platelets , Humans , Platelet Glycoprotein GPIb-IX Complex , Signal Transduction , von Willebrand Factor
10.
Nat Commun ; 6: 7737, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26185093

ABSTRACT

Immune thrombocytopenia (ITP) is a common bleeding disorder caused primarily by autoantibodies against platelet GPIIbIIIa and/or the GPIb complex. Current theory suggests that antibody-mediated platelet destruction occurs in the spleen, via macrophages through Fc-FcγR interactions. However, we and others have demonstrated that anti-GPIbα (but not GPIIbIIIa)-mediated ITP is often refractory to therapies targeting FcγR pathways. Here, we generate mouse anti-mouse monoclonal antibodies (mAbs) that recognize GPIbα and GPIIbIIIa of different species. Utilizing these unique mAbs and human ITP plasma, we find that anti-GPIbα, but not anti-GPIIbIIIa antibodies, induces Fc-independent platelet activation, sialidase neuraminidase-1 translocation and desialylation. This leads to platelet clearance in the liver via hepatocyte Ashwell-Morell receptors, which is fundamentally different from the classical Fc-FcγR-dependent macrophage phagocytosis. Importantly, sialidase inhibitors ameliorate anti-GPIbα-mediated thrombocytopenia in mice. These findings shed light on Fc-independent cytopenias, designating desialylation as a potential diagnostic biomarker and therapeutic target in the treatment of refractory ITP.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Integrin beta3/immunology , Neuraminidase/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Platelet Glycoprotein GPIb-IX Complex/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Animals , Blood Platelets , Blotting, Western , Flow Cytometry , Hepatocytes/metabolism , Humans , Immunohistochemistry , Mice , Mice, Knockout , Neuraminidase/antagonists & inhibitors
12.
Cardiovasc Hematol Disord Drug Targets ; 13(1): 50-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23082940

ABSTRACT

Phagocytes were first described by Dr. Metchnikoff in 1873. The roles of phagocytes in innate and adaptive immunity have been well established to date, although the molecular mechanisms involved in initiating phagocytosis (through Fc or other receptors) remain to be further explored. Phagocytes in the reticuloendothelial system, particularly macrophages, have been implicated in the clearance of senescent blood cells. The destruction of these cells may be primarily mediated via an Fc-independent pathway. Fc-independent phagocytosis may also play an important role in platelet clearance, including in autoimmune thrombocytopenia (ITP), and in clearance of platelet-rich emboli detached from sites of vascular injury. In ITP, the two major platelet auto-antigens have been located on glycoprotein (GP)IIbIIIa and the GPIb complex. It has been demonstrated that anti-GPIb antibodies, in contrast to anti-GPIIbIIIa, can induce thrombocytopenia in an Fc-independent manner. We further demonstrated in an animal model that intravenous IgG (IVIG) is unable to ameliorate thrombocytopenia caused by most anti-GPIb antibodies, despite its efficacy in anti- GPIIbIIIa-mediated thrombocytopenia. Our data was supported by subsequent retrospective studies with ITP patients by several independent groups. Most recently, we found that anti-GPIb-mediated ITP was also resistant to steroid therapy and that platelet activation and apoptosis induced by anti-GPIb antibodies may be involved in the Fc-independent platelet clearance. Therefore, identification of antibody specificity in patients, e.g. anti-GPIIbIIIa (Fc-dependent) versus anti-GPIb (Fc-independent), may be important for therapies against ITP, as well as other immune-mediated thrombocytopenias.


Subject(s)
Immunoglobulins, Intravenous/therapeutic use , Phagocytosis/immunology , Thrombocytopenia/drug therapy , Thrombocytopenia/immunology , Animals , Disease Models, Animal , Humans , Immunoglobulins, Intravenous/immunology , Phagocytosis/drug effects , Retrospective Studies
13.
Haematologica ; 97(10): 1514-22, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22371179

ABSTRACT

BACKGROUND: Cold storage of platelets reduces bacterial growth and preserves their hemostatic properties better than current procedures do. However, storage at 0°C induces [14-3-3ζ-glycoprotein Ibα] association, 14-3-3ζ release from phospho-Bad, Bad activation and apoptosis. DESIGN AND METHODS: We investigated whether arachidonic acid, which also binds 14-3-3ζ, contributes to coldinduced apoptosis. RESULTS: Cold storage activated P38-mitogen-activated protein kinase and released arachidonic acid, which accumulated due to cold inactivation of cyclooxygenase-1/thromboxane synthase. Accumulated arachidonic acid released 14-3-3ζ from phospho-Bad and decreased the mitochondrial membrane potential, which are steps in the induction of apoptosis. Addition of arachidonic acid did the same and its depletion made platelets resistant to cold-induced apoptosis. Incubation with biotin-arachidonic acid revealed formation of an [arachidonic acid-14-3-3ζ-glycoprotein Ibα] complex. Indomethacin promoted complex formation by accumulating arachidonic acid and released 14-3-3ζ from cyclo-oxygenase-1. Arachidonic acid depletion prevented the cold-induced reduction of platelet survival in mice. CONCLUSIONS: We conclude that cold storage induced apoptosis through an [arachidonic acid-14-3-3ζ-glycoprotein Ibα] complex, which released 14-3-3ζ from Bad in an arachidonic acid-dependent manner. Although arachidonic acid depletion reduced agonist-induced thromboxane A(2) formation and aggregation, arachidonic acid repletion restored these functions, opening ways to reduce apoptosis during storage without compromising hemostatic functions post-transfusion.


Subject(s)
14-3-3 Proteins/metabolism , Arachidonic Acid/physiology , Blood Platelets , Blood Preservation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Apoptosis/drug effects , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Blood Platelets/metabolism , Cell Survival , Cold Temperature , Cyclooxygenase 1/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Platelet Activation , Protein Binding/drug effects , bcl-Associated Death Protein/metabolism
14.
Am J Hematol ; 87(2): 206-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22139961

ABSTRACT

Immune thrombocytopenia (ITP) is characterized by platelet clearance mediated primarily by autoantibodies against the platelet GPIIbIIIa and/or GPIbα. Steroid therapy is a first-line treatment for ITP. However, some patients are refractory to this therapy and currently no method can predict which patients will respond. To evaluate whether steroids are equally efficacious in treating patients with ITP caused by anti-GPIIbIIIa versus anti-GPIbα antibodies, we performed a retrospective study on 176 newly diagnosed patients with acute ITP who had severe bleeding symptoms and were admitted as resident patients to the hospital. The patients were treated first with intravenous administration of high-dose dexamethasone (DXM), followed by oral administration of prednisone. Response to therapy was observed in a majority of patients with antibodies specific for GPIIbIIIa (31/43) or without detectable antibodies against either GPIIbIIIa or GPIbα (36/45). In contrast, the steroid response was significantly lower in patients with anti-GPIbα antibodies (9/34) or with antibodies against both GPIbα and GPIIbIIIa (16/54). The preliminary findings of this study suggest that in future prospective clinical trials including corticosteroids, the anti-GPIbα, and -GPIIbIIIa status should be assessed in order to test its potential relevance in deciding future treatments.


Subject(s)
Autoantibodies/immunology , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Membrane Glycoproteins/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Prednisone/therapeutic use , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Blood Platelets/drug effects , Blood Platelets/immunology , Blood Platelets/pathology , Dexamethasone/pharmacology , Drug Administration Schedule , Drug Monitoring , Female , Glucocorticoids/pharmacology , Humans , Male , Middle Aged , Platelet Glycoprotein GPIb-IX Complex , Prednisone/pharmacology , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/pathology , Retrospective Studies , Treatment Outcome
15.
J Clin Invest ; 121(11): 4537-47, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22019589

ABSTRACT

Fetal and neonatal immune thrombocytopenia (FNIT) is a severe bleeding disorder caused by maternal antibody-mediated destruction of fetal/neonatal platelets. It is the most common cause of severe thrombocytopenia in neonates, but the frequency of FNIT-related miscarriage is unknown, and the mechanism(s) underlying fetal mortality have not been explored. Furthermore, although platelet αIIbß3 integrin and GPIbα are the major antibody targets in immune thrombocytopenia, the reported incidence of anti-GPIbα-mediated FNIT is rare. Here, we developed mouse models of FNIT mediated by antibodies specific for GPIbα and ß3 integrin and compared their pathogenesis. We found, unexpectedly, that miscarriage occurred in the majority of pregnancies in our model of anti-GPIbα-mediated FNIT, which was far more frequent than in anti-ß3-mediated FNIT. Dams with anti-GPIbα antibodies exhibited extensive fibrin deposition and apoptosis/necrosis in their placentas, which severely impaired placental function. Furthermore, anti-GPIbα (but not anti-ß3) antiserum activated platelets and enhanced fibrin formation in vitro and thrombus formation in vivo. Importantly, treatment with either intravenous IgG or a monoclonal antibody specific for the neonatal Fc receptor efficiently prevented anti-GPIbα-mediated FNIT. Thus, the maternal immune response to fetal GPIbα causes what we believe to be a previously unidentified, nonclassical FNIT (i.e., spontaneous miscarriage but not neonatal bleeding) in mice. These results suggest that a similar pathology may have masked the severity and frequency of human anti-GPIbα-mediated FNIT, but also point to possible therapeutic interventions.


Subject(s)
Abortion, Spontaneous/etiology , Abortion, Spontaneous/prevention & control , Histocompatibility, Maternal-Fetal/immunology , Immunoglobulins, Intravenous/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/immunology , Receptors, Fc/antagonists & inhibitors , Abortion, Spontaneous/immunology , Animals , Blood Platelets/immunology , Disease Models, Animal , Female , Histocompatibility Antigens Class I/immunology , Humans , Integrin beta3/genetics , Integrin beta3/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Platelet Glycoprotein GPIb-IX Complex/genetics , Pregnancy , Receptors, Fc/immunology , Thrombocytopenia, Neonatal Alloimmune/etiology , Thrombocytopenia, Neonatal Alloimmune/immunology
16.
Thromb Haemost ; 103(5): 1033-43, 2010 May.
Article in English | MEDLINE | ID: mdl-20216992

ABSTRACT

Incubation at 0 degrees C is known to expose b- N -acetyl-D-glucosamine residues on glycoprotein (GP) Ibalpha inducing receptor clustering and alpha(M)beta(2)-mediated platelet destruction by macrophages. Here we show that incubation at 0/37 degrees C (4 hours at 0 degrees C, followed by 1 hour at 37 degrees C to mimic cold-storage and post-transfusion conditions) triggers a conformational change in the N -terminal flank (NTF, amino acids, aa 1-35) but not in aa 36-282 of GPIbalpha as detected by antibody binding. Addition of the sugar N -acetyl-D-glucosamine (GN) inhibits responses induced by 0/37 degrees C. Incubation at 0 degrees C shifts GPIbalpha from the membrane skeleton to the cytoskeleton. Different GPIbalpha conformations have little effect on VWF/ristocetin-induced aggregation, but arrest of NTF change by GN interferes with agglutination and spreading on a VWF-coated surface under flow. Strikingly, incubation at 0/37 degrees C initiates thromboxane A(2) formation through a von Willebrand factor (VWF)-independent and GPIbalpha-dependent mechanism, as confirmed in VWF- and GPIbalpha-deficient platelets. We conclude that the NTF change induced by 0/37 degrees C incubation reflects clustering of GPIbalpha supports VWF/ristocetin-induced agglutination and spreading and is sufficient to initiate platelet activation in the absence of VWF.


Subject(s)
Acetylglucosamine/metabolism , Blood Platelets/metabolism , Blood Preservation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Transfusion , von Willebrand Diseases/metabolism , Acetylglucosamine/chemistry , Antibodies, Monoclonal , Blood Platelets/pathology , Cells, Cultured , Cytoskeleton/metabolism , Humans , Macrophage Activation , Macrophage-1 Antigen/metabolism , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/immunology , Protein Binding , Protein Conformation , Protein Transport , Receptor Aggregation/physiology , Temperature , Thromboxane A2/biosynthesis , Thromboxane A2/genetics , von Willebrand Diseases/blood , von Willebrand Diseases/pathology , von Willebrand Diseases/therapy , von Willebrand Factor/metabolism
17.
Arterioscler Thromb Vasc Biol ; 29(3): 372-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19096001

ABSTRACT

OBJECTIVE: The sensitivity of platelets to aggregating agents increases when low-density lipoprotein (LDL) binds to apolipoprotein E receptor 2' (apoER2'), triggering activation of p38MAPK and formation of thromboxane A2. LDL signaling is terminated by PECAM-1 through recruitment and activation of the Ser/Thr protein phosphatase PP2A, but platelets remain unresponsive to LDL when PECAM-1 activation disappears. We report a second mechanism that halts LDL signaling and in addition lowers platelet responsiveness to aggregating agents. METHODS AND RESULTS: After a first stimulation with LDL, platelets remain unresponsive to LDL for 60 minutes, despite normal apoER2' activation by a second dose of LDL. A possible cause is persistent activation of the tyrosine phosphatases SHP-1 and SHP-2, which may not only block a second activation of p38MAPK, PECAM-1, and PP2A by LDL but also seem to reduce aggregation by TRAP, collagen, and ADP. CONCLUSION: These findings reveal that p38MAPK phosphorylation and platelet activation by LDL are suppressed by two mechanisms: (1) short activation of PECAM-1/PP2A, and (2) prolonged activation of SHP-1 and SHP-2. Activation of SHP-1 and SHP-2 is accompanied by reduced responsiveness to aggregating agents, which--if present in vivo--would make LDL an aggregation inhibitor during prolonged contact with platelets.


Subject(s)
Blood Platelets/enzymology , Lipoproteins, LDL/metabolism , Platelet Aggregation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction , Adenosine Diphosphate/metabolism , Collagen/metabolism , Down-Regulation , Humans , LDL-Receptor Related Proteins , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Phosphatase 2/metabolism , Receptors, Lipoprotein/metabolism , Receptors, Thrombin/metabolism , Thromboxane A2/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Histochem Cytochem ; 55(3): 247-54, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17101725

ABSTRACT

During early postnatal development, the myosin heavy chain (MyHC) expression pattern in equine gluteus medius muscle shows adaptation to movement and load,resulting in a decrease in the number of fast MyHC fibers and an increase in the number of slow MyHC fibers. In the present study we correlated the expression of MyHC isoforms to the expression of sarcoplasmic(endo)reticulum Ca2+-ATPase 1 and 2a (SERCA), phospholamban (PLB), calcineurin A (CnA), and calcineurin B (CnB). Gluteus medius muscle biopsies were taken at 0, 2, 4, and 48 weeks and analyzed using immunofluorescence. Both SERCA isoforms and PLB were expressed in almost all fiber types at birth. From 4 weeks of age onward, SERCA1 was exclusively expressed in fast MyHC fibers and SERCA2a and PLB in slow MyHC fibers. At all time points, CnA and CnB proteins were expressed at a basal level in all fibers, but with a higher expression level in MyHC type 1 fibers. From 4 weeks onward, expression of only CnA was also higher in MyHC type 2a and 2ad fibers. We propose a double function of calcineurin in calcium homeostasis and maintenance of slow MyHC fiber type identity. Although equine muscle is already functional at birth, expression patterns of the monitored proteins still show adaptation, depending on the MyHC fiber type.


Subject(s)
Calcineurin/biosynthesis , Calcium-Binding Proteins/biosynthesis , Calcium/metabolism , Muscle, Skeletal/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/biosynthesis , Sarcoplasmic Reticulum/metabolism , Animals , Fluorescent Antibody Technique , Horses , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/growth & development , Myosin Heavy Chains/biosynthesis , Protein Isoforms/biosynthesis , Protein Subunits/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...