Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Cell Death Dis ; 4: e611, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23618909

ABSTRACT

Immune cells and platelets maintain plasma membrane phospholipid asymmetry. Upon activation, this asymmetry is disrupted by phospholipid scrambling (PS), which is a major step during activation of immune cells, hemostasis and apoptosis. Anoctamin 6 (Ano6; TMEM16F) causes chloride (Cl(-)) and cation currents and is required for Ca(2+)-dependent PS. It is defective in blood cells from patients with Scott syndrome, a rare bleeding disorder. We examined if Cl(-) currents and PS are related, whether both processes are Ca(2+) dependent, and whether Ca(2+)-independent scrambling during intrinsic and extrinsic apoptosis is controlled by Ano6. Ca(2+) increase by ionomycin activated Ano6 Cl(-) currents and PS in normal lymphocytes, but not in B-lymphocytes from two different patients with Scott syndrome. Fas ligand (FasL) did not increase intracellular Ca(2+), but activated Cl(-) currents in normal but not in Scott lymphocytes. Whole-cell currents were inhibited by Cl(-) channel blockers and by siRNA knockdown of Ano6. In contrast, intrinsic mitochondrial apoptosis by ABT-737 did not induce Cl(-) currents in lymphocytes. PS was not inhibited by blockers of Ano6 or removal of Cl(-) ions. Remarkably, Ca(2+)-independent scrambling due to extrinsic (FasL) or intrinsic (ABT-737) apoptosis was unchanged in Scott cells. We conclude that: (i) Ano6 Cl(-) currents are activated by increase in cytosolic Ca(2+), or Ca(2+) independent by stimulation of Fas receptors; (ii) Ca(2+)-dependent PS induced by Ano6 does not require Cl(-) currents; (iii) Ca(2+)-independent PS does not require Ano6; (iv) Ano6 is necessary for Ca(2+)-dependent PS, but not by increasing intracellular Ca(2+).


Subject(s)
Calcium/metabolism , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , Anoctamins , Apoptosis/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Biphenyl Compounds/pharmacology , Blood Coagulation Disorders/physiopathology , Calcium Ionophores/pharmacology , Chloride Channels/antagonists & inhibitors , Chloride Channels/metabolism , Fas Ligand Protein/pharmacology , HEK293 Cells , Humans , Ion Transport/drug effects , Ionomycin/pharmacology , Jurkat Cells , Nitrophenols/pharmacology , Patch-Clamp Techniques , Phospholipid Transfer Proteins/antagonists & inhibitors , Phospholipid Transfer Proteins/genetics , Piperazines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Sulfonamides/pharmacology
2.
Cell Mol Life Sci ; 66(2): 314-23, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18989619

ABSTRACT

The exposure of phosphatidylserine (PS) at the cell surface plays a critical role in blood coagulation and serves as a macrophage recognition moiety for the engulfment of apoptotic cells. Previous observations have shown that a high extracellular [K(+)] and selective K(+) channel blockers inhibit PS exposure in platelets and erythrocytes. Here we show that the rate of PS exposure in erythrocytes decreases by approximately 50% when the intracellular [K(+)] increases from 0 to physiological concentrations. Using resealed erythrocyte membranes, we further show that lipid scrambling is inducible by raising the intracellular [Ca(2+)] and that K(+) ions have a direct inhibitory effect on this process. Lipid scrambling in resealed ghosts occurs in the absence of cell shrinkage and microvesicle formation, processes that are generally attributed to Ca(2+)-induced lipid scrambling in intact erythrocytes. Thus, opening of Ca(2+)-sensitive K(+) channels causes loss of intracellular K(+) that results in reduced intrinsic inhibitory effect of these ions on scramblase activity.


Subject(s)
Erythrocyte Membrane/chemistry , Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Membrane Lipids/metabolism , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Potassium/metabolism , Calcium/metabolism , Cell Shape , Erythrocyte Membrane/drug effects , Erythrocytes/cytology , Erythrocytes/drug effects , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Ions/metabolism , Sodium/metabolism , Thromboplastin/metabolism
4.
Br J Haematol ; 138(4): 527-33, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17608769

ABSTRACT

Microparticles (MPs), shed during the storage of platelets, support blood coagulation and could be helpful in restoring the haemostatic system in thrombocytopenic patients. The mechanisms by which MPs support haemostasis under flow conditions were investigated. Fluorescent-labelled MPs were perfused at shear rates of 100 and 1000/s over surfaces coated with collagen, fibrinogen, von Willebrand factor (VWF) or surface-adherent platelets. Adhesion was monitored in real-time by fluorescence microscopy. In addition, thrombin-antithrombin (TAT) complex formation was measured in flowing thrombocytopenic blood. MPs attained the capacity to firmly adhere to collagen, VWF, fibrinogen and surface-adherent platelets at high and low shear rate. Antibodies against glycoprotein Ibalpha and alpha(IIb)beta(3) were used to demonstrate the specificities of these interactions. The addition of MPs to thrombocytopenic blood did not affect platelet adhesion. TAT complex formation was increased in the presence of MPs in capillaries coated with fibrinogen, but not on collagen fibres. We confirmed that MPs adhere to a damaged vascular bed in vivo after infusion in denuded arteries in a mouse model. MPs have platelet-like adhering properties and accelerate thrombin generation. These properties strongly support the notion that MPs can be beneficial in maintaining normal haemostasis when platelet function is impaired or reduced, as in thrombocytopenic patients.


Subject(s)
Blood Coagulation , Blood Platelets/physiology , Collagen Type I/physiology , Fibrinogen/physiology , von Willebrand Factor/physiology , Animals , Antithrombin III/physiology , Capillaries , Hemorheology , Humans , Male , Mice , Mice, Inbred Strains , Microscopy, Fluorescence , Peptide Hydrolases/physiology , Stress, Mechanical , Thrombocytopenia/blood , Thrombocytopenia/therapy , Tissue Adhesions
5.
J Thromb Haemost ; 5(3): 571-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17166251

ABSTRACT

BACKGROUND: Blockade of the thrombin receptors protease-activated receptor (PAR)1 and PAR4 with pepducins, cell-penetrating lipopeptides based on the third intracellular loop of PAR1 and PAR4, effectively inhibits platelet aggregation. We have previously shown that PAR1 pepducin also exerts an anticoagulant activity by partial inhibition of the thrombin plus collagen-induced externalization of phosphatidylserine (PS) at the platelet plasma membrane. OBJECTIVE: In the present study we examined the effects of PAR1 and PAR4 pepducins on tissue factor (TF)-initiated thrombin generation in platelet-rich plasma (PRP) and the interaction between PAR4 pepducin-loaded mouse platelets and a growing thrombus to confirm the relevance of the in vitro data. RESULTS: Localization of pepducins at the inner leaflet of the plasma membrane was confirmed with a fluorescence resonance energy transfer assay. Both the PAR1 pepducin, P1pal12, and the PAR4 pepducin, P4pal10, inhibited TF-initiated thrombin generation in PRP. Concentrations of P1pal12 and P4pal10, which blocked the thrombin-induced influx of extracellular calcium ions and inhibited platelet aggregation, reduced the rate of thrombin generation during the propagation phase by 38% and 36%, respectively. Whether this anticoagulant effect is relevant in inhibiting in vivo arterial thrombin growth is uncertain because P4pal10 prevented the incorporation of platelets in a growing thrombus. CONCLUSIONS: Our findings suggest that in spite of their potential anticoagulant activities the in vivo antithrombotic effect of intracellular PAR pepducins is mainly based on inhibiting platelet-platelet interactions.


Subject(s)
Anticoagulants/pharmacology , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Lipoproteins/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Receptors, Proteinase-Activated/antagonists & inhibitors , Animals , Anticoagulants/metabolism , Anticoagulants/therapeutic use , Blood Platelets/metabolism , Carotid Artery, Common/surgery , Cell Membrane/metabolism , Cell Membrane Permeability , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/therapeutic use , Flow Cytometry , Fluorescence Resonance Energy Transfer , Humans , In Vitro Techniques , Lipoproteins/metabolism , Lipoproteins/therapeutic use , Male , Mice , Microscopy, Video , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/therapeutic use , Receptor, PAR-1/antagonists & inhibitors , Receptors, Proteinase-Activated/metabolism , Receptors, Thrombin/antagonists & inhibitors , Thrombin/metabolism , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/metabolism , Thrombosis/prevention & control , Time Factors
6.
Cell Mol Life Sci ; 62(13): 1514-25, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15971000

ABSTRACT

Platelet procoagulant activity is mainly determined by the extent of surface-exposed phosphatidylserine (PS), controlled by the activity of aminophospholipid translocase and phospholipid scramblase. Here, we studied both transport activities in single platelets upon stimulation with various agonists. Besides the formation of procoagulant microparticles, the results show that a distinct fraction of the platelets exposes PS when stimulated. The extent of PS exposure in these platelet fractions was similar to that in platelets challenged with Ca2+-ionophore, where all cells exhibit maximal attainable PS exposure. The size of the PS-exposing fraction depends on the agonist and is proportional to the platelet procoagulant activity. Scramblase activity was observed only in the PS-exposing platelet fraction, whereas translocase activity was exclusively detectable in the fraction that did not expose PS. We conclude that, irrespective of the agonist, procoagulant platelets exhibit maximal surface exposure of PS by switching on scramblase and inhibiting translocase activity.


Subject(s)
Blood Platelets/metabolism , Membrane Proteins/metabolism , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Annexin A5/metabolism , Antigens, Surface/metabolism , Collagen/pharmacology , Flow Cytometry , Fluorescein-5-isothiocyanate , Humans , Ionomycin/pharmacology , Membrane Proteins/agonists , Membrane Proteins/antagonists & inhibitors , Milk Proteins/metabolism , Phospholipid Transfer Proteins/agonists , Phospholipid Transfer Proteins/antagonists & inhibitors , Platelet Activation , Thrombin/pharmacology , Thromboplastin/metabolism
7.
Cell Mol Life Sci ; 62(9): 971-88, 2005 May.
Article in English | MEDLINE | ID: mdl-15761668

ABSTRACT

The asymmetric phospholipid distribution in plasma membranes is normally maintained by energy-dependent lipid transporters that translocate different phospholipids from one monolayer to the other against their respective concentration gradients. When cells are activated, or enter apoptosis, lipid asymmetry can be perturbed by other lipid transporters (scramblases) that shuttle phospholipids non-specifically between the two monolayers. This exposes phosphatidylserine (PS) at the cells' outer surface. Since PS promotes blood coagulation, defective scramblase activity upon platelet stimulation causes a bleeding disorder (Scott syndrome). PS exposure also plays a pivotal role in the recognition and removal of apoptotic cells via a PS-recognizing receptor on phagocytic cells. Furthermore, expression of PS at the cell surface can occur in a wide variety of disorders. This review aims at highlighting how PS expression in different cells may complicate a variety of pathological conditions, including those that promote thromboembolic complications or produce aberrations in apoptotic cell removal.


Subject(s)
Eukaryotic Cells/metabolism , Phosphatidylserines/metabolism , Antiphospholipid Syndrome/metabolism , Apoptosis/physiology , Cell Membrane/metabolism , Eukaryotic Cells/pathology , Hematologic Diseases/metabolism , Humans , Infections/metabolism , Kidney Diseases/metabolism , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/physiology , Metabolic Diseases/metabolism , Neoplasms/metabolism , Phosphatidylserines/physiology , Respiratory Tract Diseases/metabolism
9.
Biochemistry ; 40(27): 8065-72, 2001 Jul 10.
Article in English | MEDLINE | ID: mdl-11434775

ABSTRACT

In erythrocytes and platelets, activation of a nonspecific lipid flipsite termed the scramblase allows rapid, bidirectional transbilayer movement of all types of phospholipids. When applied to lymphoid cells, scramblase assays reveal a similar activity, with scrambling rates intermediate between those seen in platelets and erythrocytes. Scrambling activity initiated in lymphoid cells by elevation of intracellular Ca(2+) proceeds after a lag not noted in platelets or erythrocytes. The rates of transbilayer movement of phosphatidylserine and phosphatidylcholine analogues are similar whether the scramblase is activated by elevated internal Ca(2+) or by apoptosis. Elevation of internal Ca(2+) levels in apoptotic cells does not result in an additive increase in the rate of lipid movement. In lymphoid cells from a patient with Scott syndrome, scramblase cannot be activated by Ca(2+), but is induced normally during apoptosis. These findings suggest that Ca(2+) and apoptosis operate through different pathways to activate the same scramblase.


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Carrier Proteins/metabolism , Lymphocytes/enzymology , Membrane Proteins/metabolism , Phospholipid Transfer Proteins , Phospholipids/metabolism , 4-Chloro-7-nitrobenzofurazan/metabolism , Animals , Apoptosis/genetics , B-Lymphocytes/enzymology , B-Lymphocytes/pathology , Blood Coagulation Disorders/enzymology , Blood Coagulation Disorders/genetics , Blood Coagulation Disorders/pathology , Calcium/metabolism , Carrier Proteins/genetics , Cell Line, Transformed , Enzyme Activation/genetics , Flow Cytometry , Humans , Hybridomas , Jurkat Cells , Lymphocytes/cytology , Membrane Proteins/genetics , Mice , Mutation , Phosphatidylcholines/metabolism , Phosphatidylserines/metabolism , Spectrometry, Fluorescence , Syndrome
10.
Biochemistry ; 40(30): 8672-6, 2001 Jul 31.
Article in English | MEDLINE | ID: mdl-11467926

ABSTRACT

There is increasing evidence that endogenously generated aldehydes formed as a result of lipid peroxidation are involved in the pathophysiological effects associated with oxidative stress in cells and tissues. Malondialdehyde (MDA), a major product of lipid peroxidation, can modify amines present on the cell surface and thereby introduce negative charges that can affect the interfacial ionic layer. We show that lipid peroxidation of RBC generates MDA adducts that, similar to phosphatidylserine (PS), bind annexin V in a Ca(2+)-dependent manner. Like PS, these adducts also promote the "PS-dependent" prothrombinase assays, albeit to lower levels. These results indicate that annexin V binding cannot be used as an exclusive indicator of cell surface PS and raise the possibility that some phenomenon attributed to PS may, in fact, also involve aldehyde-lipid adducts.


Subject(s)
Annexin A5/metabolism , Lipid Peroxidation , Membrane Lipids/metabolism , Animals , Annexin A5/blood , Cattle , Erythrocytes/metabolism , Humans , Malondialdehyde/blood , Malondialdehyde/metabolism , Membrane Lipids/blood , Microscopy, Fluorescence , Phosphatidylethanolamines/blood , Phosphatidylethanolamines/metabolism , Phosphatidylserines/blood , Phosphatidylserines/metabolism , Protein Binding
11.
Br J Haematol ; 113(3): 621-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380447

ABSTRACT

We investigated the mechanism by which anti-prothrombin antibodies cause lupus anticoagulant (LAC) activity. Addition of affinity-purified anti-prothrombin antibodies from LAC-positive plasma samples (alpha-FII-LAC+) to normal plasma induced LAC activity. Upon increasing the phospholipid concentration, LAC activity was neutralized. Addition of purified alpha-FII-LAC+ to normal plasma strongly inhibited factor Xa formation. No inhibition was measured when alpha-FII-LAC+ were added to prothrombin-deficient plasma or when purified anti-prothrombin antibodies from LAC-negative plasma samples (alpha-FII-LAC-) were added. When a combination of prothrombin and alpha-FII-LAC+ was added to the purified clotting complex, a strong inhibition of factor Xa and IIa formation was seen. The alpha-FII-LAC+ alone or a combination of prothrombin and alpha-FII-LAC- did not show inhibition. Ellipsometry studies showed that, in the presence of alpha-FII-LAC+, the affinity of prothrombin for a phospholipid surface increased dramatically, whereas a much lower increase was observed with alpha-FII-LAC-. Our results show that complexes of prothrombin and anti-prothrombin antibodies with LAC activity inhibit both prothrombinase and tenase. The antibodies increase the affinity of prothrombin for the phospholipid surface, thereby competing with clotting factors for the available catalytic phospholipid surface, a mechanism similar to that of anti-beta2-glycoprotein I antibodies.


Subject(s)
Antiphospholipid Syndrome/metabolism , Autoantibodies/metabolism , Lupus Coagulation Inhibitor/metabolism , Lupus Erythematosus, Systemic/metabolism , Neoplasm Proteins , Phospholipids/metabolism , Prothrombin/immunology , Antiphospholipid Syndrome/immunology , Cysteine Endopeptidases/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Factor Xa/metabolism , Female , Humans , Lipid Bilayers/metabolism , Lupus Erythematosus, Systemic/immunology , Male , Protein Binding , Prothrombin/metabolism , Thromboplastin/metabolism
12.
Arterioscler Thromb Vasc Biol ; 21(4): 618-27, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11304481

ABSTRACT

In the final stages of activation, platelets express coagulation-promoting activity by 2 simultaneous processes: exposure of aminophospholipids, eg, phosphatidylserine (PS), at the platelet surface, and formation of membrane blebs, which may be shed as microvesicles. Contact with collagen triggers both processes via platelet glycoprotein VI (GPVI). Here, we studied the capacity of 2 GPVI ligands, collagen-related peptide (CRP) and the snake venom protein convulxin (CVX), to elicit the procoagulant platelet response. In platelets in suspension, either ligand induced full aggregation and high Ca(2+) signals but little microvesiculation or PS exposure. However, most of the platelets adhering to immobilized CRP or CVX had exposed PS and formed membrane blebs after a prolonged increase in cytosolic [Ca(2+)](i). Platelets adhering to fibrinogen responded similarly but only when exposed to soluble CRP or CVX. By scanning electron microscopic analysis, the bleb-forming platelets were detected as either round, spongelike structures with associated microparticles or as arrays of vesicular cell fragments. The phosphorylation of p38 mitogen-activated protein kinase (MAPK) elicited by CRP and CVX was enhanced in fibrinogen-adherent platelets compared with that in platelets in suspension. The p38 inhibitor SB203580 and the calpain protease inhibitor calpeptin reduced only the procoagulant bleb formation, having no effect on PS exposure. Inhibition of p38 also downregulated calpain activity. We conclude that the procoagulant response evoked by GPVI stimulation is potentiated by platelet adhesion. The sequential activation of p38 MAPK and calpain appears to regulate procoagulant membrane blebbing but not PS exposure.


Subject(s)
Blood Platelets/physiology , Carrier Proteins , Lectins, C-Type , Platelet Activation/physiology , Platelet Adhesiveness/physiology , Blood Platelets/cytology , Blood Platelets/drug effects , Blotting, Western , Calcium/metabolism , Calcium/physiology , Calpain/metabolism , Calpain/pharmacology , Crotalid Venoms/metabolism , Crotalid Venoms/pharmacology , Enzyme Activation , Flow Cytometry , Humans , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/pharmacology , Phosphatidylserines/metabolism , Phosphatidylserines/pharmacology , Phosphorylation , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/pharmacology , Proteins/metabolism , Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases
13.
Thromb Haemost ; 84(4): 653-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11057865

ABSTRACT

Anionic phospholipid membranes have a dual role in blood coagulation: they are essential for the initiation and propagation as well as for the limitation and termination of the blood coagulation process. Patients with the anti-phospholipid syndrome (APS) carrying antibodies against complexes of anionic phospholipids and plasma proteins, show in vitro inhibited phospholipid dependent coagulation reactions, whereas in vivo the presence of these antibodies is associated with an increased risk of thrombosis. In this study we focussed on the effects of these anti-phospholipid antibodies on the regulation of TF-mediated factor Xa (FXa) generation in plasma. We hypothesized that anti-phospholipid antibodies interfere with the phospholipid-dependent inhibition by tissue factor pathway inhibitor (TFPI) of TF-induced coagulation. Indeed, total-IgG, anti-cardiolipin-IgG (aCL) and anti-beta2GPI-IgG, isolated from patient plasmas, all stimulated TF-induced FXa generation in normal plasma. This enhanced FXa generation was not observed when the patient's IgG was depleted of anti-beta2GPI-IgG or when normal plasma was depleted of beta2PGPI or TFPI. Taken together, these data indicate that antibodies to beta2GPI, circulating in patients with APS, suppress TFPI-dependent inhibition of TF-induced coagulation, which results in an increased FXa generation.


Subject(s)
Antiphospholipid Syndrome/immunology , Autoantibodies/immunology , Autoantibodies/pharmacology , Glycoproteins/immunology , Lipoproteins/antagonists & inhibitors , Lipoproteins/immunology , Blood Coagulation/drug effects , Blood Coagulation/immunology , Humans , beta 2-Glycoprotein I
14.
Platelets ; 11(6): 301-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11083453

ABSTRACT

Platelets in an advanced stage of activation change from coagulation-inactive to coagulation-promoting cells. This procoagulant response is characterised by exposure of aminophospholipids, such as phosphatidylserine, to the platelet surface and by formation of microvesicles. Under specific conditions, when both signalling and adhesive platelet receptors are occupied, collagen and also thrombin are able to trigger this response. Thus, platelets express high coagulation-promoting activity only after interacting with multiple receptors.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Humans
15.
Biochem J ; 350 Pt 2: 531-5, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10947968

ABSTRACT

The role of multidrug resistance protein 1 (MRP1) in the maintenance of transbilayer lipid asymmetry in the erythrocyte membrane was investigated. The transbilayer distribution of endogenous phospholipids and [(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]hexanoyl (NBD)-labelled lipid analogues was compared in the absence and the presence of inhibitors of MRP1. At equilibrium the transbilayer distribution of the NBD analogues (in the absence of MRP1 inhibitors) was very similar to that of the endogenous lipids. Inhibition of MRP1 by verapamil or indomethacin resulted in a shift in the amount of probe that was internalized: approx. 50% of NBD-labelled phosphatidylcholine (PtdCho) and 9% of NBD-sphingomyelin (NBD-Spm) were no longer extractable by BSA in cells treated with inhibitor, in comparison with 25% and 3% for control cells respectively. To verify whether inhibition of MRP1 also affected the distribution of the endogenous phospholipids, phospholipase A2 and sphingomyelinase were used to assess the amount of each of the various lipid classes present in the membrane outer leaflet. No shift in phospholipid distribution was observed after 5 h of incubation with verapamil or indomethacin. However, after 48 h of incubation with these inhibitors, significantly smaller amounts of PtdCho and Spm were present in the outer membrane leaflet. No appreciable change was observed in the distribution of phosphatidylethanolamine or phosphatidylserine. Decreased hydrolysis of PtdCho and Spm was not due to endovesicle formation, as revealed by electron microscopy. This is the first report to show that MRP1 has a role in the maintenance of the outwards orientation of endogenous choline-containing phospholipids in the erythrocyte membrane.


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , ATP-Binding Cassette Transporters/metabolism , Aminocaproates , Cell Membrane/metabolism , Erythrocytes/metabolism , 4-Chloro-7-nitrobenzofurazan/pharmacology , Aminocaproic Acid/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Erythrocytes/ultrastructure , Fluorescent Dyes/pharmacology , Humans , Hydrolysis , Indomethacin/pharmacology , Lipid Bilayers/metabolism , Microscopy, Electron , Multidrug Resistance-Associated Proteins , Phosphatidylcholines/metabolism , Phospholipases A/metabolism , Phospholipases A2 , Phospholipids/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Time Factors , Vasodilator Agents/pharmacology , Verapamil/pharmacology
17.
Biochemistry ; 39(8): 1982-9, 2000 Feb 29.
Article in English | MEDLINE | ID: mdl-10684648

ABSTRACT

Annexin V, an intracellular protein with a calcium-dependent high affinity for anionic phospholipid membranes, acts as an inhibitor of lipid-dependent reactions of the blood coagulation. Antiphospholipid antibodies found in the plasma of patients with antiphospholipid syndrome generally do not interact with phospholipid membranes directly, but recognize (plasma) proteins associated with lipid membranes, mostly prothrombin or beta(2)-glycoprotein I (beta(2)GPI). Previously, it has been proposed that antiphospholipid antibodies may cause thrombosis by displacing annexin V from procoagulant cell surfaces. We used ellipsometry to study the binding of annexin V and of complexes of beta(2)GPI with patient-derived IgG antibodies to beta(2)GPI, commonly referred to as anticardiolipin antibodies (ACA), to phospholipid bilayers composed of phosphatidylcholine (PC) and 20% phosphatidylserine (PS). More specifically, we investigated the competition of these proteins for the binding sites at these bilayers. We show that ACA-beta(2)GPI complexes, adsorbed to PSPC bilayers, are displaced for more than 70% by annexin V and that annexin V binding is unaffected by the presence of ACA-beta(2)GPI complexes. Conversely, annexin V preadsorbed to these bilayers completely prevents adsorption of ACA-beta(2)GPI complexes, and none of the preadsorbed annexin V is displaced by ACA-beta(2)GPI complexes. Using ellipsometry, we also studied the effect of ACA-beta(2)GPI complexes on the interaction of annexin V with the membranes of ionophore-activated blood platelets as a more physiological relevant model of cell membranes. The experiments with blood platelets confirm the high-affinity binding of annexin V to these membranes and unequivocally show that annexin V binding is unaffected by the presence of ACA-beta(2)GPI. In conclusion, our data unambiguously show that ACA-beta(2)GPI complexes are unable to displace annexin V from procoagulant membranes to any significant extent, whereas annexin V does displace the majority of preadsorbed ACA-beta(2)GPI complexes from these membranes.


Subject(s)
Annexin A5/metabolism , Antibodies/metabolism , Cardiolipins/immunology , Annexin A5/chemistry , Antiphospholipid Syndrome/metabolism , Binding, Competitive , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Membrane/metabolism , Glycoproteins/metabolism , Humans , Ionophores/metabolism , Lipid Bilayers/metabolism , Phosphatidylcholines/metabolism , Phosphatidylserines/metabolism , Time Factors , beta 2-Glycoprotein I
18.
Biochim Biophys Acta ; 1439(3): 317-30, 1999 Aug 18.
Article in English | MEDLINE | ID: mdl-10446420

ABSTRACT

The plasma membrane, which forms the physical barrier between the intra- and extracellular milieu, plays a pivotal role in the communication of cells with their environment. Exchanging metabolites, transferring signals and providing a platform for the assembly of multi-protein complexes are a few of the major functions of the plasma membrane, each of which requires participation of specific membrane proteins and/or lipids. It is therefore not surprising that the two leaflets of the membrane bilayer each have their specific lipid composition. Although membrane lipid asymmetry has been known for many years, the mechanisms for maintaining or regulating the transbilayer lipid distribution are still not completely understood. Three major players have been presented over the past years: (1) an inward-directed pump specific for phosphatidylserine and phosphatidylethanolamine, known as aminophospholipid translocase; (2) an outward-directed pump referred to as 'floppase' with little selectivity for the polar headgroup of the phospholipid, but whose actual participation in transport of endogenous lipids has not been well established; and (3) a lipid scramblase, which facilitates bi-directional migration across the bilayer of all phospholipid classes, independent of the polar headgroup. Whereas a concerted action of aminophospholipid translocase and floppase could, in principle, account for the maintenance of lipid asymmetry in quiescent cells, activation of the scramblase and concomitant inhibition of the aminophospholipid translocase causes a collapse of lipid asymmetry, manifested by exposure of phosphatidylserine on the cell surface. In this article, each of these transporters will be discussed, and their physiological importance will be illustrated by the Scott syndrome, a bleeding disorder caused by impaired lipid scrambling. Finally, phosphatidylserine exposure during apoptosis will be briefly discussed in relation to inhibition of translocase and simultaneous activation of scramblase.


Subject(s)
Cell Membrane/metabolism , Lipid Bilayers/metabolism , Lipid Metabolism , Phospholipid Transfer Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Apoptosis , Carrier Proteins/metabolism , Cell Line , Humans , Membrane Proteins/metabolism , Phosphatidylserines/metabolism
19.
Lupus ; 7 Suppl 2: S126-31, 1998.
Article in English | MEDLINE | ID: mdl-9814689

ABSTRACT

The various phospholipid classes that comprise mammalian cell membranes are distributed over both leaflets of the bilayer in a non-random fashion. While a specific and ATP-dependent transporter is responsible for rapid inward movement of aminophospholipids, its inhibition does not lead to spontaneous redistribution of lipids. Conditions of cellular activation which are accompanied with increased levels of intracellular Ca2+ may cause a collapse of lipid asymmetry by switching on an ATP-independently operating scramblase, which accelerates bidirectional movement of all phospholipid classes. The most prominent change in transmembrane lipid distribution is surface exposure of phosphatidylserine (PS), the more so since conditions which activate scramblase in most if not all cases lead to inhibition of aminophospholipid translocase activity, which will prevent PS from being pumped back to the inner leaflet of the membrane. Surface-exposed PS serves at least two important physiological functions: it promotes blood coagulation and offers a recognition signal for clearance by macrophages and other cells of the reticuloendothelial system. As such, PS exposure may form an important early event in the process of apoptosis to ensure rapid removal of these cells in order to avoid release of their inflammatory contents. Defective regulation of transbilayer lipid distribution may result in clinical manifestations such as in the Scott syndrome, a bleeding disorder caused by an impaired scramblase activity. Conversely, excessive PS exposure may lead to thrombosis or may explain formation of so-called antiphospholipid antibodies as occurring in patients with antiphospholipid syndrome.


Subject(s)
Membrane Lipids/metabolism , Phospholipid Transfer Proteins , Phospholipids/metabolism , Animals , Anions , Antibodies, Antiphospholipid/immunology , Antibodies, Antiphospholipid/metabolism , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/immunology , Apoptosis , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Blood Coagulation , Calcium Signaling , Carrier Proteins/metabolism , Enzyme Activation , Humans , Lipid Bilayers , Mammals/metabolism , Membrane Proteins/metabolism , Mononuclear Phagocyte System/physiology , Phosphatidylserines/metabolism
20.
Biochim Biophys Acta ; 1376(3): 433-53, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9805008

ABSTRACT

It has long been appreciated that lipids, particularly anionic phospholipids, promote blood coagulation. The last two decades have seen an increasing insight into the kinetic and mechanistic aspects regarding the mode of action of phospholipids in blood coagulation. This essay attempts to review these developments with particular emphasis on the structure of lipid-binding domains of blood coagulation proteins, and the variable effect of phospholipid composition on the interaction with these proteins. Some examples are discussed of how lipid membranes direct the pathway of enzymatic conversions in blood coagulation complexes, also illustrating that the membrane lipid surface is more than an inert platform for the assembly of coagulation factors. Finally, the controlled exposure of procoagulant lipid on the surface of blood cells is shortly reviewed, and an example is discussed of how interference with lipid-protein interactions in blood coagulation may result in pathological phenomena.


Subject(s)
Blood Coagulation Factors/chemistry , Blood Coagulation , Membrane Lipids/chemistry , Animals , Binding Sites , Humans , Membrane Fluidity , Phospholipids/chemistry , Thrombomodulin/chemistry , Thromboplastin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...