Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Blood Coagul Fibrinolysis ; 26(5): 545-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25811451

ABSTRACT

Thrombin (FIIa) is the key enzyme in haemostasis and acts on several substrates involved in clot formation, platelet activation and feed-back regulation of its own formation. During activation of blood coagulation, FIIa is formed by proteolytic cleavage of prothrombin (FII). In the production of recombinant human FII (rhFII), a key question is whether the thrombin formed has the same properties as endogenous thrombin. We have investigated whether FIIa formed from rhFII and plasma-derived human FII (pdhFII) have the same enzymatic and haemostatic properties against a number of substrates and the same haemostatic capacity in plasma, whole blood and on platelets. Pure FIIa was isolated from rhFII and pdhFII cleaved by recombinant ecarin, and analytical methods were developed to compare the activity of FIIa against different substrates. FIIa derived from rhFII and pdhFII were found to have very similar properties in activating FVIII, FXIII, protein C, platelet aggregation and plasma or whole blood coagulation. Further, the same turnover for S-2366 was found with similar KM. However, activation of FV with rhFIIa was approximately 25% more effective than with pdhFIIa and heparin-enhanced inhibition of rhFIIa by antithrombin was significantly more efficient compared with pdhFIIa with 10% higher inhibition both at steady state and at initial rate conditions. Although differences between the two FIIa preparations using ecarin cleavage were observed, FIIa derived from rhFII administered to human would likely be very similar in activity and function as FIIa formed from endogenous FII.


Subject(s)
Blood Coagulation/physiology , Prothrombin/metabolism , Thrombin/metabolism , Hemostasis , Humans , Protein C/pharmacology
2.
Blood Coagul Fibrinolysis ; 25(8): 851-5, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25004021

ABSTRACT

In the activated partial thromboplastin time (APTT) assay, a variety of nonphysiological reagents is used to induce contact activation. The sensitivity of the APTT response for different thrombin inhibitors has previously been found to be dependent on the used reagent. Recently, infusion of prothrombin (FII) has been used in in-vivo coagulopathy models and its effect has been analyzed in different assays. Therefore, we investigated whether the FII plasma concentration might affect APTT using different commercial reagents, applying both turbidimetry and viscometry. We compared both plasma-derived human FII (pd-hFII) and recombinant human FII (r-hFII). Similar results were found for pd-hFII and r-hFII with different APTT reagents. As expected, no effect on APTT was found by increasing the plasma concentration of FII using APTT reagents consisting of ellagic acid (Actin FS or Actin). Although with Pathromtin SL, consisting of SiO2, only a slight increase was found, with most other commercial APTT reagents, consisting of SiO2 or kaolin, APTT dose-dependently increased by increasing concentration of FII. Therefore, both Pathromtin SL and Actin FS were used to compare r-hFII and pd-hFII by determining the KM at 37C using FII-depleted plasma, providing values of 6 ± 0.3 nmol/l FII for both. Thus, at normal plasma concentrations of FII, the maximal initial thrombin generation rate should be reached and no effect on the coagulation time is expected at higher FII concentrations. To completely avoid the paradoxical effect in the APTT assay at FII concentrations higher than normal, Actin or Actin FS is the preferable reagent.


Subject(s)
Ellagic Acid/chemistry , Indicators and Reagents/chemistry , Plasma/chemistry , Prothrombin/chemistry , Silicon Dioxide/chemistry , Blood Coagulation , Humans , Kinetics , Nephelometry and Turbidimetry , Partial Thromboplastin Time , Recombinant Proteins/chemistry , Thrombelastography
3.
Thromb Res ; 132(2): 248-55, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23830061

ABSTRACT

INTRODUCTION: Oral treatment is lacking for haemophilia, the rare bleeding disorders, and some severe forms of von Willebrand's disease. We have serendipitously identified a small molecule procoagulant compound (AZ10047130). This publication describes some characteristics of AZ10047130 and a systematic search for novel hits using a, human plasma-based, high-throughput screening (HTS) assay. MATERIAL AND METHODS: Coagulation, thrombin generation, chromogenic assays and surface plasmon resonance (SPR) experiments were used to characterise AZ10047130. A 1536-well formatted human plasma coagulation assay for HTS was developed. RESULTS: In the plasma clot assay (re-calcified plasma with low tissue factor) AZ10047130 shortened time to coagulation with an EC50 value of 3.9 µM (assay concentration). AZ10047130 was similarly effective in immunodepleted human and haemophilia A plasmas. SPR and chromogenic substrate experiments indicated that AZ10047130 binds to the heparin binding site of several coagulation factors. The HTS screened in excess of one million compounds. It generated some hits belonging to the same pharmacophore as AZ10047130 but also some entirely novel hits. CONCLUSION: These novel small molecule procoagulant compounds may serve as templates for discovery of oral procoagulant drugs.


Subject(s)
Benzofurans/pharmacology , Blood Chemical Analysis/methods , Blood Coagulation Factors/pharmacology , Blood Coagulation/drug effects , High-Throughput Screening Assays/methods , Sulfonamides/pharmacology , Benzofurans/chemistry , Blood Coagulation Factors/chemistry , Hemophilia A/blood , Hemophilia A/drug therapy , Hemostasis/drug effects , Humans , Sulfonamides/chemistry , Thrombin/biosynthesis
4.
Biochemistry ; 52(4): 613-26, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23290007

ABSTRACT

To improve our understanding of drug-target interactions, we explored the effect of introducing substituted amine residues with increased chain length in the P3 residue of the thrombin inhibitor melagatran. Inhibition, kinetic, and thermodynamic data obtained via stopped-flow spectroscopy (SF), isothermal microcalorimetry (ITC), and surface plasmon resonance (SPR) biosensor analysis were interpreted with the help of X-ray crystal structures of the enzyme-inhibitor complexes. The association rate became faster when the lipophilicity of the inhibitors was increased. This was coupled to an increased enthalpic component and a corresponding decreased entropic component. The dissociation rates were reduced with an increase in chain length, with only a smaller increase and a decrease in the enthalpic and entropic components, respectively. Overall, the affinity increased with an increase in chain length, with similar changes in the enthalpic and entropic components. ITC analysis confirmed the equilibrium data from SPR analysis, showing that the interaction of melagatran was the most enthalpy-driven interaction. Structural analysis of the thrombin-inhibitor complex showed that the orientation of the P1 and P2 parts of the molecules was very similar, but that there were significant differences in the interaction between the terminal part of the P3 side chain and the binding pocket. A combination of charge repulsion, H-bonds, and hydrophobic interactions could be used to explain the observed kinetic and thermodynamic profiles for the ligands. In conclusion, changes in the structure of a lead compound can have significant effects on its interaction with the target that translate directly into kinetic and thermodynamic effects. In contrast to what may be intuitively expected, hydrogen bond formation and breakage are not necessarily reflected in enthalpy gains and losses, respectively.


Subject(s)
Antithrombins/chemistry , Azetidines/chemistry , Benzylamines/chemistry , Thrombin/chemistry , Catalytic Domain , Crystallography, X-Ray , Drug Discovery , Hirudins/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Protein Binding , Surface Properties , Thermodynamics , Thrombin/antagonists & inhibitors
5.
J Biol Chem ; 288(2): 873-85, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23155046

ABSTRACT

A novel class of small molecule inhibitors for plasminogen activator inhibitor type 1 (PAI-1), represented by AZ3976, was identified in a high throughput screening campaign. AZ3976 displayed an IC(50) value of 26 µm in an enzymatic chromogenic assay. In a plasma clot lysis assay, the compound was active with an IC(50) of 16 µm. Surprisingly, AZ3976 did not bind to active PAI-1 but bound to latent PAI-1 with a K(D) of 0.29 µm at 35 °C and a binding stoichiometry of 0.94, as measured by isothermal calorimetry. Reversible binding was confirmed by surface plasmon resonance direct binding experiments. The x-ray structure of AZ3976 in complex with latent PAI-1 was determined at 2.4 Å resolution. The inhibitor was bound in the flexible joint region with the entrance to the cavity located between α-helix D and ß-strand 2A. A set of surface plasmon resonance experiments revealed that AZ3976 inhibited PAI-1 by enhancing the latency transition of active PAI-1. Because AZ3976 only had measurable affinity for latent PAI-1, we propose that its mechanism of inhibition is based on binding to a small fraction in equilibrium with active PAI-1, a latent-like prelatent form, from which latent PAI-1 is then generated more rapidly. This mode of action, with induced accelerated latency transition of active PAI-1 may, together with supporting x-ray data, provide improved opportunities for small molecule drug design in the hunt for therapeutically useful PAI-1 inhibitors.


Subject(s)
Azetidines/pharmacology , Plasminogen Activator Inhibitor 1/chemistry , Pyrimidinones/pharmacology , Animals , Azetidines/chemistry , CHO Cells , Calorimetry , Cricetinae , Cricetulus , Humans , Models, Molecular , Protein Conformation , Pyrimidinones/chemistry , Rats , Surface Plasmon Resonance , Thermodynamics
6.
Protein J ; 31(5): 353-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22528138

ABSTRACT

The snake venom protease ecarin from Echis carinatus was expressed in stable transfected CHO-S cells grown in animal component free cell culture medium. Recombinant ecarin (r-ecarin) was secreted from the suspension adapted Chinese Hamster Ovary (CHO-S) host cells as a pro-protein and activation to the mature form of r-ecarin occurred spontaneously during continued incubation of the cell culture at 37 °C after death of the host cells. Maximal ecarin activity was reached 7 days or more after cell culture viability had dropped to zero. The best producing CHO-S clone obtained produced up to 7,000 EU ecarin/litre in lab scale shaker cultures. The conversion of different concentrations of both prothrombin and prethrombin-2 as substrates for native and r-ecarin were examined with a chromogenic thrombin substrate. At low concentrations both these proteins were converted into thrombin by the two ecarin preparations with comparable rates. However, with prothrombin concentrations above 250 nM r-ecarin apparently had a two times higher turnover than native ecarin, consistent with the observed rapid complete conversion of prothrombin into thrombin by r-ecarin. With r-ecarin a K (m) value of 0.4 µM prethrombin-2 was determined but only a rough estimate could be made of the K (m) for prothrombin of 0.9 µM. In conclusion, r-ecarin was identified as a promising candidate for replacement of native ecarin in assays utilizing conversion of prothrombin to thrombin.


Subject(s)
Endopeptidases/genetics , Fibrinolytic Agents/metabolism , Viper Venoms/genetics , Viperidae/metabolism , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Endopeptidases/metabolism , Enzyme Activation , Gene Expression , Humans , Prothrombin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Viper Venoms/metabolism , Viperidae/genetics
7.
PLoS One ; 6(11): e26762, 2011.
Article in English | MEDLINE | ID: mdl-22069469

ABSTRACT

The vascular fibrinolytic system is crucial for spontaneous lysis of blood clots. Plasminogen activator inhibitor 1 (PAI-1), the principal inhibitor of the key fibrinolytic enzyme tissue-type plasminogen activator (tPA), is present in platelets at high concentrations. However, the majority of PAI-1 stored in platelets has been considered to be inactive. Our recent finding (Brogren H, et al. Blood 2004) that PAI-1 de novo synthesized in platelets remained active for over 24 h, suggested that PAI-1 stored in the α-granules might be active to a larger extent than previously reported. To re-evaluate this issue, we performed experiments where the fraction of active PAI-1 was estimated by analyzing the tPA-PAI-1 complex formation. In these experiments platelets were lysed with Triton X-100 in the presence of serial dilutions of tPA and subsequently the tPA-PAI-1 complex was evaluated by Western blot. Also, using a non-immunologic assay, tPA was labeled with (125)I, and (125)I-tPA and (125)I-tPA-PAI-1 was quantified by scintigraphy. Interestingly, both methods demonstrated that the majority (>50%) of platelet PAI-1 is active. Further analyses suggested that pre-analytical procedures used in previous studies (sonication or freezing/thawing) may have substantially reduced the activity of platelet PAI-1, which has lead to an underestimation of the proportion of active PAI-1. Our in vitro results are more compatible with the role of PAI-1 in clot stabilization as demonstrated in physiological and pathophysiological studies.


Subject(s)
Blood Platelets/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Platelet Activation/physiology , Tissue Plasminogen Activator/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Fibrinolysis , Humans , Iodine Radioisotopes , Octoxynol , Platelet Count
8.
Anal Chem ; 82(20): 8374-6, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20853853

ABSTRACT

Plasminogen is a precursor to the fibrinolytic enzyme plasmin and is known to undergo large conformational changes when subjected to low molecular lysine analogues such as tranexamic acid (TA) or ε-amino-n-caproic acid (EACA). Here, we demonstrate how well-controlled surface immobilization of biotinylated plasminogen allows for monitoring of the interaction between TA and EACA with plasminogen. The interaction was studied by the quartz crystal microbalance with dissipation monitoring (QCM-D) technique as well as by surface plasmon resonance (SPR) based sensing. QCM-D measures changes in acoustically coupled mass (by detection of changes in the resonance frequency of the crystal, Δf) and is sensitive to changes in mass adsorbed on the sensor surface including how liquid medium is associated with this material. Through the dissipation factor (i.e., changes in the energy dissipation of the crystal oscillation, ΔD), QCM-D is also sensitive to the viscoelastic properties of material adsorbed to the sensor surface. Upon binding of TA or EACA, changes in the plasminogen structure were recorded as distinct, although small, ΔD responses which were used to determine affinity constants. By comparing native and truncated plasminogen, we conclude that the observed dissipation shifts were caused by conformational changes in the proteins leading to changes in the viscoelastic properties of the protein layer on the surface. These results demonstrate a novel application of the QCM-D technique, paving the way for a whole new approach to screening of this target for novel lead structures.


Subject(s)
Elasticity , Plasminogen/analysis , Quartz Crystal Microbalance Techniques/methods , Molecular Weight , Plasminogen/chemistry , Protein Conformation , Surface Plasmon Resonance , Viscosity
9.
Thromb Haemost ; 101(6): 1051-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19492147

ABSTRACT

AZD0837 is in development as a new oral anticoagulant for use in thromboembolic disorders. In vivo, AZD0837 is converted to AR-H067637, a selective and reversible direct thrombin inhibitor. Established biochemical methods were used to assess and measure the biochemical and pharmacological properties of AR-H067637. Both direct Biacore binding studies of AR-H067637 with immobilised alpha-thrombin and inhibition studies using pre-steady state kinetics with thrombin in the fluid phase confirmed that AR-H067637 is a rapid-binding, reversible and potent (inhibition constant K(i) = 2-4 nM), competitive inhibitor of thrombin, as well as of thrombin bound to fibrin (clot-bound thrombin) or to thrombomodulin. The total amount of free thrombin generated in platelet-poor clotting plasma was inhibited concentration-dependently by AR-H067637, with a concentration giving half maximal inhibition (IC(50)) of 0.6 microM. Moreover, AR-H067637 is, with the exception of trypsin, a selective inhibitor for thrombin without inhibiting other serine proteases involved in haemostasis. Furthermore, no anticoagulant effect of the prodrug was found. AR-H067637 prolonged the clotting time concentration-dependently in a range of plasma coagulation assays including activated partial thromboplastin time, prothrombin time, prothrombinase-induced clotting time, thrombin time and ecarin clotting time. The two latter assays were found to be most sensitive for assessing the anticoagulant effect of AR-H067637 (plasma IC(50) 93 and 220 nM, respectively). AR-H067637 also inhibited thrombin-induced platelet activation (by glycoprotein IIb/IIIa exposure, IC(50) 8.4 nM) and aggregation (IC(50) 0.9 nM). In conclusion, AR-H067637 is a selective, reversible, competitive inhibitor of alpha-thrombin, with a predictable anticoagulant effect demonstrated in plasma coagulation assays.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Blood Platelets/enzymology , Platelet Activation/drug effects , Thrombin/antagonists & inhibitors , Amidines/pharmacokinetics , Amidines/pharmacology , Azetidines/pharmacokinetics , Azetidines/pharmacology , Binding, Competitive , Blood Coagulation Factors/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Inhibitory Concentration 50 , Male , Protein Binding , Whole Blood Coagulation Time
10.
J Biomater Sci Polym Ed ; 20(1): 133-40, 2009.
Article in English | MEDLINE | ID: mdl-19105905

ABSTRACT

We describe a novel method to immobilize recombinant human tissue factor (rhTF) reconstituted in phospholipid vesicles. The rhTF vesicles were immobilized in a multilayer vesicle structure using cholesterol-DNA tethers spontaneously inserted into the lipid membrane. The properties of the rhTF vesicle surface modification were characterized by surface plasmon resonance biosensor technology. As an application of this surface modification, we investigated its use as a blood coagulation activating surface. The coagulation activating capacity of the surface modification was tested by exposure to human whole blood in a flow chamber. No increase in rhTF levels in the blood was found after passage through the flow chamber, indicating that the rhTF surface modification was stable. Thrombin-antithrombin (TAT) and prothrombin fragment (PF) 1 + 2 levels increased after exposure to the surface, and decreased in a concentration-dependent way upon addition of melagatran (a direct thrombin inhibitor), i.e., coagulation activity triggered by rhTF could be suppressed by anticoagulation. The results with this new thrombogenic surface are promising, and will be further developed into a useful tool for coagulation related investigations, e.g., characterization of anticoagulants and biomaterials.


Subject(s)
Blood Coagulation/drug effects , Phospholipids/chemistry , Thromboplastin/chemistry , Thromboplastin/pharmacology , Biological Assay/instrumentation , Biological Assay/methods , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Surface Properties , Thromboplastin/genetics
11.
J Med Chem ; 51(12): 3583-7, 2008 Jun 26.
Article in English | MEDLINE | ID: mdl-18510371

ABSTRACT

Bioassay-guided fractionation of a CH2Cl2/MeOH extract of the sponge Suberea clavata using the serine protease factor XIa to detect antithrombotic activity led to the isolation of the new marine natural products, clavatadines A and B. Clavatadines A and B inhibited factor XIa with IC50's of 1.3 and 27 microM, respectively. A crystal structure of protein-inhibitor (clavatadine A) complex was obtained and revealed interesting selective binding and irreversible inhibition of factor XIa. The cocrystal structure provides guidance for the design and synthesis of future factor XIa inhibitors as antithrombotic agents.


Subject(s)
Biological Products/isolation & purification , Factor Xa Inhibitors , Fibrinolytic Agents/isolation & purification , Guanidines/isolation & purification , Phenylacetates/isolation & purification , Porifera/chemistry , Animals , Biological Products/chemistry , Chemical Fractionation , Crystallography, X-Ray , Factor Xa/chemistry , Fibrinolytic Agents/chemistry , Guanidines/chemistry , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Phenylacetates/chemistry
12.
Thromb Res ; 122(2): 271-81, 2008.
Article in English | MEDLINE | ID: mdl-18508114

ABSTRACT

The main inhibitor of intravascular fibrinolysis is plasminogen activator inhibitor 1 (PAI-1) which binds to and irreversibly inhibits tissue plasminogen activator (tPA). PAI-1 is present in blood, both in platelets and in plasma, and PAI-1 levels are associated with risk of atherothrombosis. Several tissues express PAI-1 but the source of plasma PAI-1 is not known. We recently found that platelets can de novo synthesize PAI-1 and the amount synthesized in vitro in 24 hours is 35-fold higher than required to maintain normal plasma levels. Recombinant human PAI-1 expressed in different cell types or secreted naturally by human cell lines, exhibit heterogeneous glycosylation patterns. The aim of this study was to investigate the hypothesis that platelets might be the source of plasma PAI-1 and that the cellular source of PAI-1 can be determined by its tissue-specific glycosylation pattern. PAI-1 was isolated from platelets, macrophages, endothelial cells, adipose tissue, as well as plasma from lean and obese subjects. The glycosylation was analyzed by nanoLC-MS/MS. PAI-1 isolated from cell lysates and conditioned media from macrophages, endothelial cells, and adipose tissue expressed heterogeneous glycosylation patterns. By contrast, no glycans were detected on PAI-1 isolated from plasma or platelets from healthy lean individuals. Hence, our data suggest that platelets may be the main source of plasma PAI-1 in lean individuals. Interestingly, plasma PAI-1 from obese subjects had a glycan composition similar to that of adipose tissue suggesting that obese subjects with elevated PAI-1 levels may have a major contribution from other tissues.


Subject(s)
Plasminogen Activator Inhibitor 1/chemistry , Adipose Tissue/metabolism , Adult , Blood Platelets/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Fibrinolysis , Glycosylation , Hepatocytes/cytology , Humans , Macrophages/cytology , Macrophages/metabolism , Middle Aged , Models, Biological , Monocytes/cytology , Recombinant Proteins/chemistry
13.
J Med Chem ; 50(24): 5903-11, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-17985861

ABSTRACT

Fragment-based lead generation was applied to find novel small-molecule inhibitors of beta-secretase (BACE-1), a key target for the treatment of Alzheimer's disease. Fragment hits coming from a 1D NMR screen were characterized by BIAcore, and the most promising compounds were soaked into protein crystals to help the rational design of more potent hit analogues. Problems arising due to our inability to grow BACE-1 crystals at the biologically relevant pH at which the screen was run were overcome by using endothiapepsin as a surrogate aspartyl protease. Among others, we identified 6-substituted isocytosines as a novel warhead against BACE-1, and the accompanying paper in this journal describes how these were optimized to a lead series of nanomolar inhibitors.1.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/chemistry , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/chemistry , Cytosine/analogs & derivatives , Drug Design , Enzyme Inhibitors/chemistry , Amyloid Precursor Protein Secretases/isolation & purification , Aspartic Acid Endopeptidases/isolation & purification , Cell Line , Crystallography, X-Ray , Cytosine/chemistry , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Structure-Activity Relationship
14.
J Exp Med ; 204(8): 1741-8, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17635954

ABSTRACT

Upon cerebral hypoxia-ischemia (HI), apoptosis-inducing factor (AIF) can move from mitochondria to nuclei, participate in chromatinolysis, and contribute to the execution of cell death. Previous work (Cande, C., N. Vahsen, I. Kouranti, E. Schmitt, E. Daugas, C. Spahr, J. Luban, R.T. Kroemer, F. Giordanetto, C. Garrido, et al. 2004. Oncogene. 23:1514-1521) performed in vitro suggests that AIF must interact with cyclophilin A (CypA) to form a proapoptotic DNA degradation complex. We addressed the question as to whether elimination of CypA may afford neuroprotection in vivo. 9-d-old wild-type (WT), CypA(+/-), or CypA(-/-) mice were subjected to unilateral cerebral HI. The infarct volume after HI was reduced by 47% (P = 0.0089) in CypA(-/-) mice compared with their WT littermates. Importantly, CypA(-/-) neurons failed to manifest the HI-induced nuclear translocation of AIF that was observed in WT neurons. Conversely, CypA accumulated within the nuclei of damaged neurons after HI, and this nuclear translocation of CypA was suppressed in AIF-deficient harlequin mice. Immunoprecipitation of AIF revealed coprecipitation of CypA, but only in injured, ischemic tissue. Surface plasmon resonance revealed direct molecular interactions between recombinant AIF and CypA. These data indicate that the lethal translocation of AIF to the nucleus requires interaction with CypA, suggesting a model in which two proteins that normally reside in separate cytoplasmic compartments acquire novel properties when moving together to the nucleus.


Subject(s)
Active Transport, Cell Nucleus , Apoptosis Inducing Factor/metabolism , Cyclophilin A/physiology , Hypoxia-Ischemia, Brain/metabolism , Neurons/metabolism , Animals , Apoptosis , Brain/pathology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Mice , Mice, Transgenic , Models, Biological , Oxidative Stress , Protein Binding
15.
Anal Biochem ; 362(1): 98-107, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17239338

ABSTRACT

The dynamics of the binding of human coagulation factor Xa (FXa) and prothrombin to small unilamellar vesicles (25% phosphatidylserine, 75% phosphatidylcholine) were compared and quantified by Biacore, using two immobilization techniques. The vesicles were either tagged with different molar ratios of cholesterol-DNA and attached on Au chips or fused directly on L1 chips. The diameter in solution was 145 nm, but the more DNA tags/vesicle the more compressed the immobilized vesicles became; with 30 DNA tags the calculated thickness was 88 nm and with 1 DNA tag it was 138 nm. In both models the affinity for the vesicles was higher for the activated coagulation factors than for the corresponding zymogens. FXa and prothrombin had the highest affinities. The affinity was dependent on the vesicle preparation since overall K(D) values were up to 10 times lower for N(2)-dried than for vacuum-dried phospholipids, although with apparently fewer binding sites. However, compression of the vesicles had no effect on the K(D). In contrast, the rate constants were dependent on the number of DNA tags; thus deformation of the vesicles was observed. The k(a) and k(d) for FXa were similar for vesicles attached with 30 DNA tags or fused on the L1 chip but higher with fewer tags and approximately 10 times higher if attached with 1 tag. Thus for controlled kinetic studies immobilized DNA-tagged vesicles should be used.


Subject(s)
Blood Coagulation Factors/chemistry , Cytoplasmic Vesicles/chemistry , Phospholipids/chemistry , Surface Plasmon Resonance/methods , Blood Coagulation Factors/metabolism , Cytoplasmic Vesicles/metabolism , DNA/chemistry , Humans , Kinetics , Phospholipids/metabolism , Protein Binding , Surface Plasmon Resonance/instrumentation
16.
Thromb Haemost ; 90(2): 206-17, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12888867

ABSTRACT

The serpin plasminogen activator inhibitor-1 (PAI-1) is a potential target for anti-thrombotic and anti-cancer therapy. PAI-1 has 3 potential sites for N-linked glycosylation. We demonstrate here that PAI-1 expressed recombinantly or naturally by human cell lines display a heterogeneous glycosylation pattern of the sites at N209 and N265, while that at N329 is not utilised. The IC(50)-values for inactivation of PAI-1 by 4 monoclonal antibodies differed strongly between glycosylated PAI-1 and non-glycosylated PAI-1 expressed in E. coli. For 3 antibodies, an overlap of the epitopes with the glycosylation sites could be excluded as explanation for the differential reactivity. The latency transition of non-glycosylated, but not of glycosylated PAI-1, was strongly accelerated by a non-ionic detergent. The different biochemical properties of glycosylated and non-glycosylated PAI-1 depended specifically on glycosylation of either one or the other of the utilised sites. The PAI-1-binding protein vitronectin reversed the changes associated with the lack of glycosylation at one of the sites. Our results stress the importance of the source of PAI-1 when studying the mechanisms of action of PAI-1-inactivating compounds of potential clinical importance.


Subject(s)
Glycosylation , Plasminogen Activator Inhibitor 1/metabolism , Antibodies, Monoclonal/immunology , Cell Line , Detergents/pharmacology , Genetic Variation , Half-Life , Humans , Molecular Structure , Octoxynol/pharmacology , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/immunology , Vitronectin/pharmacology
17.
J Autoimmun ; 20(4): 345-50, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12791320

ABSTRACT

The removal of beta(1)-adrenergic receptor (beta(1)AR) autoantibodies by immunoadsorption (IA) has been proposed as a potential mechanism for the improvement of the left ventricular function in dilated cardiomyopathy (DCM). In the present study, the possible association between removal of the autoantibodies against the human beta(1)AR with the hemodynamic improvement induced by IA was investigated.IA was performed in 22 DCM patients (n=22; NYHA III-IV, EF<30%, stable medication). The beta(1)AR autoantibodies from column eluents (CE) were detected by enzyme-linked immunosorbent assay (ELISA) and BIAcore methods. CE of 32% (7/22) of the patients was found to be antibody-positive with ELISA or BIAcore. In addition, a bioassay system was also used for the detection of this autoantibody. Seventy-three percent (16/22) of the patients were found to be antibody-positive by this method. However, independent of the beta(1)AR antibody detection method, both antibody-positive and antibody-negative groups showed similar acute and prolonged hemodynamic improvements during IA therapy. Furthermore, antibody-positive and -negative groups received a comparable improvement of left ventricular ejection fraction. These results suggest that different mechanisms are involved in the hemodynamic improvement induced by IA. The beneficial hemodynamic effects induced by IA are not directly associated with the removal of beta(1)AR autoantibodies.


Subject(s)
Autoantibodies/isolation & purification , Cardiomyopathy, Dilated/therapy , Receptors, Adrenergic, beta-1/immunology , Autoantibodies/blood , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/physiopathology , Case-Control Studies , Hemodynamics , Humans , Immunosorbent Techniques , Immunotherapy , Male , Middle Aged , Ventricular Function, Left
18.
Anal Biochem ; 300(2): 152-62, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11779106

ABSTRACT

The binding of a series of low-molecular-mass, active-site-directed thrombin inhibitors (399-575 Da) to human alpha-thrombin was investigated by surface plasmon resonance technology (BIACORE), stopped-flow spectrophotometry, and isothermal titration microcalorimetry (ITC). The equilibrium constants K(D) (nM to microM range) at 25 degrees C obtained from the BIACORE analysis correlated well with the inhibition constants K(i) in a chromogenic inhibition assay. The interactions between thrombin and three potent inhibitors, melagatran, inogatran, and CH-248, were further investigated at temperatures between 278 and 310K. A one-to-one binding stoichiometry found with ITC was supported by BIACORE data. K(i) and K(D) values increased with the temperature, mainly due to higher values for dissociation rate constants. The changes in enthalpy, DeltaH, and entropy, DeltaS, determined from the linear van't Hoff plots (R coefficient > 0.99), were linearly correlated by chemical compensation. Both techniques indicated clear differences in DeltaS for the three inhibitors, with a strong correlation to the number of rotational bonds. Immobilization of thrombin increased the binding stability at higher temperature and reduced the DeltaH by 20 kJ mol(-1). DeltaH values obtained from the inhibition kinetics and BIACORE were thus not identical, but correlated well with ITC data obtained at 37 degrees C. The two thermodynamic techniques allowed further differentiation between compounds of similar affinity; furthermore, kinetic analysis, hence analysis of the transition state, is complementary to ITC. A direct BIACORE binding assay might be a useful alternative to more elaborate inhibition studies.


Subject(s)
Biosensing Techniques/methods , Calorimetry/methods , Spectrophotometry/methods , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Binding Sites , Buffers , Dimethyl Sulfoxide , HEPES , Humans , Kinetics , Molecular Structure , Protein Binding , Temperature , Thermodynamics , Thrombin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...