Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Haemophilia ; 28(4): 568-577, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35467059

ABSTRACT

INTRODUCTION: Immunogenicity causing development of anti-drug antibodies (ADAs) are major challenges in the treatment of haemophilia, as well as other diseases where proteins are used for treatment. Furthermore, it is a complication for preclinical testing of such therapies in animal models. AIM: To investigate if the immunosuppressive drug CTLA4 immunoglobulin (CTLA4-Ig) can induce tolerance in haemophilia A (HA) rats receiving recombinant human coagulation factor VIII (rhFVIII) treatment. METHODS: Two different prophylactic rhFVIII compounds were given intravenously to HA rats for 4 weeks. Both rhFVIII compounds were co-administered with commercially available CTLA4-Ig or human IgG subclass 4 (hIgG4) as control, and blood samples were collected. To functionally test if pharmacological efficacy was retained, rats were subjected to a bleeding experiment under anaesthesia at end of study. RESULTS: The mean inhibitory level after 4 weeks in rats receiving rhFVIII and hIgG4 was 85.7 BU for octocog alfa and 37.4 BU for rurioctocog alfa pegol. In contrast, co-administration with CTLA4-Ig during rhFVIII therapy prevented the formation of ADAs (both binding and inhibitory) in 14/14 rats receiving octocog alfa and in 7/7 rats receiving rurioctocog alfa pegol. Moreover, we were able to show that the pharmacological efficacy of rhFVIII was preserved. CONCLUSION: In a rat model with spontaneous bleeding, co-administration of CTLA4-Ig with rhFVIII prevented antibody formation. No FVIII antibodies were detected, demonstrating that CTLA4-Ig co-administration can be applicable as a method to prevent immunogenicity, when evaluating human proteins in preclinical systems permitting continuous pharmacokinetic and pharmacodynamic assessment.


Subject(s)
Hemophilia A , Abatacept/pharmacology , Abatacept/therapeutic use , Animals , Antibodies, Neutralizing , Antibody Formation , CTLA-4 Antigen , Factor VIII , Hemophilia A/drug therapy , Hemophilia A/prevention & control , Hemorrhage/drug therapy , Humans , Rats , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
2.
J Thromb Haemost ; 20(6): 1312-1324, 2022 06.
Article in English | MEDLINE | ID: mdl-35191180

ABSTRACT

BACKGROUND: Mim8 is a novel, next-generation factor VIIIa mimetic in development for subcutaneous prophylactic treatment of patients with hemophilia A with and without inhibitors. In vitro and in vivo models indicate that Mim8 has a distinct hemostatic potential. OBJECTIVES: To test the nonclinical safety and pharmacodynamics of Mim8. METHODS: The Mim8 nonclinical safety program in cynomolgus monkeys consisted of three studies of 4-26 weeks in duration with Mim8 doses ranging from 0.3-60 mg/kg/week intravenously or subcutaneously. After sacrifice, macroscopic and microscopic pathological examinations were performed. RESULTS: Mim8 was well tolerated with no noteworthy clinical observations. No signs of excessive coagulation or pathological macroscopic or microscopic findings were observed at doses 0.3-3 mg/kg/week subcutaneous. Thrombosis-related findings were detected during histopathological examination in a small proportion of animals (16%) receiving doses ranging 6-20 mg/kg/week. Dose-dependent increases in factor X (FX) and factor IX (FIX) concentrations were observed. Shortening of activated partial thromboplastin time (APTT) and increased thrombin generation under ex vivo hemophilia A-like conditions were observed at all Mim8 dose levels. CONCLUSIONS: Thrombosis-related findings observed at doses above 6 mg/kg/week Mim8 may have been exaggerated pharmacological reactions to a procoagulant compound in normocoagulant animals. Increases in FX and FIX concentrations could be because of a half-life prolongation due to binding to Mim8, but were limited at clinically relevant exposure levels. Subcutaneous administration of up to 3 mg/kg/week (several fold greater than expected clinical exposure) for 26 weeks resulted in relevant pharmacodynamic effects, observed in thrombin generation and APTT, with no signs of thrombi or excessive coagulation activation.


Subject(s)
Hemophilia A , Thrombosis , Animals , Factor IX/metabolism , Factor X , Humans , Macaca fascicularis/metabolism , Thrombin/metabolism , Thrombosis/prevention & control
3.
Toxicol Appl Pharmacol ; 404: 115152, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32726590

ABSTRACT

Energy metabolism and reproduction are closely linked and reciprocally regulated. The detrimental effect of underweight on reproduction complicates the safety evaluation of anti-obesity drugs, making it challenging to distinguish pathological changes mediated through the intended drug-induced weight loss from direct drug effects on reproductive organs. Four-weeks dosing of normal weight Sprague Dawley rats with a glucagon-like peptide 1 (GLP-1)/glucagon receptor co-agonist induced a robust weight loss, accompanied by histological findings in prostate, seminal vesicles, mammary glands, uterus/cervix and vagina. Characterization of the hypothalamus-pituitary-gonadal (HPG) axis in male rats revealed reduced hypothalamic Kiss1 mRNA levels and decreased serum luteinizing hormone (LH) and testosterone concentrations following co-agonist dosing. These alterations resemble hypogonadotropic hypogonadism typically seen in adverse energy deprived conditions, like chronic food restriction. Concomitant daily administration of kisspeptin-52 from day 21 to the end of the four-week co-agonist dosing period evoked LH and testosterone responses without normalizing histological findings. This incomplete rescue by kisspeptin-52 may be due to the rather short kisspeptin-52 treatment period combined with a desensitization observed on testosterone responses. Concomitant leptin treatment from day 21 did not reverse co-agonist induced changes in HPG axis activity. Furthermore, a single co-agonist injection in male rats slightly elevated LH levels but left testosterone unperturbed, thereby excluding a direct acute inhibitory effect on the HPG axis. Our data suggest that the reproductive phenotype after repeated co-agonist administration was driven by the intended weight loss, however, we cannot exclude a direct organ related effect in chronically treated rats.


Subject(s)
Anti-Obesity Agents/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Kisspeptins/pharmacology , Testis/drug effects , Animals , Kisspeptins/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Testis/metabolism , Thinness , Weight Loss/drug effects
4.
J Contam Hydrol ; 226: 103517, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31280034

ABSTRACT

Assessing redox conditions in soil and groundwater is challenging because redox reactions are oxygen sensitive, hence, destructive sampling methods may provide contact with air and influence the redox state. Furthermore, commonly used redox potential sensors provide only point measurements and are prone to error. This paper assesses whether combining electrical resistivity (ER) and self-potential (SP) measurements can allow the mapping of zones affected by anaerobic degradation. We use ER imaging because anaerobic degradation can release iron and manganese ions, which decreases pore water resistivity, and produces gas, which increases resistivity. Also, electrochemical differences between anaerobic and aerobic zones may create an electron flow, forming a self-potential anomaly. In this laboratory study, with four sand tanks with constant water table heights, time-lapse ER and SP mapped changes in electrical/electron flow properties due to organic contaminant (propylene glycol) degradation. Sampled pore water mapped degradation and water chemistry. When iron and manganese oxides were available, degradation reduced resistivity, because of cation release in pore water. When iron and manganese oxides were unavailable, resistivity increased, plausibly from methane production, which reduced water saturation. To bypass the reactions producing methane and release of metallic cations, a metal pipe was installed in the sand tanks between anaerobic and aerobic zones. The degradation creates an electron surplus at the anaerobic degradation site. The metal pipe allowed electron flow from the anaerobic degradation site to the oxygen-rich near surface. The electrical current sent through the metal pipe formed an SP anomaly observable on the surface of the sand tank. Time-lapse ER demonstrates potential for mapping degradation zones under anaerobic conditions. When an electrical conductor bridges the anaerobic zone with the near surface, the electron flow causes an SP anomaly on the surface. However, electrochemical differences between anaerobic and aerobic zones alone produced no SP signal. Despite their limitations, ER and SP are promising tools for monitoring redox sensitive conditions in unsaturated sandy soils but should not be used in isolation.


Subject(s)
Groundwater , Soil Pollutants , Electricity , Oxidation-Reduction , Soil
5.
J Contam Hydrol ; 221: 1-10, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30600103

ABSTRACT

Degradation of organic chemicals in natural soils depends on oxidation-reduction conditions. To protect our groundwater resources we need to understand the degradation processes under anaerobic conditions. Available iron and manganese oxides are used as electron acceptors for anaerobic degradation and are reduced to the dissolved form of metallic cations in pore water. To monitor this process is a challenge, because anaerobic conditions are difficult to sample directly without introducing oxygen. A few studies have shown an impact of iron reduction on spectral induced polarisation (SIP) signature, often associated with bacterial growth. Our objective is to study the impact of iron and manganese oxide dissolution, caused by degradation of an organic compound, with spectral induced polarisation signatures. Twenty-six vertical columns (30 cm high, inner diameter 4.6 cm) were filled with a sand rich in oxides (manganese and iron) with a static water table in the middle. In half of the columns, a 2 cm high contaminated layer was installed just above the water table. As the contaminant degrades, the initial oxygen is consumed and anaerobic conditions form Every three days over a period of one month, spectral induced polarisation (twenty frequencies between 5mHz and 10 kHz) data were collected on six columns: three contaminated replicates and three control replicates. Chemical analysis was done on twenty columns assigned for destructive water sampling, ten contaminated columns and ten control. The results show an increase of the real conductivity associated with the degradation processes, independent of frequency. Compared with the pore water electrical conductivity in the saturated zone, the real conductivity measurement revealed the formation of surface conductivity before iron was released in the pore water. In parallel, we also observed an evolution of the imaginary conductivity in both saturated and unsaturated zones at frequencies below 1 Hz. Overall, the anaerobic reduction of iron and manganese oxide during the organic degradation increased both the conductive and polarisation component of the complex conductivity.


Subject(s)
Iron , Water Pollutants, Chemical , Anaerobiosis , Manganese Compounds , Oxidation-Reduction , Oxides , Solubility
6.
Int J Biochem Cell Biol ; 89: 34-41, 2017 08.
Article in English | MEDLINE | ID: mdl-28558995

ABSTRACT

Low density lipoprotein receptor-related protein 1 (LRP1) is involved in the catabolism of many ligands, including factor VIII (FVIII) and alpha-2-macroglobulin (α2M). Transfer of FVIII to LRP1 is currently believed to be preceded by pre-concentration on the cell surface, by interacting with a so far unidentified component. In the present study, we used confocal microscopy and flow cytometry to compare endocytosis of FVIII and α2M using U87MG cells. The results show that α2M is rapidly internalized and does not compete for LRP1 mediated internalization of FVIII. FVIII endocytosis did not occur in the presence of receptor-associated-protein (RAP), but FVIII remained visible as a striated fluorescent pattern at the cell borders. In the presence of Von Willebrand Factor (VWF), no FVIII was observed on or within the cells, suggesting that VWF blocks interaction with both cell surface and LRP1. The same dual inhibition has previously been observed for FVIII C1 domain directed monoclonal antibody KM33. Elimination of the KM33 epitope by replacing FVIII C1 residues 2091-2095 and 2155-2160 for the homologues from factor V (FV), however, did not impair FVIII endocytosis. These membrane spikes alone were insufficient for cellular uptake, because FV was neither internalized by U87MG cells nor capable of effectively competing for FVIII endocytosis. These results show that FVIII endocytosis is driven by interaction with LRP1, but at the same time involves the spikes in the C1 domain that have been implicated in lipid binding.


Subject(s)
Cell Membrane/metabolism , Endocytosis , Factor VIII/chemistry , Factor VIII/metabolism , Cell Line , Humans , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Models, Molecular , Protein Binding , Protein Domains
7.
Haematologica ; 102(4): 686-694, 2017 04.
Article in English | MEDLINE | ID: mdl-28057741

ABSTRACT

Factor VIII C-domains are believed to have specific functions in cofactor activity and in interactions with von Willebrand factor. We have previously shown that factor VIII is co-targeted with von Willebrand factor to the Weibel-Palade bodies in blood outgrowth endothelial cells, even when factor VIII carries mutations in the light chain that are associated with defective von Willebrand factor binding. In this study, we addressed the contribution of individual factor VIII C-domains in intracellular targeting, von Willebrand factor binding and cofactor activity by factor VIII/V C-domain swapping. Blood outgrowth endothelial cells were transduced with lentivirus encoding factor V, factor VIII or YFP-tagged C-domain chimeras, and examined by confocal microscopy. The same chimeras were produced in HEK293-cells for in vitro characterization and chemical foot-printing by mass spectrometry. In contrast to factor VIII, factor V did not target to Weibel-Palade bodies. The chimeras showed reduced Weibel-Palade body targeting, suggesting that this requires the factor VIII C1-C2 region. The factor VIII/V-C1 chimera did not bind von Willebrand factor and had reduced affinity for activated factor IX, whereas the factor VIII/V-C2 chimera showed a minor reduction in von Willebrand factor binding and normal interaction with activated factor IX. This suggests that mainly the C1-domain carries factor VIII-specific features in assembly with von Willebrand factor and activated factor IX. Foot-printing analysis of the chimeras revealed increased exposure of lysine residues in the A1/C2- and C1/C2-domain interface, suggesting increased C2-domain mobility and disruption of the natural C-domain tandem pair orientation. Apparently, this affects intracellular trafficking, but not extracellular function.


Subject(s)
Factor VIII/metabolism , Factor V/metabolism , Protein Interaction Domains and Motifs , Endothelial Cells/metabolism , Factor V/chemistry , Factor V/genetics , Factor VIII/chemistry , Factor VIII/genetics , Gene Expression , Humans , Intracellular Space/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Transport , Structure-Activity Relationship , von Willebrand Factor/metabolism
8.
Haematologica ; 101(3): 309-18, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26635035

ABSTRACT

It has been proposed that von Willebrand factor might affect factor VIII immunogenicity by reducing factor VIII uptake by antigen presenting cells. Here we investigate the interaction of recombinant von Willebrand factor with immature monocyte-derived dendritic cells using flow cytometry and confocal microscopy. Surprisingly, von Willebrand factor was not internalized by immature dendritic cells, but remained bound to the cell surface. As von Willebrand factor reduces the uptake of factor VIII, we investigated the repertoire of factor VIII presented peptides when in complex with von Willebrand factor. Interestingly, factor VIII-derived peptides were still abundantly presented on major histocompatibility complex class II molecules, even though a reduction of factor VIII uptake by immature dendritic cells was observed. Inspection of peptide profiles from 5 different donors showed that different core factor VIII peptide sequences were presented upon incubation with factor VIII/von Willebrand factor complex when compared to factor VIII alone. No von Willebrand factor peptides were detected when immature dendritic cells were pulsed with different concentrations of von Willebrand factor, confirming lack of von Willebrand factor endocytosis. Several von Willebrand factor derived peptides were recovered when cells were pulsed with von Willebrand factor/factor VIII complex, suggesting that factor VIII promotes endocytosis of small amounts of von Willebrand factor by immature dendritic cells. Taken together, our results establish that von Willebrand factor is poorly internalized by immature dendritic cells. We also show that von Willebrand factor modulates the internalization and presentation of factor VIII-derived peptides on major histocompatibility complex class II.


Subject(s)
Dendritic Cells/immunology , Factor VIII/immunology , HLA-DRB1 Chains/immunology , Peptides/immunology , von Willebrand Factor/immunology , Amino Acid Sequence , Antigen Presentation , Binding Sites , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endocytosis , Factor VIII/metabolism , HLA-DRB1 Chains/metabolism , Humans , Monocytes/cytology , Monocytes/immunology , Peptides/chemistry , Peptides/metabolism , Primary Cell Culture , Protein Binding , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , von Willebrand Factor/metabolism
9.
Biochem J ; 468(1): 65-72, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25728577

ABSTRACT

Tandem mass tags (TMTs) were utilized in a novel chemical footprinting approach to identify lysine residues that mediate the interaction of receptor-associated protein (RAP) with cluster II of LDL (low-density lipoprotein) receptor (LDLR)-related protein (LRP). The isolated RAP D3 domain was modified with TMT-126 and the D3 domain-cluster II complex with TMT-127. Nano-LC-MS analysis revealed reduced modification with TMT-127 of peptides including Lys(256), Lys(270) and Lys(305)-Lys(306) suggesting that these residues contribute to cluster II binding. This agrees with previous findings that Lys(256) and Lys(270) are critical for binding cluster II sub-domains [Fisher, Beglova and Blacklow (2006) Mol. Cell 22, 277-283]. Cluster II-binding studies utilizing D3 domain variants K(256)A, K(305)A and K(306)A now showed that Lys(306) contributes to cluster II binding as well. For full-length RAP, we observed that peptides including Lys(60), Lys(191), Lys(256), Lys(270) and Lys(305)-Lys(306) exhibited reduced modification with TMT in the RAP-cluster II complex. Notably, Lys(60) has previously been implicated to mediate D1 domain interaction with cluster II. Our results suggest that also Lys(191) of the D2 domain contributes to cluster II binding. Binding studies employing the RAP variants K(191)A, K(256)A, K(305)A and K(306)A, however, revealed a modest reduction in cluster II binding for the K(256)A variant only. This suggests that the other lysine residues can compensate for the absence of a single lysine residue for effective complex assembly. Collectively, novel insight has been obtained into the contribution of lysine residues of RAP to cluster II binding. In addition, we propose that TMTs can be utilized to identify lysine residues critical for protein complex formation.


Subject(s)
LDL-Receptor Related Protein-Associated Protein/chemistry , LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , Humans , LDL-Receptor Related Protein-Associated Protein/genetics , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Lysine/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Footprinting/methods , Protein Interaction Domains and Motifs , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tandem Mass Spectrometry
10.
Environ Sci Pollut Res Int ; 21(15): 8964-80, 2014.
Article in English | MEDLINE | ID: mdl-24194415

ABSTRACT

Non-invasive spatially resolved monitoring techniques may hold the key to observe heterogeneous flow and transport behavior of contaminants in soils. In this study, time-lapse electrical resistivity tomography (ERT) was employed during an infiltration experiment with deicing chemical in a small field lysimeter. Deicing chemicals like potassium formate, which frequently impact soils on airport sites, were infiltrated during snow melt. Chemical composition of seepage water and the electrical response was recorded over the spring period 2010. Time-lapse electrical resistivity tomographs are able to show the infiltration of the melt water loaded with ionic constituents of deicing chemicals and their degradation product hydrogen carbonate. The tomographs indicate early breakthrough behavior in parts of the profile. Groundtruthing with pore fluid conductivity and water content variations shows disagreement between expected and observed bulk conductivity. This was attributed to the different sampling volume of traditional methods and ERT due to a considerable fraction of immobile water in the soil. The results show that ERT can be used as a soil monitoring tool on airport sites if assisted by common soil monitoring techniques.


Subject(s)
Environmental Monitoring/methods , Soil/chemistry , Water Pollutants, Chemical/analysis , Electric Conductivity , Tomography , Water , Water Movements , Water Pollutants, Chemical/chemistry , Weather
11.
J Biol Chem ; 288(41): 29670-9, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24009077

ABSTRACT

The C1 domain of factor VIII (FVIII) has been implicated in binding to multiple constituents, including phospholipids, von Willebrand factor, and low-density lipoprotein receptor-related protein (LRP). We have previously described a human monoclonal antibody called KM33 that blocks these interactions as well as cellular uptake by LRP-expressing cells. To unambiguously identify the apparent "hot spot" on FVIII to which this antibody binds, we have employed hydrogen-deuterium exchange mass spectrometry. The results showed that KM33 protects FVIII regions 2091-2104 and 2157-2162 from hydrogen-deuterium exchange. These comprise the two C1 domain spikes 2092-2093 and 2158-2159. Spike 2092-2093 has been demonstrated recently to contribute to assembly with lipid membranes with low phosphatidylserine (PS) content. Therefore, spike 2158-2159 might serve a similar role. This was assessed by replacement of Arg-2159 for Asn, which introduces a motif for N-linked glycosylation. Binding studies revealed that the purified, glycosylated R2159N variant had lost its interaction with antibody KM33 but retained substantial binding to von Willebrand factor and LRP. Cellular uptake of the R2159N variant was reduced both by LRP-expressing U87-MG cells and by human monocyte-derived dendritic cells. FVIII activity was virtually normal on membranes containing 15% PS but reduced at low PS content. These findings suggest that the C1 domain spikes 2092-2093 and 2158-2159 together modulate FVIII membrane assembly by a subtle, PS-dependent mechanism. These findings contribute evidence in favor of an increasingly important role of the C1 domain in FVIII biology.


Subject(s)
Endocytosis , Factor VIII/chemistry , Factor VIII/metabolism , Protein Structure, Tertiary , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Binding Sites/genetics , Deuterium Exchange Measurement , Factor VIII/genetics , Glycosylation , Humans , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/metabolism , Mass Spectrometry , Models, Molecular , Mutation , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Protein Binding/drug effects , Surface Plasmon Resonance , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
12.
J Biol Chem ; 288(36): 26105-26111, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23884417

ABSTRACT

A recent chemical footprinting study in our laboratory suggested that region 1803-1818 might contribute to A2 domain retention in activated factor VIII (FVIIIa). This site has also been implicated to interact with activated factor IX (FIXa). Asn-1810 further comprises an N-linked glycan, which seems incompatible with a role of the amino acids 1803-1818 for FIXa or A2 domain binding. In the present study, FVIIIa stability and FIXa binding were evaluated in a FVIII-N1810C variant, and two FVIII variants in which residues 1803-1810 and 1811-1818 are replaced by the corresponding residues of factor V (FV). Enzyme kinetic studies showed that only FVIII/FV 1811-1818 has a decreased apparent binding affinity for FIXa. Flow cytometry analysis indicated that fluorescent FIXa exhibits impaired complex formation with only FVIII/FV 1811-1818 on lipospheres. Site-directed mutagenesis revealed that Phe-1816 contributes to the interaction with FIXa. To evaluate FVIIIa stability, the FVIII/FV chimeras were activated by thrombin, and the decline in cofactor function was followed over time. FVIII/FV 1803-1810 and FVIII/FV 1811-1818 but not FVIII-N1810C showed a decreased FVIIIa half-life. However, when the FVIII variants were activated in presence of FIXa, only FVIII/FV 1811-1818 demonstrated an enhanced decline in cofactor function. Surface plasmon resonance analysis revealed that the FVIII variants K1813A/K1818A, E1811A, and F1816A exhibit enhanced dissociation after activation. The results together demonstrate that the glycan at 1810 is not involved in FVIII cofactor function, and that Phe-1816 of region 1811-1818 contributes to FIXa binding. Both regions 1803-1810 and 1811-1818 contribute to FVIIIa stability.


Subject(s)
Factor IX/chemistry , Factor VIII/chemistry , Factor VIIIa/chemistry , Amino Acid Substitution , Binding Sites , Factor IX/genetics , Factor IX/metabolism , Factor VIII/genetics , Factor VIII/metabolism , Factor VIIIa/genetics , Factor VIIIa/metabolism , Humans , Mutagenesis, Site-Directed , Mutation, Missense , Protein Stability , Protein Structure, Tertiary
13.
Br J Haematol ; 160(3): 377-86, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23190498

ABSTRACT

During storage, erythrocytes undergo changes that alter their clearance and function after transfusion and there is increasing evidence that these changes contribute to the complications observed in transfused patients. Stored erythrocytes were incubated overnight at 37°C to mimic the temperature after transfusion. After incubation, several markers for erythrocyte damage were analysed. After overnight incubation, stored erythrocytes showed increased potassium leakage, haemolysis, PS exposure and vesicle formation, and all these effects increased with increasing storage time. Furthermore, we demonstrated that long-term stored erythrocytes develop decreased flippase activity and increased scrambling activity after overnight incubation, leading to PS exposure and the release of vesicles. Reduced intracellular potassium was identified as the cause of the decreased flippase activity. Lastly, we provide evidence that erythrocytes can return to a PS-negative state by shedding parts of their membrane as PS-containing vesicles and that these vesicles can serve as a platform for the coagulation cascade. These findings reveal that potassium leakage, a well-known phenomenon of prolonged erythrocyte storage, primes erythrocytes for PS exposure. PS exposure will lead to vesicle formation and might have an important impact on the post-transfusion function and side effects of stored erythrocytes.


Subject(s)
Erythrocytes/metabolism , Phosphatidylserines/analysis , Potassium/metabolism , Transport Vesicles/metabolism , Blood Coagulation/drug effects , Blood Coagulation/physiology , Blood Coagulation Factors/metabolism , Blood Preservation , Erythrocytes/chemistry , Hemolysis , Humans
14.
J Biol Chem ; 288(1): 393-400, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23168412

ABSTRACT

Complex formation between coagulation factor VIII (FVIII) and von Willebrand factor (VWF) is of critical importance to protect FVIII from rapid in vivo clearance and degradation. We have now employed a chemical footprinting approach to identify regions on VWF involved in FVIII binding. To this end, lysine amino acid residues of VWF were chemically modified in the presence of FVIII or activated FVIII, which does not bind VWF. Nano-LC-MS analysis showed that the lysine residues of almost all identified VWF peptides were not differentially modified upon incubation of VWF with FVIII or activated FVIII. However, Lys-773 of peptide Ser-766-Leu-774 was protected from chemical modification in the presence of FVIII. In addition, peptide Ser-764-Arg-782, which comprises the first 19 amino acid residues of mature VWF, showed a differential modification of both Lys-773 and the α-amino group of Ser-764. To verify the role of Lys-773 and the N-terminal Ser-764 in FVIII binding, we employed VWF variants in which either Lys-773 or Ser-764 was replaced with Ala. Surface plasmon resonance analysis and competition studies revealed that VWF(K773A) exhibited reduced binding to FVIII and the FVIII light chain, which harbors the VWF-binding site. In contrast, VWF(S764A) revealed more effective binding to FVIII and the FVIII light chain compared with WT VWF. The results of our study show that the N terminus of VWF is critical for the interaction with FVIII and that Ser-764 and Lys-773 have opposite roles in the binding mechanism.


Subject(s)
Factor VIII/chemistry , Lysine/chemistry , Serine/chemistry , von Willebrand Factor/chemistry , Amino Acid Sequence , Binding Sites , Dose-Response Relationship, Drug , Humans , Kinetics , Mass Spectrometry/methods , Molecular Conformation , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Surface Plasmon Resonance
15.
J Biol Chem ; 287(8): 5775-83, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22215677

ABSTRACT

The A2 domain rapidly dissociates from activated factor VIII (FVIIIa) resulting in a dampening of the activity of the activated factor X-generating complex. The amino acid residues that affect A2 domain dissociation are therefore critical for FVIII cofactor function. We have now employed chemical footprinting in conjunction with mass spectrometry to identify lysine residues that contribute to the stability of activated FVIII. We hypothesized that lysine residues, which are buried in FVIII and surface-exposed in dissociated activated FVIII (dis-FVIIIa), may contribute to interdomain interactions. Mass spectrometry analysis revealed that residues Lys(1967) and Lys(1968) of region Thr(1964)-Tyr(1971) are buried in FVIII and exposed to the surface in dis-FVIIIa. This result, combined with the observation that the FVIII variant K1967I is associated with hemophilia A, suggests that these residues contribute to the stability of activated FVIII. Kinetic analysis revealed that the FVIII variants K1967A and K1967I exhibit an almost normal cofactor activity. However, these variants also showed an increased loss in cofactor activity over time compared with that of FVIII WT. Remarkably, the cofactor activity of a K1968A variant was enhanced and sustained for a prolonged time relative to that of FVIII WT. Surface plasmon resonance analysis demonstrated that A2 domain dissociation from activated FVIII was reduced for K1968A and enhanced for K1967A. In conclusion, mass spectrometry analysis combined with site-directed mutagenesis studies revealed that the lysine couple Lys(1967)-Lys(1968) within region Thr(1964)-Tyr(1971) has an opposite contribution to the stability of FVIIIa.


Subject(s)
Factor VIIIa/chemistry , Lysine/metabolism , Mass Spectrometry , Amino Acid Sequence , Factor VIIIa/metabolism , Molecular Sequence Data , Protein Stability , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...