Disruption of PF4/Hep Multimolecular Complex Assembly Using a Minimally Anticoagulant Heparin (ODSH)

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 723-723
Author(s):  
Manali Joglekar ◽  
Pedro Quintana ◽  
Stephen Marcus ◽  
Jian Liu ◽  
Gowthami M. Arepally

Abstract Abstract 723 Recent studies indicate that multimolecular complexes of platelet factor 4 (PF4) and heparin (H) are central to the pathogenesis of Heparin-Induced Thrombocytopenia (HIT). PF4/H multimolecular complexes are recognized preferentially by HIT antibodies (Rauova, Blood 2005) and are potently immunizing in a murine immunization model (Suvarna, Blood 2005). Because PF4/H multimolecular complexes assemble through non-specific electrostatic interactions, we hypothesized that disruption of PF4/H charge-dependent interactions could reduce immune mediated complications. To test this hypothesis, we employed a minimally anticoagulant compound (2-O, 3-O desulfated heparin, or ODSH, ParinGenix, Inc.) and characterized the charge-dependent interactions of murine PF4 (mPF4), ODSH and unfractionated heparin (UFH). In chromogenic assays of thrombin (IIa) generation, UFH was >80-fold more potent than ODSH in inactivating heparin (IC50 of residual IIa generation for UFH=3.1 nM v. ODSH= 259 nM, (Figure 1A). However, when equimolar amounts of UFH or ODSH (1.7 mM) were tested in a PF4 neutralization assay (Saggin, Thrombosis and Haemostasis 1992), the amount of mPF4 required to neutralize 50% of the anticoagulant activity of ODSH (IC50) was 25μg/mL, as compared to 73μg/mL for UFH (~3-fold difference), indicating that charge-dependent interactions, but not anticoagulant activity, were preserved between PF4 and ODSH (Figure 1B). When ODSH was added at 2.5, 5 or 10 fold molar excess to a fixed amount of UFH (6nM) in the PF4 neutralization assay, a proportionate increase in the amount of PF4 was needed to neutralize UFH, indicating that ODSH promotes the anticoagulant effect of UFH through preferential binding of PF4. To further characterize the biophysical interactions of PF4, ODSH and UFH, we used spectrophotometry and zeta potential to study the multimolecular complex formation (Suvarna, Blood 2007). We noted that mPF4 and ODSH formed multimolecular complexes at molar ratios of 2:1, whereas mPF4 and UFH complexes occurred at molar ratios of 1:1. When increasing concentrations of ODSH were added to pre-formed PF4/H multimolecular complexes, we noted a decrease in absorbance with increasing amounts of ODSH, indicating disruption of PF4/H multimolecular complexes (Figure 1C). However, when increasing amounts of UFH was added to preformed PF4/ODSH multimolecular complexes, a plateau in signal was noted, suggesting a higher affinity of ODSH for PF4. In PF4/H immunoassays, incubation of ODSH (1μg/mL) with HIT antibodies was effective in reducing antibody binding by >50% as compared to wells without ODSH. HIT antibodies did not recognize hPF4 (10mg/mL) in complex with ODSH (0.4-3.2 mg/mL), indicating minimal cross-reactivity of HIT antibodies with PF4/ODSH complexes (Figure 1D). In summary, we show that ODSH, a minimally anticoagulant heparin, can disrupt PF4/H multimolecular complex formation through charge dependent interactions and interfere with HIT antibody binding. These studies suggest that manipulation of PF4:H charge interactions can be a potential therapeutic strategy in the management of HIT. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1316-1316
Author(s):  
Shalini Chudasama ◽  
Benjamin Espinasse ◽  
Mark R Wiesner ◽  
Gowthami M Arepally

Abstract Abstract 1316 Poster Board I-340 The immune response in Heparin-Induced Thrombocytopenia (HIT) is directed to and initiated by large multimolecular complexes of Platelet Factor 4 (PF4) and heparin. We, and others, have previously shown that PF4 and heparin multimolecular assembly occurs through colloidal interactions, wherein heparin, a negatively charged polymeric compound, facilitates macromolecular assembly by binding and neutralizing PF4's positive charge (Suvarna, Blood 2007). In these same studies, we also demonstrated that changes in the molar ratios of the two reactants result in PF4/heparin (PF4:H) complexes with markedly altered biophysical properties and immunogenicity. Because PF4:H electrostatic interactions are non-specific, we hypothesized that other positively charged proteins would exhibit similar colloidal interactions with heparin. To test this hypothesis, we selected two positively charged proteins (protamine and lysozyme) and studied heparin-dependent complex formation by spectrophotometry (A280nm), and zeta potential (Zeta Sizer, Malvern, UK). Protamine sulfate (250, 125, 62.5, 31.2, 15.6 and 7.8 mcg/mL; Mw 5.1kDa) and lysozyme (1000, 500, 250 and 125 mcg/mL; Mw 14.3kDa) were mixed with various heparin concentrations (0-160 U/mL; activity 140U/mg; Mw 12kDa) and biophysical properties characterized by both instruments. Both protamine and lysozyme showed heparin-dependent complex formation, with peak particle formation occurring over a range of heparin concentrations (2-25 U/mL ) for both compounds. For protamine, particle formation was maximal at protamine:heparin (Pr:H) molar ratios of ∼2.5-3:1, whereas lysozyme formed peak particles at lysozyme;heparin (Ly:H) molar ratios of ∼5:1 (See figure). As with PF4:H complexes, size of complexes was dependent on mass amounts of protamine or lysozyme, with particle size increasing or decreasing in proportion to the amounts of protamine or lysozyme available for complex formation. These findings indicate that heparin is capable of forming macromolecular complexes with other proteins through charge dependent interactions. Additional in vitro and in vivo studies are underway to determine if Pr:H or Lys:H complexes exhibit cross-reactivity with PF4/heparin antibodies and if complex formation is associated with immunological consequences. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3391-3391
Author(s):  
Georgios Pongas ◽  
Swapan Dasgupta ◽  
Perumal Thiagarajan

Abstract Abstract 3391 Introduction The anti-platelet factor 4(PF4)/heparin antibodies, arising as a result of previous heparin exposure, are causally related to the procoagulant state due to platelet and monocyte activation. Formation of these antibodies with subsequent thrombocytopenia or thrombosis has also been described in patients, who have not been previously exposed to heparin. The presence of anti-PF4/heparin antibodies in individuals correlates with the severity of periodontal disease, implying that their occurrence may be triggered by periodontal pathogens. In this study, we determined the presence of anti-PF4/heparin antibodies in gram-negative bacteremic patients in a hospital setting and propose a pathophysiologic mechanism of their presence. Method We developed an in house ELISA for quantifying anti-PF4/heparin antibodies using therapeutic heparin and PF4 isolated from platelets. We used serum from a patient with high optical density as a standard and assigned an unit of 100 arbitrarily to construct a standard curve. We tested the sera from gram negative bacteremic patients (n= 34) in the quantitative ELISA along with normal controls (n=10). We also developed an in house ELISA for studying cross reactivity between anti-PF4/heparin antibodies and lipopolysaccharide (LPS)/PF4. We tested the sera from patients (n=5) with heparin induced thrombocytopenia in this cross reactivity ELISA. To test the interaction of LPS with PF4, we labeled PF4 with Alexa488 and measured its binding to LPS by monitoring the changes in fluorescence emission spectrum following excitation at λ480. Results Patients with bacteremia had higher titers of antiPF4/heparin antibodies compared to normal controls (26.4 ± SD 33 units, N=34 versus 6.3 ± SD 2.38 units, N=10, P=0.032). Bacterial LPS interacted with alexa488-labeled PF4 in a concentration-dependent manner, as measured by the quenching of the excitation spectrum. Patients with ant-PF4/heparin antibodies also reacted with LPS/PF4 complex in ELISA. Prior absorption of serum with PF4/heparin complex coated on ELISA plates decreased the reactivity of the serum towards PF4/LPS complex (19–46%) in two out of the five patients tested suggesting some were cross-reaction between PF4/Heparin and PF4/LPS complex. Conclusions PF4 forms a complex with lipopolysaccharide and this complex is immunogenic. Antibodies to PF4/LPS complex can cross-react with PF4/heparin complex raising the possibility that these antibodies may be responsible for the detection of PF4/heparin in individuals never been exposed to heparin previously. These antibodies may also be at least partly responsible for increased thrombosis associated with infection. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3461-3461 ◽  
Author(s):  
Grace M. Lee ◽  
Manali Joglekar ◽  
Sanjay Khandelwal ◽  
Rui Qi ◽  
Lubica Rauova ◽  
...  

Abstract Protamine/heparin (PRT/H) antibodies (Abs) are a newly described class of heparin-dependent antibodies found in ~25% of patients exposed to protamine and heparin during cardiopulmonary bypass surgery (CPB). Although recent studies show that PRT/H Abs have several serologic properties similar to platelet factor 4 (PF4)/heparin Abs, the clinical significance of PRT/H Abs is unknown. To understand their clinical significance, we undertook studies to characterize the biologic effects of PRT/H Abs in vitro. Using a previously described murine monoclonal antibody to PRT/H complexes (IgG3 isotype, ADA) and patient-derived PRT/H Abs, we examined antibody cross-reactivity with histones and nuclear proteins as well as functional effects of PRT/H Abs on neutrophil activation. Using a commercial ANA immunofluorescence assay (ImmuGlow Hep-2 Cells Anti-nuclear Antibody IFA kit), we first examined cross-reactivity of anti-PRT/H on nuclear proteins. As seen in Figure 1, both ADA and patient-derived PRT/H Abs (depicted as α-PRT/H (+) in Figure 1D) showed significant binding to Hep-2 cells. In contrast, no reactivity was seen when Hep-2 cells were incubated with plasma from CPB patients who were seronegative for PRT/H antibodies (depicted as α-PRT/H (-) in Figure 1D) or with IgG3 isotype (data not shown). To confirm that PRT/H Abs were binding to nuclear antigens, we examined the cross-reactivity of monoclonal and polyclonal anti-PRT/H on individual nuclear binding proteins, including Single Stranded Binding Protein, RO-52, JO-1, (Sigma; St. Louis, MO, USA) and nucleosomes (New England Biolabs; UK) by ELISA. As shown in Table 1, ADA showed significantly higher binding to all nuclear antigens as compared to isotype control. Polyclonal PRT/H Abs from CPB patients also showed increased binding to nuclear antigens relative to control plasma, but did not achieve statistical significance. To determine if PRT/H Abs activate neutrophils, we isolated neutrophils by gradient centrifugation and incubated cells with 100 ug/mL of ADA or isotype in the presence or absence of antigen (PRT 31 ug/mL + H4 U/mL) and measured release of myeloperoxidase (MPO). As shown in Figure 2, ADA alone or isotype control showed minimal MPO release. In the presence of PRT or PRT/H, ADA, but not isotype control, showed significant MPO release. Taken together, these studies demonstrate that PRT/H Abs cross-react with nuclear antigens and can trigger neutrophil activation. These findings suggest that PRT/H Abs cross-react with a closely related class of antigens (histones) and enhance inflammation through cross-reactivity with nuclear antigens and/or through functional effects on neutrophil activation. Table 1. Antibody SSBP RO52 JO-1 nucleosomes ADA v. Isotype 1.36 ± 0.06 v. 0.13 ± 0.01 1.76 ± 0.06 v. 0.08 ± 0.01 1.64 ± 0.03 v. 0.09 ± 0.01 0.73 ± 0.03 v. 0.07 ± 0.01 Polyclonal PRT/H Abs v. control plasma 1.01 ± 0.23 v. 0.54 ± 0.05 1.15 ± 0.21 v. 0.72 ± 0.02 0.92 ± 0.16 v. 0.49 ± 0.04 0.81 ± 0.14 v. 0.75 ± 0.3 Figure 1. Figure 1. Figure 2. Figure 2. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2204-2204
Author(s):  
Benjamin Espinasse ◽  
Manali Joglekar ◽  
Giancarlo Valiente ◽  
Gowthami M. Arepally

Abstract Abstract 2204 Electrostatic interactions between Platelet factor 4 (PF4), a cationic protein, and heparin, an anionic carbohydrate result in the formation of ultra-large complexes (ULCs) that are immunogenic in mice (Suvarna, Blood 2007) and contribute to the immune pathogenesis of Heparin-induced thrombocytopenia (HIT). Previous studies (Rauova, Blood 2005; Greinacher, Arterioscler Thromb Vasc Biol, 2006) have shown that the size of ULCs is determined by the concentration and the molar ratios of PF4:H (PHRs) of each compound. Size determination of PF4/H complexes has been problematic due to technical limitations of two commonly employed methods for sizing complexes, photon correlation spectroscopy (PCS) and size exclusion chromatography (SEC). PCS is a technique for measuring particles in solution using laser illumination is based on principles of Brownian motion. PCS performs optimally with monodisperse populations and is biased by the presence of large aggregates. SEC, a liquid chromatography method, is technically cumbersome, requires sample labeling and not feasible for measuring large numbers of samples. To address these limitations, we examined two novel approaches for measuring a broad range of PF4/H complex size (100–3000 nm) in vitro: Nanosight and flow cytometry (FC). Nanosight (Nanosight Ltd, Wiltshire, United Kingdom),was employed for measuring small-sized complexes using physiologic concentrations of hPF4 (10 ug/mL). Nanosight uses proprietary software to track nanoparticles (range 10–1000nm) in solution by laser illumination with real-time tracking of the motion of individual particles by a camera. Analysis parameters provided by the software include: 1) Particle size distributions displayed as histograms 2) direct visualization of particles 3) particle counting and sizing and 4) particle scatter intensity vs. count and size. For measuring intermediate to large sized particles, formed at high hPF4 concentrations (95 ug/mL), we used flow cytometry calibrated with sizing beads on side scatter channel (SSC). FC was performed using a BD LSRII cell analyzer (Becton Dickinson, Franklin Lakes, NJ), a high throughput flow analyzer with the threshold channel for SSC set to 200 and a flow rate of 1 ul per second. The instrument was calibrated using sizing beads ranging from 0.3–6 μm in size (Figure A). For both techniques, PF4/H ULCs were formed by adding hPF4 (10 or 95 ug/mL)and various UFH concentrations in HBSS to yield the indicated PHRs. Complexes were incubated for 60 minutes and measured by NanoSight or FC. Results of experiments using Nanosight are shown in Table 1 with results showing size and particle counts for each PHR. Results of FC are shown in Figure B and Table 2 (median, 5% and 95% size in nm). Both studies showed reproducibility for measurements for a given concentration and showed changes in complex size as a function of PHR (Figure B). Both methodologies are technically simple and provide complementary approaches to PCS for PF4/H complex size determination. Disclosures: No relevant conflicts of interest to declare.


2012 ◽  
Vol 107 (04) ◽  
pp. 717-725 ◽  
Author(s):  
Manali V. Joglekar ◽  
Pedro M. Diez ◽  
Stephen Marcus ◽  
Rui Qi ◽  
Benjamin Espinasse ◽  
...  

SummaryRecent studies have shown that ultra-large complexes (ULCs) of platelet factor 4 (PF4) and heparin (H) play an essential role in the pathogenesis of heparin-induced thrombocytopenia (HIT), an immune-mediated disorder caused by PF4/H antibodies. Because antigenic PF4/H ULCs assemble through non-specific electrostatic interactions, we reasoned that disruption of charge-based interactions can modulate the immune response to antigen. We tested a minimally anticoagulant compound (2-O, 3-O desulfated heparin, ODSH) with preserved charge to disrupt PF4/H complex formation and immunogenicity. We show that ODSH disrupts complexes when added to pre-formed PF4/H ULCs and prevents ULC formation when incubated simultaneously with PF4 and UFH. In other studies, we show that excess ODSH reduces HIT antibody (Ab) binding in immunoassays and that PF4/ODSH complexes do not cross-react with HIT Abs. When ODSH and unfractionated heparin (UFH) are mixed at equimolar concentrations, we show that there is a negligible effect on amount of protamine required for heparin neutralisation and reduced immunogenicity of PF4/UFH in the presence of ODSH. Taken together, these studies suggest that ODSH can be used concurrently with UFH to disrupt PF4/H charge interactions and provides a novel strategy to reduce antibody mediated complications in HIT.Presented in part at the 52nd American Society of Hematology Annual Meeting and Exposition, December 6th, 2010, Orlando, Florida, USA.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3403-3403
Author(s):  
Anny Appert-Flory ◽  
Florence Fischer ◽  
Didier Jambou ◽  
Sylvain Buvat ◽  
Marie-Helene Mahagne ◽  
...  

Background: Unfractionated heparin (UFH) has been in clinical use for more than half a century. Monitoring UFH treatments is mandatory due to large inter- and intra-individual variations in its anticoagulant activity, with a risk of thrombosis in case of under-dosing and a risk of hemorrhage in case of over-dosing. Laboratory monitoring of UFH treatments is usually based on the prolongation of activated partial thromboplastin time (aPTT), or on the measurement of plasma anti-Xa activity. As UFH present in the patient's blood sample could be neutralized by platelet factor 4 (PF4) released by in-vitro activated platelets, it is recommended to perform the tests aimed at evaluating its anticoagulant activity as soon as possible after blood collection. Actually, the current guidelines recommend a maximum delay of 2 hours between blood sampling and testing for anti-Xa activity or aPTT prescribed for monitoring treatments by UFH, when blood is collected into citrated tubes. As such a short delay could be an issue, particularly for multisite centres, we evaluated the potential impact of a longer delay on test results. Design of the study: For that purpose, 2 citrated evacuated tubes containing 0.109 M tri-Na citrate (1 vol./9 vol.) were collected from patients on UFH: one was centrifuged and tested within 1h after blood collection (T1h) and one was stored for 4h at room temperature (+22°C) before being centrifuged and immediately analyzed (T4h). Methods: Anti-Xa activity was evaluated using 2 reagents: Biophen Heparin LRT (Hyphen Biomed, Neuville-sur-Oise, France) and HemosIL Liquid Heparin (Instrumentation Laboratory, IL, Bedford, MA, USA). aPTT was evaluated using the HemosIL SynthASil reagent (IL). All assays were automated on an ACL TOP 700 CTS (IL). As the distributions of the data were not found to be normal, anti-Xa and aPTT results were expressed as the median values (with ranges), and comparison of test results obtained at T1h and T4h performed using the Wilcoxon test. Test results were also compared according to Bland-Altman. Results: A total of 123 paired tubes were investigated. Analytical comparison of anti-Xa activity demonstrated a significant decrease (p<0.0001) after a 4 h-storage at room temperature (T4h) vs. a <1h-delay (T1h), for the two evaluated reagents (Table). The mean bias between test results obtained at T4h and T1h, evaluated according to Bland and Altman, was <0.05 IU/mL (in absolute value) for the two reagents, and identical for anti-Xa activities below or above 0.50 IU/mL. Such a value was below the imprecision of the techniques. There were 12 cases of discrepancy as whether test results were within the therapeutic range (0.30 - 0.70 IU/mL) or not when evaluated at T4h vs. T1h using the Hyphen reagent (9.8%), and 12.9% with the IL reagent. In most cases, these discrepancies were found for anti-Xa activities close to the lower limit of the therapeutic range, and would not have induced any change in the management of anticoagulation in these patients. APTT was significantly shortened (p<0.0001) after a 4h- vs. a 1h-storage at room temperature, with a mean bias of -8.1 sec, corresponding to a -0.25 decrease in ratio [relative change of -12.1% (95%CI= -34.8 +10.6)]. If considering the aPTT therapeutic range corresponding to anti-Xa activity between 0.30 and 0.70 IU/ml, 29 cases of discordant test results (23.6%) were observed that could have induced changes in the management of anticoagulation in 10% of the patients. Finally, the concordance whether test results measured at T4h and T0 were within or outside the therapeutic range was excellent for anti-Xa activity (kappa=0.813) and good for aPTT (kappa=0.661). Conclusions: These results suggest that extending to 4h the delay between blood sampling and measurement of anti-Xa activity prescribed for monitoring UFH treatments when blood is collected into citrated tubes is acceptable and safe, at least when evaluated with the two tested reagents. As change in aPTT results was found to be of a greater order of magnitude and more relevant, it could be justified recommending a shorter delay. Table Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2351-2351
Author(s):  
Jeanine Walenga ◽  
Walter Jeske ◽  
Sabrina Bertini ◽  
Giulia Risi ◽  
Michelle Sung ◽  
...  

Heparin, an anticoagulant widely used in numerous medical applications, is considered an essential medicine by the WHO. Due to its high volume use and that it is the parent material for low molecular weight heparins, there is potential for the raw material to be in short supply. The African swine fever epidemic in China, ongoing since August 2018, has added further restraints on heparin source material supply. At present medical grade heparin in the US is only derived from porcine intestinal mucosa; however, there are explorations into using bovine, ovine, and other sources. Bovine heparin, once common place in the US pharmaceutical sector, is again under consideration by the US FDA. This study focused on the primary immunogenic activity associated with heparin, that is heparin-induced thrombocytopenia (HIT), and how the interaction of bovine heparin with functional HIT antibodies compares to that of porcine heparin. Materials and Methods Bovine unfractionated heparin from multiple manufacturers was compared to commercial medical grade porcine heparin obtained from US pharmacies. The US Pharmacopeia porcine heparin standard was used to determine potency equivalence. Antibodies to the complex of platelet factor 4/heparin (PF4/heparin) from banked clinically confirmed HIT patient apheresis fluids were combined with heparin and donor platelet rich plasma (PRP; blood collected in citrate from volunteers after signing a consent document). Heparins were tested at final concentrations of 0.1, 0.4, 0.8, 1, and 100 U/mL. The platelet activation response was determined on the BioData PAP-8 Platelet Aggregometer and quantitated in terms of primary slope (PS), area under the curve (AUC), maximum aggregation (MA), and final aggregation (FA). Characterization of the biophysical interaction between varying molar ratios of human PF4 and heparin was performed using photon correlation spectroscopy (PCS) and zeta potential (Zp) measurements of PF4/heparin complexes using Zetasizer Nano ZS instrumentation and software. Differences between bovine and porcine heparin were assessed by t-test or Mann-Whitney test. Concentration-response curves were analyzed by two-way ANOVA followed by the Holm-Sidak multiple comparison test using SigmaPlot software. Results Platelet activation to PF4/heparin antibodies at bovine and porcine heparin concentrations of 0.1 U/mL (56 ± 9 % vs. 54 ± 11 % MA) and 0.4 U/mL (59 ± 10 % vs. 65 ± 8 % MA) were the same with the expected inhibition (9 ± 4% MA) at the supra-therapeutic concentration of 100 U/mL. Consistent responses were obtained across 21 lots of bovine heparin, 30 lots of porcine heparin, and 38 platelet-HIT antibody combinations. The HIT potential of bovine heparin and porcine heparin was not statistically different (p>0.05). At higher medical use doses, the platelet aggregation response in the presence of HIT antibodies was actually lower for bovine heparin than porcine heparin (0.8 U/mL, 49 ± 10 % vs. 64 ± 9 % MA, p<0.05; and 1.0 U/mL, 45 ± 11 % vs. 62 ± 9 % MA, p<0.05). By PCS, it was observed that the maximal aggregation between PF4 and either porcine or bovine heparin occurred at comparable molar ratios (7.3 ± 1.5 vs. 6.4 ± 0). Although the porcine and bovine heparins exhibited comparable molecular weights (16,333 ± 153 vs. 16,790 ± 230 Da) and polydispersities (1.19 ± 0.02 vs. 1.15 ± 0.01), porcine heparin formed somewhat larger complexes with PF4 (1113 ± 65 nm) than did bovine heparin (863 ± 68 nm). The molar ratios of PF4 to heparin at which the charge of the complex was fully neutralized (Zp = 0) was comparable for porcine and bovine heparin (9.04 ± 0.19 vs. 9.97 ± 0.65). Consistent responses were obtained across 4 lots of bovine heparin and 3 lots of porcine heparin. Conclusions Bovine heparin and porcine heparin had the same in vitro functional platelet activation response in the presence of HIT antibodies, the same potential to form complexes with human PF4, and the same associated features that make PF4 immunogenic. This investigation demonstrates that bovine heparins should have a similar immunogenic response as porcine heparin at equi-unit dosing. Current refinements in the manufacturing process for bovine and porcine heparins have led to well-characterized and purified products which is reflected in their comparable biological behavior. Disclosures No relevant conflicts of interest to declare.


1993 ◽  
Vol 70 (04) ◽  
pp. 625-630 ◽  
Author(s):  
Edward Young ◽  
Benilde Cosmi ◽  
Jeffrey Weitz ◽  
Jack Hirsh

SummaryThe non-specific binding of anticoagulantly-active heparin to plasma proteins may influence its anticoagulant effect. We used low affinity heparin (LAH) essentially devoid of anti-factor Xa activity to investigate the extent and possible mechanism of this non-specific binding. The addition of excess LAH to platelet-poor plasma containing a fixed amount of unfractionated heparin doubled the anti-factor Xa activity presumably because it displaces anticoagulantly-active heparin from plasma proteins. Although dextran sulfates of varying molecular weights also increased the anti-factor Xa activity, less sulfated heparin-like polysaccharides had no effect. These findings suggest that the ability to displace active heparin from plasma protein binding sites is related to charge and may be independent of molecular size. In contrast to its effect in plasma containing unfractionated heparin, there was little augmentation in anti-factor Xa activity when LAH was added to plasma containing low molecular weight heparin (LMWH), indicating that LMWH binds less to plasma proteins than unfractionated heparin. This concept is supported by studies comparing the anticoagulant activity of unfractionated heparin and LMWH in plasma with that in buffer containing antithrombin III. The anti-factor Xa activity of unfractionated heparin was 2-fold less in plasma than in the purified system. In contrast, LMWH had identical anti-factor Xa activity in both plasma and buffer, respectively. These findings may be clinically relevant because the recovered anti-factor Xa activity of unfractionated heparin was 33% lower in plasma from patients with suspected venous thrombosis than in plasma from healthy volunteers. The reduced heparin recovery in patient plasma reflects increased heparin binding to plasma proteins because the addition of LAH augmented the anti-factor Xa activity. In contrast to unfractionated heparin, there was complete recovery of LMWH added to patient plasma and little increase of anti-factor Xa activity after the addition of LAH. These findings may explain why LMWH gives a more predictable dose response than unfractionated heparin.


Vaccines ◽  
2021 ◽  
Vol 9 (6) ◽  
pp. 559
Author(s):  
Piotr Rzymski ◽  
Bartłomiej Perek ◽  
Robert Flisiak

The rollout of COVID-19 vaccines brings hope for successful pandemic mitigation and getting the transmission of SARS-CoV-2 under control. The vaccines authorized in Europe displayed a good safety profile in the clinical trials. However, during their post-authorization use, unusual thrombotic events associated with thrombocytopenia have rarely been reported for vector vaccines. This led to the temporary suspension of the AZD1222 vaccine (Oxford/AstraZeneca) in various European countries and the Ad26.COV2 vaccine (Janssen/Johnson&Johnson) in the United States, with regulatory bodies launching investigations into potential causal associations. The thromboembolic reactions were also rarely reported after mRNA vaccines. The exact cause of these adverse effects remains to be elucidated. The present paper outlines the hypotheses on the mechanisms behind the very rare thrombotic thrombocytopenia reported after the COVID-19 vaccination, along with currently existing evidence and future research prospects. The following are discussed: (i) the role of antibodies against platelet factor 4 (PF4), (ii) the direct interaction between adenoviral vector and platelets, (iii) the cross-reactivity of antibodies against SARS-CoV-2 spike protein with PF4, (iv) cross-reactivity of anti-adenovirus antibodies and PF4, (v) interaction between spike protein and platelets, (vi) the platelet expression of spike protein and subsequent immune response, and (vii) the platelet expression of other adenoviral proteins and subsequent reactions. It is also plausible that thrombotic thrombocytopenia after the COVID-19 vaccine is multifactorial. The elucidation of the causes of these adverse events is pivotal in taking precautionary measures and managing vaccine hesitancy. It needs to be stressed, however, that the reported cases are currently sporadic and that the benefits of COVID-19 vaccines vastly outweigh their potential risks.


Symmetry ◽  
2021 ◽  
Vol 13 (5) ◽  
pp. 766
Author(s):  
Barbara Bankiewicz ◽  
Marcin Palusiak

The DFT calculations have been performed on a series of two-element complexes formed by substituted 2,6-diaminopyridine (R−PDA) and pyridine (R−Pyr) with X−CY3 molecules (where X = Cl, Br and Y = H, F). The primary aim of this study was to examine the intermolecular hydrogen and halogen bonds in the condition of their mutual coexistence. Symmetry/antisymmetry of the interrelation between three individual interactions is addressed. It appears that halogen bonds play the main role in the stabilization of the structures of the selected systems. However, the occurrence of one or two hydrogen bonds was associated with the favourable geometry of the complexes. Moreover, the impact of different substituent groups attached in the para position to the aromatic ring of the 2,6-diaminopyridine and pyridine on the character of the intermolecular hydrogen and halogen bonds was examined. The results indicate that the presence of electron-donating substituents strengthens the bonds. In turn, the presence of electron-withdrawing substituents reduces the strength of halogen bonds. Additionally, when hydrogen and halogen bonds lose their leading role in the complex formation, the nonspecific electrostatic interactions between dipole moments take their place. Analysis was based on geometric, energetic, and topological parameters of the studied systems.


Sign in / Sign up

Export Citation Format

Share Document