GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Thrombosis and Haemostasis, Georg Thieme Verlag KG, Vol. 107, No. 04 ( 2012), p. 717-725
    Abstract: Recent 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.
    Type of Medium: Online Resource
    ISSN: 0340-6245 , 2567-689X
    Language: English
    Publisher: Georg Thieme Verlag KG
    Publication Date: 2012
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2017
    In:  Blood Advances Vol. 1, No. 11 ( 2017-04-25), p. 644-651
    In: Blood Advances, American Society of Hematology, Vol. 1, No. 11 ( 2017-04-25), p. 644-651
    Abstract: Monoclonal and polyclonal anti-PRT/heparin antibodies are serologically distinct from anti-PF4/heparin antibodies. Binding of anti-PRT/heparin antibodies to PRT/dextran complexes correlates with PRT/GAG reactivity.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2017
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2015
    In:  Blood Vol. 126, No. 23 ( 2015-12-03), p. 3461-3461
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3461-3461
    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.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2014
    In:  Blood Vol. 124, No. 21 ( 2014-12-06), p. 2784-2784
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2784-2784
    Abstract: Protamine/heparin (PRT/H) antibodies (Ab) are a newly described class of heparin-dependent Abs found in ~25% of patients exposed to protamine and heparin during cardiopulmonary bypass surgery (CPB). Studies indicate that PRT/H Abs may be biologically similar to PF4/H Abs, the causative antibodies in heparin-induced thrombocytopenia (HIT). Like PF4/H Abs, PRT/H Abs show heparin-dependent reactivity and elicit platelet activation. These recent studies also showed that PRT/H Abs may be associated with adverse outcomes in the setting of protamine re-exposure (Lee, Blood 2013; Bakchoul, Blood 2013; Singla, Transfusion 2013). To understand the mechanisms of PRT/H Ab pathogenicity, we examined the biologic activities of both patient-derived polyclonal antibodies and a recently developed monoclonal antibody to PRT/H complexes (ADA). With Duke IRB approval, patient-derived PRT/H Abs (designated + CPB Abs) or control samples (- CPB Abs) were obtained from the HIT 5801 Study, a prospective multi-center study of patients with HIT undergoing cardiac surgery. ADA, an IgG3 monoclonal PRT/H Ab, was developed after immunization of BALB/c mice with PRT/H complexes and fusion of splenocytes with P3U.1 murine myeloma cells. A hybridoma clone showing reactivity to PRT/H 〉 〉 PRT was identified among 152 screened hybridomas and further subcloned by limiting dilution. As shown in Figure 1A, ADA was specific for PRT/H complexes and did not bind to other positively charged proteins (Figure 1B). Using these reagents, we performed a series of experiments to examine the biologic activities of PRT/H Abs. HIT Abs show limited binding to PF4/H complexes over a narrow stoichiometric range of PF4 and heparin concentrations (Kelton, Blood 1994). To determine if PRT/H Abs show similar protamine or heparin-dependent reactivity, binding of CPB Abs and ADA was measured at fixed concentrations of heparin (4 U/mL) and varying protamine concentration (0-200 mcg/mL; Figure 2A) or at a fixed concentration of protamine (31 mcg/mL) and varying heparin (0-50 U/mL; Figure 2B). Both ADA and CPB Abs did not bind to heparin alone. ADA did not bind to protamine alone while CPB Abs demonstrated binding to protamine alone. However, unlike PF4/H Abs, which lose binding at high heparin concentrations, neither ADA nor CPB Abs showed loss of reactivity at high protamine or heparin concentrations. Because protamines and histones are highly homologous proteins, we next examined the binding properties of ADA and CPB Abs to histones by protein immunoblot. For these studies, 1 ug of protamine, histones (H1, H2a, H2b, H3.1 and H4), nucleosomes and core histones were separated by protein electrophoresis. After transfer, cross-reactivity was examined using CPB Abs or ADA. As shown in Figure 3A, we noted that CPB Abs bind to protamine, histones H1, H2b, H3.1, H4 and core histones while ADA, which does not bind to protamine, binds to all histones, including H2a and native nucleosomes (Figure 3B). By contrast, no binding was seen with – CPB Abs or IgG3 isotype control. In other preliminary studies, we show that both CPB Abs and ADA activate neutrophils in the presence of protamine and PRT/H, as measured by neutrophil degranulation and myeloperoxidase release. Taken together, these studies show that the biologic activities of ADA, a new monoclonal PRT/H Ab, mirrors that of polyclonal patient-derived PRT/H Abs. As well, we demonstrate that PRT/H Abs cross-react with histones and may have pathogenic potential in inflammatory conditions associated with neutrophil activation and histone release. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2007
    In:  Blood Vol. 110, No. 13 ( 2007-12-15), p. 4253-4260
    In: Blood, American Society of Hematology, Vol. 110, No. 13 ( 2007-12-15), p. 4253-4260
    Abstract: Heparin-induced thrombocytopenia (HIT) is an antibody-mediated disorder that occurs with variable frequency in patients exposed to heparin. HIT antibodies preferentially recognize large macromolecular complexes formed between PF4 and heparin over a narrow range of molar ratios, but the biophysical properties of complexes that initiate antibody production are unknown. To identify structural determinants underlying PF4/heparin immunogenicity, we characterized the in vitro interactions of murine PF4 (mPF4) and heparin with respect to light absorption, size, and surface charge (zeta potential). We show that PF4/heparin macromolecular assembly occurs through colloidal interactions, wherein heparin facilitates the growth of complexes through charge neutralization. The size of PF4/heparin macromolecules is governed by the molar ratios of the reactants. Maximal complex size occurs at molar ratios of PF4/heparin at which surface charge is neutral. When mice are immunized with complexes that differ in size and/or zeta potential, antibody formation varies inversely with heparin concentration and is most robust in animals immunized with complexes displaying a net positive zeta-potential. These studies suggest that the clinical heterogeneity in the HIT immune response may be due in part to requirements for specific biophysical parameters of the PF4/heparin complexes that occur in settings of intense platelet activation and PF4 release.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2006
    In:  Blood Vol. 108, No. 11 ( 2006-11-16), p. 96-96
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 96-96
    Abstract: Heparin-Induced Thrombocytopenia (HIT) is a life-threatening immune-mediated reaction caused by platelet factor 4 (PF4)/heparin complexes. In recent in vitro studies, we have shown that human and monoclonal HIT antibodies recognize and optimally bind to ultra-large complexes (ULC; MW & gt;670kDa) formed at equimolar ratios of PF4 and heparin. We undertook the following studies to determine if the in vivo immunogenicity of PF4/heparin complexes showed similar requirements for stoichiometric molar ratios of PF4:heparin (PHR). Using a previously described murine immunization model, mice were immunized with antigen by retro-orbital injection daily for 5 days without adjuvant. Blood was collected at baseline and at weekly intervals for 4–6 weeks. Plasma was tested for antibody development by a murine PF4/heparin (mP+H) ELISA. To determine the effects of PHR on mP+H antibody production in vivo, PF4 was mixed with unfractionated heparin (UFH) at molar ratios of 2:1, 1:1 or 1:2 and injected into mice (n=5/cohort). Antibody levels (mean A450nm ± SD) of mice injected with PHR 2:1 (0.79 ± 0.77) were higher than mice injected with PHR 1:1 (0.23± 0. 2), or PHR 1:2 (0.09 ± 0.02, p & lt;0.05 for PHR 2:1 v. PHR 1:2). Because in vitro studies showed that UFH was far more capable of generating ULCs when mixed with PF4 than low-molecular weight heparin (LMWH) or fondaparinux, mice (n=5/cohort) were injected with buffer, or mPF4 mixed with UFH, or enoxaparin (a LMWH) or the pentasaccharide, fondaparinux at doses that are therapeutically equivalent to doses given to humans. Antibody levels (mean A450nm ± SD) were significantly higher in mice injected with UFH (3.340 + 0.15) than in mice injected with buffer (0.0451 ± 0.013, p & lt;0.001 for buffer v UFH), LMWH (0.8526 ± 0.44, p & lt;0.001 for LMWH v UFH) or the pentasaccharide (0.80 ± 0.0.7, p & lt;0.001 for pentasaccharide v UFH). Because PF4/heparin ULCs are composed of repeating structural antigenic units, we next investigated the role of toll-like receptors (TLRs), a family of pattern recognition receptors (PRRs) that recognize repeating structural determinants on microbes and foreign antigens. To determine if mP+H complexes engage TLRs, we injected mP+H into wild-type (WT, n=5) or mice deficient in MyD88 (n=7), an adaptor molecule critical for intracellular signaling by most TLRs. Antibody levels (mean A450nm ± SD) did not significantly differ between WT (1.5 ± 0.6) and MyD88 null mice (0.9 ± 0.5). In summary, these studies indicate that in vivo antibody responses to mP+H complexes are critically dependent on stoichiometric ratios favoring ULC formation and that immune activation by PF4/heparin is independent of MyD88 signaling pathways.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2008
    In:  Blood Vol. 112, No. 11 ( 2008-11-16), p. 270-270
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 270-270
    Abstract: Heparin-Induced Thrombocytopenia (HIT) is an immune-mediated thrombotic disorder caused by antibodies that recognize complexes of Platelet Factor 4 (PF4) and heparin. We have previously shown that exogenous administration of murine PF4 (mPF4) and heparin (mP+H) daily for five days into mice leads to development of high titer PF4/heparin autoantibodies (anti-mP+H; Suvarna Blood, 2007). While this experimental model has been extremely useful for understanding various biological aspects of heparin sensitization, this murine model requires extremely high-doses of mP+H to elicit antibody production. To circumvent this non-physiologic approach, we studied effects of in vivo platelet activation in mice with or without concomitant heparin administration. After receiving approval from our Institutional Animal Care and Use Committee, we initially performed dose-finding studies of various platelet agonists {arachidonic acid (AA), ADP, collagen and PAR-4} to identify the maximal tolerated dose associated with in vivo platelet activation (assays of circulating PF4 levels) and associated with increased morbidity. Once optimal doses of platelet agonists were identified, we injected agonists in the presence or absence of heparin daily for five days and monitored for development of PF4/heparin antibodies as previously described (Suvarna, Blood, 2005). Of the various agonists tested, we noted development of anti-mP+H in mice injected with collagen (coll), but not in mice injected with AA, ADP and PAR-4. Moreover, anti-mP+H responses in mice injected with collagen showed heparin dependence, as shown in the Table below. Mean A450nm Collagen (coll) alone (n=5) 0.04 ± 0.01 coll + 0.25 U/mL hep (n=5) 0.1 ± 0.04 coll + 0.5 U/mL hep (n=5) 0.1 ± 0.02 coll + 1 U/mL hep (n=5) 0.32 ± 0.27* (P & lt;0.05 v collagen alone) Additional cohorts injected with coll + 1U/mL heparin showed significantly higher rates of seroconversion (mean A450nm= 0.25 ± 0.06, n=15) as compared to mice injected with coll alone (mean A450nm= 0.07 ± 0.02, n=15; p=0.007 for coll+hep v. coll alone, See Figure). In mice treated with coll+ 1U/mL Hep, anti-mP+H were specific for mP+H (mean A450nm= 0.358 ± 0.133) as compared to mPF4 alone (mean A450nm= 0.06±0.007). Anti-mP+H did not recognize rat albumin (mean A450nm= 0.015 ±0.006) or ELISA wells coated with PBS (mean A450nm=0.019 ±0.003). In summary, these studies indicate that anti-mP+H can be elicited in mice after platelet activation and heparin exposure. Further refinements of this model will be important in advancing our understanding the physiologic basis of heparin sensitization. Figure Figure
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1435-1435
    Abstract: Abstract 1435 Platelet Factor 4 (PF4)/heparin (H) multimolecular complexes initiate an immune response that can ultimately lead to complications of Heparin-Induced Thrombocytopenia (HIT), a life-threatening prothrombotic disorder. We have previously shown that PF4:H multimolecular complexes assemble through non-specific electrostatic interactions and that other unrelated positively-charged proteins such as protamine (PRT) and lysozyme (Lys) exhibit similar biophysical interactions with heparin (ASH 2009; abstract # 1316). In these earlier studies, we showed that PRT/H and Lys/H, like PF4/H, show heparin-dependent binding over a range of heparin concentrations and that formation of multimolecular complexes occurs at distinct stoichiometric ratios (PRT/H at 3:1 and Lys/H at 5:1 molar ratios). We now extend these observations in vivo to show relevance to human disease. Using a murine immunization model, we show that mice injected with PRT/H and Lys/H multimolecular complexes, but not PRT alone, Lys alone or buffer, develop antigen-specific immune responses. In additional studies, we show that the immune response to PRT/H or Lys/H shares important biologic similarities with the humoral response to murine (m) PF4/H multimolecular complexes. Specifically, we demonstrate that antibody formation to PRT/H and Lys/H is heparin-dependent (occurs optimally at certain stoichiometric ratios) dose-dependent (requires threshold amounts of multimolecular complexes) and shows serologic transience. To demonstrate the clinical relevance of our findings, we examined patients undergoing cardiopulmonary bypass (CPB) for development of PRT/H antibodies. For these studies, we assayed the plasma from healthy subjects (n=45) and patients undergoing CPB (n=15) at three time points {baseline (BL), 5 days (5D) and 30 days (30D) after CPB} for the presence of PRT/H antibodies. As shown Figure 1A, plasma from normal subjects and patients undergoing CPB patients at BL and D5 displayed minimal reactivity in the PRT/H ELISA. However, by 30D, we observed that 4/15 patients (27%) developed significantly elevated levels of antibodies to PRT/H as compared to normals, or their respective samples obtained at baseline or 5D after surgery. Seropositive patients (filled symbols, n=4) as compared to seronegative patients (open symbols, n=3) recognized PRT/H and to some extent, PRT alone, but did not cross-react with other antigens including PRT/H, BSA, Lys, Lys/H or human PF4/H, Figure 1B; p 〈 0.001). To identify the mechanism by which protein/heparin multimolecular complexes triggered immune activation, we incubated murine dendritic cells from non-immunized C57Bl/6 mice with heparin or buffer, protein (mPF4, PRT or Lys), or protein/H complexes and measured IL-12, a marker of dendritic cell activation. As shown in Figure 1C, we demonstrated that IL-12 levels were significantly increased in wells containing protein/H complexes as compared to wells containing uncomplexed protein, buffer or heparin. Taken together, these studies indicate that heparin significantly alters the biophysical and biological properties of positively-charged compounds through formation of macromolecular complexes that lead to dendritic cell activation and trigger immune responses in vivo. Disclosures: Arepally: Glaxo Smith Kline: Speakers Bureau; Paringenix: Research Funding; University Of New Mexico: Patents & Royalties; Amgen: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Wiley ; 2008
    In:  British Journal of Haematology Vol. 142, No. 4 ( 2008-08), p. 671-673
    In: British Journal of Haematology, Wiley, Vol. 142, No. 4 ( 2008-08), p. 671-673
    Type of Medium: Online Resource
    ISSN: 0007-1048
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2008
    detail.hit.zdb_id: 1475751-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2012
    In:  Blood Vol. 120, No. 21 ( 2012-11-16), p. 269-269
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 269-269
    Abstract: Abstract 269 Mice injected with ultra-large complexes (ULCs) of platelet factor-4 (PF4) and heparin (PF4/H) exhibit a strong immune response which mimics that seen in patients developing Heparin induced thrombocytopenia (HIT)(Suvarna, Blood 2005). Previous studies with this murine immunization model have shown that murine (m) PF4/H ULCs potently activate dendritic cells (DCs) (Chudasama, Blood 2010) and initiate a T-cell dependent immune response (Suvarna, Blood 2005). To examine cellular/anatomic basis of PF4/H antibody formation, we examined the role of the spleen and germinal center formation in mice injected with mPF4/H complexes. To evaluate the role of the spleen, we immunized splenectomized mice (SPL) (n=15) and wild type (WT) mice (n=15) retro-orbitally with 100 mg/mL mPF4 and 5 U/mL Hep for 5 days. Antibodies to mPF4/H complexes were detected by an ELISA developed in our laboratory (Suvarna, Blood 2005; Suvarana, Blood 2007). As shown in Figure 1, SPL animals showed a marked reduction in Ab formation compared to WT animals (mean A450nm ± SD; SPL 0.206 ± 0.253 vs. WT 0.76 ± 0.788, p 〈 0.003). We next asked if antibody responses to PF4/H were associated with germinal center (GC) formation. The GC is a hallmark of T-cell dependent (TD) immune responses and is considered vital for development of high-affinity and isotype-switched antibodies. For these studies, mice were injected with a high dose of PF4/H (400 ug/mL PF4: 10 U/mL heparin) (n=2), standard protocol dose of PF4/H (100 ug/mL: 5 U/mL) and or buffer (Hank's Balanced Salt Solution, n=3) daily for 5 days via retro-orbital injection. As a positive control for GC formation, we injected two mice with a conventional antigen, nitrophenyl conjugated chicken gamma-globulin (NP-CGG). Mice expressing high antibody titers on Day 16 (mean A450nm± SD; high dose- 1.112 ± 0.5767; standard dose- 1.186 ± 0.7853) were sacrificed, spleens harvested and frozen in an embedding medium. Splenic sections (5m, longitudinal) were obtained as frozen sections and stained with immunohistochemical markers for GCs (GL-7 FITC; 1:400, counter-stain- FITC Alexa Fluor 488; 1:200), T-cells (TCR-β PE; 1:200) and B-cell follicles (B220 Biotin; 1:400, counter-stain- Streptavidin Alexa Fluor 350; 1:200). Images were obtained using Carl Zeiss AxioVert 200M microscope and analyzed with AxioVision Rel 4.6 software. Fluorescent images were processed with Adobe photoshop CS5.1. As shown in Figure 2, we noted that mice injected with NP-CGG, had ∼ 22 GC's, mice injected with buffer had 2 GC's and mice injected with high and standard dose of PF4/H, had ∼20–22 and 15–17 GC's respectively. These studies show that mPF4/H complexes elicit GC formation in association with secretion of mPF4/H Abs and that GC formation is comparable to that seen with conventional antigen. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...