Desialylation is a mechanism of Fc-independent platelet clearance and a therapeutic target in immune thrombocytopenia

June Li, Dianne E van der Wal, Guangheng Zhu, Miao Xu, Issaka Yougbare, Li Ma, Brian Vadasz, Naadiya Carrim, Renata Grozovsky, Min Ruan, Lingyan Zhu, Qingshu Zeng, Lili Tao, Zhi-min Zhai, Jun Peng, Ming Hou, Valery Leytin, John Freedman, Karin M Hoffmeister, Heyu Ni, June Li, Dianne E van der Wal, Guangheng Zhu, Miao Xu, Issaka Yougbare, Li Ma, Brian Vadasz, Naadiya Carrim, Renata Grozovsky, Min Ruan, Lingyan Zhu, Qingshu Zeng, Lili Tao, Zhi-min Zhai, Jun Peng, Ming Hou, Valery Leytin, John Freedman, Karin M Hoffmeister, Heyu Ni

Abstract

Immune thrombocytopenia (ITP) is a common bleeding disorder caused primarily by autoantibodies against platelet GPIIbIIIa and/or the GPIb complex. Current theory suggests that antibody-mediated platelet destruction occurs in the spleen, via macrophages through Fc-FcγR interactions. However, we and others have demonstrated that anti-GPIbα (but not GPIIbIIIa)-mediated ITP is often refractory to therapies targeting FcγR pathways. Here, we generate mouse anti-mouse monoclonal antibodies (mAbs) that recognize GPIbα and GPIIbIIIa of different species. Utilizing these unique mAbs and human ITP plasma, we find that anti-GPIbα, but not anti-GPIIbIIIa antibodies, induces Fc-independent platelet activation, sialidase neuraminidase-1 translocation and desialylation. This leads to platelet clearance in the liver via hepatocyte Ashwell-Morell receptors, which is fundamentally different from the classical Fc-FcγR-dependent macrophage phagocytosis. Importantly, sialidase inhibitors ameliorate anti-GPIbα-mediated thrombocytopenia in mice. These findings shed light on Fc-independent cytopenias, designating desialylation as a potential diagnostic biomarker and therapeutic target in the treatment of refractory ITP.

Figures

Figure 1. Anti-GPIbα antibodies induce platelet activation.
Figure 1. Anti-GPIbα antibodies induce platelet activation.
Surface P-selectin expression on murine (a,b) or human platelets (e,g,h) and JON/A binding on murine (c,d) or PAC-1 binding on human platelets (f) were measured by flow cytometry following incubation with mAbs (a,c,e,f,g) or antisera (b,d). (g) FcγRIIa/III blocker IV.3 was incubated with mAb 9D2 and platelets, and platelet P-selectin was assessed following. Only the healthy donor platelets that were significantly activated in the presence of 9D2 were tested; n=3. (h) P-selectin expression was measured in healthy human platelets following incubation with anti-GPIbα (ITP-1 to ITP-12) or anti-GPIIbIIIa (ITP-a to ITP-l) antibody-positive ITP patient plasma. All flow cytometry data are expressed as fold change from nonspecific murine IgG (murine)- or IVIG (human)-treated control platelets (CTRL). Anti-GPIbα mAbs shown as mean±s.e.m. of individual mAbs. *P<0.05, **P<0.01, ***P<0.001 versus CTRL as analysed by the Student's t-test (af) or one-way analysis of variance followed by Bonferroni post hoc analysis (g,h). NS, not significant.
Figure 2. Antibody-mediated platelet desialylation occurs mainly…
Figure 2. Antibody-mediated platelet desialylation occurs mainly on the GPIbα subunit.
Galactose exposure was detected with RCA-1 binding and measured by flow cytometry in murine (ad) and human (ej) platelets following incubation with mAbs (a,b,d,e,fh,j,k) or antisera (c) n=10–20. (d,g) Co-incubation with sialidase inhibitor DANA prior to addition of mAbs to murine (d) or human (g) platelets; n=8. (h) FcγRII/III blocker IV.3 was incubated with mAbs (9D2, M1 or HUTA B) and platelets, RCA-1 binding was assessed following. Only the healthy donor platelets that were significantly desialylated in the presence of these mAbs were tested; n=4. (i) RCA-1 binding was measured in healthy human platelets following incubation with anti-GPIbα (ITP-1 to ITP-12) or anti-GPIIbIIIa (ITP-a to ITP-l) antibody-positive ITP patient plasma. (j) anti-GPIbα-mediated RCA-1 binding was assessed following removal of GPIbα with OSGE. (k) Representative western blot of RCA-1 binding (left), and probing with commercial anti-GPIbα antibody (right) to confirm the identity of the RCA-1-positive band following incubation with anti-GPIbα mAb (NIT F). RCA-1 binding on GPIbα was also quantified by protein densitometry in the presence of DANA. All flow cytometry data are expressed as fold change from nonspecific murine IgG (murine)- or IVIG (human)-treated control platelets (CTRL). Anti-GPIbα mAbs shown as mean±s.e.m. of individual mAbs. *P<0.05, **P<0.01, ***P<0.001 versus CTRL as analysed by the Student's t-test (ac,e,f) or one-way analysis of variance followed by Bonferroni post hoc (d,gk). NS, not significant.
Figure 3. Anti-GPIbα antibodies induce surface expression…
Figure 3. Anti-GPIbα antibodies induce surface expression of NEU1.
Representative confocal images of surface expression of NEU1 on murine (a) and human (d) platelets stained with anti-NEU1 and anti-CD61 following incubations with anti-GPIbα mAb (NIT G (murine), NIT B (human)) or anti-GPIbα sera (murine). All other mAbs were also tested with similar results; n=5–8. Total NEU1 was detected in permeabilized human platelets; n=2. White scale bars, 5 μM; yellow scale bars, 2 μM. (b,c,e,f) Flow cytometric analysis of surface NEU1 expression on murine (b,c) and human (e,f) platelets following incubations with anti-GPIbα mAb or sera. Anti-GPIbα mAbs shown as mean±s.e.m. of individual mAbs; n=5–8. All flow cytometry data are expressed as fold change from nonspecific murine IgG (murine)- or IVIG (human)-treated control platelets (CTRL). Anti-GPIbα mAbs shown as mean±s.e.m. of individual mAbs. *P<0.05, **P<0.01, ***P<0.001 versus CTRL as analysed by the Student's t-test.
Figure 4. Anti-GPIbα platelet activation and desialylation…
Figure 4. Anti-GPIbα platelet activation and desialylation is a positive feedback loop.
(a,d) Human platelets were pre-incubated with inhibitors of platelet activation including intracellular Ca2+ flux (BAPTA-AM), phosphorylation of P38MAPK (SB203580) and Src kinase (PP1) prior to addition of anti-GPIbα mAbs. Following which, platelet activation (P-selectin expression) (a) or desialylation (RCA-1 binding) (d) was detected via flow cytometry. # and ## indicate comparison with CTRL for a only; n=8. (b) Representative western blots of whole-platelet lysate following incubations with GPIbα mAbs with or without indicated inhibitors. Membranes were probed for phosphorylated p38MAPK (p-p38MAPK) then stripped and re-probed for total pMAP38K. (c) Densitometry protein quantification of p-P38MAPK detected in b. Data representative of three separate experiments. (e) Sialidase inhibitor DANA was added prior to anti-GPIbα mAb incubation. DANA-mediated inhibition of antibody-induced platelet activation was measured by P-selectin expression and detected via flow cytometry. n=6. (f) Schematic flow chart illustrating platelet activation–desialylation-positive feedback loop. Following initial anti-GPIbα antibody crosslinking of GPIbα subunits, activation signalling occurs leading to surface translocation of NEU1. NEU1 cleavage of terminal sialic residues on a GPIbα subunit facilitates receptor clustering, resulting in amplification of platelet activation. (g,h) NIT A, NIT B and NIT F Fab fragments were generated and their effects on platelet activation and desialylation on murine (g) and human platelets (h) were analysed by flow cytometry. n=5. All flow cytometry data are expressed as fold change from nonspecific murine IgG (murine)- or IVIG (human)-treated control platelets (CTRL), unless otherwise indicated. Anti-GPIbα mAbs shown as mean±s.e.m. of individual mAbs. *,#P<0.05, **,##P<0.01, ***P<0.001 as assessed by one-way analysis of variance followed by Bonferroni post hoc analysis. NS, not significant.
Figure 5. Anti-GPIbα mAbs induce platelet activation…
Figure 5. Anti-GPIbα mAbs induce platelet activation and desialylation in vivo.
(a,b) ITP was induced by intraperitoneal injection of anti-GPIbα mAbs (NIT A, NIT B, NIT E, NIT F, NIT G and NIT H) or sera. At 16 h post injection, platelets isolated from the mice were examined for activation (P-selectin) and desialylation (RCA-1) by flow cytometry; n=3 per mAb. (a) Representative dot plots of isolated washed platelets from anti-GPIbα mAb (NIT E)- and sera-injected mice double-stained for anti-mouse IgG (against anti-GPIbα antibodies) (x axis) and RCA-1/P-selectin (y axis).
Figure 6. Fc-independent anti-GPIbα-mediated thrombocytopenia occurs via…
Figure 6. Fc-independent anti-GPIbα-mediated thrombocytopenia occurs via the AMR.
Mice were co-injected with (a) anti-GPIbα (NIT G) or (b) anti-GPIIbIIIa mAb (9D2)- or polyclonal sera-opsonized CFMDA-labelled platelets with asialofetuin (AMR inhibitor) or fetuin (a nonspecific control). Mice were bled at indicated time points and the percentage of fluorescently labelled platelets remaining in circulation was assessed with flow cytometry; n=6. *P<0.05 versus Fetuin-injected group at same time point as analysed by two-way analysis of variance followed by Bonferroni post hoc analysis. (ce) Same doses of indicated mAb or anti-GPIbα or anti-GPIIbIIIa mAbs (equal ratio mixture of individual antibody clones) were injected into age-matched FcγR−/− (c,d) or Aspgr−/− (e) or WT control mice. Platelet counts were taken immediately prior to antibody injection (time 0) and 24 h later (time 24). (e) Data are represented as a percentage calculated from [(time 0 platelet count−time 24 platelet count)/time 0 platelet count] × 100. *P<0.05, **P<0.01, ***P<0.001 as determined by the Student's t-test.
Figure 7. Anti-GPIbα platelet clearance in macrophage-depleted…
Figure 7. Anti-GPIbα platelet clearance in macrophage-depleted mice is via the AMR.
(a-e) Macrophages were depleted with clondrate liposomes via intravenous injection prior to study, and CFMDA-labelled antisera-opsonized platelets were injected with asialofeuin or fetuin and circulating platelets were quantified (a) as described in Fig. 6. n=5. ^^P<0.01 versus fetuin-injected group at the same time point. *P<0.05, **P<0.01 versus baseline as determined by two-way analysis of variance followed by Bonferroni post hoc analysis. (bd) Tissue sections of the spleen and liver harvested from normal or macrophage-depleted mice following the above circulation studies and were stained with anti-ASPGR1/2 (liver; red) (b,c) or anti-F4/80 (spleen; red) (d). Nuclei were counterstained with 4′,6-diamidino-2-phenylindole (blue). Arrows indicate co-localization of CFMDA-labelled platelets with ASPGR on hepatocytes. (e) Fluorescent platelet (green) localization was assessed with immunofluorescent microscopy at × 60 magnification and quantified with Image J. White scale bars, 10 μM. *P<0.05 as assessed by the Student's t-test. Data are representative of five randomly selected fluorescent images.
Figure 8. Sialidase inhibition rescues thrombocytopenia in…
Figure 8. Sialidase inhibition rescues thrombocytopenia in a murine model of ITP.
(ac) DANA or PBS was injected intraperitoneally immediately prior to anti-GPIbα or anti-GPIIbIIIa antibody injection (mAb or sera) to induce thrombocyotopenia. (a) Platelets from antibody-injected mice were analysed for desialylation (RCA-1 binding) via flow cytometry. (b) Platelet numbers were enumerated and compared between DANA-treated or mock (PBS)-treated groups. (c) Cumulative platelet counts from all anti-GPIbα and anti-GPIIbIIIa antibody (9D2, M1, PSI C1 and antisera)-injected mice with or without DANA treatment. For individual platelet counts of anti-GPIIbIIIa-injected mice, please see Supplementary Fig. 7. (d) Oseltamivir phosphate was injected intravenously 2 h after anti-GPIbα mAbs injection (equal ratio mixture of individual mAb clones). Platelet enumeration was as described above. Data are shown as mean±s.e.m. platelet counts of individual mAbs. *P<0.05, ***P<0.001 as determined by the Student's t-test.

References

    1. Rodeghiero F. et al.. Standardization of terminology, definitions and outcome criteria in immune thrombocytopenic purpura of adults and children: report from an international working group. Blood 113, 2386–2393 (2009).
    1. McMillan R. The pathogenesis of chronic immune thrombocytopenic purpura. Semin. Hematol. 44, S3–S11 (2007).
    1. Cines D. B., Bussel J. B., Liebman H. A. & Luning Prak E. T. The ITP syndrome: pathogenic and clinical diversity. Blood 113, 6511–6521 (2009).
    1. Mathias S. D. et al.. Impact of chronic Immune Thrombocytopenic Purpura (ITP) on health-related quality of life: a conceptual model starting with the patient perspective. Health Qual. Life Outcomes 6, 13 (2008).
    1. Provan D. et al.. International consensus report on the investigation and management of primary immune thrombocytopenia. Blood 115, 168–186 (2010).
    1. Vianelli N. et al.. Efficacy and safety of splenectomy in immune thrombocytopenic purpura: long-term results of 402 cases. Haematologica 90, 72–77 (2005).
    1. Neunert C. et al.. The American Society of Hematology 2011 evidence-based practice guideline for immune thrombocytopenia. Blood 117, 4190–4207 (2011).
    1. Harrington W. J., Minnich V., Hollingsworth J. W. & Moore C. V. Demonstration of a thrombocytopenic factor in the blood of patients with thrombocytopenic purpura. J. Lab. Clin. Med. 38, 1–10 (1951).
    1. Beardsley D. S. & Ertem M. Platelet autoantibodies in immune thrombocytopenic purpura. Transfus. Sci. 19, 237–244 (1998).
    1. Bussel J. B. Fc receptor blockade and immune thrombocytopenic purpura. Semin. Hematol. 37, 261–266 (2000).
    1. Nieswandt B., Bergmeier W., Rackebrandt K., Gessner J. E. & Zirngibl H. Identification of critical antigen-specific mechanisms in the development of immune thrombocytopenic purpura in mice. Blood 96, 2520–2527 (2000).
    1. Webster M. L. et al.. Relative efficacy of intravenous immunoglobulin G in ameliorating thrombocytopenia induced by antiplatelet GPIIbIIIa versus GPIbalpha antibodies. Blood 108, 943–946 (2006).
    1. Go R. S., Johnston K. L. & Bruden K. C. The association between platelet autoantibody specificity and response to intravenous immunoglobulin G in the treatment of patients with immune thrombocytopenia. Haematologica 92, 283–284 (2007).
    1. Peng J. et al.. Association of autoantibody specificity and response to intravenous immunoglobulin G therapy in immune thrombocytopenia: a multicenter cohort study. J. Thromb. Haemost. 12, 497–504 (2014).
    1. Chang M. et al.. Immune thrombocytopenic purpura (ITP) plasma and purified ITP monoclonal autoantibodies inhibit megakaryocytopoiesis in vitro. Blood 102, 887–895 (2003).
    1. Zeng Q. et al.. Relative efficacy of steroid therapy in immune thrombocytopenia mediated by anti-platelet GPIIbIIIa versus GPIbalpha antibodies. Am. J. Hematol. 87, 206–208 (2012).
    1. Ozaki Y., Asazuma N., Suzuki-Inoue K. & Berndt M. C. Platelet GPIb-IX-V-dependent signaling. J. Thromb. Haemost. 3, 1745–1751 (2005).
    1. Shattil S. J. & Newman P. J. Integrins: dynamic scaffolds for adhesion and signaling in platelets. Blood 104, 1606–1615 (2004).
    1. Du X. Signaling and regulation of the platelet glycoprotein Ib-IX-V complex. Curr. Opin. Hematol. 14, 262–269 (2007).
    1. Okumura I., Lombart C. & Jamieson G. A. Platelet glycocalicin. II. Purification and characterization. J. Biol. Chem. 251, 5950–5955 (1976).
    1. Solum N. O., Hagen I., Filion-Myklebust C. & Stabaek T. Platelet glycocalicin. Its membrane association and solubilization in aqueous media. Biochim. Biophys. Acta 597, 235–246 (1980).
    1. Rumjantseva V. et al.. Dual roles for hepatic lectin receptors in the clearance of chilled platelets. Nat. Med. 15, 1273–1280 (2009).
    1. Wandall H. H. et al.. Galactosylation does not prevent the rapid clearance of long-term, 4 degrees C-stored platelets. Blood 111, 3249–3256 (2008).
    1. Sorensen A. L. et al.. Role of sialic acid for platelet life span: exposure of beta-galactose results in the rapid clearance of platelets from the circulation by asialoglycoprotein receptor-expressing liver macrophages and hepatocytes. Blood 114, 1645–1654 (2009).
    1. Li C. et al.. The maternal immune response to fetal platelet GPIbalpha causes frequent miscarriage in mice that can be prevented by intravenous IgG and anti-FcRn therapies. J. Clin. Invest. 121, 4537–4547 (2011).
    1. Lei X. et al.. Anfibatide, a novel GPIb complex antagonist, inhibits platelet adhesion and thrombus formation in vitro and in vivo in murine models of thrombosis. Thromb. Haemost. 111, 279–289 (2014).
    1. Yang H. et al.. Fibrinogen and von Willebrand factor-independent platelet aggregation in vitro and in vivo. J. Thromb. Haemost. 4, 2230–2237 (2006).
    1. Reheman A. et al.. Plasma fibronectin depletion enhances platelet aggregation and thrombus formation in mice lacking fibrinogen and von Willebrand factor. Blood 113, 1809–1817 (2009).
    1. Wang Y. et al.. Plasma fibronectin supports hemostasis and regulates thrombosis. J. Clin. Invest. 124, 4281–4293 (2014).
    1. Yanabu M. et al.. Tyrosine phosphorylation and p72syk activation by an anti-glycoprotein Ib monoclonal antibody. Blood 89, 1590–1598 (1997).
    1. Cauwenberghs N. et al.. Fc-receptor dependent platelet aggregation induced by monoclonal antibodies against platelet glycoprotein Ib or von Willebrand factor. Thromb. Haemost. 85, 679–685 (2001).
    1. Shattil S. J., Hoxie J. A., Cunningham M. & Brass L. F. Changes in the platelet membrane glycoprotein IIb.IIIa complex during platelet activation. J. Biol. Chem. 260, 11107–11114 (1985).
    1. Bergmeier W. et al.. Flow cytometric detection of activated mouse integrin alphaIIbbeta3 with a novel monoclonal antibody. Cytometry 48, 80–86 (2002).
    1. King M., McDermott P. & Schreiber A. D. Characterization of the Fc gamma receptor on human platelets. Cell. Immunol. 128, 462–479 (1990).
    1. Pfueller S. L. & Luscher E. F. The effects of aggregated immunoglobulins on human blood platelets in relation to their complement-fixing abilities. II. Structural requirements of the immunoglobulin. J. Immunol. 109, 526–533 (1972).
    1. Hoffmeister K. M. et al.. Glycosylation restores survival of chilled blood platelets. Science 301, 1531–1534 (2003).
    1. Zhang Y. et al.. Identification of selective inhibitors for human neuraminidase isoenzymes using C4,C7-modified 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA) analogues. J. Med. Chem. 56, 2948–2958 (2013).
    1. Judson P. A., Anstee D. J. & Clamp J. R. Isolation and characterization of the major oligosaccharide of human platelet membrane glycoprotein GPIb. Biochem. J. 205, 81–90 (1982).
    1. Kinlough-Rathbone R. L., Perry D. W., Rand M. L. & Packham M. A. Responses to aggregating agents after cleavage of GPIb of human platelets by the O-sialoglycoprotein endoprotease from Pasteurella haemolytica- potential surrogates for Bernard-Soulier platelets? Thromb. Res. 99, 165–172 (2000).
    1. Jansen A. J. et al.. Desialylation accelerates platelet clearance after refrigeration and initiates GPIbalpha metalloproteinase-mediated cleavage in mice. Blood 119, 1263–1273 (2012).
    1. van der Wal D. E. et al.. Role of glycoprotein Ibalpha mobility in platelet function. Thromb. Haemost. 103, 1033–1043 (2010).
    1. Gitz E. et al.. Improved platelet survival after cold storage by prevention of glycoprotein Ibalpha clustering in lipid rafts. Haematologica 97, 1873–1881 (2012).
    1. van der Wal D. E. et al.. Platelet apoptosis by cold-induced glycoprotein Ibalpha clustering. J. Thromb. Haemost. 8, 2554–2562 (2010).
    1. Kasirer-Friede A. et al.. Lateral clustering of platelet GP Ib-IX complexes leads to up-regulation of the adhesive function of integrin alpha IIbbeta 3. J. Biol. Chem. 277, 11949–11956 (2002).
    1. Marshall J. S. et al.. Measurement of circulating desialylated glycoproteins and correlation with hepatocellular damage. J. Clin. Invest. 54, 555–562 (1974).
    1. Grozovsky R. et al.. The Ashwell-Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling. Nat. Med. 21, 47–54 (2015).
    1. Alimardani G., Guichard J., Fichelson S. & Cramer E. M. Pathogenic effects of anti-glycoprotein Ib antibodies on megakaryocytes and platelets. Thromb. Haemost. 88, 1039–1046 (2002).
    1. Cadroy Y. et al.. Relative antithrombotic effects of monoclonal antibodies targeting different platelet glycoprotein-adhesive molecule interactions in nonhuman primates. Blood 83, 3218–3224 (1994).
    1. Cauwenberghs N. et al.. Antithrombotic effect of platelet glycoprotein Ib-blocking monoclonal antibody Fab fragments in nonhuman primates. Arterioscler. Thromb. Vasc. Biol. 20, 1347–1353 (2000).
    1. Rubinstein E., Kouns W. C., Jennings L. K., Boucheix C. & Carroll R. C. Interaction of two GPIIb/IIIa monoclonal antibodies with platelet Fc receptor (Fc gamma RII). Br. J. Haematol. 78, 80–86 (1991).
    1. Sullam P. M. et al.. Physical proximity and functional interplay of the glycoprotein Ib-IX-V complex and the Fc receptor FcgammaRIIA on the platelet plasma membrane. J. Biol. Chem. 273, 5331–5336 (1998).
    1. Shrimpton C. N. et al.. Localization of the adhesion receptor glycoprotein Ib-IX-V complex to lipid rafts is required for platelet adhesion and activation. J. Exp. Med. 196, 1057–1066 (2002).
    1. Urbanus R. T. et al.. Patient autoantibodies induce platelet destruction signals via raft-associated glycoprotein Ibalpha and FcgammaRIIa in immune thrombocytopenia. Haematologica 98, e70–e72 (2013).
    1. Sarpatwari A. et al.. Autologous 111 In-labelled platelet sequestration studies in patients with primary immune thrombocytopenia (ITP) prior to splenectomy: a report from the United Kingdom ITP Registry. Br. J. Haematol. 151, 477–487 (2010).
    1. Ghanima W., Godeau B., Cines D. B. & Bussel J. B. How I treat immune thrombocytopenia: the choice between splenectomy or a medical therapy as a second-line treatment. Blood 120, 960–969 (2012).
    1. Enger C. et al.. Hepatobiliary laboratory abnormalities among patients with chronic or persistent immune thrombocytopenia (ITP). Ann. Hepatol. 10, 188–195 (2011).
    1. Davidson C. J., Tuddenham E. G. & McVey J. H. 450 million years of hemostasis. J. Thromb. Haemost. 1, 1487–1494 (2003).
    1. Li J. et al.. Severe platelet desialylation in a patient with glycoprotein Ib/IX antibody-mediated immune thrombocytopenia and fatal pulmonary hemorrhage. Haematologica 99, e61–e63 (2014).
    1. Jansen A. J., Peng J., Zhao H. G., Hou M. & Ni H. Sialidase inhibition to increase platelet counts: a new treatment option for thrombocytopenia. Am. J. Hematol. 90, E94–E95 (2015).
    1. Shao L. et al.. Successful treatment with oseltamivir phosphate in a patient with chronic immune thrombocytopenia positive for anti-GPIb/IX autoantibody. Platelets 26, 495–497 (2014).
    1. Nan X., Carubelli I. & Stamatos N. M. Sialidase expression in activated human T lymphocytes influences production of IFN-gamma. J. Leukoc. Biol. 81, 284–296 (2007).
    1. Chen X. P., Enioutina E. Y. & Daynes R. A. The control of IL-4 gene expression in activated murine T lymphocytes: a novel role for neu-1 sialidase. J. Immunol. 158, 3070–3080 (1997).
    1. Seyrantepe V. et al.. Regulation of phagocytosis in macrophages by neuraminidase 1. J. Biol. Chem. 285, 206–215 (2010).
    1. Grewal P. K. et al.. The Ashwell receptor mitigates the lethal coagulopathy of sepsis. Nature medicine 14, 648–655 (2008).
    1. Tribulatti M. V., Mucci J., Van Rooijen N., Leguizamon M. S. & Campetella O. The trans-sialidase from Trypanosoma cruzi induces thrombocytopenia during acute Chagas' disease by reducing the platelet sialic acid contents. Infect. Immun. 73, 201–207 (2005).
    1. Alioglu B., Tasar A., Ozen C., Selver B. & Dallar Y. An experience of oseltamivir phosphate (tamiflu) in a pediatric patient with chronic idiopathic thrombocytopenic purpura: a case report. Pathophysiol. Haemost. Thromb. 37, 55–58 (2010).
    1. Mandic R., Opper C., Krappe J. & Wesemann W. Platelet sialic acid as a potential pathogenic factor in coronary heart disease. Thromb. Res. 106, 137–141 (2002).
    1. Butun I. I. et al.. Preliminary study showing the relationship between platelet fibronectin, sialic acid, and ADP-induced aggregation levels in coronary heart disease. Clin. Appl. Thromb. Hemost. 13, 308–312 (2007).
    1. McGill D. A. & Ardlie N. G. Abnormal platelet reactivity in men with premature coronary heart disease. Coron. Artery Dis. 5, 889–900 (1994).
    1. Wang Y. et al.. Tyrosine phosphatase MEG2 modulates murine development and platelet and lymphocyte activation through secretory vesicle function. J. Exp. Med. 202, 1587–1597 (2005).

Source: PubMed

Подписаться