Semin Thromb Hemost 2021; 47(03): 254-260
DOI: 10.1055/s-0041-1725065
Review Article

Effect of Heparanase and Heparan Sulfate Chains in Hemostasis

Yona Nadir
1   Thrombosis and Hemostasis Unit, Rambam Health Care Campus, The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
› Author Affiliations

Abstract

Heparanase, the only mammalian enzyme known to degrade heparan sulfate chains, affects the hemostatic system through several mechanisms. Along with the degrading effect, heparanase engenders release of syndecan-1 from the cell surface and directly enhances the activity of the blood coagulation initiator, tissue factor, in the coagulation system. Upregulation of tissue factor and release of tissue factor pathway inhibitor from the cell surface contribute to the prothrombotic effect. Tissue factor pathway inhibitor and the strongest physiological anticoagulant antithrombin are attached to the endothelial cell surface by heparan sulfate. Hence, degradation of heparan sulfate induces further release of these two natural anticoagulants from endothelial cells. Elevated heparanase procoagulant activity and heparan sulfate chain levels in plasma, demonstrated in cancer, pregnancy, oral contraceptive use, and aging, could suggest a potential mechanism for increased risk of thrombosis in these clinical settings. In contrast to the blood circulation, accumulation of heparan sulfate chains in transudate and exudate pleural effusions induces a local anticoagulant milieu. The anticoagulant effect of heparan sulfate chains in other closed spaces such as peritoneal or subdural cavities should be further investigated.



Publication History

Article published online:
01 April 2021

© 2021. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Vlodavsky I, Elkin M, Abboud-Jarrous G. et al. Heparanase: one molecule with multiple functions in cancer progression. Connect Tissue Res 2008; 49 (03) 207-210
  • 2 Sanderson RD, Elkin M, Rapraeger AC, Ilan N, Vlodavsky I. Heparanase regulation of cancer, autophagy and inflammation: new mechanisms and targets for therapy. FEBS J 2017; 284 (01) 42-55
  • 3 Nadir Y, Vlodavsky I, Brenner B. Heparanase, tissue factor, and cancer. Semin Thromb Hemost 2008; 34 (02) 187-194
  • 4 Vlodavsky I, Eldor A, Haimovitz-Friedman A. et al. Expression of heparanase by platelets and circulating cells of the immune system: possible involvement in diapedesis and extravasation. Invasion Metastasis 1992; 12 (02) 112-127
  • 5 Parish CR, Freeman C, Hulett MD. Heparanase: a key enzyme involved in cell invasion. Biochim Biophys Acta 2001; 1471 (03) M99-M108
  • 6 Freeman C, Parish CR. Human platelet heparanase: purification, characterization and catalytic activity. Biochem J 1998; 330 (Pt 3): 1341-1350
  • 7 Goshen R, Hochberg AA, Korner G. et al. Purification and characterization of placental heparanase and its expression by cultured cytotrophoblasts. Mol Hum Reprod 1996; 2 (09) 679-684
  • 8 Tatour M, Shapira M, Axelman E. et al. Thrombin is a selective inducer of heparanase release from platelets and granulocytes via protease-activated receptor-1. Thromb Haemost 2017; 117 (07) 1391-1401
  • 9 Teien AN, Abildgaard U, Höök M. The anticoagulant effect of heparan sulfate and dermatan sulfate. Thromb Res 1976; 8 (06) 859-867
  • 10 Chappell D, Jacob M, Hofmann-Kiefer K. et al. Antithrombin reduces shedding of the endothelial glycocalyx following ischaemia/reperfusion. Cardiovasc Res 2009; 83 (02) 388-396
  • 11 Lwaleed BA, Bass PS. Tissue factor pathway inhibitor: structure, biology and involvement in disease. J Pathol 2006; 208 (03) 327-339
  • 12 Valentin S, Larnkjer A, Ostergaard P, Nielsen JI, Nordfang O. Characterization of the binding between tissue factor pathway inhibitor and glycosaminoglycans. Thromb Res 1994; 75 (02) 173-183
  • 13 Nadir Y, Brenner B, Gingis-Velitski S. et al. Heparanase induces tissue factor pathway inhibitor expression and extracellular accumulation in endothelial and tumor cells. Thromb Haemost 2008; 99 (01) 133-141
  • 14 Nadir Y, Brenner B, Zetser A. et al. Heparanase induces tissue factor expression in vascular endothelial and cancer cells. J Thromb Haemost 2006; 4 (11) 2443-2451
  • 15 Nadir Y, Brenner B, Fux L, Shafat I, Attias J, Vlodavsky I. Heparanase enhances the generation of activated factor X in the presence of tissue factor and activated factor VII. Haematologica 2010; 95 (11) 1927-1934
  • 16 Baker AB, Gibson WJ, Kolachalama VB. et al. Heparanase regulates thrombosis in vascular injury and stent-induced flow disturbance. J Am Coll Cardiol 2012; 59 (17) 1551-1560
  • 17 Nadir Y, Henig I, Naroditzky I, Paz B, Vlodavsky I, Brenner B. Involvement of Heparanase in early pregnancy losses. Thromb Res 2010; 125 (05) e251-e257
  • 18 Nadir Y, Kenig Y, Drugan A, Zcharia E, Brenner B. Involvement of heparanase in vaginal and cesarean section deliveries. Thromb Res 2010; 126 (06) e444-e450
  • 19 Nadir Y, Kenig Y, Drugan A, Shafat I, Brenner B. An assay to evaluate heparanase procoagulant activity. Thromb Res 2011; 128 (04) e3-e8
  • 20 Matan M, Axelman E, Brenner B, Nadir Y. Heparanase procoagulant activity is elevated in women using oral contraceptives. Hum Reprod 2013; 28 (09) 2372-2380
  • 21 Nadir Y, Sarig G, Axelman E. et al. Heparanase procoagulant activity is elevated and predicts survival in non-small cell lung cancer patients. Thromb Res 2014; 134 (03) 639-642
  • 22 Peled E, Rovitsky A, Axelman E, Norman D, Brenner B, Nadir Y. Increased heparanase level and procoagulant activity in orthopedic surgery patients receiving prophylactic dose of enoxaparin. Thromb Res 2012; 130 (01) 129-134
  • 23 Peled E, Melamed E, Portal TB. et al. Heparanase procoagulant activity as a predictor of wound necrosis following diabetic foot amputation. Thromb Res 2016; 139: 148-153
  • 24 Nadir Y, Saharov G, Hoffman R. et al. Heparanase procoagulant activity, factor Xa, and plasminogen activator inhibitor 1 are increased in shift work female nurses. Ann Hematol 2015; 94 (07) 1213-1219
  • 25 Hu Y, Atik A, Yu H. et al. Serum heparanase concentration and heparanase activity in patients with retinal vein occlusion. Acta Ophthalmol 2017; 95 (01) e62-e66
  • 26 Bayam E, Kalçık M, Gürbüz AS. et al. The relationship between heparanase levels, thrombus burden and thromboembolism in patients receiving unfractionated heparin treatment for prosthetic valve thrombosis. Thromb Res 2018; 171: 103-110
  • 27 Levi M. Pathogenesis and diagnosis of disseminated intravascular coagulation. Int J Lab Hematol 2018; 40 (Suppl. 01) 15-20
  • 28 Matan M, King D, Peled E. et al. Heparanase level and procoagulant activity are reduced in severe sepsis. Eur J Haematol 2018; 100 (02) 182-188
  • 29 Martin L, De Santis R, Koczera P. et al. The synthetic antimicrobial peptide 19-2.5 interacts with heparanase and heparan sulfate in murine and human sepsis. PLoS One 2015; 10 (11) e0143583
  • 30 Hofmann-Kiefer KF, Kemming GI, Chappell D. et al. Serum heparan sulfate levels are elevated in endotoxemia. Eur J Med Res 2009; 14: 526-531
  • 31 Xu X, Quiros RM, Maxhimer JB. et al. Inverse correlation between heparan sulfate composition and heparanase-1 gene expression in thyroid papillary carcinomas: a potential role in tumor metastasis. Clin Cancer Res 2003; 9 (16, Pt 1): 5968-5979
  • 32 Jung O, Trapp-Stamborski V, Purushothaman A. et al. Heparanase-induced shedding of syndecan-1/CD138 in myeloma and endothelial cells activates VEGFR2 and an invasive phenotype: prevention by novel synstatins. Oncogenesis 2016; 5: e202
  • 33 Yang Y, Macleod V, Miao HQ. et al. Heparanase enhances syndecan-1 shedding: a novel mechanism for stimulation of tumor growth and metastasis. J Biol Chem 2007; 282 (18) 13326-13333
  • 34 Maurice-Dror C, Litvak M, Keren-Politansky A, Ackerman S, Haim N, Nadir Y. Circulating heparan sulfate chains and body weight contribute to anti-Xa levels in cancer patients using the prophylactic dose of enoxaparin. J Thromb Thrombolysis 2020; 50 (01) 112-122
  • 35 Nevo N, Ghanem S, Crispel Y. et al. Heparanase level in the microcirculation as a possible modulator of the metastatic process. Am J Pathol 2019; 189 (08) 1654-1663
  • 36 Axelman E, Henig I, Crispel Y. et al. Novel peptides that inhibit heparanase activation of the coagulation system. Thromb Haemost 2014; 112 (03) 466-477
  • 37 Crispel Y, Axelman E, Tatour M. et al. Peptides inhibiting heparanase procoagulant activity significantly reduce tumour growth and vascularisation in a mouse model. Thromb Haemost 2016; 116 (04) 669-678
  • 38 Lanir N, Aharon A, Brenner B. Procoagulant and anticoagulant mechanisms in human placenta. Semin Thromb Hemost 2003; 29 (02) 175-184
  • 39 Haimov-Kochman R, Friedmann Y, Prus D. et al. Localization of heparanase in normal and pathological human placenta. Mol Hum Reprod 2002; 8 (06) 566-573
  • 40 McCarthy C, Cotter FE, McElwaine S. et al. Altered gene expression patterns in intrauterine growth restriction: potential role of hypoxia. Am J Obstet Gynecol 2007; 196 (01) 70.e1-70.e6
  • 41 Udagawa K, Yasumitsu H, Esaki M. et al. Subcellular localization of PP5/TFPI-2 in human placenta: a possible role of PP5/TFPI-2 as an anti-coagulant on the surface of syncytiotrophoblasts. Placenta 2002; 23 (2-3): 145-153
  • 42 Salem HT, Westergaard JG, Hindersson P, Lee JN, Grudzinskas JG, Chard T. Maternal serum levels of placental protein 5 in complications of late pregnancy. Obstet Gynecol 1982; 59 (04) 467-471
  • 43 Deitcher SR, Gomes MP. The risk of venous thromboembolic disease associated with adjuvant hormone therapy for breast carcinoma: a systematic review. Cancer 2004; 101 (03) 439-449
  • 44 Rosing J, Middeldorp S, Curvers J. et al. Low-dose oral contraceptives and acquired resistance to activated protein C: a randomised cross-over study. Lancet 1999; 354 (9195): 2036-2040
  • 45 Elkin M, Cohen I, Zcharia E. et al. Regulation of heparanase gene expression by estrogen in breast cancer. Cancer Res 2003; 63 (24) 8821-8826
  • 46 Treger S, Ackerman S, Kaplan V, Ghanem S, Nadir Y. Progesterone type affects the increase of heparanase level and pro-coagulant activity mediated by the estrogen receptor. Human Reproduction 2021; 36: 61-69
  • 47 Gunatillake T, Chui A, Fitzpatrick E. et al. Decreased placental glypican expression is associated with human fetal growth restriction. Placenta 2019; 76: 6-9
  • 48 Hofmann-Kiefer KF, Knabl J, Martinoff N. et al. Increased serum concentrations of circulating glycocalyx components in HELLP syndrome compared to healthy pregnancy: an observational study. Reprod Sci 2013; 20 (03) 318-325
  • 49 Oh JH, Kim YK, Jung JY, Shin JE, Chung JH. Changes in glycosaminoglycans and related proteoglycans in intrinsically aged human skin in vivo. Exp Dermatol 2011; 20 (05) 454-456
  • 50 Kurdykowski S, Mine S, Bardey V. et al. Ultraviolet-B irradiation induces epidermal up-regulation of heparanase expression and activity. J Photochem Photobiol B 2012; 106: 107-112
  • 51 Iriyama S, Matsunaga Y, Takahashi K, Matsuzaki K, Kumagai N, Amano S. Activation of heparanase by ultraviolet B irradiation leads to functional loss of basement membrane at the dermal-epidermal junction in human skin. Arch Dermatol Res 2011; 303 (04) 253-261
  • 52 Konno K, Arai H, Motomiya M. et al. A biochemical study on glycosaminoglycans (mucopolysaccharides) in emphysematous and in aged lungs. Am Rev Respir Dis 1982; 126 (05) 797-801
  • 53 Nishiguchi KM, Ushida H, Tomida D, Kachi S, Kondo M, Terasaki H. Age-dependent alteration of intraocular soluble heparan sulfate levels and its implications for proliferative diabetic retinopathy. Mol Vis 2013; 19: 1125-1131
  • 54 Nitschmann E, Berry L, Bridge S. et al. Morphological and biochemical features affecting the antithrombotic properties of the aorta in adult rabbits and rabbit pups. Thromb Haemost 1998; 79 (05) 1034-1040
  • 55 Nitschmann E, Berry L, Bridge S. et al. Morphologic and biochemical features affecting the antithrombotic properties of the inferior vena cava of rabbit pups and adult rabbits. Pediatr Res 1998; 43 (01) 62-67
  • 56 Campisi J. Aging, tumor suppression and cancer: high wire-act!. Mech Ageing Dev 2005; 126 (01) 51-58
  • 57 Bochenek ML, Bauer T, Gogiraju R. et al. The endothelial tumor suppressor p53 is essential for venous thrombus formation in aged mice. Blood Adv 2018; 2 (11) 1300-1314
  • 58 Hardak E, Peled E, Crispel Y. et al. Heparan sulfate chains contribute to the anticoagulant milieu in malignant pleural effusion. Thorax 2020; 75 (02) 143-152