1
|
Atiq F, Rawley O, O'Sullivan JM, Özbil M, Doherty D, Cooke N, Terraube V, Chion A, Amin A, Hulshof AM, Baci B, Byrne C, Aburawi HE, Lillicrap D, O'Donnell JS. R1205H (Vicenza) causes conformational changes in the von Willebrand factor D'D3 domains and enhances von Willebrand factor binding to clearance receptors LRP1 and SR-AI. J Thromb Haemost 2024; 22:2752-2760. [PMID: 38996914 PMCID: PMC11533894 DOI: 10.1016/j.jtha.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND von Willebrand factor (VWF)-R1205H variant (Vicenza) results in markedly enhanced VWF clearance in humans that has been shown to be largely macrophage-mediated. However, the biological mechanisms underlying this enhanced clearance remain poorly understood. OBJECTIVES This study aimed to investigate the roles of (i) specific VWF domains and (ii) different macrophage receptors in regulating enhanced VWF-R1205H clearance. METHODS In vivo clearance of full-length and truncated wild-type (WT)-VWF and VWF with R1205 substitutions was investigated in VWF-/- mice. Plate-binding assays were employed to characterize VWF binding to purified scavenger receptor class A member 1 (SR-AI), low-density lipoprotein receptor-related protein-1 (LRP1) cluster II or cluster IV receptors, and macrophage galactose-type lectin. RESULTS In full-length VWF missing the A1 domain, introduction of R1205H led to significantly enhanced clearance in VWF-/- mice compared with WT-VWF missing the A1 domain. Importantly, R1205H in a truncated VWF-D'D3 fragment also triggered increased clearance compared with WT-VWF-D'D3. Additional in vivo studies demonstrated that VWF-R1205K (which preserves the positive charge at 1205) exhibited normal clearance, whereas VWF-R1205E (which results in loss of the positive charge) caused significantly enhanced clearance, pinpointing the importance of the positive charge at VWF-R1205. In vitro plate-binding studies confirmed increased VWF-R1205H interaction with SR-AI compared with WT-VWF. Furthermore, significantly enhanced VWF-R1205H binding to LRP1 cluster IV (P < .001) and less marked enhanced binding to LRP1 cluster II (P = .034) was observed. In contrast, VWF-R1205H and WT-VWF demonstrated no difference in binding affinity to macrophage galactose-type lectin. CONCLUSION Disruption of the positive charge at amino acid R1205 causes conformational changes in the VWF-D'D3 domains and triggers enhanced LRP1-mediated and SR-AI-mediated clearance.
Collapse
Affiliation(s)
- Ferdows Atiq
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Orla Rawley
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mehmet Özbil
- Computational Biochemistry Group, Gebze Technical University, Institute of Biotechnology, Gebze, Kocaeli, Turkey
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Niamh Cooke
- BioMedicine Design, Pfizer, Grange Castle, Dublin, Ireland
| | | | - Alain Chion
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aamir Amin
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Anne-Marije Hulshof
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bogdan Baci
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ciara Byrne
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hanan E Aburawi
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; National Coagulation Centre, St James's Hospital, Dublin, Ireland.
| |
Collapse
|
2
|
Karampini E, Doherty D, Bürgisser PE, Garre M, Schoen I, Elliott S, Bierings R, O’Donnell JS. O-glycan determinants regulate VWF trafficking to Weibel-Palade bodies. Blood Adv 2024; 8:3254-3266. [PMID: 38640438 PMCID: PMC11226974 DOI: 10.1182/bloodadvances.2023012499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT von Willebrand factor (VWF) undergoes complex posttranslational modification within endothelial cells (ECs) before secretion. This includes significant N- and O-linked glycosylation. Previous studies have demonstrated that changes in N-linked glycan structures significantly influence VWF biosynthesis. In contrast, although abnormalities in VWF O-linked glycans (OLGs) have been associated with enhanced VWF clearance, their effect on VWF biosynthesis remains poorly explored. Herein, we report a novel role for OLG determinants in regulating VWF biosynthesis and trafficking within ECs. We demonstrate that alterations in OLGs (notably reduced terminal sialylation) lead to activation of the A1 domain of VWF within EC. In the presence of altered OLG, VWF multimerization is reduced and Weibel-Palade body (WPB) formation significantly impaired. Consistently, the amount of VWF secreted from WPB after EC activation was significantly reduced in the context of O-glycosylation inhibition. Finally, altered OLG on VWF not only reduced the amount of VWF secreted after EC activation but also affected its hemostatic efficacy. Notably, VWF secreted after WPB exocytosis consisted predominantly of low molecular weight multimers, and the length of tethered VWF string formation on the surface of activated ECs was significantly reduced. In conclusion, our data therefore support the hypothesis that alterations in O-glycosylation pathways directly affect VWF trafficking within human EC. These findings are interesting given that previous studies have reported altered OLG on plasma VWF (notably increased T-antigen expression) in patients with von Willebrand disease.
Collapse
Affiliation(s)
- Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Petra E. Bürgisser
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Massimiliano Garre
- Super-Resolution Imaging Consortium, Department of Chemistry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stephanie Elliott
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ruben Bierings
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Coagulation Centre, St James’s Hospital, Dublin, Ireland
| |
Collapse
|
3
|
Sialic acid-containing glycans play a role in the activity of snake venom proteases. Biochimie 2023; 204:140-153. [PMID: 36210615 DOI: 10.1016/j.biochi.2022.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/31/2022] [Accepted: 09/14/2022] [Indexed: 01/12/2023]
Abstract
Structural variability is a feature of snake venom proteins, and glycosylation is a post-translational modification that contributes to the diversification of venom proteomes. Studies by our group have shown that Bothrops venoms are distinctly defined by their glycoprotein content, and that most hybrid/complex N-glycans identified in these venoms contain sialic acid. Considering that metalloproteases and serine proteases are abundant components of Bothrops venoms and essential in the envenomation process, and that these enzymes contain several glycosylation sites, the role of sialic acid in venom proteolytic activity was evaluated. Here we show that removal of sialic acid by treatment of nine Bothrops venoms with neuraminidase (i) altered the pattern of gelatinolysis in zymography of most venoms and reduced the gelatinolytic activity of all venoms, (ii) decreased the proteolytic activity of some venoms on fibrinogen and the clotting activity of human plasma of all venoms, and (iii) altered the proteolysis profile of plasma proteins by B. jararaca venom, suggesting that sialic acid may play a role in the interaction of proteases with their protein substrates. In contrast, the profile of venom amidolytic activity on Bz-Arg-pNA did not change after removal of sialic acid, indicating that this monosaccharide is not essential in N-glycans of serine proteases acting on small substrates. In summary, these results expand the knowledge about the variability of the subproteomes of Bothrops venom proteases, and for the first time point to the importance of carbohydrate chains containing sialic acid in the enzymatic activities of venom proteases relevant in human envenomation.
Collapse
|
4
|
Akiyama M, Eura Y, Kokame K. Siglec-5 and Siglec-14 mediate the endocytosis of ADAMTS13. Thromb Res 2022; 219:49-59. [PMID: 36116391 DOI: 10.1016/j.thromres.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND The plasma metalloprotease ADAMTS13 regulates the thrombotic activity of the von Willebrand factor (VWF). ADAMTS13 is highly glycosylated and its carbohydrate chains are capped with sialic acid (SA). Thus, ADAMTS13 may interact with carbohydrate- and/or SA-binding plasma membrane receptors that are involved in the clearance of various plasma proteins. We have investigated ADAMTS13 endocytosis via Siglecs, which were originally identified as SA-binding immunoreceptor family proteins expressed on leukocytes and are also known as endocytic receptors. MATERIALS AND METHODS Endocytic internalization of fluorescently labeled ADAMTS13 into HEK293 cells expressing Siglecs was examined via fluorescence microscopy. In vitro binding of ADAMTS13 to the extracellular region of Siglec-5 was examined. Plasma ADAMTS13 activity in human Siglec-5-expressing mice was measured. RESULTS AND CONCLUSIONS Siglec-5- and Siglec-14-expressing cells internalized not only full-length ADAMTS13 (FL) but also the truncated form (MDTCS) at least partly in an SA-independent manner. Replacement of the V-set domain of Siglec-14 with that of Siglec-3 abrogated the internalization of ADAMTS13. ADAMTS13 directly bound to the extracellular region of Siglec-5 in vitro. Expression of Siglec-5 in the mouse liver resulted in a significant decrease in plasma ADAMTS13 activity. These results suggest that Siglec-5 and Siglec-14, which have nearly identical ligand-binding domains, may contribute to the regulation of plasma ADAMTS13 levels as endocytic receptors for ADAMTS13.
Collapse
Affiliation(s)
- Masashi Akiyama
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yuka Eura
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Koichi Kokame
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.
| |
Collapse
|
5
|
Huang X, Yao M, Tian P, Wong JYY, Li Z, Liu Z, Zhao JV. Shared genetic etiology and causality between COVID-19 and venous thromboembolism: evidence from genome-wide cross trait analysis and bi-directional Mendelian randomization study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.05.21.22275413. [PMID: 35665015 PMCID: PMC9164523 DOI: 10.1101/2022.05.21.22275413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Venous thromboembolism (VTE) occurs in up to one third patients with COVID-19. VTE and COVID-19 may share a common genetic architecture, which has not been clarified yet. To fill this gap, we leveraged summary-level genetic data from the latest COVID-19 host genetics consortium and UK Biobank and examined the shared genetic etiology and causal relationship between COVID-19 and VTE. The cross-trait analysis identified 8, 11, and 7 shared loci between VTE and severe COVID-19, COVID-19 hospitalization, SARS-CoV-2 infection respectively, in 13 genes involved in coagulation and immune function and enriched in the lung. Co-localization analysis identified eight shared loci in ABO, ADAMTS13 and FUT2 genes. Bi-direction Mendelian randomization suggested that VTE was associated with higher risks of all COVID-19 related traits, and SARS-CoV-2 infection was associated with higher risk of VTE. Our study provided timely evidence and novel insights into the genetic etiology between COVID-19 and VTE.
Collapse
|
6
|
Ward SE, O'Sullivan JM, Moran AB, Spencer DIR, Gardner RA, Sharma J, Fazavana J, Monopoli M, McKinnon TAJ, Chion A, Haberichter S, O'Donnell JS. Sialylation on O-linked glycans protects von Willebrand factor from macrophage galactose lectin-mediated clearance. Haematologica 2022; 107:668-679. [PMID: 33763999 PMCID: PMC8883566 DOI: 10.3324/haematol.2020.274720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Terminal sialylation determines the plasma half-life of von Willebrand factor (VWF). A role for macrophage galactose lectin (MGL) in regulating hyposialylated VWF clearance has recently been proposed. In this study, we showed that MGL influences physiological plasma VWF clearance. MGL inhibition was associated with a significantly extended mean residence time and 3-fold increase in endogenous plasma VWF antigen levels (P<0.05). Using a series of VWF truncations, we further demonstrated that the A1 domain of VWF is predominantly responsible for enabling the MGL interaction. Binding of both full-length and VWF-A1-A2-A3 to MGL was significantly enhanced in the presence of ristocetin (P<0.05), suggesting that the MGL-binding site in A1 is not fully accessible in globular VWF. Additional studies using different VWF glycoforms demonstrated that VWF O-linked glycans, clustered at either end of the A1 domain, play a key role in protecting VWF against MGLmediated clearance. Reduced sialylation has been associated with pathological, increased clearance of VWF in patients with von Willebrand disease. Herein, we demonstrate that specific loss of α2-3 linked sialylation from O-glycans results in markedly increased MGL-binding in vitro, and markedly enhanced MGL-mediated clearance of VWF in vivo. Our data further show that the asialoglycoprotein receptor (ASGPR) does not have a significant role in mediating the increased clearance of VWF following loss of O-sialylation. Conversely however, we observed that loss of N-linked sialylation from VWF drives enhanced circulatory clearance predominantly via the ASGPR. Collectively, our data support the hypothesis that in addition to regulating physiological VWF clearance, the MGL receptor works in tandem with ASGPR to modulate enhanced clearance of aberrantly sialylated VWF in the pathogenesis of von Willebrand disease.
Collapse
Affiliation(s)
- Soracha E Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Alan B Moran
- Ludger, Ltd., Culham Science Centre, Abingdon, Oxfordshire OX14 3EB, United Kingdom; Leiden University Medical Centre, Centre for Proteomics and Metabolomics, 2300 RC Leiden
| | | | | | - Jyotika Sharma
- Department of Basic Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota
| | - Judicael Fazavana
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Marco Monopoli
- Department of Chemistry, RCSI, 123 St. Stephen's Green, Dublin 2
| | - Thomas A J McKinnon
- Faculty of Medicine, Imperial College, Hammersmith Hospital, Ducane Road, London
| | - Alain Chion
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | | | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; National Coagulation Centre, St James's Hospital, Dublin.
| |
Collapse
|
7
|
O’Donnell AS, Fazavana J, O’Donnell JS. The von Willebrand factor - ADAMTS-13 axis in malaria. Res Pract Thromb Haemost 2022; 6:e12641. [PMID: 35128300 PMCID: PMC8804941 DOI: 10.1002/rth2.12641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 12/19/2022] Open
Abstract
Cerebral malaria (CM) continues to be associated with major morbidity and mortality, particularly in children aged <5 years in sub-Saharan Africa. Although the biological mechanisms underpinning severe malaria pathophysiology remain incompletely understood, studies have shown that cytoadhesion of malaria-infected erythrocytes to endothelial cells (ECs) within the cerebral microvasculature represents a key step in this process. Furthermore, these studies have also highlighted that marked EC activation, with secretion of Weibel-Palade bodies (WPBs), occurs at a remarkably early stage following malaria infection. As a result, plasma levels of proteins normally stored within WPBs (including high-molecular-weight von Willebrand factor [VWF] multimers, VWF propeptide, and angiopoietin-2) are significantly elevated. In this review, we provide an overview of recent studies that have identified novel roles through which these secreted WPB glycoproteins may directly facilitate malaria pathogenesis through a number of different platelet-dependent and platelet-independent pathways. Collectively, these emerging insights suggest that hemostatic dysfunction, and in particular disruption of the normal VWF-ADAMTS-13 axis, may be of specific importance in triggering cerebral microangiopathy. Defining the molecular mechanisms involved may offer the opportunity to develop novel targeted therapeutic approaches, which are urgently needed as the mortality rate associated with CM remains in the order of 20%.
Collapse
Affiliation(s)
- Andrew S. O’Donnell
- Department of PaediatricsUniversity Maternity Hospital LimerickLimerickIreland
| | - Judicael Fazavana
- Irish Centre for Vascular BiologySchool of Pharmacy & Biomolecular SciencesRoyal College of Surgeons in IrelandDublin 2Ireland
| | - James S. O’Donnell
- Irish Centre for Vascular BiologySchool of Pharmacy & Biomolecular SciencesRoyal College of Surgeons in IrelandDublin 2Ireland
- National Coagulation CentreSt James’s HospitalDublinIreland
- National Children’s Research CentreOur Lady’s Children’s Hospital CrumlinDublinIreland
| |
Collapse
|
8
|
Ward SE, Fogarty H, Karampini E, Lavin M, Schneppenheim S, Dittmer R, Morrin H, Glavey S, Ni Cheallaigh C, Bergin C, Martin-Loeches I, Mallon PW, Curley GF, Baker RI, Budde U, O'Sullivan JM, O'Donnell JS. ADAMTS13 regulation of VWF multimer distribution in severe COVID-19. J Thromb Haemost 2021; 19:1914-1921. [PMID: 34053187 PMCID: PMC8237059 DOI: 10.1111/jth.15409] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/11/2021] [Accepted: 05/27/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Consistent with fulminant endothelial cell activation, elevated plasma von Willebrand factor (VWF) antigen levels have been reported in patients with COVID-19. The multimeric size and function of VWF are normally regulated through A Disintegrin And Metalloprotease with ThrombSpondin Motif type 1 motif, member 13 (ADAMTS-13)--mediated proteolysis. OBJECTIVES This study investigated the hypothesis that ADAMTS-13 regulation of VWF multimer distribution may be impaired in severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection contributing to the observed microvascular thrombosis. PATIENTS AND METHODS Patients with COVID-19 (n = 23) were recruited from the Beaumont Hospital Intensive Care Unit (ICU) in Dublin. Plasma VWF antigen, multimer distribution, ADAMTS-13 activity, and known inhibitors thereof were assessed. RESULTS We observed markedly increased VWF collagen-binding activity in patients with severe COVID-19 compared to controls (median 509.1 versus 94.3 IU/dl). Conversely, plasma ADAMTS-13 activity was significantly reduced (median 68.2 IU/dl). In keeping with an increase in VWF:ADAMTS-13 ratio, abnormalities in VWF multimer distribution were common in patients with COVID-19, with reductions in high molecular weight VWF multimers. Terminal sialylation regulates VWF susceptibility to proteolysis by ADAMTS-13 and other proteases. We observed that both N- and O-linked sialylation were altered in severe COVID-19. Furthermore, plasma levels of the ADAMTS-13 inhibitors interleukin-6, thrombospondin-1, and platelet factor 4 were significantly elevated. CONCLUSIONS These findings support the hypothesis that SARS-CoV-2 is associated with profound quantitative and qualitative increases in plasma VWF levels, and a multifactorial down-regulation in ADAMTS-13 function. Further studies will be required to determine whether therapeutic interventions to correct ADAMTS-13-VWF multimer dysfunction may be useful in COVID-microvascular thrombosis and angiopathy.
Collapse
Affiliation(s)
- Soracha E Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Helen Fogarty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michelle Lavin
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Sonja Schneppenheim
- Department of Hämostaseology, Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Rita Dittmer
- Department of Hämostaseology, Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Hannah Morrin
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Siobhan Glavey
- Department of Haematology, Beaumont Hospital, Dublin, Ireland
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Colm Bergin
- St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Ignacio Martin-Loeches
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Patrick W Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Ireland
- St Vincent's University Hospital, Dublin, Ireland
| | - Gerard F Curley
- Department of Anaesthesia and Critical Care, RCSI, Dublin, Ireland
| | - Ross I Baker
- Western Australia Centre for Thrombosis and Haemostasis, Perth Blood Institute, Murdoch University, Perth, Australia
| | - Ulrich Budde
- Department of Hämostaseology, Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
9
|
Mobayen G, Dhutia A, Clarke C, Prendecki M, McAdoo S, Keniyopoullos R, Malik T, Laffan M, Willicombe M, McKinnon T. Severe COVID-19 is associated with endothelial activation and abnormal glycosylation of von Willebrand factor in patients undergoing hemodialysis. Res Pract Thromb Haemost 2021; 5:e12582. [PMID: 34532629 PMCID: PMC8435526 DOI: 10.1002/rth2.12582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND A major clinical feature of severe coronavirus diease 2019 (COVID-19) is microvascular thrombosis linked to endothelial cell activation. Consistent with this, a number of studies have shown that patients with severe COVID-19 have highly elevated plasma levels of von Willebrand Factor (VWF) that may contribute to the prothrombotic phenotype. In the current study, we investigated the extent of endothelial activation in patients receiving hemodialysis who had either mild or severe COVID-19. METHODS Plasma VWF, ADAMTS-13, angiopoietin-2 (Ang2), and syndecan-1 levels were determined by ELISA. The sialic acid content of VWF was investigated using a modified ELISA to measure elderberry bark lectin, specific for sialic acid residues, binding to VWF. RESULTS Patients receiving hemodialysis with severe COVID-19 had significantly higher plasma levels of VWF and lower ADAMTS-13. VWF levels peaked and were sustained during the first 10 days after positive confirmation of infection. While Ang2 trended toward being higher in severely ill patients, this did not reach significance; however, severely ill patients had significantly higher soluble syndecan-1 levels, with high levels related to risk of death. Finally, higher VWF levels in severely ill patients were correlated with lower VWF sialic acid content. CONCLUSIONS Severe COVID-19 in patients undergoing hemodialysis is associated with both acute and sustained activation of the endothelium, leading to alteration of the VWF/ADAMTS-13 axis. Lower VWF sialic acid content represents altered VWF processing and further confirms the disturbance caused to the endothelium in COVID-19.
Collapse
Affiliation(s)
- Golzar Mobayen
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Amrita Dhutia
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Candice Clarke
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Maria Prendecki
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Stephen McAdoo
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Renos Keniyopoullos
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Talat Malik
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Michael Laffan
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Michelle Willicombe
- Department of Immunology and InflammationCentre for Inflammatory DiseaseCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| | - Thomas McKinnon
- Department of Immunology and InflammationCentre for HaematologyCommonwealth BuildingHammersmith CampusImperial College of Science Technology and MedicineLondonUK
| |
Collapse
|
10
|
Ward S, O'Sullivan JM, O'Donnell JS. The Biological Significance of von Willebrand Factor O-Linked Glycosylation. Semin Thromb Hemost 2021; 47:855-861. [PMID: 34130346 DOI: 10.1055/s-0041-1726373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glycosylation is a key posttranslational modification, known to occur on more than half of all secreted proteins in man. As such, the role of N- and O-linked glycan structures in modulating various aspects of protein biology is an area of much research. Given their prevalence, it is perhaps unsurprising that variations in glycan structures have been demonstrated to play critical roles in modulating protein function and have been implicated in the pathophysiology of human diseases. von Willebrand factor (VWF), a plasma glycoprotein that is essential for normal hemostasis, is heavily glycosylated, containing 13 N-linked and 10 O-linked glycans. Together, these carbohydrate chains account for 20% of VWF monomeric mass, and have been shown to modulate VWF structure, function, and half-life. In this review, we focus on the specific role played by O-linked glycans in modulating VWF biology. Specifically, VWF O-linked glycans have been shown to modulate tertiary protein structure, susceptibility to ADAMTS13 proteolysis, platelet tethering, and VWF circulatory half-life.
Collapse
Affiliation(s)
- Soracha Ward
- Haemostasis Research Group, Irish Centre for Vascular Biology, School of Pharmacy and Bimolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jamie M O'Sullivan
- Haemostasis Research Group, Irish Centre for Vascular Biology, School of Pharmacy and Bimolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - James S O'Donnell
- Haemostasis Research Group, Irish Centre for Vascular Biology, School of Pharmacy and Bimolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Coagulation Centre, St James's Hospital, Dublin, Ireland
| |
Collapse
|
11
|
Goumidi L, Thibord F, Wiggins KL, Li-Gao R, Brown MR, van Hylckama Vlieg A, Souto JC, Soria JM, Ibrahim-Kosta M, Saut N, Daian D, Olaso R, Amouyel P, Debette S, Boland A, Bailly P, Morrison AC, Mook-Kanamori DO, Deleuze JF, Johnson A, de Vries PS, Sabater-Lleal M, Chiaroni J, Smith NL, Rosendaal FR, Chasman DI, Trégouët DA, Morange PE. Association between ABO haplotypes and the risk of venous thrombosis: impact on disease risk estimation. Blood 2021; 137:2394-2402. [PMID: 33512453 PMCID: PMC8085481 DOI: 10.1182/blood.2020008997] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Genetic risk score (GRS) analysis is a popular approach to derive individual risk prediction models for complex diseases. In venous thrombosis (VT), such type of analysis shall integrate information at the ABO blood group locus, which is one of the major susceptibility loci. However, there is no consensus about which single nucleotide polymorphisms (SNPs) must be investigated when properly assessing association between ABO locus and VT risk. Using comprehensive haplotype analyses of ABO blood group tagging SNPs in 5425 cases and 8445 controls from 6 studies, we demonstrate that using only rs8176719 (tagging O1) to correctly assess the impact of ABO locus on VT risk is suboptimal, because 5% of rs8176719-delG carriers do not have an increased risk of developing VT. Instead, we recommend the use of 4 SNPs, rs2519093 (tagging A1), rs1053878 (A2), rs8176743 (B), and rs41302905 (O2), when assessing the impact of ABO locus on VT risk to avoid any risk misestimation. Compared with the O1 haplotype, the A2 haplotype is associated with a modest increase in VT risk (odds ratio, ∼1.2), the A1 and B haplotypes are associated with an ∼1.8-fold increased risk, whereas the O2 haplotype tends to be slightly protective (odds ratio, ∼0.80). In addition, although the A1 and B blood groups are associated with increased von Willebrand factor and factor VIII plasma levels, only the A1 blood group is associated with ICAM levels, but in an opposite direction, leaving additional avenues to be explored to fully understand the spectrum of biological effects mediated by ABO locus on cardiovascular traits.
Collapse
Affiliation(s)
- Louisa Goumidi
- Aix Marseille University, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Centre de Recherche en CardioVasculaire et Nutrition, Marseille, France
| | - Florian Thibord
- INSERM U1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
- Laboratory of Excellence (LabEx) Genomique Médicale, Evry, France
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mickael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | | | - Joan-Carles Souto
- Thrombosis and Hemostasis Research Group, Sant Pau Institute of Biomedical Research (IIB Sant Pau), Barcelona, Spain
- Unit of Hemostasis and Thrombosis, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - José-Manuel Soria
- Unit of Genomic of Complex Disease, Institut de Recerca Hospital de la Sant Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain
| | - Manal Ibrahim-Kosta
- Aix Marseille University, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Centre de Recherche en CardioVasculaire et Nutrition, Marseille, France
- Hematology Laboratory, La Timone University Hospital of Marseille, Marseille, France
| | - Noémie Saut
- Aix Marseille University, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Centre de Recherche en CardioVasculaire et Nutrition, Marseille, France
- Hematology Laboratory, La Timone University Hospital of Marseille, Marseille, France
| | - Delphine Daian
- Laboratory of Excellence (LabEx) Genomique Médicale, Evry, France
- Université Paris-Saclay, Commissariat à l'Energie Atomique, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Robert Olaso
- Laboratory of Excellence (LabEx) Genomique Médicale, Evry, France
- Université Paris-Saclay, Commissariat à l'Energie Atomique, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Philippe Amouyel
- Lille University, INSERM, Institut Pasteur de Lille, Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement (RID-AGE), LabEx Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease (DISTALZ), Lille, France
- Lille University, INSERM, Centre Hospitalier Universitaire (CHU) Lille, Institut Pasteur de Lille, RID-AGE, Lille, France
| | - Stéphanie Debette
- INSERM U1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
- Department of Neurology, CHU de Bordeaux, Bordeaux, France
| | - Anne Boland
- Laboratory of Excellence (LabEx) Genomique Médicale, Evry, France
- Université Paris-Saclay, Commissariat à l'Energie Atomique, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Pascal Bailly
- Etablissement Français du Sang Provence-Alpes-Côte d'Azur-Corse "Biologie des Groupes Sanguins," Marseille, France
- Aix Marseille University, Etablissement Français du Sang, Centre National pour la Recherche Scientifique, Anthropologie Bio-Culturelle, Droit, Ethique et Santé, "Biologie des Groupes Sanguins," Marseille, France
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | - Denis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean-François Deleuze
- Laboratory of Excellence (LabEx) Genomique Médicale, Evry, France
- Université Paris-Saclay, Commissariat à l'Energie Atomique, Centre National de Recherche en Génomique Humaine, Evry, France
- Centre d'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Andrew Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | - Maria Sabater-Lleal
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, Barcelona, Spain
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jacques Chiaroni
- Etablissement Français du Sang Provence-Alpes-Côte d'Azur-Corse "Biologie des Groupes Sanguins," Marseille, France
- Aix Marseille University, Etablissement Français du Sang, Centre National pour la Recherche Scientifique, Anthropologie Bio-Culturelle, Droit, Ethique et Santé, "Biologie des Groupes Sanguins," Marseille, France
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Unit, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA; and
- Department of Medicine, Harvard Medical School, Boston, MA
| | - David-Alexandre Trégouët
- INSERM U1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
- Laboratory of Excellence (LabEx) Genomique Médicale, Evry, France
| | - Pierre-Emmanuel Morange
- Aix Marseille University, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Centre de Recherche en CardioVasculaire et Nutrition, Marseille, France
- Laboratory of Excellence (LabEx) Genomique Médicale, Evry, France
- Hematology Laboratory, La Timone University Hospital of Marseille, Marseille, France
| |
Collapse
|
12
|
The relationship between ABO blood group, von Willebrand factor, and primary hemostasis. Blood 2021; 136:2864-2874. [PMID: 32785650 DOI: 10.1182/blood.2020005843] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
Numerous studies have reported significant associations between ABO blood group and risk of cardiovascular disease. These studies have consistently demonstrated that thrombotic risk is significantly reduced in individuals in blood group O. Nevertheless, the biological mechanisms through which ABO influences hemostasis have remained poorly understood. Exciting recent data have provided novel insights into how these ABO effects are modulated and have highlighted that ABO group significantly influences platelet plug formation at sites of vascular injury (primary hemostasis). In particular, ABO affects multiple aspects of von Willebrand factor (VWF) biology. In keeping with their reduced thrombotic risk, plasma VWF levels are ∼25% lower in healthy group O compared with healthy group non-O individuals. In addition, blood group O VWF demonstrates enhanced susceptibility to ADAMTS13 proteolysis. Finally, preliminary findings suggest that the interaction of group O VWF with platelets may also be reduced. Although the molecular mechanisms underlying these ABO effects on VWF have not been fully elucidated, it seems likely that they are mediated in large part by the ABO(H) carbohydrate structures that are carried on both the N- and O-linked glycans of VWF. Interestingly, ABO(H) determinants are also expressed on several different platelet surface glycoprotein receptors. Recent studies support the hypothesis that ABO group not only exerts major quantitative and qualitative effects on VWF, but also affect specific aspects of platelet function. Given the severe morbidity and the mortality associated with thrombotic disorders, defining the mechanisms underlying these ABO effects is not only of scientific interest, but also of direct clinical importance.
Collapse
|
13
|
Kragh T, Pekrul I, Ott HW, Spannagl M, Möhnle P. A novel approach to laboratory assessment and reporting of platelet von Willebrand factor. Platelets 2021; 33:242-248. [PMID: 33427003 DOI: 10.1080/09537104.2020.1869715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The interaction of platelets with von Willebrand factor is essential for primary hemostasis. Concentration and activity of plasma von Willebrand factor are routine parameters in the assessment of hemostasis disorders. In addition to plasma von Willebrand factor, platelet von Willebrand factor, synthesized in megakaryocytes and stored in α-granules of circulating platelets, is known to contribute to primary hemostasis and the microenvironment of thrombus formation. The laboratory assessment of platelet von Willebrand factor however is cumbersome and not widely established as a routine parameter. We here propose a method for laboratory assessment and reporting of platelet von Willebrand factor potentially useful for laboratory routines in specialized laboratories. Our model allows to describe platelet von Willebrand factor as 1. the concentration of platelet von Willebrand factor in whole blood, 2. the amount of platelet von Willebrand factor in a sample with a defined concentration of 1000 platelets/nl, and 3. the concentration of platelet von Willebrand factor in one platelet. According to our results in healthy individuals, the proportion of platelet von Willebrand factor activity is estimated to be about 10% of total von Willebrand factor in human plasma under physiological circumstances. The concentration of platelet von Willebrand factor is estimated to be 0.4 IU/ml in a sample with a defined concentration of 1000 platelets/nl and to be about 42 IU/ml in one platelet (both expressed as VWF:Ag).
Collapse
Affiliation(s)
- Thorsten Kragh
- Department of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU Klinikum, München, Germany
| | - Isabell Pekrul
- Department of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU Klinikum, München, Germany.,Department of Anaesthesiology, LMU Klinikum, München, Germany
| | | | - Michael Spannagl
- Department of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU Klinikum, München, Germany
| | - Patrick Möhnle
- Department of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU Klinikum, München, Germany
| |
Collapse
|
14
|
Yu X, Shi H, Li Y, Guo Y, Zhang P, Wang G, Li L, Chen X, Ding L, Ju H. Thermally Triggered, Cell-Specific Enzymatic Glyco-Editing: In Situ Regulation of Lectin Recognition and Immune Response on Target Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:54387-54398. [PMID: 33236873 DOI: 10.1021/acsami.0c15212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In situ glyco-editing on the cell surface can endow cellular glycoforms with new structures and properties; however, the lack of cell specificity and dependence on cells' endogenous functions plague the revelation of cellular glycan recognition properties and hamper the application of glyco-editing in complicated authentic biosystems. Herein, we develop a thermally triggered, cell-specific glyco-editing method for regulation of lectin recognition on target live cells in both single- and cocultured settings. The method relies on the aptamer-mediated anchoring of microgel-encapsulated neuraminidase on target cells and subsequent thermally triggered enzyme release for localized sialic acid (Sia) trimming. This temperature-based enzyme accessibility modulation strategy exempts genetic or metabolic engineering operations and, thus for the first time, enables tumor-specific desialylation on complicated tissue slices. The proposed method also provides an unprecedented opportunity to potentiate the innate immune response of natural killer cells toward target tumor cells through thermally triggered cell-specific desialylation, which paves the way for in vivo glycoimmune-checkpoint-targeted cancer therapeutic intervention.
Collapse
Affiliation(s)
- Xiaofei Yu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Huifang Shi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yuna Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Peiwen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Guyu Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Lei Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xian Chen
- Jiangsu Province Blood Center, Nanjing 210008, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
15
|
Matsui T, Nakamura Y. Von Willebrand Factor and ABO Blood Group. TRENDS GLYCOSCI GLYC 2020. [DOI: 10.4052/tigg.1842.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Taei Matsui
- Department of Molecular Laboratory Medicine, Fujita Health University Graduate School of Health Sciences
| | - Yuta Nakamura
- Department of Molecular Laboratory Medicine, Fujita Health University Graduate School of Health Sciences
| |
Collapse
|
16
|
Affiliation(s)
- Taei Matsui
- Department of Molecular Laboratory Medicine, Fujita Health University Graduate School of Health Sciences
| | - Yuta Nakamura
- Department of Molecular Laboratory Medicine, Fujita Health University Graduate School of Health Sciences
| |
Collapse
|
17
|
Ibrahim-Kosta M, Bailly P, Silvy M, Saut N, Suchon P, Morange PE, Chiaroni J, Trégouët DA, Goumidi L. ABO blood group, glycosyltransferase activity and risk of venous thromboembolism. Thromb Res 2020; 193:31-35. [DOI: 10.1016/j.thromres.2020.05.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/20/2020] [Accepted: 05/29/2020] [Indexed: 01/06/2023]
|
18
|
Common Genetic Variants in ABO and CLEC4M Modulate the Pharmacokinetics of Recombinant FVIII in Severe Hemophilia A Patients. Thromb Haemost 2020; 120:1395-1406. [PMID: 32726853 DOI: 10.1055/s-0040-1714214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The pharmacokinetic (PK) response of severe hemophilia A (HA) patients to infused factor VIII (FVIII) shows substantial variability. Several environmental and genetic factors are associated with changes in FVIII plasma levels and infused FVIII PK. Based on the hypothesis that factors influencing endogenous FVIII can affect FVIII PK, the contribution of single-nucleotide variants (SNVs) in candidate genes was investigated in 51 severe HA patients. The effects of blood group, F8 variant type, von Willebrand factor antigen and activity levels, age, and weight were also explored. The myPKFiT device was used to estimate individual PK parameters, and SNVs and clinically reportable F8 variants were simultaneously analyzed in an Illumina MiSeq instrument, using the microfluidics-based Fluidigm Access Array system. The contribution of SNVs to FVIII half-life and clearance was addressed by robust regression modeling, taking into account other modulators. In line with previous studies, we provide robust evidence that age, body weight, and blood group, as well as SNVs in ABO and CLEC4M, participate in the variability of FVIII PK in HA patients. Main results: each copy of the rs7853989 (ABO) allele increases FVIII half-life by 1.4 hours (p = 0.0131) and decreases clearance by 0.5 mL/h/kg (p = 5.57E-03), whereas each additional rs868875 (CLEC4M) allele reduces FVIII half-life by 1.1 hours (p = 2.90E-05) and increases clearance by 0.3 mL/h/kg (p = 1.01E-03). These results contribute to advancing efforts to improve FVIII replacement therapies by adjusting to each patient's PK profile based on pharmacogenomic data. This personalized medicine will decrease the burden of treatment and maximize the benefits obtained.
Collapse
|
19
|
Kang T, Boland BB, Jensen P, Alarcon C, Nawrocki A, Grimsby JS, Rhodes CJ, Larsen MR. Characterization of Signaling Pathways Associated with Pancreatic β-cell Adaptive Flexibility in Compensation of Obesity-linked Diabetes in db/db Mice. Mol Cell Proteomics 2020; 19:971-993. [PMID: 32265294 PMCID: PMC7261816 DOI: 10.1074/mcp.ra119.001882] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/03/2020] [Indexed: 12/20/2022] Open
Abstract
The onset of obesity-linked type 2 diabetes (T2D) is marked by an eventual failure in pancreatic β-cell function and mass that is no longer able to compensate for the inherent insulin resistance and increased metabolic load intrinsic to obesity. However, in a commonly used model of T2D, the db/db mouse, β-cells have an inbuilt adaptive flexibility enabling them to effectively adjust insulin production rates relative to the metabolic demand. Pancreatic β-cells from these animals have markedly reduced intracellular insulin stores, yet high rates of (pro)insulin secretion, together with a substantial increase in proinsulin biosynthesis highlighted by expanded rough endoplasmic reticulum and Golgi apparatus. However, when the metabolic overload and/or hyperglycemia is normalized, β-cells from db/db mice quickly restore their insulin stores and normalize secretory function. This demonstrates the β-cell's adaptive flexibility and indicates that therapeutic approaches applied to encourage β-cell rest are capable of restoring endogenous β-cell function. However, mechanisms that regulate β-cell adaptive flexibility are essentially unknown. To gain deeper mechanistic insight into the molecular events underlying β-cell adaptive flexibility in db/db β-cells, we conducted a combined proteomic and post-translational modification specific proteomic (PTMomics) approach on islets from db/db mice and wild-type controls (WT) with or without prior exposure to normal glucose levels. We identified differential modifications of proteins involved in redox homeostasis, protein refolding, K48-linked deubiquitination, mRNA/protein export, focal adhesion, ERK1/2 signaling, and renin-angiotensin-aldosterone signaling, as well as sialyltransferase activity, associated with β-cell adaptive flexibility. These proteins are all related to proinsulin biosynthesis and processing, maturation of insulin secretory granules, and vesicular trafficking-core pathways involved in the adaptation of insulin production to meet metabolic demand. Collectively, this study outlines a novel and comprehensive global PTMome signaling map that highlights important molecular mechanisms related to the adaptive flexibility of β-cell function, providing improved insight into disease pathogenesis of T2D.
Collapse
Affiliation(s)
- Taewook Kang
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark; The Danish Diabetes Academy, Odense, Denmark
| | - Brandon B Boland
- The Kovler Diabetes Center, Department of Medicine Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, Illinois 60637; Cardiovascular, Renal and Metabolic Disease, BioPharmaceuticals Research and Development, AstraZeneca Gaithersburg, Maryland 20878
| | - Pia Jensen
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Cristina Alarcon
- The Kovler Diabetes Center, Department of Medicine Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, Illinois 60637
| | - Arkadiusz Nawrocki
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Joseph S Grimsby
- Cardiovascular, Renal and Metabolic Disease, BioPharmaceuticals Research and Development, AstraZeneca Gaithersburg, Maryland 20878
| | - Christopher J Rhodes
- The Kovler Diabetes Center, Department of Medicine Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, Illinois 60637; Cardiovascular, Renal and Metabolic Disease, BioPharmaceuticals Research and Development, AstraZeneca Gaithersburg, Maryland 20878
| | - Martin R Larsen
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark.
| |
Collapse
|
20
|
Johnston I, Sarkar A, Hayes V, Koma GT, Arepally GM, Chen J, Chung DW, López JA, Cines DB, Rauova L, Poncz M. Recognition of PF4-VWF complexes by heparin-induced thrombocytopenia antibodies contributes to thrombus propagation. Blood 2020; 135:1270-1280. [PMID: 32077913 PMCID: PMC7146020 DOI: 10.1182/blood.2018881607] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/23/2020] [Indexed: 01/19/2023] Open
Abstract
Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder mediated by complexes between platelet factor 4 (PF4) and heparin or other polyanions, but the risk of thrombosis extends beyond exposure to heparin implicating other PF4 partners. We recently reported that peri-thrombus endothelium is targeted by HIT antibodies, but the binding site(s) has not been identified. We now show that PF4 binds at multiple discrete sites along the surface of extended strings of von Willebrand factor (VWF) released from the endothelium following photochemical injury in an endothelialized microfluidic system under flow. The HIT-like monoclonal antibody KKO and HIT patient antibodies recognize PF4-VWF complexes, promoting platelet adhesion and enlargement of thrombi within the microfluidic channels. Platelet adhesion to the PF4-VWF-HIT antibody complexes is inhibited by antibodies that block FcγRIIA or the glycoprotein Ib-IX complex on platelets. Disruption of PF4-VWF-HIT antibody complexes by drugs that prevent or block VWF oligomerization attenuate thrombus formation in a murine model of HIT. Together, these studies demonstrate assembly of HIT immune complexes along VWF strings released by injured endothelium that might propagate the risk of thrombosis in HIT. Disruption of PF4-VWF complex formation may provide a new therapeutic approach to HIT.
Collapse
Affiliation(s)
- Ian Johnston
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pharmacology and
| | - Amrita Sarkar
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Vincent Hayes
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
- Departments of Pathology and Laboratory Medicine and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gavin T Koma
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | | | | | - Dominic W Chung
- Bloodworks Northwest, Seattle, WA
- Departments of Medicine and Biochemistry, University of Washington, Seattle, WA; and
| | - José A López
- Bloodworks Northwest, Seattle, WA
- Departments of Medicine and Biochemistry, University of Washington, Seattle, WA; and
| | - Douglas B Cines
- Departments of Pathology and Laboratory Medicine and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Lubica Rauova
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mortimer Poncz
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
21
|
Ward S, O'Sullivan JM, O'Donnell JS. von Willebrand factor sialylation-A critical regulator of biological function. J Thromb Haemost 2019; 17:1018-1029. [PMID: 31055873 DOI: 10.1111/jth.14471] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/24/2019] [Indexed: 12/29/2022]
Abstract
Essentials Von Willebrand Factor (VWF) is extensively glycosylated with serial studies demonstrating that these carbohydrate determinants play critical roles in regulating multiple aspects of VWF biology. Terminal sialic acid residues, expressed on both the N- and O-linked glycans of VWF, regulate VWF functional activity, susceptibility to proteolysis and plasma clearance in vivo. Quantitative and qualitative variations in VWF sialylation have been reported in patients with von Willebrand Disease, as well as in a number of other physiological and pathological states. Further studies are warranted to define the molecular mechanisms through which N- and O-linked sialylation impacts upon the multiple biological activities of VWF. von Willebrand factor (VWF) undergoes complex post-translational modification prior to its secretion into the plasma. Consequently, VWF monomers contain complex N-glycan and O-glycan structures that, together, account for approximately 20% of the final monomeric mass. An increasing body of evidence has confirmed that these carbohydrate determinants play critical roles in regulating multiple aspects of VWF biology. In particular, studies have demonstrated that terminal ABO blood group has an important effect on plasma VWF levels. This effect is interesting, given that only 15% of the N-glycans and 1% of the O-glycans of VWF actually express terminal ABO(H) determinants. In contrast, the vast majority of the N-glycans and O-glycans on human VWF are capped by terminal negatively charged sialic acid residues. Recent data suggest that sialylation significantly regulates VWF functional activity, susceptibility to proteolysis, and clearance, through a number of independent pathways. These findings are of direct clinical relevence, in that quantitative and qualitative variations in VWF sialylation have been described in patients with VWD, as well as in patients with a number of other physiologic and pathologic conditions. Moreover, platelet-derived VWF is significantly hyposialylated as compared with plasma-derived VWF, whereas the recently licensed recombinant VWF therapeutic is hypersialylated. In this review, we examine the evidence supporting the hypothesis that VWF sialylation plays multiple biological roles. In addition, we consider data suggesting that quantitative and qualitative variations in VWF sialylation may play specific roles in the pathogenesis of VWD, and that sialic acid expression on VWF may also differ across a number of other physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Soracha Ward
- Haemostasis Research Group, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jamie M O'Sullivan
- Haemostasis Research Group, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - James S O'Donnell
- Haemostasis Research Group, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| |
Collapse
|
22
|
Stowell SR, Stowell CP. Biologic roles of the ABH and Lewis histo-blood group antigens part II: thrombosis, cardiovascular disease and metabolism. Vox Sang 2019; 114:535-552. [PMID: 31090093 DOI: 10.1111/vox.12786] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
The ABH and Lewis antigens were among the first of the human red blood cell polymorphisms to be identified and, in the case of the former, play a dominant role in transfusion and transplantation. But these two therapies are largely twentieth-century innovations, and the ABH and related carbohydrate antigens are not only expressed on a very wide range of human tissues, but were present in primates long before modern humans evolved. Although we have learned a great deal about the biochemistry and genetics of these structures, the biological roles that they play in human health and disease are incompletely understood. This review and its companion, which appeared in a previous issue of Vox Sanguinis, will focus on a few of the biologic and pathologic processes which appear to be affected by histo-blood group phenotype. The first of the two reviews explored the interactions of two bacteria with the ABH and Lewis glycoconjugates of their human host cells, and described the possible connections between the immune response of the human host to infection and the development of the AB-isoagglutinins. This second review will describe the relationship between ABO phenotype and thromboembolic disease, cardiovascular disease states, and general metabolism.
Collapse
Affiliation(s)
- Sean R Stowell
- Center for Apheresis, Center for Transfusion and Cellular Therapies, Emory Hospital, Emory University School of Medicine, Atlanta, GA, USA.,Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher P Stowell
- Blood Transfusion Service, Massachusetts General Hospital, Boston, MA, USA.,Department of Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Increased galactose expression and enhanced clearance in patients with low von Willebrand factor. Blood 2019; 133:1585-1596. [DOI: 10.1182/blood-2018-09-874636] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/06/2019] [Indexed: 11/20/2022] Open
Abstract
Abstract
Glycan determinants on von Willebrand factor (VWF) play critical roles in regulating its susceptibility to proteolysis and clearance. Abnormal glycosylation has been shown to cause von Willebrand disease (VWD) in a number of different mouse models. However, because of the significant technical challenges associated with accurate assessment of VWF glycan composition, the importance of carbohydrates in human VWD pathogenesis remains largely unexplored. To address this, we developed a novel lectin-binding panel to enable human VWF glycan characterization. This methodology was then used to study glycan expression in a cohort of 110 patients with low VWF compared with O blood group-matched healthy controls. Interestingly, significant interindividual heterogeneity in VWF glycan expression was seen in the healthy control population. This variation included terminal sialylation and ABO(H) blood group expression on VWF. Importantly, we also observed evidence of aberrant glycosylation in a subgroup of patients with low VWF. In particular, terminal α(2-6)-linked sialylation was reduced in patients with low VWF, with a secondary increase in galactose (Gal) exposure. Furthermore, an inverse correlation between Gal exposure and estimated VWF half-life was observed in those patients with enhanced VWF clearance. Together, these findings support the hypothesis that loss of terminal sialylation contributes to the pathophysiology underpinning low VWF in at least a subgroup of patients by promoting enhanced clearance. In addition, alterations in VWF carbohydrate expression are likely to contribute to quantitative and qualitative variations in VWF levels in the normal population. This trial was registered at www.clinicaltrials.gov as #NCT03167320.
Collapse
|
24
|
Blood group alters platelet binding kinetics to von Willebrand factor and consequently platelet function. Blood 2019; 133:1371-1377. [PMID: 30642918 DOI: 10.1182/blood-2018-06-855528] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 01/03/2019] [Indexed: 01/29/2023] Open
Abstract
Blood type O is associated with a lower risk of myocardial infarction. Platelets play a critical role in myocardial infarction. It is not known whether the expression of blood group antigens on platelet proteins alters platelet function; we hypothesized that platelet function would be different between donors with blood type O and those with non-O. To address this hypothesis, we perfused blood from healthy type O donors (n = 33) or non-O donors (n = 54) over pooled plasma derived von Willebrand factor (VWF) protein and purified blood type-specific VWF at arterial shear and measured platelet translocation dynamics. We demonstrate for the first time that type O platelets travel farther at greater speeds before forming stable bonds with VWF. To further characterize these findings, we used a novel analytical model of platelet interaction. Modeling revealed that the kinetics for GPIb/VWF binding rate are significantly lower for type O compared with non-O platelets. Our results demonstrate that platelets from type O donors interact less with VWF at arterial shear than non-O platelets. Our results suggest a potential mechanism for the reduced risk of myocardial infarction associated with blood type O.
Collapse
|
25
|
O'Sullivan JM, Ward S, Lavin M, O'Donnell JS. von Willebrand factor clearance - biological mechanisms and clinical significance. Br J Haematol 2018; 183:185-195. [DOI: 10.1111/bjh.15565] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jamie M. O'Sullivan
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Soracha Ward
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Michelle Lavin
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
- National Coagulation Centre; St James's Hospital; Dublin Ireland
| | - James S. O'Donnell
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
- National Coagulation Centre; St James's Hospital; Dublin Ireland
| |
Collapse
|
26
|
van der Vorm LN, Remijn JA, de Laat B, Huskens D. Effects of Plasmin on von Willebrand Factor and Platelets: A Narrative Review. TH OPEN 2018; 2:e218-e228. [PMID: 31249945 PMCID: PMC6524877 DOI: 10.1055/s-0038-1660505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/03/2018] [Indexed: 02/01/2023] Open
Abstract
Plasmin is the major fibrinolytic protease responsible for dissolving thrombi by cleavage of its primary substrate fibrin. In addition, emerging evidence points to other roles of plasmin: (1) as a back-up for ADAMTS13 in proteolysis of ultra-large von Willebrand factor (VWF) multimers and (2) as an activator of platelets. Although the molecular mechanisms of fibrinolysis are well defined, insights on the effects of plasmin on VWF and platelets are relatively scarce and sometimes conflicting. Hence, this review provides an overview of the literature on the effects of plasmin on VWF multimeric structures, on VWF binding to platelets, and on platelet activation. This information is placed in the context of possible applications of thrombolytic therapy for the condition thrombotic thrombocytopenic purpura.
Collapse
Affiliation(s)
- Lisa N van der Vorm
- Synapse Research Institute, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Chemistry and Hematology, Gelre Hospitals, Apeldoorn, The Netherlands
| | - Jasper A Remijn
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Chemistry and Hematology, Gelre Hospitals, Apeldoorn, The Netherlands
| | - Bas de Laat
- Synapse Research Institute, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Chemistry and Hematology, Gelre Hospitals, Apeldoorn, The Netherlands
| | - Dana Huskens
- Synapse Research Institute, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
27
|
A novel role for the macrophage galactose-type lectin receptor in mediating von Willebrand factor clearance. Blood 2017; 131:911-916. [PMID: 29282218 DOI: 10.1182/blood-2017-06-787853] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/12/2017] [Indexed: 01/22/2023] Open
Abstract
Previous studies have shown that loss of terminal sialic acid causes enhanced von Willebrand factor (VWF) clearance through the Ashwell-Morrell receptor (AMR). In this study, we investigated (1) the specific importance of N- vs O-linked sialic acid in protecting against VWF clearance and (2) whether additional receptors contribute to the reduced half-life of hyposialylated VWF. α2-3-linked sialic acid accounts for <20% of total sialic acid and is predominantly expressed on VWF O-glycans. Nevertheless, specific digestion with α2-3 neuraminidase (α2-3Neu-VWF) was sufficient to cause markedly enhanced VWF clearance. Interestingly, in vivo clearance experiments in dual VWF-/-/Asgr1-/- mice demonstrated enhanced clearance of α2-3Neu-VWF even in the absence of the AMR. The macrophage galactose-type lectin (MGL) is a C-type lectin that binds to glycoproteins expressing terminal N-acetylgalactosamine or galactose residues. Importantly, the markedly enhanced clearance of hyposialylated VWF in VWF-/-/Asgr1-/- mice was significantly attenuated in the presence of an anti-MGL inhibitory antibody. Furthermore, dose-dependent binding of human VWF to purified recombinant human MGL was confirmed using surface plasmon resonance. Additionally, plasma VWF:Ag levels were significantly elevated in MGL1-/- mice compared with controls. Collectively, these findings identify MGL as a novel macrophage receptor for VWF that significantly contributes to the clearance of both wild-type and hyposialylated VWF.
Collapse
|
28
|
Long Z, Du Y, Li H, Han B. Polymorphism of the ABO gene associate with thrombosis risk in patients with paroxysmal nocturnal hemoglobinuria. Oncotarget 2017; 8:92411-92419. [PMID: 29190926 PMCID: PMC5696192 DOI: 10.18632/oncotarget.21361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/08/2017] [Indexed: 11/25/2022] Open
Abstract
Thrombosis is one of the most common causes of mortality in Paroxysmal nocturnal hemoglobinuria (PNH), but the predisposing factors for thrombosis are yet to be defined. In this study, we outline the clinical characters and the susceptible genes which lead to thrombotic formation in 104 patients with PNH. The results displayed that the genotypes with minor alleles of rs495828 or rs2519093 in the ABO gene were associated with high risk to thrombus formation (OR 5.95, 95% CI 1.90-18.65 and OR 6.3, 95% CI 2.01-19.79, respectively). Further, the TT haplotype was associated with a significant increased risk of thrombosis (OR=3.25, 95%CI 1.42-7.39). Multivariate regression analysis showed larger PNH clone and genotypes with rs495828/rs2519093 minor allele as independent risk factors for thrombosis in PNH. Some patients who came back for follow-up were tested for the plasma levels of vWF and factor VIII. Patients carrying the rs495828/rs2519093 minor allele had a significant higher level of vWF and factor VIII compared with those carrying the major allele. Therefore, we found for the first time that the rs495828/rs2519093 polymorphism represent an independent prognostic factor in PNH patients for thrombus formation, probably by increasing the vWF and factor VIII.
Collapse
Affiliation(s)
- Zhangbiao Long
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yali Du
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hongmin Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
29
|
Gashash EA, Aloor A, Li D, Zhu H, Xu XQ, Xiao C, Zhang J, Parameswaran A, Song J, Ma C, Xiao W, Wang PG. An Insight into Glyco-Microheterogeneity of Plasma von Willebrand Factor by Mass Spectrometry. J Proteome Res 2017; 16:3348-3362. [PMID: 28696719 PMCID: PMC6309539 DOI: 10.1021/acs.jproteome.7b00359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human plasma von Willebrand Factor (VWF) plays essential roles in primary hemostasis in cooperation with other coagulations factors. There is ample indication that glycosylation affects many biological phases during the protein life cycle. However, comprehensive characterization of all probable N-glycosites simultaneous with O-glycosites is still not fully revealed. Thus, the intention of this exploration was to estimate the occupancy of all canonical N-glycosites besides simultaneous characterization of N- and O-glycoforms. An RP-LC-MS/MS system functionalized with CID and HCD tandem mass was utilized to analyze VWF. N-Glycosite occupancy varied along the protein backbone chain. Out of 257 HCD spectra, 181 characterized glycoforms were specified as either N- or O-glycosites. Sequential cleavage of glycosidic bonds along with Human Database mass matching have confirmed the glycoform structures. A total of 173 glycoforms represented most commonly biantennary and infrequently tri- and tetra-antennary N-glycans beside high mannose, hybrid, ABH antigen-terminated, and sulfated N-glycans. Many glycoforms were common across all N-sites. Noteworthy, previously unreported N-glycosites within domain D'(TIL'-E') showed glycosylation. Moreover, sialylated core 1 and core 2 O-glycans were detected on 2298T. Given subtle characterization of site-specific glycoforms, we can attain a profound understanding of the biological roles of VWF as well as facilitate the production of VWF-based therapeutics.
Collapse
Affiliation(s)
- Ebtesam A. Gashash
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
- Department of Chemistry, College of Science, Albaha University, Baljurashi, Albaha 65635, Saudi Arabia
| | - Arya Aloor
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Dong Li
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, 200065 Shanghai, China
| | - He Zhu
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Xiao-Qian Xu
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Cong Xiao
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Junping Zhang
- Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Aishwarya Parameswaran
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Jing Song
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Cheng Ma
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Weidong Xiao
- Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Peng George Wang
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| |
Collapse
|
30
|
Nowak AA, O'Brien HER, Henne P, Doerr A, Vanhoorelbeke K, Laffan MA, McKinnon TAJ. ADAMTS-13 glycans and conformation-dependent activity. J Thromb Haemost 2017; 15:1155-1166. [PMID: 28370891 DOI: 10.1111/jth.13688] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Indexed: 11/30/2022]
Abstract
Essentials The impact of N-linked glycosylation on ADAMTS-13 function has not been fully explored. The activity of glycan modified ADAMTS-13 was investigated under static and shear stress conditions. Terminal sialic acid on the metalloprotease domain glycans are important for ADAMTS-13 activity. The CUB domain glycans modulate ADAMTS-13 activity. SUMMARY Background ADAMTS-13 activity can be regulated by its conformation, whereby interactions between the C-terminal CUB domains and the spacer domain maintain ADAMTS-13 in a closed conformation. ADAMTS-13 contains 10 N-linked glycans, with four sites present in theTSP2 through to CUB domains that may contribute to its conformation. Objectives/Methods We hypothesized that glycosylation contributes to ADAMTS-13 conformation and function. The proteolytic activity of glycan-modified ADAMTS-13 was assessed under static and shear stress conditions. Results Enzymatic removal of terminal silaic acid or entire N-linked glycan chains decreased activity against FRETS-VWF73 at pH 7.4 and against full-length von Willebrand factor (VWF) under shear stress. Using truncated ADAMTS-13, we demonstrated that this was attributable to loss of sialic acid from the glycans in the metalloprotease domain and an effect of N-linked glycosylation in the TSP2 through to CUB domains. Mutation of the N-linked glycan sites in the MDTCS domains reduced or abolished protein expression. However, the N707Q, N828Q, N1235Q and N1354Q (TSP2, TSP4, CUB1, and CUB2 domains, respectively) variants were expressed normally. Interestingly, the N707Q and N828Q variants showed reduced activity against FRETS-VWF73, but normal activity under flow conditions. In contrast, the N1235Q and N1354Q variants had enhanced activity against FRETS-VWF73 and VWF under shear stress. Immunoprecipitation experiments confirmed that loss of N-linked glycans in the CUB domains significantly reduced the interaction with the spacer domain and enhanced binding to the 6A6 anti-ADAMTS-13 antibody, which recognizes a cryptic epitope in the metalloprotease domain. Conclusions Together, these data demonstrate that the N-linked glycans of ADAMTS-13 play a crucial role in regulating ADAMTS-13 activity.
Collapse
Affiliation(s)
- A A Nowak
- Department of Haematology, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - H E R O'Brien
- Department of Haematology, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - P Henne
- Department of Haematology, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - A Doerr
- Department of Haematology, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - K Vanhoorelbeke
- Laboratory for Thrombosis Research, Catholic University Leuven, Leuven, Belgium
| | - M A Laffan
- Department of Haematology, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - T A J McKinnon
- Department of Haematology, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, London, UK
| |
Collapse
|
31
|
Brophy TM, Ward SE, McGimsey TR, Schneppenheim S, Drakeford C, O’Sullivan JM, Chion A, Budde U, O’Donnell JS. Plasmin Cleaves Von Willebrand Factor at K1491-R1492 in the A1–A2 Linker Region in a Shear- and Glycan-Dependent Manner In Vitro. Arterioscler Thromb Vasc Biol 2017; 37:845-855. [DOI: 10.1161/atvbaha.116.308524] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022]
Abstract
Objective—
Previous studies have demonstrated a role for plasmin in regulating plasma von Willebrand factor (VWF) multimer composition. Moreover, emerging data have shown that plasmin-induced cleavage of VWF is of particular importance in specific pathological states. Interestingly, plasmin has been successfully used as an alternative to ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif) in a mouse model of thrombotic thrombocytopenic purpura. Consequently, elucidating the molecular mechanisms through which plasmin binds and cleaves VWF is not only of basic scientific interest but also of direct clinical importance. Our aim was to investigate factors that modulate the susceptibility of human VWF to proteolysis by plasmin.
Approach and Results—
We have adapted the VWF vortex proteolysis assay to allow for time-dependent shear exposure studies. We show that globular VWF is resistant to plasmin cleavage under static conditions, but is readily cleaved by plasmin under shear. Although both plasmin and ADAMTS13 cleave VWF in a shear-dependent manner, plasmin does not cleave at the Tyr1605-Met1606 ADAMTS13 proteolytic site in the A2 domain. Rather under shear stress conditions, or in the presence of denaturants, such as urea or ristocetin, plasmin cleaves the K1491-R1492 peptide bond within the VWF A1–A2 linker region. Finally, we demonstrate that VWF susceptibility to plasmin proteolysis at K1491-R1492 is modulated by local N-linked glycan expression within A1A2A3, and specifically inhibited by heparin binding to the A1 domain.
Conclusions—
Improved understanding of the plasmin–VWF interaction offers exciting opportunities to develop novel adjunctive therapies for the treatment of refractory thrombotic thrombocytopenic purpura.
Collapse
Affiliation(s)
- Teresa M. Brophy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Soracha E. Ward
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Thomas R. McGimsey
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Sonja Schneppenheim
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Clive Drakeford
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Jamie M. O’Sullivan
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Alain Chion
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Ulrich Budde
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - James S. O’Donnell
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| |
Collapse
|
32
|
Characterizing the O-glycosylation landscape of human plasma, platelets, and endothelial cells. Blood Adv 2017; 1:429-442. [PMID: 29296958 DOI: 10.1182/bloodadvances.2016002121] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/17/2017] [Indexed: 01/26/2023] Open
Abstract
The hemostatic system comprises platelet aggregation, coagulation, and fibrinolysis, and is critical to the maintenance of vascular integrity. Multiple studies indicate that glycans play important roles in the hemostatic system; however, most investigations have focused on N-glycans because of the complexity of O-glycan analysis. Here we performed the first systematic analysis of native-O-glycosylation using lectin affinity chromatography coupled to liquid chromatography mass spectrometry (LC-MS)/MS to determine the precise location of O-glycans in human plasma, platelets, and endothelial cells, which coordinately regulate hemostasis. We identified the hitherto largest O-glycoproteome from native tissue with a total of 649 glycoproteins and 1123 nonambiguous O-glycosites, demonstrating that O-glycosylation is a ubiquitous modification of extracellular proteins. Investigation of the general properties of O-glycosylation established that it is a heterogeneous modification, frequently occurring at low density within disordered regions in a cell-dependent manner. Using an unbiased screen to identify associations between O-glycosites and protein annotations we found that O-glycans were over-represented close (± 15 amino acids) to tandem repeat regions, protease cleavage sites, within propeptides, and located on a select group of protein domains. The importance of O-glycosites in proximity to proteolytic cleavage sites was further supported by in vitro peptide assays demonstrating that proteolysis of key hemostatic proteins can be inhibited by the presence of O-glycans. Collectively, these data illustrate the global properties of native O-glycosylation and provide the requisite roadmap for future biomarker and structure-function studies.
Collapse
|
33
|
Hasnain SZ, Dawson PA, Lourie R, Hutson P, Tong H, Grencis RK, McGuckin MA, Thornton DJ. Immune-driven alterations in mucin sulphation is an important mediator of Trichuris muris helminth expulsion. PLoS Pathog 2017; 13:e1006218. [PMID: 28192541 PMCID: PMC5325613 DOI: 10.1371/journal.ppat.1006218] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/24/2017] [Accepted: 02/03/2017] [Indexed: 12/21/2022] Open
Abstract
Mucins are heavily glycosylated proteins that give mucus its gel-like properties. Moreover, the glycans decorating the mucin protein core can alter the protective properties of the mucus barrier. To investigate whether these alterations could be parasite-induced we utilized the Trichuris muris (T. muris) infection model, using different infection doses and strains of mice that are resistant (high dose infection in BALB/c and C57BL6 mice) or susceptible (high dose infection in AKR and low dose infection in BALB/c mice) to chronic infection by T. muris. During chronicity, within the immediate vicinity of the T. muris helminth the goblet cell thecae contained mainly sialylated mucins. In contrast, the goblet cells within the epithelial crypts in the resistant models contained mainly sulphated mucins. Maintained mucin sulphation was promoted by TH2-immune responses, in particular IL-13, and contributed to the protective properties of the mucus layer, making it less vulnerable to degradation by T. muris excretory secretory products. Mucin sulphation was markedly reduced in the caecal goblet cells in the sulphate anion transporter-1 (Sat-1) deficient mice. We found that Sat-1 deficient mice were susceptible to chronic infection despite a strong TH2-immune response. Lower sulphation levels lead to decreased efficiency of establishment of T. muris infection, independent of egg hatching. This study highlights the complex process by which immune-regulated alterations in mucin glycosylation occur following T. muris infection, which contributes to clearance of parasitic infection. Approximately 2 billion people are infected with worms every year, causing physical, nutritional and cognitive impairment particularly in children. Mucins are large sugar-coated (glycosylated) proteins that form the intestinal mucus layer. This mucus layer protects our ‘insides’ from external insults and plays an important role during worm infection. We discovered that there is a difference in the glycosylation of mucins in people infected with worms compared to uninfected individuals. Therefore, using different mouse models we investigated the role of glycosylation, and in particular sulphation of mucins in infection. We found that mucin glycosylation is controlled by the immune response and increased sulphation correlated with the expulsion of the worm from the host. Highly sulphated mucins were protected from degradation by the worm. Moreover, mice lacking a sulphate transporter had significantly lower sulphation levels on mucins, which resulted in a reduction in the establishment of the worms and chronic infection.
Collapse
Affiliation(s)
- Sumaira Z. Hasnain
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Australia
- * E-mail:
| | - Paul A. Dawson
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Rohan Lourie
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Australia
- Mater Pathology Services, Mater Hospitals, South Brisbane, Queensland, Australia
| | - Peter Hutson
- Mater Pathology Services, Mater Hospitals, South Brisbane, Queensland, Australia
| | - Hui Tong
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Richard K. Grencis
- Manchester Immunology Group Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Michael A. McGuckin
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - David J. Thornton
- Manchester Immunology Group Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
34
|
O'Sullivan JM, Aguila S, McRae E, Ward SE, Rawley O, Fallon PG, Brophy TM, Preston RJS, Brady L, Sheils O, Chion A, O'Donnell JS. N-linked glycan truncation causes enhanced clearance of plasma-derived von Willebrand factor. J Thromb Haemost 2016; 14:2446-2457. [PMID: 27732771 DOI: 10.1111/jth.13537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 12/31/2022]
Abstract
Essentials von Willebrands factor (VWF) glycosylation plays a key role in modulating in vivo clearance. VWF glycoforms were used to examine the role of specific glycan moieties in regulating clearance. Reduction in sialylation resulted in enhanced VWF clearance through asialoglycoprotein receptor. Progressive VWF N-linked glycan trimming resulted in increased macrophage-mediated clearance. Click to hear Dr Denis discuss clearance of von Willebrand factor in a free presentation from the ISTH Academy SUMMARY: Background Enhanced von Willebrand factor (VWF) clearance is important in the etiology of both type 1 and type 2 von Willebrand disease (VWD). In addition, previous studies have demonstrated that VWF glycans play a key role in regulating in vivo clearance. However, the molecular mechanisms underlying VWF clearance remain poorly understood. Objective To define the molecular mechanisms through which VWF N-linked glycan structures influence in vivo clearance. Methods By use of a series of exoglycosidases, different plasma-derived VWF (pd-VWF) glycoforms were generated. In vivo clearance of these glycoforms was then assessed in VWF-/- mice in the presence or absence of inhibitors of asialoglycoprotein receptor (ASGPR), or following clodronate-induced macrophage depletion. Results Reduced amounts of N-linked and O-linked sialylation resulted in enhanced pd-VWF clearance modulated via ASGPR. In addition to this role of terminal sialylation, we further observed that progressive N-linked glycan trimming also resulted in markedly enhanced VWF clearance. Furthermore, these additional N-linked glycan effects on clearance were ASGPR-independent, and instead involved enhanced macrophage clearance that was mediated, at least in part, through LDL receptor-related protein 1. Conclusion The carbohydrate determinants expressed on VWF regulate susceptibility to proteolysis by ADAMTS-13. In addition, our findings now further demonstrate that non-sialic acid carbohydrate determinants expressed on VWF also play an unexpectedly important role in modulating in vivo clearance through both hepatic ASGPR-dependent and macrophage-dependent pathways. In addition, these data further support the hypothesis that variation in VWF glycosylation may be important in the pathophysiology underlying type 1C VWD.
Collapse
Affiliation(s)
- J M O'Sullivan
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - S Aguila
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - E McRae
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - S E Ward
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - O Rawley
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - P G Fallon
- Inflammation and Immunity Research Group, Institute of Molecular Medicine, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - T M Brophy
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - R J S Preston
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - L Brady
- Department of Histopathology, Sir Patrick Dun Research Laboratory, Trinity College Dublin, St James's Hospital Dublin, Dublin, Ireland
| | - O Sheils
- Department of Histopathology, Sir Patrick Dun Research Laboratory, Trinity College Dublin, St James's Hospital Dublin, Dublin, Ireland
| | - A Chion
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - J S O'Donnell
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
- National Centre for Hereditary Coagulation Disorders, St James's Hospital, Dublin, Ireland
| |
Collapse
|
35
|
Brehm MA. Von Willebrand factor processing. Hamostaseologie 2016; 37:59-72. [PMID: 28139814 DOI: 10.5482/hamo-16-06-0018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/03/2016] [Indexed: 11/05/2022] Open
Abstract
Von Willebrand factor (VWF) is a multimeric glycoprotein essential for primary haemostasis that is produced only in endothelial cells and megakaryocytes. Key to VWF's function in recruitment of platelets to the site of vascular injury is its multimeric structure. The individual steps of VWF multimer biosynthesis rely on distinct posttranslational modifications at specific pH conditions, which are realized by spatial separation of the involved processes to different cell organelles. Production of multimers starts with translocation and modification of the VWF prepropolypeptide in the endoplasmic reticulum to produce dimers primed for glycosylation. In the Golgi apparatus they are further processed to multimers that carry more than 300 complex glycan structures functionalized by sialylation, sulfation and blood group determinants. Of special importance is the sequential formation of disulfide bonds with different functions in structural support of VWF multimers, which are packaged, stored and further processed after secretion. Here, all these processes are being reviewed in detail including background information on the occurring biochemical reactions.
Collapse
Affiliation(s)
- Maria A Brehm
- PD Dr. Maria A. Brehm, Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 22399 Hamburg, Germany, Tel.: +49 40 7410 58523, Fax: +49 40 7410 54601, E-Mail:
| |
Collapse
|
36
|
Song J, Xue C, Preisser JS, Cramer DW, Houck KL, Liu G, Folsom AR, Couper D, Yu F, Dong JF. Association of Single Nucleotide Polymorphisms in the ST3GAL4 Gene with VWF Antigen and Factor VIII Activity. PLoS One 2016; 11:e0160757. [PMID: 27584569 PMCID: PMC5008807 DOI: 10.1371/journal.pone.0160757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/25/2016] [Indexed: 02/05/2023] Open
Abstract
VWF is extensively glycosylated with biantennary core fucosylated glycans. Most N-linked and O-linked glycans on VWF are sialylated. FVIII is also glycosylated, with a glycan structure similar to that of VWF. ST3GAL sialyltransferases catalyze the transfer of sialic acids in the α2,3 linkage to termini of N- and O-glycans. This sialic acid modification is critical for VWF synthesis and activity. We analyzed genetic and phenotypic data from the Atherosclerosis Risk in Communities (ARIC) study for the association of single nucleotide polymorphisms (SNPs) in the ST3GAL4 gene with plasma VWF levels and FVIII activity in 12,117 subjects. We also analyzed ST3GAL4 SNPs found in 2,535 subjects of 26 ethnicities from the 1000 Genomes (1000G) project for ethnic diversity, SNP imputation, and ST3GAL4 haplotypes. We identified 14 and 1,714 ST3GAL4 variants in the ARIC GWAS and 1000G databases respectively, with 46% being ethnically diverse in their allele frequencies. Among the 14 ST3GAL4 SNPs found in ARIC GWAS, the intronic rs2186717, rs7928391, and rs11220465 were associated with VWF levels and with FVIII activity after adjustment for age, BMI, hypertension, diabetes, ever-smoking status, and ABO. This study illustrates the power of next-generation sequencing in the discovery of new genetic variants and a significant ethnic diversity in the ST3GAL4 gene. We discuss potential mechanisms through which these intronic SNPs regulate ST3GAL4 biosynthesis and the activity that affects VWF and FVIII.
Collapse
Affiliation(s)
- Jaewoo Song
- BloodWorks Northwest Research Institute, Seattle, WA, United States of America
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Cheng Xue
- Human Genome Sequencing Center, Molecular and Human Genetics Department, Baylor College of Medicine, Houston, TX, 77030, United States of America
| | - John S. Preisser
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, United States of America
| | - Drake W. Cramer
- BloodWorks Northwest Research Institute, Seattle, WA, United States of America
| | - Katie L. Houck
- BloodWorks Northwest Research Institute, Seattle, WA, United States of America
| | - Guo Liu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Aaron R. Folsom
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| | - David Couper
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, United States of America
| | - Fuli Yu
- Human Genome Sequencing Center, Molecular and Human Genetics Department, Baylor College of Medicine, Houston, TX, 77030, United States of America
- Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- * E-mail: (JFD); (FY)
| | - Jing-fei Dong
- BloodWorks Northwest Research Institute, Seattle, WA, United States of America
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States of America
- * E-mail: (JFD); (FY)
| |
Collapse
|
37
|
N-linked glycans within the A2 domain of von Willebrand factor modulate macrophage-mediated clearance. Blood 2016; 128:1959-1968. [PMID: 27554083 DOI: 10.1182/blood-2016-04-709436] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022] Open
Abstract
Enhanced von Willebrand factor (VWF) clearance is important in the etiology of von Willebrand disease. However, the molecular mechanisms underlying VWF clearance remain poorly understood. In this study, we investigated the role of VWF domains and specific glycan moieties in regulating in vivo clearance. Our findings demonstrate that the A1 domain of VWF contains a receptor-recognition site that plays a key role in regulating the interaction of VWF with macrophages. In A1-A2-A3 and full-length VWF, this macrophage-binding site is cryptic but becomes exposed following exposure to shear or ristocetin. Previous studies have demonstrated that the N-linked glycans within the A2 domain play an important role in modulating susceptibility to ADAMTS13 proteolysis. We further demonstrate that these glycans presented at N1515 and N1574 also play a critical role in protecting VWF against macrophage binding and clearance. Indeed, loss of the N-glycan at N1515 resulted in markedly enhanced VWF clearance that was significantly faster than that observed with any previously described VWF mutations. In addition, A1-A2-A3 fragments containing the N1515Q or N1574Q substitutions also demonstrated significantly enhanced clearance. Importantly, clodronate-induced macrophage depletion significantly attenuated the increased clearance observed with N1515Q and N1574Q in both full-length VWF and A1-A2-A3. Finally, we further demonstrate that loss of these N-linked glycans does not enhance clearance in VWF in the presence of a structurally constrained A2 domain. Collectively, these novel findings support the hypothesis that conformation of the VWF A domains plays a critical role in modulating macrophage-mediated clearance of VWF in vivo.
Collapse
|
38
|
Indian Bombay phenotype: it is different! BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2016; 15:74-76. [PMID: 27416578 DOI: 10.2450/2016.0266-15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/04/2016] [Indexed: 11/21/2022]
|
39
|
O'Sullivan JM, Jenkins PV, Rawley O, Gegenbauer K, Chion A, Lavin M, Byrne B, O'Kennedy R, Preston RJS, Brophy TM, O'Donnell JS. Galectin-1 and Galectin-3 Constitute Novel-Binding Partners for Factor VIII. Arterioscler Thromb Vasc Biol 2016; 36:855-63. [PMID: 27013611 DOI: 10.1161/atvbaha.115.306915] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 03/14/2016] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Recent studies have demonstrated that galectin-1 (Gal-1) and galectin-3 (Gal-3) can bind von Willebrand factor and directly modulate von Willebrand factor-dependent early thrombus formation in vivo. Because the glycans expressed on human factor VIII (FVIII) are similar to those of von Willebrand factor, we investigated whether galectins might also bind and modulate the activity of FVIII. APPROACH AND RESULTS Immunosorbant assays and surface plasmon resonance analysis confirmed that Gal-1 and Gal-3 bound purified FVIII with high affinity. Exoglycosidase removal of FVIII N-linked glycans significantly reduced binding to both Gal-1 and Gal-3. Moreover, combined removal of both the N- and O-glycans of FVIII further attenuated Gal-3 binding. Notably, specific digestion of FVIII high-mannose glycans at N239 and N2118 significantly impaired FVIII affinity for Gal-1. Importantly Gal-1, but not Gal-3, bound to free FVIII in the plasma milieu, and significantly inhibited FVIII functional activity. Interestingly, commercial recombinant FVIII (rFVIII) concentrates are manufactured in different cell lines and differ in their glycosylation profiles. Although the biological mechanism has not been defined, recent studies in previously untreated patients with severe hemophilia A reported significant differences in inhibitor development associated with different rFVIII products. Interestingly, Gal-1 and Gal-3 both displayed enhanced affinity for BHK-rFVIII compared with CHO-rFVIII. Furthermore, binding of Gal-1 and Gal-3 to BDD-FVIII was markedly reduced compared with full-length rFVIII. CONCLUSIONS We have identified Gal-1 and Gal-3 as novel-binding partners for human FVIII and demonstrated that Gal-1 binding can influence the procoagulant activity of FVIII.
Collapse
Affiliation(s)
- Jamie M O'Sullivan
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - P Vince Jenkins
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Orla Rawley
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Kristina Gegenbauer
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Alain Chion
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Michelle Lavin
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Barry Byrne
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Richard O'Kennedy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Roger J S Preston
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Teresa M Brophy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - James S O'Donnell
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.).
| |
Collapse
|
40
|
O'Regan N, Moxon C, Gegenbauer K, O'Sullivan JM, Chion A, Smith OP, Preston RJS, Brophy TM, Craig AG, O'Donnell JS. Marked elevation in plasma osteoprotegerin constitutes an early and consistent feature of cerebral malaria. Thromb Haemost 2016; 115:773-80. [PMID: 26766771 PMCID: PMC4990170 DOI: 10.1160/th15-10-0796] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/22/2015] [Indexed: 11/21/2022]
Abstract
Adherence of infected erythrocytes to vascular endothelium causes acute endothelial cell (EC) activation during Plasmodium falciparum infection. Consequently, proteins stored in Weibel-Palade (WP) bodies within EC are secreted into the plasma. Osteoprotegerin (OPG) binds to VWF and consequently is stored within WP bodies. Given the critical role of EC activation in the pathogenesis of severe malaria, we investigated plasma OPG levels in children with P. falciparum malaria. At presentation, plasma OPG levels were significantly elevated in children with cerebral malaria (CM) compared to healthy controls (means 16.0 vs 0.8 ng/ml; p<0.01). Importantly, OPG levels were also significantly higher in children with CM who had a fatal outcome, compared to children with CM who survived. Finally, in children with CM, plasma OPG levels correlated with other established prognostic indices (including plasma lactate levels and peripheral parasite density). To further investigate the relationship between severe malaria and OPG, we utilised a murine model of experimental CM in which C57BL/6J mice were infected with P. berghei ANKA. Interestingly, plasma OPG levels were increased 4.6 fold within 24 hours following P. berghei inoculation. This early marked elevation in OPG levels was observed before any objective clinical signs were apparent, and preceded the development of peripheral blood parasitaemia. As the mice became increasingly unwell, plasma OPG levels progressively increased. Collectively, these data suggest that OPG constitutes a novel biomarker with prognostic significance in patients with severe malaria. In addition, further studies are required to determine whether OPG plays a role in modulating malaria pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - James S O'Donnell
- Prof. James O'Donnell, Haemostasis Research Group, Institute of Molecular Medicine, Trinity College, Dublin, Ireland, Tel.: +353 1 416 2141, Fax: +353 1 410 3570, E-mail:
| |
Collapse
|
41
|
A novel role for von Willebrand factor in the pathogenesis of experimental cerebral malaria. Blood 2015; 127:1192-201. [PMID: 26511133 DOI: 10.1182/blood-2015-07-654921] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/19/2015] [Indexed: 01/28/2023] Open
Abstract
Plasmodium falciparum malaria infection is associated with an early marked increase in plasma von Willebrand factor (VWF) levels, together with a pathological accumulation of hyperreactive ultra-large VWF (UL-VWF) multimers. Given the established critical role of platelets in malaria pathogenesis, these increases in plasma VWF raise the intriguing possibility that VWF may play a direct role in modulating malaria pathogenesis. To address this hypothesis, we used an established murine model of experimental cerebral malaria (ECM), in which wild-type (WT) C57BL/6J mice were infected with Plasmodium berghei ANKA. In keeping with findings in children with P falciparum malaria, acute endothelial cell activation was an early and consistent feature in the murine model of cerebral malaria (CM), resulting in significantly increased plasma VWF levels. Despite the fact that murine plasma ADAMTS13 levels were not significantly reduced, pathological UL-VWF multimers were also observed in murine plasma following P berghei infection. To determine whether VWF plays a role in modulating the pathogenesis of CM in vivo, we further investigated P berghei infection in VWF(-/-) C57BL/6J mice. Clinical ECM progression was delayed, and overall survival was significantly prolonged in VWF(-/-) mice compared with WT controls. Despite this protection against ECM, no significant differences in platelet counts or blood parasitemia levels were observed between VWF(-/-) and WT mice. Interestingly, however, the degree of ECM-associated enhanced blood-brain barrier permeability was significantly attenuated in VWF(-/-) mice compared with WT controls. Given the significant morbidity and mortality associated with CM, these novel data may have direct translational significance.
Collapse
|
42
|
Zhong M, Zhang H, Reilly JP, Chrisitie JD, Ishihara M, Kumagai T, Azadi P, Reilly MP. ABO Blood Group as a Model for Platelet Glycan Modification in Arterial Thrombosis. Arterioscler Thromb Vasc Biol 2015; 35:1570-8. [PMID: 26044584 DOI: 10.1161/atvbaha.115.305337] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 05/22/2015] [Indexed: 01/02/2023]
Abstract
ABO blood groups have long been associated with cardiovascular disease, thrombosis, and acute coronary syndromes. Many studies over the years have shown type O blood group to be associated with lower risk of cardiovascular disease than non-type O blood groups. However, the mechanisms underlying this association remain unclear. Although ABO blood group is associated with variations in concentrations of circulating von Willebrand Factor and other endothelial cell adhesion molecules, ABO antigens are also present on several platelet surface glycoproteins and glycosphingolipids. As we highlight in this platelet-centric review, these glycomic modifications may affect platelet function in arterial thrombosis. More broadly, improving our understanding of the role of platelet glycan modifications in acute coronary syndromes may inform future diagnostics and therapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Ming Zhong
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.)
| | - Hanrui Zhang
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.)
| | - John P Reilly
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.)
| | - Jason D Chrisitie
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.)
| | - Mayumi Ishihara
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.)
| | - Tadahiro Kumagai
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.)
| | - Parastoo Azadi
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.)
| | - Muredach P Reilly
- From the Cardiology Division, Department of Medicine, Cardiovascular Institute (M.Z., H.Z., M.P.R.) and Pulmonology, Allergy, and Critical Care Division, Department of Medicine (J.P.R., J.D.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Complex Carbohydrate Research Center, University of Georgia, Athens (M.I., T.K., P.A.).
| |
Collapse
|
43
|
Rawley O, O'Sullivan JM, Chion A, Keyes S, Lavin M, van Rooijen N, Brophy TM, Fallon P, Preston RJS, O'Donnell JS. von Willebrand factor arginine 1205 substitution results in accelerated macrophage-dependent clearance in vivo. J Thromb Haemost 2015; 13:821-6. [PMID: 25690668 DOI: 10.1111/jth.12875] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/04/2015] [Indexed: 01/23/2023]
Abstract
BACKGROUND Enhanced von Willebrand factor (VWF) clearance is important in the etiology of type 1 and type 2 von Willebrand disease (VWD). More than 20 different VWF point mutations have already been reported in patients with enhanced clearance. These include the VWD-Vicenza variant, which is characterized by an Arg1205His substitution in the VWF D3 domain. Critically, however, the molecular mechanisms through which single amino acid substitutions in VWF result in enhanced clearance of this complex multimeric glycoprotein have not been defined. OBJECTIVES In this study, we have investigated the biological basis underlying the enhanced clearance of the VWF-R1205H variant. METHODS Using VWF(-/-) mice, in vivo clearance rates were determined for a series of full-length and truncated recombinant VWF variants. In addition, the role of macrophages in modulating enhanced VWD-Vicenza clearance was investigated using clodronate liposome administration. RESULTS Our findings demonstrate that substitutions of R1205 with histidine, cysteine or serine all result in markedly reduced survival of full-length recombinant VWF. Importantly, D'A3 fragments containing these same R1205 substitutions also demonstrated significantly enhanced clearance. In contrast to the reduced in vivo survival observed with R1205H, clearance of R1204H was not enhanced. Recent studies have demonstrated that hepatic and splenic macrophages play key roles in regulating VWF clearance. Importantly, macrophage-depletion also served to markedly attenuate the enhanced clearance phenotypes associated with VWF-R1205H, VWF-R1205S and VWF-R1205C. CONCLUSIONS Collectively, these novel findings demonstrate a specific and critical role for the R1205 residue in modulating macrophage-mediated clearance of VWF in vivo.
Collapse
Affiliation(s)
- O Rawley
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Podoplanin requires sialylated O-glycans for stable expression on lymphatic endothelial cells and for interaction with platelets. Blood 2014; 124:3656-65. [PMID: 25336627 DOI: 10.1182/blood-2014-04-572107] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
O-glycosylation of podoplanin (PDPN) on lymphatic endothelial cells is critical for the separation of blood and lymphatic systems by interacting with platelet C-type lectin-like receptor 2 during development. However, how O-glycosylation controls endothelial PDPN function and expression remains unclear. In this study, we report that core 1 O-glycan-deficient or desialylated PDPN was highly susceptible to proteolytic degradation by various proteases, including metalloproteinases (MMP)-2/9. We found that the lymph contained activated MMP-2/9 and incubation of the lymph reduced surface levels of PDPN on core 1 O-glycan-deficient endothelial cells, but not on wild-type ECs. The lymph from mice with sepsis induced by cecal ligation and puncture, which contained bacteria-derived sialidase, reduced PDPN levels on wild-type ECs. The MMP inhibitor, GM6001, rescued these reductions. Additionally, GM6001 treatment rescued the reduction of PDPN level on lymphatic endothelial cells in mice lacking endothelial core 1 O-glycan or cecal ligation and puncture-treated mice. Furthermore, core 1 O-glycan-deficient or desialylated PDPN impaired platelet interaction under physiological flow. These data indicate that sialylated O-glycans of PDPN are essential for platelet adhesion and prevent PDPN from proteolytic degradation primarily mediated by MMPs in the lymph.
Collapse
|
45
|
Franchini M, Mannucci PM. ABO blood group and thrombotic vascular disease. Thromb Haemost 2014; 112:1103-9. [PMID: 25187297 DOI: 10.1160/th14-05-0457] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/22/2014] [Indexed: 01/15/2023]
Abstract
ABO blood group antigens are complex carbohydrate molecules expressed on red blood cells and a variety of tissues. The ABO blood type is implicated in the development of a number of human diseases and there is increasing evidence regarding its involvement in the pathogenesis of cardiovascular disorders, mainly through its effect on von Willebrand factor levels. In this review, after a brief analysis of the potential molecular mechanisms by which the blood group influences haemostasis, we focus on the clinical implications of such interaction. Overall, the literature data document the close relationship between venous thromboembolism (VTE) and non-O blood type, which is associated with an approximately two-fold increased risk of venous thrombosis. A supra-additive effect on VTE risk is observed when an inherited thrombophilic condition is associated with non-O blood group. A weaker association exists between non-O blood type and arterial thrombosis, which needs to be further investigated.
Collapse
Affiliation(s)
- M Franchini
- Massimo Franchini, MD, Director, Dipartimento di Medicina Trasfusionale ed Ematologia, Azienda Ospedaliera Carlo Poma, Mantova, Italy, Tel.: +39 0376 201234, Fax: +39 0376 220144, E-mail:
| | | |
Collapse
|
46
|
Kragh T, Napoleone M, Fallah MA, Gritsch H, Schneider MF, Reininger AJ. High shear dependent von Willebrand factor self-assembly fostered by platelet interaction and controlled by ADAMTS13. Thromb Res 2014; 133:1079-87. [PMID: 24681085 DOI: 10.1016/j.thromres.2014.03.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 02/06/2014] [Accepted: 03/03/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION The paradigm of activation induced platelet aggregation has recently been refuted under blood flow conditions with shear rates exceeding 20,000s(-1). These lead to reversible rolling platelet aggregates, which were dependent on the presence of immobilized and soluble von Willebrand factor. MATERIAL AND METHODS In vitro experiments using direct fluorescence video-microscopy were performed in wall parallel and stagnation point flow chambers with shear rates raised from 20,000 to 50,000s(-1). Washed blood cell suspension containing recombinant von Willebrand factor (rVWF) was perfused over rVWF or collagen coated surfaces. RESULTS Here we show for the first time with the visualization of rVWF that not only colloid and polymer, i.e. platelets and VWF, form a composite, but that VWF itself is capable of entirely reversible self-assembly. On a collagen surface the platelet-VWF-conglomerates did not roll but VWF nets bound permanently to the collagen fibers and captured and immobilized platelets from the flow. Lowering the shear rate below the threshold of 20,000s(-1) no longer dissolved these deposits. Ultralarge multimer containing rVWF was most effective compared to normal sized rVWF. The presence of ADAMTS13 limited rolling aggregate and platelet-VWF-conglomerate formation to a time window of 7-8minutes. Changing wall parallel flow to stagnation point flow halved the required shear rate threshold. CONCLUSION We conclude that flow dynamics can trigger reversible von Willebrand factor self-assembly and platelet-VWF-conglomerate accrual, which are regulated by ADAMTS13 to a time span needed by coagulation to stabilize it, e.g. in case of vessel injury.
Collapse
Affiliation(s)
- Thorsten Kragh
- Department of Transfusion Medicine, Cell Therapeutics and Hemostasis, Medical Center of the University of Munich, Germany.
| | - Marina Napoleone
- Department of Transfusion Medicine, Cell Therapeutics and Hemostasis, Medical Center of the University of Munich, Germany
| | - Mohammad A Fallah
- Department of Biophysical Chemistry, University of Konstanz, Germany
| | | | | | | |
Collapse
|
47
|
Nowak AA, McKinnon TAJ, Hughes JM, Chion ACK, Laffan MA. The O-linked glycans of human von Willebrand factor modulate its interaction with ADAMTS-13. J Thromb Haemost 2014; 12:54-61. [PMID: 24406064 DOI: 10.1111/jth.12451] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND O-linked glycans (OLGs) are clustered on either side of the von Willebrand factor (VWF) A1 domain and modulate its interaction with platelets; however, their influence on the VWF interaction with ADAMTS-13 is unknown. OBJECTIVES To assess the role of the OLGs in VWF susceptibility to ADAMTS-13 proteolysis, which would help to explain their specific distribution. METHODS OLG sites were mutated individually and as clusters on either and both sides of the A1 domain, and expressed in HEK293T cells. First, their proteolysis by ADAMTS-13 was assayed in the presence of urea. Next, a parallel-flow chamber was used to analyze VWF-mediated platelet capture on collagen in the presence and absence of ADAMTS-13 under a shear stress of 1500 s(-1) . The decrease in platelet capture in the presence ADAMTS-13 was used as a measure of VWF proteolysis. RESULTS Initially, we found that, under denaturing conditions, the C-terminal S1486A and Cluster 2 and double cluster (DC) variants were less susceptible to ADAMTS-13 proteolysis than wild-type VWF. Next, we showed that addition of ADAMTS-13 diminished VWF-mediated platelet capture on collagen under flow; surprisingly, this was more pronounced with the S1486A, Cluster 2 and DC variants than with wild-type VWF, indicating that these are proteolyzed more rapidly under shear flow. CONCLUSIONS OLGs provide rigidity to peptide backbones, and our findings suggest that OLG in the A1-A2 linker region regulates VWF conformational changes under shear. Importantly, the impact of OLGs on ADAMTS-13 cleavage under shear stress is the opposite of that under denaturing conditions, highlighting the non-physiologic nature of in vitro cleavage assays.
Collapse
Affiliation(s)
- A A Nowak
- Department of Haematology, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College, London, UK
| | | | | | | | | |
Collapse
|
48
|
Investigating the influence of age, gender and ABO blood group on ADAMTS-13 antigen and activity levels in healthy Arabs. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2013; 12:138-40. [PMID: 24333090 DOI: 10.2450/2013.0155-13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/09/2013] [Indexed: 11/21/2022]
|
49
|
Altered glycosylation of platelet-derived von Willebrand factor confers resistance to ADAMTS13 proteolysis. Blood 2013; 122:4107-10. [DOI: 10.1182/blood-2013-04-496851] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Key Points
Platelet-VWF exists as a distinct natural glycoform. Platelet-VWF is resistant to ADAMTS13 proteolysis.
Collapse
|
50
|
Berntorp E, Fuchs B, Makris M, Montgomery R, Flood V, O'Donnell JS, Federici AB, Lillicrap D, James P, Budde U, Morfini M, Petrini P, Austin S, Kannicht C, Jiménez-Yuste V, Lee C. Third Åland islands conference on von Willebrand disease, 26-28 September 2012: meeting report. Haemophilia 2013; 19 Suppl 3:1-18. [PMID: 23383607 DOI: 10.1111/hae.12078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2012] [Indexed: 01/03/2023]
Abstract
The first meeting of international specialists in the field of von Willebrand disease (VWD) was held in the Åland islands in 1998 where Erik von Willebrand had first observed a bleeding disorder in some members of a family from Föglö and a summary of the meeting was published in 1999. The second meeting was held in 2010 and a report of the meeting was published in 2012. Topics covered included progress in understanding of VWD over the last 50 years; multimers; classification of VWD; pharmacokinetics and laboratory assays; genetics; treating the paediatric patient; prophylaxis; geriatrics; gene therapy and treatment guidelines. This third meeting held over 3 days covered the structure and function of von Willebrand factor (VWF); type 1 VWD, the most common form of the disease; a lifespan of pharmacokinetics in VWD; detecting inhibitors in VWD patients; and special challenges in understanding and treating the female VWD patient.
Collapse
Affiliation(s)
- E Berntorp
- Department of Hematology and Coagulation Disorders, Lund University, Skåne University Hospital, Malmö, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|