1
|
Childers KC, Cowper B, Vaughan JD, McGill JR, Davulcu O, Lollar P, Doering CB, Coxon CH, Spiegel PC. Structural basis for inhibition of coagulation factor VIII reveals a shared antigenic hotspot on the C1 domain. J Thromb Haemost 2024; 22:2449-2459. [PMID: 38849084 PMCID: PMC11343672 DOI: 10.1016/j.jtha.2024.05.024] [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: 11/02/2023] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Hemophilia A arises from dysfunctional or deficient coagulation factor (F)VIII and leads to inefficient fibrin clot formation and uncontrolled bleeding events. The development of antibody inhibitors is a clinical complication in hemophilia A patients receiving FVIII replacement therapy. LE2E9 is an anti-C1 domain inhibitor previously isolated from a mild/moderate hemophilia A patient and disrupts FVIII interactions with von Willebrand factor and FIXa, though the intermolecular contacts that underpin LE2E9-mediated FVIII neutralization are undefined. OBJECTIVES To determine the structure of the complex between FVIII and LE2E9 and characterize its mechanism of inhibition. METHODS FVIII was bound to the antigen binding fragment (Fab) of NB2E9, a recombinant construct of LE2E9, and its structure was determined by cryogenic electron microscopy. RESULTS This report communicates the 3.46 Å structure of FVIII bound to NB2E9, with its epitope comprising FVIII residues S2040 to Y2043, K2065 to W2070, and R2150 to H2155. Structural analysis reveals that the LE2E9 epitope overlaps with portions of the epitope for 2A9, a murine-derived inhibitor, suggesting that these residues represent a shared antigenic region on the C1 domain between FVIII-/- mice and hemophilia A patients. Furthermore, the FVIII:NB2E9 structure elucidates the orientation of the LE2E9 glycan, illustrating how the glycan sterically blocks interactions between the FVIII C1 domain and the von Willebrand factor D' domain. A putative model of the FVIIIa:FIXa complex suggests potential clashing between the NB2E9 glycan and FIXa light chain. CONCLUSION These results describe an antigenic "hotspot" on the FVIII C1 domain and provide a structural basis for engineering FVIII replacement therapeutics with reduced antigenicity.
Collapse
Affiliation(s)
- Kenneth C Childers
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - Ben Cowper
- Medicines and Healthcare products Regulatory Agency, South Mimms Laboratories, Potters Bar, Hertfordshire, UK
| | - Jordan D Vaughan
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - Juliet R McGill
- Chemistry Department, Western Washington University, Bellingham, Washington, USA
| | - Omar Davulcu
- Pacific Northwest Center for Cryo-EM, Oregon Health & Science University, Portland, Oregon, USA; Pacific Northwest National Laboratory, Environmental Molecular Sciences Laboratory, Richland, Washington, USA
| | - Pete Lollar
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Christopher B Doering
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University, Atlanta, Georgia, USA; Expression Therapeutics, Inc, Tucker, Georgia, USA
| | - Carmen H Coxon
- Medicines and Healthcare products Regulatory Agency, South Mimms Laboratories, Potters Bar, Hertfordshire, UK
| | - Paul C Spiegel
- Chemistry Department, Western Washington University, Bellingham, Washington, USA.
| |
Collapse
|
2
|
Shestopal SA, Parunov LA, Olivares P, Chun H, Ovanesov MV, Pettersson JR, Sarafanov AG. Isolated Variable Domains of an Antibody Can Assemble on Blood Coagulation Factor VIII into a Functional Fv-like Complex. Int J Mol Sci 2022; 23:ijms23158134. [PMID: 35897712 PMCID: PMC9330781 DOI: 10.3390/ijms23158134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022] Open
Abstract
Single-chain variable fragments (scFv) are antigen-recognizing variable fragments of antibodies (FV) where both subunits (VL and VH) are connected via an artificial linker. One particular scFv, iKM33, directed against blood coagulation factor VIII (FVIII) was shown to inhibit major FVIII functions and is useful in FVIII research. We aimed to investigate the properties of iKM33 enabled with protease-dependent disintegration. Three variants of iKM33 bearing thrombin cleavage sites within the linker were expressed using a baculovirus system and purified by two-step chromatography. All proteins retained strong binding to FVIII by surface plasmon resonance, and upon thrombin cleavage, dissociated into VL and VH as shown by size-exclusion chromatography. However, in FVIII activity and low-density lipoprotein receptor-related protein 1 binding assays, the thrombin-cleaved iKM33 variants were still inhibitory. In a pull-down assay using an FVIII-affinity sorbent, the isolated VH, a mixture of VL and VH, and intact iKM33 were carried over via FVIII analyzed by electrophoresis. We concluded that the isolated VL and VH assembled into scFv-like heterodimer on FVIII, and the isolated VH alone also bound FVIII. We discuss the potential use of both protease-cleavable scFvs and isolated Fv subunits retaining high affinity to the antigens in various practical applications such as therapeutics, diagnostics, and research.
Collapse
|
3
|
Freato N, van Alphen FPJ, Boon‐Spijker M, van den Biggelaar M, Meijer AB, Mertens K, Ebberink EHTM. Probing activation-driven changes in coagulation factor IX by mass spectrometry. J Thromb Haemost 2021; 19:1447-1459. [PMID: 33687765 PMCID: PMC8252100 DOI: 10.1111/jth.15288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/21/2021] [Accepted: 02/10/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Activated factor IX (FIXa) is an inefficient enzyme that needs activated factor VIII (FVIII) for full activity. Recently, we identified a network of FVIII-driven changes in FIXa employing hydrogen-deuterium eXchange mass spectrometry (HDX-MS). Some changes also occurred in active-site inhibited FIXa, but others were not cofactor-driven, in particular those within the 220-loop (in chymotrypsin numbering). OBJECTIVE The aim of this work is to better understand the zymogen-to-enzyme transition in FIX, with specific focus on substrate-driven changes at the catalytic site. METHODS Footprinting mass spectrometry by HDX and Tandem-Mass Tags (TMT) labelling were used to explore changes occurring upon the conversion from FIX into FIXa. Mutagenesis and kinetic studies served to assess the role of the 220-loop. RESULTS HDX-MS displayed remarkably few differences between FIX and FIXa. In comparison with FIX, FIXa did exhibit decreased deuterium uptake at the N-terminus region. This was more prominent when the FIXa active site was occupied by an irreversible inhibitor. TMT-labelling showed that the N-terminus is largely protected from labelling, and that inhibitor binding increases protection to a minor extent. Occupation of the active site also reduced deuterium uptake within the 220-loop backbone. Mutagenesis within the 220-loop revealed that a putative H-bond network contributes to FIXa activity. TMT labeling of the N-terminus suggested that these 220-loop variants are more zymogen-like than wild-type FIXa. CONCLUSION In the absence of cofactor and substrate, FIXa is predominantly zymogen-like. Stabilization in its enzyme-like form involves, apart from FVIII-binding, also interplay between the 220-loop, N-terminus, and the substrate binding site.
Collapse
Affiliation(s)
- Nadia Freato
- Department of Molecular and Cellular HemostasisSanquin ResearchAmsterdamThe Netherlands
| | | | - Mariëtte Boon‐Spijker
- Department of Molecular and Cellular HemostasisSanquin ResearchAmsterdamThe Netherlands
| | | | - Alexander B. Meijer
- Department of Molecular and Cellular HemostasisSanquin ResearchAmsterdamThe Netherlands
- Department of Biomolecular Mass Spectrometry and ProteomicsUtrecht Institute for Pharmaceutical Sciences (UIPS)Utrecht UniversityUtrechtThe Netherlands
| | - Koen Mertens
- Department of Molecular and Cellular HemostasisSanquin ResearchAmsterdamThe Netherlands
- Department of PharmaceuticsUtrecht Institute for Pharmaceutical Sciences (UIPS)Utrecht UniversityUtrechtThe Netherlands
| | | |
Collapse
|
4
|
Factor VIII-driven changes in activated factor IX explored by hydrogen-deuterium exchange mass spectrometry. Blood 2021; 136:2703-2714. [PMID: 32678887 DOI: 10.1182/blood.2020005593] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/29/2020] [Indexed: 11/20/2022] Open
Abstract
The assembly of the enzyme-activated factor IX (FIXa) with its cofactor, activated factor VIII (FVIIIa) is a crucial event in the coagulation cascade. The absence or dysfunction of either enzyme or cofactor severely compromises hemostasis and causes hemophilia. FIXa is a notoriously inefficient enzyme that needs FVIIIa to drive its hemostatic potential, by a mechanism that has remained largely elusive to date. In this study, we employed hydrogen-deuterium exchange-mass spectrometry (HDX-MS) to investigate how FIXa responds to assembly with FVIIIa in the presence of phospholipids. This revealed a complex pattern of changes that partially overlaps with those changes that occur upon occupation of the substrate-binding site by an active site-directed inhibitor. Among the changes driven by both cofactor and substrate, HDX-MS highlighted several surface loops that have been implicated in allosteric networks in related coagulation enzymes. Inspection of FVIIIa-specific changes indicated that 3 helices are involved in FIXa-FVIIIa assembly. These are part of a basic interface that is also known as exosite II. Mutagenesis of basic residues herein, followed by functional studies, identified this interface as an extended FVIIIa-interactive patch. HDX-MS was also applied to recombinant FIXa variants that are associated with severe hemophilia B. This revealed that single amino acid substitutions can silence the extended network of FVIIIa-driven allosteric changes. We conclude that HDX-MS has the potential to visualize the functional impact of disease-associated mutations on enzyme-cofactor complexes in the hemostatic system.
Collapse
|
5
|
Molecular determinants of the factor VIII/von Willebrand factor complex revealed by BIVV001 cryo-electron microscopy. Blood 2021; 137:2970-2980. [PMID: 33569592 DOI: 10.1182/blood.2020009197] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/01/2021] [Indexed: 12/15/2022] Open
Abstract
Interaction of factor VIII (FVIII) with von Willebrand factor (VWF) is mediated by the VWF D'D3 domains and thrombin-mediated release is essential for hemostasis after vascular injury. VWF-D'D3 mutations resulting in loss of FVIII binding are the underlying cause of von Willebrand disease (VWD) type 2N. Furthermore, the FVIII-VWF interaction has significant implications for the development of therapeutics for bleeding disorders, particularly hemophilia A, in which endogenous VWF clearance imposes a half-life ceiling on replacement FVIII therapy. To understand the structural basis of FVIII engagement by VWF, we solved the structure of BIVV001 by cryo-electron microscopy to 2.9 Å resolution. BIVV001 is a bioengineered clinical-stage FVIII molecule for the treatment of hemophilia A. In BIVV001, VWF-D'D3 is covalently linked to an Fc domain of a B domain-deleted recombinant FVIII (rFVIII) Fc fusion protein, resulting in a stabilized rFVIII/VWF-D'D3 complex. Our rFVIII/VWF structure resolves BIVV001 architecture and provides a detailed spatial understanding of previous biochemical and clinical observations related to FVIII-VWF engagement. Notably, the FVIII acidic a3 peptide region (FVIII-a3), established as a critical determinant of FVIII/VWF complex formation, inserts into a basic groove formed at the VWF-D'/rFVIII interface. Our structure shows direct interaction of sulfated Y1680 in FVIII-a3 and VWF-R816 that, when mutated, leads to severe hemophilia A or VWD type 2N, respectively. These results provide insight on this key coagulation complex, explain the structural basis of many hemophilia A and VWD type 2N mutations, and inform studies to further elucidate how VWF dissociates rapidly from FVIII upon activation.
Collapse
|
6
|
van Galen J, Freato N, Przeradzka MA, Ebberink EHTM, Boon-Spijker M, van der Zwaan C, van den Biggelaar M, Meijer AB. Hydrogen-Deuterium Exchange Mass Spectrometry Identifies Activated Factor IX-Induced molecular Changes in Activated Factor VIII. Thromb Haemost 2020; 121:594-602. [PMID: 33302303 DOI: 10.1055/s-0040-1721422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) was employed to gain insight into the changes in factor VIII (FVIII) that occur upon its activation and assembly with activated factor IX (FIXa) on phospholipid membranes. HDX-MS analysis of thrombin-activated FVIII (FVIIIa) revealed a marked increase in deuterium incorporation of amino acid residues along the A1-A2 and A2-A3 interface. Rapid dissociation of the A2 domain from FVIIIa can explain this observation. In the presence of FIXa, enhanced deuterium incorporation at the interface of FVIIIa was similar to that of FVIII. This is compatible with the previous finding that FIXa contributes to A2 domain retention in FVIIIa. A2 domain region Leu631-Tyr637, which is not part of the interface between the A domains, also showed a marked increase in deuterium incorporation in FVIIIa compared with FVIII. Deuterium uptake of this region was decreased in the presence of FIXa beyond that observed in FVIII. This implies that FIXa alters the conformation or directly interacts with this region in FVIIIa. Replacement of Val634 in FVIII by alanine using site-directed mutagenesis almost completely impaired the ability of the activated cofactor to enhance the activity of FIXa. Surface plasmon resonance analysis revealed that the rates of A2 domain dissociation from FVIIIa and FVIIIa-Val634Ala were indistinguishable. HDX-MS analysis showed, however, that FIXa was unable to retain the A2 domain in FVIIIa-Val634Ala. The combined results of this study suggest that the local structure of Leu631-Tyr637 is altered by FIXa and that this region contributes to the cofactor function of FVIII.
Collapse
Affiliation(s)
- Josse van Galen
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands
| | - Nadia Freato
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands
| | - Małgorzata A Przeradzka
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands
| | - Eduard H T M Ebberink
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands
| | - Mariëtte Boon-Spijker
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands
| | - Carmen van der Zwaan
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands
| | | | - Alexander B Meijer
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands.,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
7
|
Przeradzka MA, Freato N, Boon-Spijker M, van Galen J, van der Zwaan C, Mertens K, van den Biggelaar M, Meijer AB. Unique surface-exposed hydrophobic residues in the C1 domain of factor VIII contribute to cofactor function and von Willebrand factor binding. J Thromb Haemost 2020; 18:364-372. [PMID: 31675465 DOI: 10.1111/jth.14668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/29/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND The identity of the amino acid regions of factor VIII (FVIII) that contribute to factor IXa (FIXa) and von Willebrand factor (VWF) binding has not been fully resolved. Previously, we observed that replacing the FVIII C1 domain for the one of factor V (FV) markedly reduces VWF binding and cofactor function. Compared to the FV C1 domain, this implies that the FVIII C1 domain comprises unique surface-exposed elements involved in VWF and FIXa interaction. OBJECTIVE The aim of this study is to identify residues in the FVIII C1 domain that contribute to VWF and FIXa binding. METHODS Structures and primary sequences of FVIII and FV were compared to identify surface-exposed residues unique to the FVIII C1 domain. The identified residues were replaced with alanine residues to identify their role in FIXa and VWF interaction. This role was assessed employing surface plasmon resonance analysis studies and enzyme kinetic assays. RESULTS Five surface-exposed hydrophobic residues unique to the FVIII C1 domain, ie, F2035, F2068, F2127, V2130, I2139 were identified. Functional analysis indicated that residues F2068, V2130, and especially F2127 contribute to VWF and/or FIXa interaction. Substitution into alanine of the also surface-exposed V2125, which is spatially next to F2127, affected only VWF binding. CONCLUSION The surface-exposed hydrophobic residues in C1 domain contribute to cofactor function and VWF binding. These findings provide novel information on the fundamental role of the C1 domain in FVIII life cycle.
Collapse
Affiliation(s)
- Małgorzata A Przeradzka
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, the Netherlands
| | - Nadia Freato
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, the Netherlands
| | - Mariëtte Boon-Spijker
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, the Netherlands
| | - Josse van Galen
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, the Netherlands
| | - Carmen van der Zwaan
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, the Netherlands
| | - Koen Mertens
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, the Netherlands
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | | | - Alexander B Meijer
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, the Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
8
|
Przeradzka MA, van Galen J, Ebberink EHTM, Hoogendijk AJ, van der Zwaan C, Mertens K, van den Biggelaar M, Meijer AB. D' domain region Arg782-Cys799 of von Willebrand factor contributes to factor VIII binding. Haematologica 2019; 105:1695-1703. [PMID: 31558672 PMCID: PMC7271574 DOI: 10.3324/haematol.2019.221994] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022] Open
Abstract
In the complex with von Willebrand factor (VWF) factor VIII (FVIII) is protected from rapid clearance from circulation. Although it has been established that the FVIII binding site resides in the N-terminal D'-D3 domains of VWF, detailed information about the amino acid regions that contribute to FVIII binding is still lacking. In the present study, hydrogen-deuterium exchange mass spectrometry was employed to gain insight into the FVIII binding region on VWF. To this end, time-dependent deuterium incorporation was assessed in D'-D3 and the FVIII-D'-D3 complex. Data showed reduced deuterium incorporation in the D' region Arg782-Cys799 in the FVIII-D'-D3 complex compared to D'-D3. This implies that this region interacts with FVIII. Site-directed mutagenesis of the six charged amino acids in Arg782-Cys799 into alanine residues followed by surface plasmon resonance analysis and solid phase binding studies revealed that replacement of Asp796 affected FVIII binding. A marked decrease in FVIII binding was observed for the D'-D3 Glu787Ala variant. The same was observed for D'-D3 variants in which Asp796 and Glu787 were replaced by Asn796 and Gln787. Site-directed mutagenesis of Leu786, which together with Glu787 and Cys789 forms a short helical region in the crystal structure of D'-D3, also had a marked impact on FVIII binding. The combined results show that the amino acid region Arg782-Cys799 is part of a FVIII binding surface. Our study provides new insight into FVIII-VWF complex formation and defects therein that may be associated with bleeding caused by markedly reduced levels of FVIII.
Collapse
Affiliation(s)
| | - Josse van Galen
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam
| | | | - Arie J Hoogendijk
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam
| | | | - Koen Mertens
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam
| | | | - Alexander B Meijer
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam .,Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
9
|
Cheng J, Geng F, Hu J, Lü J. Single-molecule measurement and bioinformatics analysis suggest a preferred orientation of human coagulation factor VIII on hydrophobic interfaces. Biophys Chem 2019; 248:9-15. [PMID: 30901531 DOI: 10.1016/j.bpc.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/04/2019] [Accepted: 03/13/2019] [Indexed: 11/28/2022]
Abstract
Investigating the adsorption behavior of coagulation proteins on interfaces will contribute to better understating blood clotting and to the development of biocompatible materials. In this work, atomic force microscopy (AFM)-based peakforce quantitative nanomechanical mapping (PF-QNM) was combined with bioinformatics tool to study the adsorption and orientation of coagulation factor VIII (FVIII) on both hydrophilic and hydrophobic interfaces by the height and mechanical measurement of single protein molecules. We found that interfacial hydrophilicity/hydrophobicity greatly influence the heights and Young's modulus of individual proteins. Compared to on the hydrophilic mica surface, FVIII proteins appear bigger vertical sizes while similar lateral sizes on the HOPG surface. The water accessible surface area analysis indicate stronger apolar properties C1 and C2 domains than others, suggesting a preferred orientation through the strong hydrophobic interactions between HOPG and the hydrophobic residues interface of the protein domains. These results provide novel insights on the adsorption and binding mechanism of the FVIII on cell membrane and will be helpful for the design of anticoagulant materials.
Collapse
Affiliation(s)
- Jie Cheng
- Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China; CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Geng
- School of Pharmacy, Binzhou Medicine University, Yantai 300064, China
| | - Jun Hu
- Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China; CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Junhong Lü
- Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China; CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
| |
Collapse
|
10
|
Bloem E, Karpf DM, Nørby PL, Johansen PB, Loftager M, Rahbek-Nielsen H, Petersen HH, Blouse GE, Thim L, Kjalke M, Bolt G. Factor VIII with a 237 amino acid B-domain has an extended half-life in F8-knockout mice. J Thromb Haemost 2019; 17:350-360. [PMID: 30525289 DOI: 10.1111/jth.14355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Indexed: 12/30/2022]
Abstract
Essentials Factor (F)VIII with an intermediate-length B-domain showed higher levels in murine gene therapy. FVIII with different B-domain lengths were analysed. FVIII variants with B-domains between 186 and 240 amino acids (aa) have extended half-life in mice. Reduced cell binding of FVIII with a 237aa B-domain may explain the extended half-life. SUMMARY: Background Factor VIII consists of the A1-domain, A2-domain, B-domain, A3-domain, C1-domain, and C2-domain. FVIII with an intermediate-length B-domain of 226 amino acids (aa) has previously been evaluated in murine gene therapy studies. Objective To characterize FVIII with intermediate-length B-domains in vitro and in vivo in F8-knockout (KO) mice. Methods and results FVIII molecules with B-domains of 186-240aa had longer half-lives in F8-KO mice than FVIII molecules with shorter or longer B-domains. FVIII with a B-domain containing the 225 N-terminal aa fused to the 12 C-terminal aa of the wild-type B-domain (FVIII-237) had a 1.6-fold extended half-life in F8-KO mice as compared with FVIII with a 21aa B-domain (FVIII-21). The in vitro and in vivo activity of FVIII-237 were comparable to those of FVIII-21, as was binding to von Willebrand factor. Cell binding to LDL receptor-related protein 1 (LRP-1)-expressing cells was markedly reduced for FVIII-237 as compared with FVIII-21, whereas the affinity for LRP-1 was not reduced in surface plasmon resonance (SPR) studies. FVIII-21 cell binding and internalization could be inhibited by a fragment consisting of the 226 N-terminal aa of the FVIII B-domain, and SPR analysis suggested that this B-domain fragment might bind with weak affinity to FVIII-21. Conclusion Reduced cell binding of FVIII-237 might explain the observed extended half-life in F8-KO mice. This may contribute to the increased FVIII levels measured in murine gene therapy studies using FVIII constructs with similar B-domain lengths.
Collapse
Affiliation(s)
- E Bloem
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - D M Karpf
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - P L Nørby
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - P B Johansen
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - M Loftager
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | | | - H H Petersen
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - G E Blouse
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - L Thim
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - M Kjalke
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - G Bolt
- Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| |
Collapse
|
11
|
The evolving understanding of factor VIII binding sites and implications for the treatment of hemophilia A. Blood Rev 2019; 33:1-5. [DOI: 10.1016/j.blre.2018.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/29/2018] [Accepted: 05/22/2018] [Indexed: 11/21/2022]
|
12
|
The D' domain of von Willebrand factor requires the presence of the D3 domain for optimal factor VIII binding. Biochem J 2018; 475:2819-2830. [PMID: 30111575 DOI: 10.1042/bcj20180431] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/07/2018] [Accepted: 08/14/2018] [Indexed: 11/17/2022]
Abstract
The D'-D3 fragment of von Willebrand factor (VWF) can be divided into TIL'-E'-VWD3-C8_3-TIL3-E3 subdomains of which TIL'-E'-VWD3 comprises the main factor VIII (FVIII)-binding region. Yet, von Willebrand disease (VWD) Type 2 Normandy (2N) mutations, associated with impaired FVIII interaction, have been identified in C8_3-TIL3-E3. We now assessed the role of the VWF (sub)domains for FVIII binding using isolated D', D3 and monomeric C-terminal subdomain truncation variants of D'-D3. Competitive binding assays and surface plasmon resonance analysis revealed that D' requires the presence of D3 for effective interaction with FVIII. The isolated D3 domain, however, did not show any FVIII binding. Results indicated that the E3 subdomain is dispensable for FVIII binding. Subsequent deletion of the other subdomains from D3 resulted in a progressive decrease in FVIII-binding affinity. Chemical footprinting mass spectrometry suggested increased conformational changes at the N-terminal side of D3 upon subsequent subdomain deletions at the C-terminal side of the D3. A D'-D3 variant with a VWD type 2N mutation in VWD3 (D879N) or C8_3 (C1060R) also revealed conformational changes in D3, which were proportional to a decrease in FVIII-binding affinity. A D'-D3 variant with a putative VWD type 2N mutation in the E3 subdomain (C1225G) showed, however, normal binding. This implies that the designation VWD type 2N is incorrect for this variant. Results together imply that a structurally intact D3 in D'-D3 is indispensable for effective interaction between D' and FVIII explaining why specific mutations in D3 can impair FVIII binding.
Collapse
|
13
|
Castro-Núñez L, Koornneef JM, Rondaij MG, Bloem E, van der Zwaan C, Mertens K, Meijer AB, Meems H. Cellular uptake of coagulation factor VIII: Elusive role of the membrane-binding spikes in the C1 domain. Int J Biochem Cell Biol 2017; 89:34-41. [DOI: 10.1016/j.biocel.2017.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 03/31/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
|
14
|
Hartholt RB, van Velzen AS, Peyron I, Ten Brinke A, Fijnvandraat K, Voorberg J. To serve and protect: The modulatory role of von Willebrand factor on factor VIII immunogenicity. Blood Rev 2017; 31:339-347. [PMID: 28716211 DOI: 10.1016/j.blre.2017.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/26/2017] [Accepted: 07/03/2017] [Indexed: 12/23/2022]
Abstract
Hemophilia A is a bleeding disorder characterized by the absence or dysfunction of blood coagulation factor VIII (FVIII). Patients are treated with regular infusions of FVIII concentrate. In response to treatment, approximately 30% of patients with severe hemophilia A develop inhibitory antibodies targeting FVIII. Both patient and treatment related risk factors for inhibitor development have been described. Multiple studies comparing the immunogenicity of recombinant and plasma-derived FVIII have yielded conflicting results. The randomized controlled SIPPET (Survey of Inhibitors in Plasma-Product Exposed Toddlers) trial demonstrated an increased risk of inhibitor development of recombinant FVIII when compared to von Willebrand factor (VWF)-containing plasma-derived FVIII. Presently, it is unclear which mechanism underlies the reduced immunogenicity of plasma-derived FVIII. In this review we address the potential role of VWF on FVIII immunogenicity and we discuss how VWF affects the immune recognition, processing and presentation of FVIII. We also briefly discuss the potential impact of glycan-composition on FVIII immunogenicity. It is well established that VWF shields the uptake of FVIII by antigen presenting cells. We have recently shown that VWF binds to the surface of dendritic cells. Here, we present a novel model in which surface bound FVIII-VWF complexes regulate the internalization of FVIII. Binding of FVIII to VWF is critically dependent on sulfation of Tyr1699 (HVGS numbering) in the light chain of FVIII. Incomplete sulfation of Tyr1699 has been suggested to occur in several recombinant FVIII products resulting in a loss of VWF binding. We hypothesize that this results in alternative pathways of FVIII internalization by antigen presenting cells which are not regulated by VWF. This hypothetical mechanism may explain the reduced immunogenicity of VWF containing plasma-derived FVIII concentrates as found in the SIPPET study.
Collapse
Affiliation(s)
- Robin B Hartholt
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Alice S van Velzen
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | - Ivan Peyron
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Anja Ten Brinke
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Karin Fijnvandraat
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | - Jan Voorberg
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Ebberink EHTM, Bouwens EAM, Bloem E, Boon-Spijker M, van den Biggelaar M, Voorberg J, Meijer AB, Mertens K. Factor VIII/V C-domain swaps reveal discrete C-domain roles in factor VIII function and intracellular trafficking. Haematologica 2017; 102:686-694. [PMID: 28057741 PMCID: PMC5395109 DOI: 10.3324/haematol.2016.153163] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/23/2016] [Indexed: 01/28/2023] Open
Abstract
Factor VIII C-domains are believed to have specific functions in cofactor activity and in interactions with von Willebrand factor. We have previously shown that factor VIII is co-targeted with von Willebrand factor to the Weibel-Palade bodies in blood outgrowth endothelial cells, even when factor VIII carries mutations in the light chain that are associated with defective von Willebrand factor binding. In this study, we addressed the contribution of individual factor VIII C-domains in intracellular targeting, von Willebrand factor binding and cofactor activity by factor VIII/V C-domain swapping. Blood outgrowth endothelial cells were transduced with lentivirus encoding factor V, factor VIII or YFP-tagged C-domain chimeras, and examined by confocal microscopy. The same chimeras were produced in HEK293-cells for in vitro characterization and chemical foot-printing by mass spectrometry. In contrast to factor VIII, factor V did not target to Weibel-Palade bodies. The chimeras showed reduced Weibel-Palade body targeting, suggesting that this requires the factor VIII C1–C2 region. The factor VIII/V-C1 chimera did not bind von Willebrand factor and had reduced affinity for activated factor IX, whereas the factor VIII/V-C2 chimera showed a minor reduction in von Willebrand factor binding and normal interaction with activated factor IX. This suggests that mainly the C1-domain carries factor VIII-specific features in assembly with von Willebrand factor and activated factor IX. Foot-printing analysis of the chimeras revealed increased exposure of lysine residues in the A1/C2- and C1/C2-domain interface, suggesting increased C2-domain mobility and disruption of the natural C-domain tandem pair orientation. Apparently, this affects intracellular trafficking, but not extracellular function.
Collapse
Affiliation(s)
| | | | - Esther Bloem
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands
| | | | | | - Jan Voorberg
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands.,Landsteiner Laboratory of AMC and Sanquin, University of Amsterdam, the Netherlands
| | - Alexander B Meijer
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands.,Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| | - Koen Mertens
- Department of Plasma Proteins, Sanquin Research, Amsterdam, the Netherlands .,Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| |
Collapse
|
16
|
Gangadharan B, Ing M, Delignat S, Peyron I, Teyssandier M, Kaveri SV, Lacroix-Desmazes S. The C1 and C2 domains of blood coagulation factor VIII mediate its endocytosis by dendritic cells. Haematologica 2016; 102:271-281. [PMID: 27758819 DOI: 10.3324/haematol.2016.148502] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 10/03/2016] [Indexed: 02/01/2023] Open
Abstract
The development of inhibitory antibodies to therapeutic factor VIII is the major complication of replacement therapy in patients with hemophilia A. The first step in the initiation of the anti-factor VIII immune response is factor VIII interaction with receptor(s) on antigen-presenting cells, followed by endocytosis and presentation to naïve CD4+ T cells. Recent studies indicate a role for the C1 domain in factor VIII uptake. We investigated whether charged residues in the C2 domain participate in immunogenic factor VIII uptake. Co-incubation of factor VIII with BO2C11, a monoclonal C2-specific immunoglobulin G, reduced factor VIII endocytosis by dendritic cells and presentation to CD4+ T cells, and diminished factor VIII immunogenicity in factor VIII-deficient mice. The mutation of basic residues within the BO2C11 epitope of C2 replicated reduced in vitro immunogenic uptake, but failed to prevent factor VIII immunogenicity in mice. BO2C11 prevents factor VIII binding to von Willebrand factor, thus potentially biasing factor VIII immunogenicity by perturbing its half-life. Interestingly, a factor VIIIY1680C mutant, that does not bind von Willebrand factor, demonstrated unaltered endocytosis by dendritic cells as well as immunogenicity in factor VIII-deficient mice. Co-incubation of factor VIIIY1680C with BO2C11, however, resulted in decreased factor VIII immunogenicity in vivo In addition, a previously described triple C1 mutant showed decreased uptake in vitro, and reduced immunogenicity in vivo, but only in the absence of endogenous von Willebrand factor. Taken together, the results indicate that residues in the C1 and/or C2 domains of factor VIII are implicated in immunogenic factor VIII uptake, at least in vitro Conversely, in vivo, the binding to endogenous von Willebrand factor masks the reducing effect of mutations in the C domains on factor VIII immunogenicity.
Collapse
Affiliation(s)
- Bagirath Gangadharan
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,INSERM, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Mathieu Ing
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,INSERM, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Sandrine Delignat
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,INSERM, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Ivan Peyron
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,INSERM, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Maud Teyssandier
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,INSERM, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Srinivas V Kaveri
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,INSERM, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Sébastien Lacroix-Desmazes
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France .,INSERM, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| |
Collapse
|
17
|
Doherty CM, Visse R, Dinakarpandian D, Strickland DK, Nagase H, Troeberg L. Engineered Tissue Inhibitor of Metalloproteinases-3 Variants Resistant to Endocytosis Have Prolonged Chondroprotective Activity. J Biol Chem 2016; 291:22160-22172. [PMID: 27582494 PMCID: PMC5063997 DOI: 10.1074/jbc.m116.733261] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Indexed: 01/03/2023] Open
Abstract
Tissue inhibitor of metalloproteinases-3 (TIMP-3) is a central inhibitor of matrix-degrading and sheddase families of metalloproteinases. Extracellular levels of the inhibitor are regulated by the balance between its retention on the extracellular matrix and its endocytic clearance by the scavenger receptor low density lipoprotein receptor-related protein 1 (LRP1). Here, we used molecular modeling to predict TIMP-3 residues potentially involved in binding to LRP1 based on the proposed LRP1 binding motif of 2 lysine residues separated by about 21 Å and mutated the candidate lysine residues to alanine individually and in pairs. Of the 22 mutants generated, 13 displayed a reduced rate of uptake by HTB94 chondrosarcoma cells. The two mutants (TIMP-3 K26A/K45A and K42A/K110A) with lowest rates of uptake were further evaluated and found to display reduced binding to LRP1 and unaltered inhibitory activity against prototypic metalloproteinases. TIMP-3 K26A/K45A retained higher affinity for sulfated glycosaminoglycans than K42A/K110A and exhibited increased affinity for ADAMTS-5 in the presence of heparin. Both mutants inhibited metalloproteinase-mediated degradation of cartilage at lower concentrations and for longer than wild-type TIMP-3, indicating that their increased half-lives improved their ability to protect cartilage. These mutants may be useful in treating connective tissue diseases associated with increased metalloproteinase activity.
Collapse
Affiliation(s)
- Christine M Doherty
- From the Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom
| | - Robert Visse
- From the Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom
| | - Deendayal Dinakarpandian
- the School of Computing and Engineering, University of Missouri, Kansas City, Missouri 64111, and
| | | | - Hideaki Nagase
- From the Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom
| | - Linda Troeberg
- From the Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom,
| |
Collapse
|
18
|
Madsen JJ, Ohkubo YZ, Peters GH, Faber JH, Tajkhorshid E, Olsen OH. Membrane Interaction of the Factor VIIIa Discoidin Domains in Atomistic Detail. Biochemistry 2015; 54:6123-31. [PMID: 26346528 DOI: 10.1021/acs.biochem.5b00417] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A recently developed membrane-mimetic model was applied to study membrane interaction and binding of the two anchoring C2-like discoidin domains of human coagulation factor VIIIa (FVIIIa), the C1 and C2 domains. Both individual domains, FVIII C1 and FVIII C2, were observed to bind the phospholipid membrane by partial or full insertion of their extruding loops (the spikes). However, the two domains adopted different molecular orientations in their membrane-bound states; FVIII C2 roughly was positioned normal to the membrane plane, while FVIII C1 displayed a multitude of tilted orientations. The results indicate that FVIII C1 may be important in modulating the orientation of the FVIIIa molecule to optimize the interaction with FIXa, which is anchored to the membrane via its γ-carboxyglutamic acid-rich (Gla) domain. Additionally, a structural change was observed in FVIII C1 in the coiled main chain leading the first spike. A tight interaction with one lipid per domain, similar to what has been suggested for the homologous FVa C2, is characterized. Finally, we rationalize known FVIII antibody epitopes and the scarcity of documented hemophilic missense mutations related to improper membrane binding of FVIIIa, based on the prevalent nonspecificity of ionic interactions in the simulated membrane-bound states of FVIII C1 and FVIII C2.
Collapse
Affiliation(s)
- Jesper J Madsen
- Global Research, Novo Nordisk A/S , DK-2760 Måløv, Denmark.,Department of Chemistry, Technical University of Denmark , DK-2800 Kgs. Lyngby, Denmark
| | | | - Günther H Peters
- Department of Chemistry, Technical University of Denmark , DK-2800 Kgs. Lyngby, Denmark
| | - Johan H Faber
- Global Research, Novo Nordisk A/S , DK-2760 Måløv, Denmark
| | | | - Ole H Olsen
- Global Research, Novo Nordisk A/S , DK-2760 Måløv, Denmark
| |
Collapse
|
19
|
A novel chemical footprinting approach identifies critical lysine residues involved in the binding of receptor-associated protein to cluster II of LDL receptor-related protein. Biochem J 2015; 468:65-72. [PMID: 25728577 DOI: 10.1042/bj20140977] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Tandem mass tags (TMTs) were utilized in a novel chemical footprinting approach to identify lysine residues that mediate the interaction of receptor-associated protein (RAP) with cluster II of LDL (low-density lipoprotein) receptor (LDLR)-related protein (LRP). The isolated RAP D3 domain was modified with TMT-126 and the D3 domain-cluster II complex with TMT-127. Nano-LC-MS analysis revealed reduced modification with TMT-127 of peptides including Lys(256), Lys(270) and Lys(305)-Lys(306) suggesting that these residues contribute to cluster II binding. This agrees with previous findings that Lys(256) and Lys(270) are critical for binding cluster II sub-domains [Fisher, Beglova and Blacklow (2006) Mol. Cell 22, 277-283]. Cluster II-binding studies utilizing D3 domain variants K(256)A, K(305)A and K(306)A now showed that Lys(306) contributes to cluster II binding as well. For full-length RAP, we observed that peptides including Lys(60), Lys(191), Lys(256), Lys(270) and Lys(305)-Lys(306) exhibited reduced modification with TMT in the RAP-cluster II complex. Notably, Lys(60) has previously been implicated to mediate D1 domain interaction with cluster II. Our results suggest that also Lys(191) of the D2 domain contributes to cluster II binding. Binding studies employing the RAP variants K(191)A, K(256)A, K(305)A and K(306)A, however, revealed a modest reduction in cluster II binding for the K(256)A variant only. This suggests that the other lysine residues can compensate for the absence of a single lysine residue for effective complex assembly. Collectively, novel insight has been obtained into the contribution of lysine residues of RAP to cluster II binding. In addition, we propose that TMTs can be utilized to identify lysine residues critical for protein complex formation.
Collapse
|
20
|
van den Biggelaar M, Madsen JJ, Faber JH, Zuurveld MG, van der Zwaan C, Olsen OH, Stennicke HR, Mertens K, Meijer AB. Factor VIII Interacts with the Endocytic Receptor Low-density Lipoprotein Receptor-related Protein 1 via an Extended Surface Comprising "Hot-Spot" Lysine Residues. J Biol Chem 2015; 290:16463-76. [PMID: 25903134 DOI: 10.1074/jbc.m115.650911] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Indexed: 11/06/2022] Open
Abstract
Lysine residues are implicated in driving the ligand binding to the LDL receptor family. However, it has remained unclear how specificity is regulated. Using coagulation factor VIII as a model ligand, we now study the contribution of individual lysine residues in the interaction with the largest member of the LDL receptor family, low-density lipoprotein receptor-related protein (LRP1). Using hydrogen-deuterium exchange mass spectrometry (HDX-MS) and SPR interaction analysis on a library of lysine replacement variants as two independent approaches, we demonstrate that the interaction between factor VIII (FVIII) and LRP1 occurs over an extended surface containing multiple lysine residues. None of the individual lysine residues account completely for LRP1 binding, suggesting an additive binding model. Together with structural docking studies, our data suggest that FVIII interacts with LRP1 via an extended surface of multiple lysine residues that starts at the bottom of the C1 domain and winds around the FVIII molecule.
Collapse
Affiliation(s)
- Maartje van den Biggelaar
- From the Department of Plasma Proteins, Sanquin Blood Supply Foundation, 1066 CX Amsterdam, The Netherlands,
| | - Jesper J Madsen
- Global Research, Novo Nordisk A/S, DK-2760 Måløv, Denmark, and
| | - Johan H Faber
- Global Research, Novo Nordisk A/S, DK-2760 Måløv, Denmark, and
| | - Marleen G Zuurveld
- From the Department of Plasma Proteins, Sanquin Blood Supply Foundation, 1066 CX Amsterdam, The Netherlands
| | - Carmen van der Zwaan
- From the Department of Plasma Proteins, Sanquin Blood Supply Foundation, 1066 CX Amsterdam, The Netherlands
| | - Ole H Olsen
- Global Research, Novo Nordisk A/S, DK-2760 Måløv, Denmark, and
| | | | - Koen Mertens
- From the Department of Plasma Proteins, Sanquin Blood Supply Foundation, 1066 CX Amsterdam, The Netherlands, the Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TC Utrecht, The Netherlands
| | - Alexander B Meijer
- From the Department of Plasma Proteins, Sanquin Blood Supply Foundation, 1066 CX Amsterdam, The Netherlands, the Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TC Utrecht, The Netherlands
| |
Collapse
|
21
|
Kurasawa JH, Shestopal SA, Woodle SA, Ovanesov MV, Lee TK, Sarafanov AG. Cluster III of low-density lipoprotein receptor-related protein 1 binds activated blood coagulation factor VIII. Biochemistry 2014; 54:481-9. [PMID: 25486042 DOI: 10.1021/bi5011688] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP) mediates clearance of blood coagulation factor VIII (FVIII). In LRP, FVIII binds the complement-type repeats (CRs) of clusters II and IV, which also bind a majority of other LRP ligands. No ligand is known for LRP cluster I, and only three ligands, including the LRP chaperone alpha-2 macroglobulin receptor-associated protein (RAP), bind cluster III. Using surface plasmon resonance, we found that in addition to clusters II and IV, activated FVIII (FVIIIa) binds cluster III. The specificity of this interaction was confirmed using an anti-FVIII antibody fragment, which inhibited the binding. Recombinant fragments of cluster III and its site-directed mutagenesis were used to localize the cluster's site for binding FVIIIa to CR.14-19. The interactive site of FVIIIa was localized within its A1/A3'-C1-C2 heterodimer (HDa), which is a major physiological remnant of FVIIIa. In mice, the clearance of HDa was faster than that of FVIII and prolonged in the presence of RAP, which is known to inhibit interactions of LRP with its ligands. In accordance with this, the cluster III site for RAP (CR.15-19) was found to overlap that for FVIIIa. Altogether, our findings support the involvement of LRP in FVIIIa catabolism and suggest a greater significance of the biological role of cluster III compared to that previously known.
Collapse
Affiliation(s)
- James H Kurasawa
- Center for Biologics Evaluation and Research, Food and Drug Administration , Silver Spring, Maryland 20993-0002, United States
| | | | | | | | | | | |
Collapse
|
22
|
Bloem E, van den Biggelaar M, Wroblewska A, Voorberg J, Faber JH, Kjalke M, Stennicke HR, Mertens K, Meijer AB. Factor VIII C1 domain spikes 2092-2093 and 2158-2159 comprise regions that modulate cofactor function and cellular uptake. J Biol Chem 2013; 288:29670-9. [PMID: 24009077 PMCID: PMC3795264 DOI: 10.1074/jbc.m113.473116] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/30/2013] [Indexed: 01/05/2023] Open
Abstract
The C1 domain of factor VIII (FVIII) has been implicated in binding to multiple constituents, including phospholipids, von Willebrand factor, and low-density lipoprotein receptor-related protein (LRP). We have previously described a human monoclonal antibody called KM33 that blocks these interactions as well as cellular uptake by LRP-expressing cells. To unambiguously identify the apparent "hot spot" on FVIII to which this antibody binds, we have employed hydrogen-deuterium exchange mass spectrometry. The results showed that KM33 protects FVIII regions 2091-2104 and 2157-2162 from hydrogen-deuterium exchange. These comprise the two C1 domain spikes 2092-2093 and 2158-2159. Spike 2092-2093 has been demonstrated recently to contribute to assembly with lipid membranes with low phosphatidylserine (PS) content. Therefore, spike 2158-2159 might serve a similar role. This was assessed by replacement of Arg-2159 for Asn, which introduces a motif for N-linked glycosylation. Binding studies revealed that the purified, glycosylated R2159N variant had lost its interaction with antibody KM33 but retained substantial binding to von Willebrand factor and LRP. Cellular uptake of the R2159N variant was reduced both by LRP-expressing U87-MG cells and by human monocyte-derived dendritic cells. FVIII activity was virtually normal on membranes containing 15% PS but reduced at low PS content. These findings suggest that the C1 domain spikes 2092-2093 and 2158-2159 together modulate FVIII membrane assembly by a subtle, PS-dependent mechanism. These findings contribute evidence in favor of an increasingly important role of the C1 domain in FVIII biology.
Collapse
Affiliation(s)
- Esther Bloem
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | | | - Aleksandra Wroblewska
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | - Jan Voorberg
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | - Johan H. Faber
- the Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Måløv, Denmark, and
| | - Marianne Kjalke
- the Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Måløv, Denmark, and
| | - Henning R. Stennicke
- the Biopharmaceutical Research Unit, Novo Nordisk A/S, DK-2760 Måløv, Denmark, and
| | - Koen Mertens
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands
- the Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Alexander B. Meijer
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands
- the Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
23
|
Bloem E, Meems H, van den Biggelaar M, Mertens K, Meijer AB. A3 domain region 1803-1818 contributes to the stability of activated factor VIII and includes a binding site for activated factor IX. J Biol Chem 2013; 288:26105-26111. [PMID: 23884417 DOI: 10.1074/jbc.m113.500884] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A recent chemical footprinting study in our laboratory suggested that region 1803-1818 might contribute to A2 domain retention in activated factor VIII (FVIIIa). This site has also been implicated to interact with activated factor IX (FIXa). Asn-1810 further comprises an N-linked glycan, which seems incompatible with a role of the amino acids 1803-1818 for FIXa or A2 domain binding. In the present study, FVIIIa stability and FIXa binding were evaluated in a FVIII-N1810C variant, and two FVIII variants in which residues 1803-1810 and 1811-1818 are replaced by the corresponding residues of factor V (FV). Enzyme kinetic studies showed that only FVIII/FV 1811-1818 has a decreased apparent binding affinity for FIXa. Flow cytometry analysis indicated that fluorescent FIXa exhibits impaired complex formation with only FVIII/FV 1811-1818 on lipospheres. Site-directed mutagenesis revealed that Phe-1816 contributes to the interaction with FIXa. To evaluate FVIIIa stability, the FVIII/FV chimeras were activated by thrombin, and the decline in cofactor function was followed over time. FVIII/FV 1803-1810 and FVIII/FV 1811-1818 but not FVIII-N1810C showed a decreased FVIIIa half-life. However, when the FVIII variants were activated in presence of FIXa, only FVIII/FV 1811-1818 demonstrated an enhanced decline in cofactor function. Surface plasmon resonance analysis revealed that the FVIII variants K1813A/K1818A, E1811A, and F1816A exhibit enhanced dissociation after activation. The results together demonstrate that the glycan at 1810 is not involved in FVIII cofactor function, and that Phe-1816 of region 1811-1818 contributes to FIXa binding. Both regions 1803-1810 and 1811-1818 contribute to FVIIIa stability.
Collapse
Affiliation(s)
- Esther Bloem
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
| | - Henriet Meems
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
| | | | - Koen Mertens
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and; Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 1066 CX Amsterdam, The Netherlands
| | - Alexander B Meijer
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and; Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Kurasawa JH, Shestopal SA, Karnaukhova E, Struble EB, Lee TK, Sarafanov AG. Mapping the binding region on the low density lipoprotein receptor for blood coagulation factor VIII. J Biol Chem 2013; 288:22033-41. [PMID: 23754288 DOI: 10.1074/jbc.m113.468108] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Low density lipoprotein receptor (LDLR) was shown to mediate clearance of blood coagulation factor VIII (FVIII) from the circulation. To elucidate the mechanism of interaction of LDLR and FVIII, our objective was to identify the region of the receptor necessary for binding FVIII. Using surface plasmon resonance, we found that LDLR exodomain and its cluster of complement-type repeats (CRs) bind FVIII in the same mode. This indicated that the LDLR site for FVIII is located within the LDLR cluster. Similar results were obtained for another ligand of LDLR, α-2-macroglobulin receptor-associated protein (RAP), a common ligand of receptors from the LDLR family. We further generated a set of recombinant fragments of the LDLR cluster and assessed their structural integrity by binding to RAP and by circular dichroism. A number of fragments overlapping CR.2-5 of the cluster were positive for binding RAP and FVIII. The specificity of these interactions was tested by site-directed mutagenesis of conserved tryptophans within the LDLR fragments. For FVIII, the specificity was also tested using a single-chain variable antibody fragment directed against the FVIII light chain as a competitor. Both cases resulted in decreased binding, thus confirming its specificity. The mutagenic study also showed an importance of the conserved tryptophans in LDLR for both ligands, and the competitive binding results showed an involvement of the light chain of FVIII in its interaction with LDLR. In conclusion, the region of CR.2-5 of LDLR was defined as the binding site for FVIII and RAP.
Collapse
Affiliation(s)
- James H Kurasawa
- Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland 20852, USA
| | | | | | | | | | | |
Collapse
|
25
|
Kurasawa JH, Shestopal SA, Jha NK, Ovanesov MV, Lee TK, Sarafanov AG. Insect cell-based expression and characterization of a single-chain variable antibody fragment directed against blood coagulation factor VIII. Protein Expr Purif 2013; 88:201-6. [PMID: 23306063 DOI: 10.1016/j.pep.2012.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 12/20/2012] [Accepted: 12/25/2012] [Indexed: 10/27/2022]
Abstract
A recombinant single-chain variable antibody fragment (scFv) KM33 was previously described as a ligand that can inhibit the function of blood coagulation factor VIII (FVIII). This scFv was previously derived from an individual with anti-FVIII antibodies manifested in FVIII functional deficiency (Hemophilia A) and expressed in bacteria. In the present work, we describe an alternative approach for fast and easy production of KM33 in insect cells (Spodoptera frugiperda). The KM33 gene was codon-optimized and expressed in secreted form using a baculovirus system. The protein was isolated using metal-affinity and size-exclusion chromatography to purity of about 96% and yield of 0.4-1.2 mg per 120 mL of culture, based on several independent expression experiments. In a binding assay using surface plasmon resonance, the insect cell-derived KM33 (iKM33) was qualified as a high-affinity ligand for FVIII. Epitope specificity of iKM33 on FVIII (C1 domain) was confirmed by testing the binding with a relevant mutant of FVIII. In several FVIII functional tests (factor Xa generation, APTT clotting, thrombin generation and video microscopy clot growth assays), iKM33 strongly inhibited FVIII activity in accordance with the clinical effect of the parental antibody. Therefore, the expressed protein was concluded to be fully functional and applicable in various assays with FVIII.
Collapse
Affiliation(s)
- James H Kurasawa
- Division of Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Bethesda, MD 20852, USA
| | | | | | | | | | | |
Collapse
|
26
|
Wroblewska A, Reipert BM, Pratt KP, Voorberg J. Dangerous liaisons: how the immune system deals with factor VIII. J Thromb Haemost 2013; 11:47-55. [PMID: 23140211 DOI: 10.1111/jth.12065] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Only a fraction of patients with hemophilia A develop a neutralizing antibody (inhibitor) response to therapeutic infusions of factor VIII. Our present understanding of the underlying causes of the immunogenicity of this protein is limited. In the past few years, insights into the uptake and processing of FVIII by antigen-presenting cells (APCs) have expanded significantly. Although the mechanism of endocytosis remains unclear, current data indicate that FVIII enters APCs via its C1 domain. Its subsequent processing within endolysosomes allows for presentation of a heterogeneous collection of FVIII-derived peptides on major histocompatibility complex (MHC) class II, and this peptide-MHC class II complex may then be recognized by cognate effector CD4(+) T cells, leading to anti-FVIII antibody production. Here we aim to summarize recent knowledge gained about FVIII processing and presentation by APCs, as well as the diversity of the FVIII-specific T-cell repertoire in mice and humans. Moreover, we discuss possible factors that can drive FVIII immunogenicity. We believe that increasing understanding of the immune recognition of FVIII and the cellular mechanisms of anti-FVIII antibody production will lead to novel therapeutic approaches to prevent inhibitor formation in patients with hemophilia A.
Collapse
Affiliation(s)
- A Wroblewska
- Department of Plasma Proteins, Sanquin-AMC Landsteiner Laboratory and van Creveld Laboratory, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
27
|
Castro-Núñez L, Bloem E, Boon-Spijker MG, van der Zwaan C, van den Biggelaar M, Mertens K, Meijer AB. Distinct roles of Ser-764 and Lys-773 at the N terminus of von Willebrand factor in complex assembly with coagulation factor VIII. J Biol Chem 2012; 288:393-400. [PMID: 23168412 DOI: 10.1074/jbc.m112.400572] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Complex formation between coagulation factor VIII (FVIII) and von Willebrand factor (VWF) is of critical importance to protect FVIII from rapid in vivo clearance and degradation. We have now employed a chemical footprinting approach to identify regions on VWF involved in FVIII binding. To this end, lysine amino acid residues of VWF were chemically modified in the presence of FVIII or activated FVIII, which does not bind VWF. Nano-LC-MS analysis showed that the lysine residues of almost all identified VWF peptides were not differentially modified upon incubation of VWF with FVIII or activated FVIII. However, Lys-773 of peptide Ser-766-Leu-774 was protected from chemical modification in the presence of FVIII. In addition, peptide Ser-764-Arg-782, which comprises the first 19 amino acid residues of mature VWF, showed a differential modification of both Lys-773 and the α-amino group of Ser-764. To verify the role of Lys-773 and the N-terminal Ser-764 in FVIII binding, we employed VWF variants in which either Lys-773 or Ser-764 was replaced with Ala. Surface plasmon resonance analysis and competition studies revealed that VWF(K773A) exhibited reduced binding to FVIII and the FVIII light chain, which harbors the VWF-binding site. In contrast, VWF(S764A) revealed more effective binding to FVIII and the FVIII light chain compared with WT VWF. The results of our study show that the N terminus of VWF is critical for the interaction with FVIII and that Ser-764 and Lys-773 have opposite roles in the binding mechanism.
Collapse
Affiliation(s)
- Lydia Castro-Núñez
- Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
28
|
Castro-Núñez L, Dienava-Verdoold I, Herczenik E, Mertens K, Meijer AB. Shear stress is required for the endocytic uptake of the factor VIII-von Willebrand factor complex by macrophages. J Thromb Haemost 2012; 10:1929-37. [PMID: 22812646 DOI: 10.1111/j.1538-7836.2012.04860.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Low-density lipoprotein (LDL) receptor family members contribute to the cellular uptake of factor VIII. How von Willebrand factor fits into this endocytic pathway has remained poorly understood. OBJECTIVES It has been suggested that macrophages contribute to the clearance of the factor VIII (FVIII)-von Willebrand factor (VWF) complex. We now assessed the mechanisms of uptake employing human monocyte-derived macrophages. METHODS A confocal microscopy study was employed to study the uptake by monocyte-derived macrophages of a functional green fluorescent FVIII-GFP derivative in the presence and absence of VWF. RESULTS The results revealed that FVIII-GFP is internalized by macrophages. We found that FVIII-GFP co-localizes with LDL receptor-related protein (LRP), and that the LRP antagonist Receptor Associated Protein (RAP) blocks the uptake of FVIII-GFP. However, FVIII-GFP was not detected in the macrophages in the presence of VWF, suggesting that the FVIII-VWF complex is not internalized by these cells at all. Apart from static conditions, we also investigated the effect of shear stress on the uptake of FVIII-GFP in presence of VWF. Immunofluorescence studies demonstrated that VWF does not block endocytosis of FVIII-GFP under flow conditions. Moreover, VWF itself was also internalized by the macrophages. Strikingly, in the presence of RAP, endocytosis of FVIII-GFP and VWF was inhibited. CONCLUSION The results show that shear stress is required for macrophages to internalize both constituents of the FVIII-VWF complex.
Collapse
Affiliation(s)
- L Castro-Núñez
- Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
29
|
A major determinant of the immunogenicity of factor VIII in a murine model is independent of its procoagulant function. Blood 2012; 120:2512-20. [PMID: 22855607 DOI: 10.1182/blood-2012-02-412361] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A main complication of treatment of patients with hemophilia A is the development of anti-factor VIII (fVIII) antibodies. The immunogenicity of fVIII potentially is a function of its procoagulant activity, which may result in danger signals that drive the immune response. Alternatively, intrinsic structural elements in fVIII may be particularly immunogenic. Finally, VWF, the carrier protein for fVIII in plasma, may play a role in immune recognition. We compared the immunogenicity of wild-type (wt) B domain-deleted fVIII and 2 inactive fVIII molecules, R372A/R1689A fVIII and V634M fVIII in fVIII(-/-) and fVIII(-/-)/VWF(-/-) mice. R372A/R1689A fVIII lacks proteolytic recognition sites and is not released from VWF. In contrast, V634M fVIII undergoes proteolytic cleavage and dissociation from VWF. No significant difference was observed in the immunogenicity of wt fVIII and V634M fVIII. R372A/R1689A fVIII was slightly less immunogenic in a subset of immunization regimens tested. High doses of wt fVIII were required to produce an immune response in fVIII(-/-)/VWF(-/-) mice. Our results indicate that a main component of the immune response to fVIII is independent of its procoagulant function, is both positively and negatively affected by its association with VWF, and may involve intrinsic elements of fVIII structure.
Collapse
|
30
|
Modification of an exposed loop in the C1 domain reduces immune responses to factor VIII in hemophilia A mice. Blood 2012; 119:5294-300. [PMID: 22498747 DOI: 10.1182/blood-2011-11-391680] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Development of neutralizing Abs to blood coagulation factor VIII (FVIII) provides a major complication in hemophilia care. In this study we explored whether modulation of the uptake of FVIII by APCs can reduce its intrinsic immunogenicity. Endocytosis of FVIII by professional APCs is significantly blocked by mAb KM33, directed toward the C1 domain of FVIII. We created a C1 domain variant (FVIII-R2090A/K2092A/F2093A), which showed only minimal binding to KM33 and retained its activity as measured by chromogenic assay. FVIII-R2090A/K2092A/F2093A displayed a strongly reduced internalization by human monocyte-derived dendritic cells and macrophages, as well as murine BM-derived dendritic cells. We subsequently investigated the ability of this variant to induce an immune response in FVIII-deficient mice. We show that mice treated with FVIII-R2090A/K2092A/F2093A have significantly lower anti-FVIII Ab titers and FVIII-specific CD4(+) T-cell responses compared with mice treated with wild-type FVIII. These data show that alanine substitutions at positions 2090, 2092, and 2093 reduce the immunogenicity of FVIII. According to our findings we hypothesize that FVIII variants displaying a reduced uptake by APCs provide a novel therapeutic approach to reduce inhibitor development in hemophilia A.
Collapse
|
31
|
Bloem E, Meems H, van den Biggelaar M, van der Zwaan C, Mertens K, Meijer AB. Mass spectrometry-assisted study reveals that lysine residues 1967 and 1968 have opposite contribution to stability of activated factor VIII. J Biol Chem 2012; 287:5775-83. [PMID: 22215677 PMCID: PMC3285348 DOI: 10.1074/jbc.m111.308627] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 12/13/2011] [Indexed: 11/06/2022] Open
Abstract
The A2 domain rapidly dissociates from activated factor VIII (FVIIIa) resulting in a dampening of the activity of the activated factor X-generating complex. The amino acid residues that affect A2 domain dissociation are therefore critical for FVIII cofactor function. We have now employed chemical footprinting in conjunction with mass spectrometry to identify lysine residues that contribute to the stability of activated FVIII. We hypothesized that lysine residues, which are buried in FVIII and surface-exposed in dissociated activated FVIII (dis-FVIIIa), may contribute to interdomain interactions. Mass spectrometry analysis revealed that residues Lys(1967) and Lys(1968) of region Thr(1964)-Tyr(1971) are buried in FVIII and exposed to the surface in dis-FVIIIa. This result, combined with the observation that the FVIII variant K1967I is associated with hemophilia A, suggests that these residues contribute to the stability of activated FVIII. Kinetic analysis revealed that the FVIII variants K1967A and K1967I exhibit an almost normal cofactor activity. However, these variants also showed an increased loss in cofactor activity over time compared with that of FVIII WT. Remarkably, the cofactor activity of a K1968A variant was enhanced and sustained for a prolonged time relative to that of FVIII WT. Surface plasmon resonance analysis demonstrated that A2 domain dissociation from activated FVIII was reduced for K1968A and enhanced for K1967A. In conclusion, mass spectrometry analysis combined with site-directed mutagenesis studies revealed that the lysine couple Lys(1967)-Lys(1968) within region Thr(1964)-Tyr(1971) has an opposite contribution to the stability of FVIIIa.
Collapse
Affiliation(s)
- Esther Bloem
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
| | - Henriet Meems
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
| | | | - Carmen van der Zwaan
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
| | - Koen Mertens
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
- Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Alexander B. Meijer
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
- Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
32
|
Zappelli C, van der Zwaan C, Thijssen-Timmer DC, Mertens K, Meijer AB. Novel role for galectin-8 protein as mediator of coagulation factor V endocytosis by megakaryocytes. J Biol Chem 2012; 287:8327-35. [PMID: 22267735 DOI: 10.1074/jbc.m111.305151] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Galectin-8 (Gal8) interacts with β-galactoside-containing glycoproteins and has recently been implicated to play a role in platelet activation. It has been suggested that Gal8 may also interact with platelet coagulation factor V (FV). This indispensable cofactor is stored in α-granules of platelets via a poorly understood endocytic mechanism that only exists in megakaryocytes (platelet precursor cells). In this study, we now assessed the putative role of Gal8 for FV biology. Surface plasmon resonance analysis and a solid phase binding assay revealed that Gal8 binds FV. The data further show that β-galactosides block the interaction between FV and Gal8. These findings indicate that Gal8 specifically interacts with FV in a carbohydrate-dependent manner. Confocal microscopy studies and flow cytometry analysis demonstrated that megakaryocytic DAMI cells internalize FV. Flow cytometry showed that these cells express Gal8 on their cell surface. Reducing the functional presence of Gal8 on the cells either by an anti-Gal8 antibody or by siRNA technology markedly impaired the endocytic uptake of FV. Compatible with the apparent role of Gal8 for FV uptake, endocytosis of FV was also affected in the presence of β-galactosides. Strikingly, thrombopoietin-differentiated DAMI cells, which represent a more mature megakaryocytic state, not only lose the capacity to express cell-surface bound Gal8 but also lose the ability to internalize FV. Collectively, our data reveal a novel role for the tandem repeat Gal8 in promoting FV endocytosis.
Collapse
Affiliation(s)
- Claudia Zappelli
- Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
33
|
Factor VIII alters tubular organization and functional properties of von Willebrand factor stored in Weibel-Palade bodies. Blood 2011; 118:5947-56. [DOI: 10.1182/blood-2011-05-355354] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abstract
In endothelial cells, von Willebrand factor (VWF) multimers are packaged into tubules that direct biogenesis of elongated Weibel-Palade bodies (WPBs). WPB release results in unfurling of VWF tubules and assembly into strings that serve to recruit platelets. By confocal microscopy, we have previously observed a rounded morphology of WPBs in blood outgrowth endothelial cells transduced to express factor VIII (FVIII). Using correlative light-electron microscopy and tomography, we now demonstrate that FVIII-containing WPBs have disorganized, short VWF tubules. Whereas normal FVIII and FVIII Y1680F interfered with formation of ultra-large VWF multimers, release of the WPBs resulted in VWF strings of equal length as those from nontransduced blood outgrowth endothelial cells. After release, both WPB-derived FVIII and FVIII Y1680F remained bound to VWF strings, which however had largely lost their ability to recruit platelets. Strings from nontransduced cells, however, were capable of simultaneously recruiting exogenous FVIII and platelets. These findings suggest that the interaction of FVIII with VWF during WPB formation is independent of Y1680, is maintained after WPB release in FVIII-covered VWF strings, and impairs recruitment of platelets. Apparently, intra-cellular and extracellular assembly of FVIII-VWF complex involves distinct mechanisms, which differ with regard to their implications for platelet binding to released VWF strings.
Collapse
|