92 results on '"Meijer, AB"'
Search Results
2. Monitoring storage induced changes in the platelet proteome employing label free quantitative mass spectrometry
- Author
-
Rijkers, M, van den Eshof, BL, van der Meer, PF, van Alphen, FPJ, de Korte, D, Leebeek, Frank, Meijer, AB, Voorberg, J, Jansen, AJG, Rijkers, M, van den Eshof, BL, van der Meer, PF, van Alphen, FPJ, de Korte, D, Leebeek, Frank, Meijer, AB, Voorberg, J, and Jansen, AJG
- Published
- 2017
3. Integrative phosphoproteomic analyses reveal hemostatic-endothelial signaling interplay.
- Author
-
Groten SA, van den Eshof BL, van Alphen FPJ, Meijer AB, van den Biggelaar M, and Hoogendijk AJ
- Abstract
Background: The vascular endothelial cell (EC) monolayer plays a crucial part in maintaining hemostasis. An extensive array of G protein-coupled receptors allows ECs to dynamically act on key hemostatic stimuli such as thrombin and histamine. The impact of these individual stimuli on EC signal transduction has been the subject of various studies, but insight into discordant and concordant EC signaling between different G protein-coupled receptors remains limited., Objectives: To elucidate histamine and protease-activated receptor (PAR1-4) signaling cascades in ECs, discern overlapping and diverging regulation between these stimuli and their effect on the EC monolayer., Methods: We employed stable isotope labeling by amino acids in cell culture mass spectrometry-based phosphoproteomics on in vitro cultured blood outgrowth ECs stimulated with histamine and different PAR1 to 4 peptides. We investigated key phosphosites through immuno(fluorescence) staining and determined effects on barrier function through transendothelial resistance assays., Results: EC histamine activation initiated an extensive (kinase) signaling network (including MAPK3, STAT3, and CTNND1). PAR1 and PAR2 receptors induced highly similar signaling cascades, whereas PAR3 and PAR4 induced minimal phospho-regulation. Integration of all applied stimuli indicated uniquely activated proteins between both stimuli, as well as a general overlapping activation of cell junction and actin cytoskeletal proteins., Conclusion: We provide an integrative phosphoproteomic analysis of histamine and PAR agonists in the endothelium that highlights the endothelial response programs that are at the basis of regulating hemostasis., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Plasma Profiling of Acute Myeloid Leukemia With Fever- and Infection-Related Complications During Chemotherapy-Induced Neutropenia.
- Author
-
Kreft IC, van de Geer A, Smit ER, van der Zwaan C, van Alphen FPJ, Meijer AB, Nur E, Hoogendijk AJ, Kuijpers TW, and van den Biggelaar M
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Neutropenia chemically induced, Neutropenia blood, Neutropenia diagnosis, Chemotherapy-Induced Febrile Neutropenia blood, Chemotherapy-Induced Febrile Neutropenia diagnosis, Chemotherapy-Induced Febrile Neutropenia etiology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Longitudinal Studies, Infections blood, Infections diagnosis, C-Reactive Protein analysis, Young Adult, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute blood, Fever chemically induced, Fever blood, Fever diagnosis
- Abstract
Background: Acute myeloid leukemia (AML) is a heterogenous and complex blood cancer requiring aggressive treatment. Early identification and prediction of the complications following treatment is vital for effective disease management., Aims: We explored associations between plasma protein levels and fever- and infection-related complications in 26 AML patients during chemotherapy-induced neutropenia., Material and Methods: Longitudinal plasma profiling was conducted using data-dependent mass spectrometry analysis., Results: Mass spectrometry-based plasma profiling data correlated well with laboratory parameters, including C-reactive protein, and revealed a broader inflammation protein network associated with fever- and infection-related complications., Discussion and Conclusion: These data indicate the potential of longitudinal plasma profiling in AML patients for identifying and predicting complications that may aid in improved disease monitoring and treatment., (© 2024 The Author(s). Cancer Reports published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
5. Variant mapping using mass spectrometry-based proteotyping as a diagnostic tool in von Willebrand disease.
- Author
-
Kreft IC, van Duijl TT, van Kwawegen C, Atiq F, Phan W, Schuller MBP, Boon-Spijker M, van der Zwaan C, Meijer AB, Hoogendijk AJ, Bierings R, Eikenboom JCJ, Leebeek FWG, and van den Biggelaar M
- Subjects
- Humans, Netherlands, Phenotype, Female, Factor VIII genetics, Factor VIII analysis, Factor VIII metabolism, Mass Spectrometry, Male, Predictive Value of Tests, von Willebrand Factor genetics, von Willebrand Factor analysis, von Willebrand Factor metabolism, von Willebrand Diseases diagnosis, von Willebrand Diseases blood, von Willebrand Diseases genetics, Proteomics methods
- Abstract
Background: von Willebrand disease (VWD) is the most common inherited bleeding disorder, characterized by either partial or complete von Willebrand factor (VWF) deficiency or by the occurrence of VWF proteoforms of altered functionality. The gene encoding VWF is highly polymorphic, giving rise to a variety of proteoforms with varying plasma concentrations and clinical significance., Objectives: To address this complexity, we translated genomic variation in VWF to corresponding VWF proteoforms circulating in blood., Methods: VWF was characterized in VWD patients (n = 64) participating in the Willebrand in the Netherlands study by conventional laboratory testing, DNA sequencing and complementary discovery, and targeted mass spectrometry-based plasma proteomic strategies., Results: Unbiased plasma profiling combined with immune enrichment of VWF verified VWF and its binding partner factor VIII as key determinants of VWD and revealed a remarkable heterogeneity in VWF amino acid sequence coverage among patients. Subsequent VWF proteotyping enabled identification of both polymorphisms (eg, p.Thr789Ala, p.Gln852Arg, and p.Thr1381Ala), as well as pathogenic variants (n = 16) along with their corresponding canonical sequences. Targeted proteomics using stable isotope-labeled peptides confirmed unbiased proteotyping for 5 selected variants and suggested differential proteoform quantities in plasma. The variant-to-wild-type peptide ratio was determined in 6 type 2B patients heterozygous for p.Arg1306Trp, confirming the relatively low proteoform concentration of the pathogenic variant. The elevated VWF propeptide/VWF ratio indicated increased clearance of specific VWF proteoforms., Conclusion: This study highlights how VWF proteotyping from plasma could be the first step to bridge the gap between genotyping and functional testing in VWD., Competing Interests: Declaration of competing interests There are no competing interests to disclose., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Platelet proteomic profiling in sitosterolemia suggests thrombocytopenia is driven by lipid disorder and not platelet aberrations.
- Author
-
Del Castillo J, Tool ATJ, van Leeuwen K, van Alphen FPJ, Brands MM, Suijker MH, Meijer AB, Hoogendijk AJ, and Kuijpers TW
- Subjects
- Humans, ATP Binding Cassette Transporter, Subfamily G, Member 8 genetics, Lipoproteins, Pedigree, Proteome, ATP Binding Cassette Transporter, Subfamily G, Member 5 genetics, Blood Platelets metabolism, Blood Platelets pathology, Hypercholesterolemia blood, Hypercholesterolemia genetics, Hypercholesterolemia complications, Intestinal Diseases blood, Intestinal Diseases diagnosis, Intestinal Diseases genetics, Intestinal Diseases etiology, Intestinal Diseases metabolism, Lipid Metabolism, Inborn Errors diagnosis, Lipid Metabolism, Inborn Errors genetics, Lipid Metabolism, Inborn Errors blood, Lipid Metabolism, Inborn Errors complications, Phytosterols adverse effects, Phytosterols blood, Proteomics methods, Thrombocytopenia diagnosis, Thrombocytopenia blood, Thrombocytopenia etiology, Thrombocytopenia metabolism
- Abstract
Abstract: Sitosterolemia is a rare autosomal recessive genetic disorder in which patients develop hypercholesterolemia and may exhibit abnormal hematologic and/or liver test results. In this disease, dysfunction of either ABCG5 or ABCG8 results in the intestinal hyperabsorption of all sterols, including cholesterol and, more specifically, plant sterols or xenosterols, as well as in the impaired ability to excrete xenosterols into the bile. It remains unknown how and why some patients develop hematologic abnormalities. Only a few unrelated patients with hematologic abnormalities at the time of diagnosis have been reported. Here, we report on 2 unrelated pedigrees who were believed to have chronic immune thrombocytopenia as their most prominent feature. Both consanguineous families showed recessive gene variants in ABCG5, which were associated with the disease by in silico protein structure analysis and clinical segregation. Hepatosplenomegaly was absent. Thrombopoietin levels and megakaryocyte numbers in the bone marrow were normal. Metabolic analysis confirmed the presence of strongly elevated plasma levels of xenosterols. Potential platelet proteomic aberrations were longitudinally assessed following dietary restrictions combined with administration of the sterol absorption inhibitor ezetimibe. No significant effects on platelet protein content before and after the onset of treatment were demonstrated. Although we cannot exclude that lipotoxicity has a direct and platelet-specific impact in patients with sitosterolemia, our data suggest that thrombocytopenia is neither caused by a lack of megakaryocytes nor driven by proteomic aberrations in the platelets themselves., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Multi-omics delineation of cytokine-induced endothelial inflammatory states.
- Author
-
Groten SA, Smit ER, Janssen EFJ, van den Eshof BL, van Alphen FPJ, van der Zwaan C, Meijer AB, Hoogendijk AJ, and van den Biggelaar M
- Subjects
- Humans, Endothelial Cells metabolism, Proteomics, Multiomics, Inflammation metabolism, Endothelium, Vascular, Cytokines metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Vascular endothelial cells (ECs) form a dynamic interface between blood and tissue and play a crucial role in the progression of vascular inflammation. Here, we aim to dissect the system-wide molecular mechanisms of inflammatory endothelial-cytokine responses. Applying an unbiased cytokine library, we determined that TNFα and IFNγ induced the largest EC response resulting in distinct proteomic inflammatory signatures. Notably, combined TNFα + IFNγ stimulation induced an additional synergetic inflammatory signature. We employed a multi-omics approach to dissect these inflammatory states, combining (phospho-) proteome, transcriptome and secretome and found, depending on the stimulus, a wide-array of altered immune-modulating processes, including complement proteins, MHC complexes and distinct secretory cytokines. Synergy resulted in cooperative activation of transcript induction. This resource describes the intricate molecular mechanisms that are at the basis of endothelial inflammation and supports the adaptive immunomodulatory role of the endothelium in host defense and vascular inflammation., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. Mass spectrometry-based analysis on the impact of whole blood donation on the global plasma proteome.
- Author
-
Kreft IC, Hoogendijk AJ, van der Zwaan C, van Alphen FPJ, Boon-Spijker M, Prinsze F, Meijer AB, de Korte D, van den Hurk K, and van den Biggelaar M
- Subjects
- Humans, Male, Female, Proteomics, Erythrocytes chemistry, Blood Donors, Ferritins, Hemoglobins analysis, Proteome, Blood Donation
- Abstract
Background: Biomonitoring may provide important insights into the impact of a whole blood donation for individual blood donors., Study Design and Methods: Here, we used unbiased mass spectrometry (MS)-based proteomics to assess longitudinal changes in the global plasma proteome, after a single blood donation for new and regular donors. Subsequently, we compared plasma proteomes of 76 male and female whole blood donors, that were grouped based on their ferritin and hemoglobin (Hb) levels., Results: The longitudinal analysis showed limited changes in the plasma proteomes of new and regular donors after a whole blood donation during a 180-day follow-up period, apart from a significant short-term decrease in fibronectin. No differences were observed in the plasma proteomes of donors with high versus low Hb and/or ferritin levels. Plasma proteins with the highest variation between and within donors included pregnancy zone protein, which was associated with sex, Alfa 1-antitrypsin which was associated with the allelic variation, and Immunoglobulin D. Coexpression analysis revealed clustering of proteins that are associated with platelet, red cell, and neutrophil signatures as well as with the complement system and immune responses, including a prominent correlating cluster of immunoglobulin M (IgM), immunoglobulin J chain (JCHAIN), and CD5 antigen-like (CD5L)., Discussion: Overall, our proteomic approach shows that whole blood donation has a limited impact on the plasma proteins measured. Our findings suggest that plasma profiling can be successfully employed to consistently detect proteins and protein complexes that reflect the functionality and integrity of platelets, red blood cells, and immune cells in blood donors and thus highlights its potential use for donor health monitoring., (© 2023 The Authors. Transfusion published by Wiley Periodicals LLC on behalf of AABB.)
- Published
- 2023
- Full Text
- View/download PDF
9. Proteomic landscapes of inherited platelet disorders with different etiologies.
- Author
-
Kreft IC, Huisman EJ, Cnossen MH, van Alphen FPJ, van der Zwaan C, van Leeuwen K, van Spaendonk R, Porcelijn L, Veen CSB, van den Biggelaar M, de Haas M, Meijer AB, and Hoogendijk AJ
- Subjects
- Humans, Proteome metabolism, Blood Platelets metabolism, Thrombopoiesis, Proteomics, Blood Platelet Disorders diagnosis, Blood Platelet Disorders genetics, Blood Platelet Disorders metabolism
- Abstract
Background: Inherited platelet disorders (IPDs) are a heterogeneous group of rare diseases that are caused by the defects in early megakaryopoiesis, proplatelet formation, and/or mature platelet function. Although genomic sequencing is increasingly used to identify genetic variants underlying IPD, this technique does not disclose resulting molecular changes that impact platelet function. Proteins are the functional units that shape platelet function; however, insights into how variants that cause IPDs impact platelet proteomes are limited., Objectives: The objective of this study was to profile the platelet proteomics signatures of IPDs., Methods: We performed unbiased label-free quantitative mass spectrometry (MS)-based proteome profiling on platelets of 34 patients with IPDs with variants in 13 ISTH TIER1 genes that affect different stages of platelet development., Results: In line with the phenotypical heterogeneity between IPDs, proteomes were diverse between IPDs. We observed extensive proteomic alterations in patients with a GFI1B variant and for genetic variants in genes encoding proteins that impact cytoskeletal processes (MYH9, TUBB1, and WAS). Using the diversity between IPDs, we clustered protein dynamics, revealing disrupted protein-protein complexes. This analysis furthermore grouped proteins with similar cellular function and location, classifying mitochondrial protein constituents and identifying both known and putative novel alpha granule associated proteins., Conclusions: With this study, we demonstrate a MS-based proteomics perspective to IPDs. By integrating the effects of IPDs that impact different aspects of platelet function, we dissected the biological contexts of protein alterations to gain further insights into the biology of platelet (dys)function., (Copyright © 2022 International Society on Thrombosis and Haemostasis. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Nanobodies against factor XI apple 3 domain inhibit binding of factor IX and reveal a novel binding site for high molecular weight kininogen.
- Author
-
Bar Barroeta A, Marquart JA, Bakhtiari K, Meijer AB, Urbanus RT, and Meijers JCM
- Subjects
- Humans, Anticoagulants, Binding Sites, Deuterium, Epitopes, Factor IX metabolism, Factor XI metabolism, Kininogen, High-Molecular-Weight metabolism, Single-Domain Antibodies
- Abstract
Background: Factor XI (FXI) is a promising target for novel anticoagulants because it shows a strong relation to thromboembolic diseases, while fulfilling a mostly supportive role in hemostasis. Anticoagulants targeting FXI could therefore reduce the risk for thrombosis, without increasing the chance of bleeding side effects., Objectives: To generate nanobodies that can interfere with FXIa mediated activation of factor IX (FIX)., Methods: Nanobodies were selected for binding to the apple 3 domain of FXI and their effects on FXI and coagulation were measured in purified protein systems as well as in plasma-based coagulation assays. Additionally, the binding epitope of selected nanobodies was assessed by hydrogen-deuterium exchange mass spectrometry., Results: We have identified five nanobodies that inhibit FIX activation by FXI by competing with the FIX binding site on FXI. Interestingly, a sixth nanobody was found to target a different binding epitope in the apple 3 domain, resulting in competition with the FXI-high molecular weight kininogen (HK) interaction., Conclusions: We have characterized a nanobody targeting the FXI apple 3 domain that elucidates the binding orientation of HK on FXI. Moreover, we have produced five nanobodies that can inhibit the FXI-FIX interaction., (© 2022 The Authors. Journal of Thrombosis and Haemostasis published by Wiley Periodicals LLC on behalf of International Society on Thrombosis and Haemostasis.)
- Published
- 2022
- Full Text
- View/download PDF
11. SYMPHONY consortium: Orchestrating personalized treatment for patients with bleeding disorders.
- Author
-
Cnossen MH, van Moort I, Reitsma SH, de Maat MPM, Schutgens REG, Urbanus RT, Lingsma HF, Mathot RAA, Gouw SC, Meijer K, Bredenoord AL, van der Graaf R, Fijnvandraat K, Meijer AB, van den Akker E, Bierings R, Eikenboom JCJ, van den Biggelaar M, de Haas M, Voorberg J, and Leebeek FWG
- Abstract
Background: Treatment choices for individual patients with an inborn bleeding disorder are increasingly challenging due to increasing options and rising costs for society. We have initiated an integrated interdisciplinary national research programme., Objectives: The SYMPHONY consortium strives to orchestrate personalized treatment in patients with an inborn bleeding disorder, by unravelling the mechanisms behind inter-individual variations of bleeding phenotype., Patients: The SYMPHONY consortium will investigate patients with an inborn bleeding disorder, both diagnosed and not yet diagnosed., Results: Research questions are categorized under the themes: 1) Diagnosis; 2) Treatment; and 3) Fundamental research and consist of workpackages addressing specific domains. Importantly, collaborations between patients and talented researchers from different areas of expertise promise to augment the impact of the SYMPHONY consortium, leading to unique interactions and intellectual property., Conclusions: SYMPHONY will perform research on all aspects of care, treatment individualization in patients with inborn bleeding disorders as well as diagnostic innovations and results of molecular genetics and cellular model technology with regard to the hemostatic process. We believe that these research investments will lead to health care innovations with long-term clinical and societal impact. This consortium has been made possible by a governmental, competitive grant from the Netherlands Organization for Scientific Research (NWO) within the framework of the NWA-ORC Call grant agreement NWA.1160.18.038., (This article is protected by copyright. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
12. Neutrophil azurophilic granule glycoproteins are distinctively decorated by atypical pauci- and phosphomannose glycans.
- Author
-
Reiding KR, Lin YH, van Alphen FPJ, Meijer AB, and Heck AJR
- Subjects
- Chromatography, Liquid, Glycosylation, Humans, Tandem Mass Spectrometry, Cytoplasmic Granules metabolism, Glycoproteins metabolism, Neutrophils metabolism, Polysaccharides metabolism, Proteome metabolism
- Abstract
While neutrophils are critical first-responders of the immune system, they also cause tissue damage and act in a variety of autoimmune diseases. Many neutrophil proteins are N-glycosylated, a post-translational modification that may affect, among others, enzymatic activity, receptor interaction, and protein backbone accessibility. So far, a handful neutrophil proteins were reported to be decorated with atypical small glycans (paucimannose and smaller) and phosphomannosylated glycans. To elucidate the occurrence of these atypical glycoforms across the neutrophil proteome, we performed LC-MS/MS-based (glyco)proteomics of pooled neutrophils from healthy donors, obtaining site-specific N-glycan characterisation of >200 glycoproteins. We found that glycoproteins that are typically membrane-bound to be mostly decorated with high-mannose/complex N-glycans, while secreted proteins mainly harboured complex N-glycans. In contrast, proteins inferred to originate from azurophilic granules carried distinct and abundant paucimannosylation, asymmetric/hybrid glycans, and glycan phosphomannosylation. As these same proteins are often autoantigenic, uncovering their atypical glycosylation characteristics is an important step towards understanding autoimmune disease and improving treatment., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
13. Specific proteome changes in platelets from individuals with GATA1-, GFI1B-, and RUNX1-linked bleeding disorders.
- Author
-
Van Bergen MGJM, Marneth AE, Hoogendijk AJ, Van Alphen FPJ, Van den Akker E, Laros-Van Gorkom BAP, Hoeks M, Simons A, De Munnik SA, Janssen JJWM, Martens JHA, Jansen JH, Meijer AB, and Van der Reijden BA
- Subjects
- Homeostasis, Humans, Mutation genetics, Signal Transduction, Transcription Factors genetics, Transcription Factors metabolism, Blood Platelet Disorders metabolism, Blood Platelets metabolism, Core Binding Factor Alpha 2 Subunit metabolism, GATA1 Transcription Factor metabolism, Proteome metabolism, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism
- Abstract
Mutations in the transcription factors GATA binding factor 1 (GATA1), growth factor independence 1B (GFI1B), and Runt-related transcription factor 1 (RUNX1) cause familial platelet and bleeding disorders. Mutant platelets exhibit common abnormalities including an α-granule reduction resulting in a grayish appearance in blood smears. This suggests that similar pathways are deregulated by different transcription factor mutations. To identify common factors, full platelet proteomes from 11 individuals with mutant GATA1R216Q, GFI1BQ287*, RUNX1Q154Rfs, or RUNX1TD2-6 and 28 healthy controls were examined by label-free quantitative mass spectrometry. In total, 2875 platelet proteins were reliably quantified. Clustering analysis of more than 300 differentially expressed proteins revealed profound differences between cases and controls. Among cases, 44 of 143 significantly downregulated proteins were assigned to platelet function, hemostasis, and granule biology, in line with platelet dysfunction and bleedings. Remarkably, none of these proteins were significantly diminished in all affected cases. Similarly, no proteins were commonly overrepresented in all affected cases compared with controls. These data indicate that the studied transcription factor mutations alter platelet proteomes in distinct largely nonoverlapping manners. This work provides the quantitative landscape of proteins that affect platelet function when deregulated by mutated transcription factors in inherited bleeding disorders., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
14. Probing activation-driven changes in coagulation factor IX by mass spectrometry.
- Author
-
Freato N, van Alphen FPJ, Boon-Spijker M, van den Biggelaar M, Meijer AB, Mertens K, and Ebberink EHTM
- Subjects
- Factor VIIIa, Humans, Kinetics, Mass Spectrometry, Factor IX genetics, Factor IX metabolism, Factor IXa metabolism
- 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., (© 2021 The Authors. Journal of Thrombosis and Haemostasis published by Wiley Periodicals LLC on behalf of International Society on Thrombosis and Haemostasis.)
- Published
- 2021
- Full Text
- View/download PDF
15. Hydrogen-Deuterium Exchange Mass Spectrometry Identifies Activated Factor IX-Induced molecular Changes in Activated Factor VIII.
- Author
-
van Galen J, Freato N, Przeradzka MA, Ebberink EHTM, Boon-Spijker M, van der Zwaan C, van den Biggelaar M, and Meijer AB
- Subjects
- Factor IXa genetics, Humans, Leucine, Mass Spectrometry, Molecular Conformation, Mutagenesis, Site-Directed, Protein Binding, Surface Plasmon Resonance, Tyrosine, Blood Coagulation genetics, Deuterium chemistry, Deuterium Exchange Measurement methods, Factor IXa chemistry, Factor VIIIa chemistry, Hemophilia A genetics
- 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., Competing Interests: None declared., (Thieme. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
16. Neutrophil specific granule and NETosis defects in gray platelet syndrome.
- Author
-
Aarts CEM, Downes K, Hoogendijk AJ, Sprenkeler EGG, Gazendam RP, Favier R, Favier M, Tool ATJ, van Hamme JL, Kostadima MA, Waller K, Zieger B, van Bergen MGJM, Langemeijer SMC, van der Reijden BA, Janssen H, van den Berg TK, van Bruggen R, Meijer AB, Ouwehand WH, and Kuijpers TW
- Subjects
- Animals, Blood Platelets, Blood Proteins, Cytoplasmic Granules, Humans, Mice, Neutrophils, Gray Platelet Syndrome genetics
- Abstract
Gray platelet syndrome (GPS) is an autosomal recessive bleeding disorder characterized by a lack of α-granules in platelets and progressive myelofibrosis. Rare loss-of-function variants in neurobeachin-like 2 (NBEAL2), a member of the family of beige and Chédiak-Higashi (BEACH) genes, are causal of GPS. It is suggested that BEACH domain containing proteins are involved in fusion, fission, and trafficking of vesicles and granules. Studies in knockout mice suggest that NBEAL2 may control the formation and retention of granules in neutrophils. We found that neutrophils obtained from the peripheral blood from 13 patients with GPS have a normal distribution of azurophilic granules but show a deficiency of specific granules (SGs), as confirmed by immunoelectron microscopy and mass spectrometry proteomics analyses. CD34+ hematopoietic stem cells (HSCs) from patients with GPS differentiated into mature neutrophils also lacked NBEAL2 expression but showed similar SG protein expression as control cells. This is indicative of normal granulopoiesis in GPS and identifies NBEAL2 as a potentially important regulator of granule release. Patient neutrophil functions, including production of reactive oxygen species, chemotaxis, and killing of bacteria and fungi, were intact. NETosis was absent in circulating GPS neutrophils. Lack of NETosis is suggested to be independent of NBEAL2 expression but associated with SG defects instead, as indicated by comparison with HSC-derived neutrophils. Since patients with GPS do not excessively suffer from infections, the consequence of the reduced SG content and lack of NETosis for innate immunity remains to be explored., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
17. Factor VIII-driven changes in activated factor IX explored by hydrogen-deuterium exchange mass spectrometry.
- Author
-
Freato N, Ebberink EHTM, van Galen J, Fribourg C, Boon-Spijker M, van Alphen FPJ, Meijer AB, van den Biggelaar M, and Mertens K
- Subjects
- Allosteric Regulation genetics, Factor IXa genetics, Factor IXa metabolism, Factor VIII genetics, Factor VIII metabolism, Hemophilia B genetics, Hemophilia B metabolism, Humans, Protein Conformation, alpha-Helical, Protein Domains, Deuterium Exchange Measurement, Factor IXa chemistry, Factor VIII chemistry, Mass Spectrometry, Mutation
- 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., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
18. Hemolysis in the spleen drives erythrocyte turnover.
- Author
-
Klei TRL, Dalimot J, Nota B, Veldthuis M, Mul FPJ, Rademakers T, Hoogenboezem M, Nagelkerke SQ, van IJcken WFJ, Oole E, Svendsen P, Moestrup SK, van Alphen FPJ, Meijer AB, Kuijpers TW, van Zwieten R, and van Bruggen R
- Subjects
- Animals, Biomarkers, Erythrocyte Aging drug effects, Erythrocyte Deformability, Erythrocyte Membrane, Erythrocyte Transfusion, Erythrocytes drug effects, Female, Gene Expression Profiling, Histocytochemistry, Humans, Immunophenotyping, Laminin pharmacology, Macrophages metabolism, Mice, Phagocytosis, Erythrocytes metabolism, Hemolysis, Spleen metabolism, Spleen physiopathology
- Abstract
Red pulp macrophages (RPMs) of the spleen mediate turnover of billions of senescent erythrocytes per day. However, the molecular mechanisms involved in sequestration of senescent erythrocytes, their recognition, and their subsequent degradation by RPMs remain unclear. In this study, we provide evidence that the splenic environment is of substantial importance in facilitating erythrocyte turnover through induction of hemolysis. Upon isolating human spleen RPMs, we noted a substantial lack of macrophages that were in the process of phagocytosing intact erythrocytes. Detailed characterization of erythrocyte and macrophage subpopulations from human spleen tissue led to the identification of erythrocytes that are devoid of hemoglobin, so-called erythrocyte ghosts. By using in vivo imaging and transfusion experiments, we further confirmed that senescent erythrocytes that are retained in the spleen are subject to hemolysis. In addition, we showed that erythrocyte adhesion molecules, which are specifically activated on aged erythrocytes, cause senescent erythrocytes to interact with extracellular matrix proteins that are exposed within the splenic architecture. Such adhesion molecule-driven retention of senescent erythrocytes under low shear conditions was found to result in steady shrinkage of the cell and ultimately resulted in hemolysis. In contrast to intact senescent erythrocytes, the remnant erythrocyte ghost shells were prone to recognition and breakdown by RPMs. These data identify hemolysis as a key event in the turnover of senescent erythrocytes, which alters our current understanding of how erythrocyte degradation is regulated., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
19. Flow-induced Reorganization of Laminin-integrin Networks Within the Endothelial Basement Membrane Uncovered by Proteomics.
- Author
-
Béguin EP, Janssen EFJ, Hoogenboezem M, Meijer AB, Hoogendijk AJ, and van den Biggelaar M
- Subjects
- Cells, Cultured, Chromatography, Liquid, Endothelial Cells cytology, Endothelial Cells drug effects, Extracellular Matrix metabolism, Fluorescent Antibody Technique, Gene Expression Profiling, Gene Ontology, Hemodynamics, Humans, Integrin alpha Chains metabolism, Integrin alpha6 metabolism, Integrin beta Chains metabolism, Integrin beta4 metabolism, Protein Interaction Maps physiology, Proteomics, Tandem Mass Spectrometry, Tumor Necrosis Factor-alpha pharmacology, Basement Membrane metabolism, Blood Circulation physiology, Endothelial Cells metabolism, Integrins metabolism, Laminin metabolism
- Abstract
The vessel wall is continuously exposed to hemodynamic forces generated by blood flow. Endothelial mechanosensors perceive and translate mechanical signals via cellular signaling pathways into biological processes that control endothelial development, phenotype and function. To assess the hemodynamic effects on the endothelium on a system-wide level, we applied a quantitative mass spectrometry approach combined with cell surface chemical footprinting. SILAC-labeled endothelial cells were subjected to flow-induced shear stress for 0, 24 or 48 h, followed by chemical labeling of surface proteins using a non-membrane permeable biotin label, and analysis of the whole proteome and the cell surface proteome by LC-MS/MS analysis. These studies revealed that of the >5000 quantified proteins 104 were altered, which were highly enriched for extracellular matrix proteins and proteins involved in cell-matrix adhesion. Cell surface proteomics indicated that LAMA4 was proteolytically processed upon flow-exposure, which corresponded to the decreased LAMA4 mass observed on immunoblot. Immunofluorescence microscopy studies highlighted that the endothelial basement membrane was drastically remodeled upon flow exposure. We observed a network-like pattern of LAMA4 and LAMA5, which corresponded to the localization of laminin-adhesion molecules ITGA6 and ITGB4. Furthermore, the adaptation to flow-exposure did not affect the inflammatory response to tumor necrosis factor α, indicating that inflammation and flow trigger fundamentally distinct endothelial signaling pathways with limited reciprocity and synergy. Taken together, this study uncovers the blood flow-induced remodeling of the basement membrane and stresses the importance of the subendothelial basement membrane in vascular homeostasis., Competing Interests: Conflict of interest—Authors declare no competing interests., (© 2020 Béguin et al.)
- Published
- 2020
- Full Text
- View/download PDF
20. D' domain region Arg782-Cys799 of von Willebrand factor contributes to factor VIII binding.
- Author
-
Przeradzka MA, van Galen J, Ebberink EHTM, Hoogendijk AJ, van der Zwaan C, Mertens K, van den Biggelaar M, and Meijer AB
- Subjects
- Binding Sites, Hemorrhage, Humans, Protein Domains, Factor VIII genetics, von Willebrand Factor genetics
- 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., (Copyright© 2020 Ferrata Storti Foundation.)
- Published
- 2020
- Full Text
- View/download PDF
21. Endocytosis by macrophages: interplay of macrophage scavenger receptor-1 and LDL receptor-related protein-1.
- Author
-
Béguin EP, Przeradzka MA, Janssen EFJ, Meems H, Sedek M, van der Zwaan C, Mertens K, van den Biggelaar M, Meijer AB, and Mourik MJ
- Subjects
- Humans, Lipoproteins, LDL, Endocytosis, Low Density Lipoprotein Receptor-Related Protein-1 genetics, Macrophages cytology, Scavenger Receptors, Class A genetics
- Published
- 2020
- Full Text
- View/download PDF
22. Unique surface-exposed hydrophobic residues in the C1 domain of factor VIII contribute to cofactor function and von Willebrand factor binding.
- Author
-
Przeradzka MA, Freato N, Boon-Spijker M, van Galen J, van der Zwaan C, Mertens K, van den Biggelaar M, and Meijer AB
- Subjects
- Factor IXa, Factor VIII, Humans, Protein Domains, Hemostatics, von Willebrand Factor
- 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., (© 2019 International Society on Thrombosis and Haemostasis.)
- Published
- 2020
- Full Text
- View/download PDF
23. Missing regions within the molecular architecture of human fibrin clots structurally resolved by XL-MS and integrative structural modeling.
- Author
-
Klykov O, van der Zwaan C, Heck AJR, Meijer AB, and Scheltema RA
- Subjects
- Albumins chemistry, Fibrin genetics, Humans, Mutation, Protein Conformation, Albumins metabolism, Cross-Linking Reagents metabolism, Fibrin chemistry, Fibrin metabolism, Mass Spectrometry methods, Models, Structural, Thrombosis physiopathology
- Abstract
Upon activation, fibrinogen forms large fibrin biopolymers that coalesce into clots which assist in wound healing. Limited insights into their molecular architecture, due to the sheer size and the insoluble character of fibrin clots, have restricted our ability to develop novel treatments for clotting diseases. The, so far resolved, disparate structural details have provided insights into linear elongation; however, molecular details like the C-terminal domain of the α-chain, the heparin-binding domain on the β-chain, and other functional domains remain elusive. To illuminate these dark areas, we applied cross-linking mass spectrometry (XL-MS) to obtain biochemical evidence in the form of over 300 distance constraints and combined this with structural modeling. These restraints additionally define the interaction network of the clots and provide molecular details for the interaction with human serum albumin (HSA). We were able to construct the structural models of the fibrinogen α-chain (excluding two highly flexible regions) and the N termini of the β-chain, confirm these models with known structural arrangements, and map how the structure laterally aggregates to form intricate lattices together with the γ-chain. We validate the final model by mapping mutations leading to impaired clot formation. From a list of 22 mutations, we uncovered structural features for all, including a crucial role for βArg'169 (UniProt: 196) in lateral aggregation. The resulting model can potentially serve for research on dysfibrinogenemia and amyloidosis as it provides insights into the molecular mechanisms of thrombosis and bleeding disorders related to fibrinogen variants. The structure is provided in the PDB-DEV repository (PDBDEV_00000030)., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
24. Hydrogen-deuterium exchange mass spectrometry highlights conformational changes induced by factor XI activation and binding of factor IX to factor XIa.
- Author
-
Bar Barroeta A, van Galen J, Stroo I, Marquart JA, Meijer AB, and Meijers JCM
- Subjects
- Enzyme Activation, Factor IX chemistry, Factor XI chemistry, Factor XI genetics, Factor XIa chemistry, Factor XIa genetics, HEK293 Cells, Humans, Models, Molecular, Protein Binding, Protein Interaction Domains and Motifs, Structure-Activity Relationship, Factor IX metabolism, Factor XI metabolism, Factor XIa metabolism, Hemostasis, Hydrogen Deuterium Exchange-Mass Spectrometry
- Abstract
Background: Factor XI (FXI) is a zymogen in the coagulation pathway that, once activated, promotes haemostasis by activating factor IX (FIX). Substitution studies using apple domains of the homologous protein prekallikrein have identified that FIX binds to the apple 3 domain of FXI. However, the molecular changes upon activation of FXI or binding of FIX to FXIa have remained largely unresolved., Objectives: This study aimed to gain more insight in the FXI activation mechanism by identifying the molecular differences between FXI and FXIa, and in the conformational changes in FXIa induced by binding of FIX., Methods: Hydrogen-deuterium exchange mass spectrometry was performed on FXI, FXIa, and FXIa in complex with FIX., Results: Both activation and binding to FIX induced conformational changes at the interface between the catalytic domain and the apple domains of FXI(a)-more specifically at the loops connecting the apple domains. Moreover, introduction of FIX uniquely induced a reduction of deuterium uptake in the beginning of the apple 3 domain., Conclusions: We propose that the conformational changes of the catalytic domain upon activation increase the accessibility to the apple 3 domain to enable FIX binding. Moreover, our HDX MS results support the location of the proposed FIX binding site at the beginning of the apple 3 domain and suggest a mediating role in FIX binding for both loops adjacent to the apple 3 domain., (© 2019 The Authors. Journal of Thrombosis and Haemostasis published by Wiley Periodicals, Inc. on behalf of International Society on Thrombosis and Haemostasis.)
- Published
- 2019
- Full Text
- View/download PDF
25. Activated neutrophils exert myeloid-derived suppressor cell activity damaging T cells beyond repair.
- Author
-
Aarts CEM, Hiemstra IH, Béguin EP, Hoogendijk AJ, Bouchmal S, van Houdt M, Tool ATJ, Mul E, Jansen MH, Janssen H, van Alphen FPJ, de Boer JP, Zuur CL, Meijer AB, van den Berg TK, and Kuijpers TW
- Subjects
- Apoptosis, Biomarkers, Cell Communication immunology, Cell Degranulation immunology, Cytokines metabolism, Humans, Immunomodulation, Immunophenotyping, Lymphocyte Activation immunology, Reactive Oxygen Species metabolism, Signal Transduction, Myeloid-Derived Suppressor Cells immunology, Myeloid-Derived Suppressor Cells metabolism, Neutrophil Activation immunology, Neutrophils immunology, Neutrophils metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Myeloid-derived suppressor cells (MDSCs) have the capacity to suppress T-cell-mediated immune responses and impact the clinical outcome of cancer, infections, and transplantation settings. Although MDSCs were initially described as bone marrow-derived immature myeloid cells (either monocytic or granulocytic MDSCs), mature neutrophils have been shown to exert MDSC activity toward T cells in ways that remain unclear. In this study, we demonstrated that human neutrophils from both healthy donors and cancer patients do not exert MDSC activity unless they are activated. By using neutrophils with genetically well-defined defects, we found that reactive oxygen species (ROS) and granule-derived constituents are required for MDSC activity after direct CD11b-dependent interactions between neutrophils and T cells. In addition to these cellular interactions, neutrophils are engaged in the uptake of pieces of T-cell membrane, a process called trogocytosis. Together, these interactions led to changes in T-cell morphology, mitochondrial dysfunction, and adenosine triphosphate depletion, as indicated by electron microscopy, mass spectrometry, and metabolic parameters. Our studies characterize the different steps by which activated mature neutrophils induce functional T-cell nonresponsiveness and irreparable cell damage., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
26. Dynamic Transcriptome-Proteome Correlation Networks Reveal Human Myeloid Differentiation and Neutrophil-Specific Programming.
- Author
-
Hoogendijk AJ, Pourfarzad F, Aarts CEM, Tool ATJ, Hiemstra IH, Grassi L, Frontini M, Meijer AB, van den Biggelaar M, and Kuijpers TW
- Subjects
- Granulocytes cytology, Granulocytes metabolism, Hematopoiesis genetics, Hematopoiesis physiology, Humans, Proteomics methods, Neutrophils cytology, Neutrophils metabolism, Proteome metabolism, Transcriptome genetics
- Abstract
Human neutrophilic granulocytes form the largest pool of innate immune cells for host defense against bacterial and fungal pathogens. The dynamic changes that accompany the metamorphosis from a proliferating myeloid progenitor cell in the bone marrow into a mature non-dividing polymorphonuclear blood cell have remained poorly defined. Using mass spectrometry-based quantitative proteomics combined with transcriptomic data, we report on the dynamic changes of five developmental stages in the bone marrow and blood. Integration of transcriptomes and proteome unveils highly dynamic and differential interactions between RNA and protein kinetics during human neutrophil development, which can be linked to functional maturation of typical end-stage blood neutrophil killing activities., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
27. Molecular mechanisms of bleeding disorderassociated GFI1B Q287* mutation and its affected pathways in megakaryocytes and platelets.
- Author
-
van Oorschot R, Hansen M, Koornneef JM, Marneth AE, Bergevoet SM, van Bergen MGJM, van Alphen FPJ, van der Zwaan C, Martens JHA, Vermeulen M, Jansen PWTC, Baltissen MPA, Gorkom BAPL, Janssen H, Jansen JH, von Lindern M, Meijer AB, van den Akker E, and van der Reijden BA
- Subjects
- Blood Coagulation Disorders genetics, Blood Coagulation Disorders metabolism, Blood Platelets metabolism, Cell Differentiation, Co-Repressor Proteins genetics, Co-Repressor Proteins metabolism, Histone Deacetylase 1 genetics, Histone Deacetylase 1 metabolism, Histone Deacetylase 2 genetics, Histone Deacetylase 2 metabolism, Histone Demethylases genetics, Histone Demethylases metabolism, Humans, Induced Pluripotent Stem Cells metabolism, Megakaryocytes metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Phenotype, Protein Interaction Maps, Proteome analysis, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism, Blood Coagulation Disorders pathology, Blood Platelets pathology, Gene Expression Regulation, Induced Pluripotent Stem Cells pathology, Megakaryocytes pathology, Mutation, Proto-Oncogene Proteins genetics, Repressor Proteins genetics
- Abstract
Dominant-negative mutations in the transcription factor Growth Factor Independence-1B (GFI1B), such as GFI1B
Q287* , cause a bleeding disorder characterized by a plethora of megakaryocyte and platelet abnormalities. The deregulated molecular mechanisms and pathways are unknown. Here we show that both normal and Q287* mutant GFI1B interacted most strongly with the lysine specific demethylase-1 - REST corepressor - histone deacetylase (LSD1-RCOR-HDAC) complex in megakaryoblasts. Sequestration of this complex by GFI1BQ287* and chemical separation of GFI1B from LSD1 induced abnormalities in normal megakaryocytes comparable to those seen in patients. Megakaryocytes derived from GFI1BQ287* -induced pluripotent stem cells also phenocopied abnormalities seen in patients. Proteome studies on normal and mutant-induced pluripotent stem cell-derived megakaryocytes identified a multitude of deregulated pathways downstream of GFI1BQ287* including cell division and interferon signaling. Proteome studies on platelets from GFI1BQ287* patients showed reduced expression of proteins implicated in platelet function, and elevated expression of proteins normally downregulated during megakaryocyte differentiation. Thus, GFI1B and LSD1 regulate a broad developmental program during megakaryopoiesis, and GFI1BQ287* deregulates this program through LSD1-RCOR-HDAC sequestering., (Copyright© 2019 Ferrata Storti Foundation.)- Published
- 2019
- Full Text
- View/download PDF
28. A combined immunodeficiency with severe infections, inflammation, and allergy caused by ARPC1B deficiency.
- Author
-
Volpi S, Cicalese MP, Tuijnenburg P, Tool ATJ, Cuadrado E, Abu-Halaweh M, Ahanchian H, Alzyoud R, Akdemir ZC, Barzaghi F, Blank A, Boisson B, Bottino C, Brigida I, Caorsi R, Casanova JL, Chiesa S, Chinn IK, Dückers G, Enders A, Erichsen HC, Forbes LR, Gambin T, Gattorno M, Karimiani EG, Giliani S, Gold MS, Jacobsen EM, Jansen MH, King JR, Laxer RM, Lupski JR, Mace E, Marcenaro S, Maroofian R, Meijer AB, Niehues T, Notarangelo LD, Orange J, Pannicke U, Pearson C, Picco P, Quinn PJ, Schulz A, Seeborg F, Stray-Pedersen A, Tawamie H, van Leeuwen EMM, Aiuti A, Yeung R, Schwarz K, and Kuijpers TW
- Subjects
- Actin-Related Protein 2-3 Complex genetics, Adolescent, Child, Child, Preschool, Female, Humans, Hypersensitivity genetics, Hypersensitivity immunology, Immunologic Deficiency Syndromes immunology, Infant, Infections genetics, Infections immunology, Inflammation genetics, Inflammation immunology, Male, Mutation, Actin-Related Protein 2-3 Complex deficiency, Immunologic Deficiency Syndromes genetics
- Published
- 2019
- Full Text
- View/download PDF
29. A Homozygous Mutation on the HBA1 Gene Coding for Hb Charlieu (HBA1: c.320T>C) Together with β-Thalassemia Trait Results in Severe Hemolytic Anemia.
- Author
-
Klei TRL, Kheradmand Kia S, Veldthuis M, Dehbozorgian J, Karimi M, Geissler J, Sellink E, Thiel-Valkhof M, Burger P, van Alphen F, Meijer AB, van Bruggen R, and van Zwieten R
- Subjects
- Child, Preschool, Humans, Male, Homozygote, Mutation genetics
- Abstract
A 4-year-old boy, a β-thalassemia (β-thal) carrier, with an unexplained severe chronic microcytic anemia was referred to us. Sequencing of the α-globin genes revealed a Hb Charlieu [α106(G13)Leu→Pro, HBA1 : c.320T>C, p.Leu107Pro] mutation present on both HBA1 genes. Quantitative polymerase chain reaction (qPCR) confirmed α
Charlieu mRNA in the proband and his parents, showing that the mutation does not affect mRNA stability. However, we were unable to detect the Hb Charlieu protein by capillary electrophoresis (CE), reverse phase electrophoresis, cation exchange electrophoresis or isoelectric focusing. Mass spectrometry (MS) allowed us to confirm the presence of the Hb Charlieu peptide in erythrocyte progenitors. These findings suggest that the mutation affects the stability of αCharlieu . As hemoglobin (Hb) heat stability tests showed no abnormalities in erythrocytes, we speculated that αCharlieu is already degraded during red blood cell (RBC) development. The clinical severity in the proband and the presence of new methylene blue-stained aggregates in his reticulocytes indicates that incorporation of αCharlieu destabilizes Hb. This, combined with an excess of unstable free α-globins as the result of β-thal minor, results in severely impaired erythropoiesis and, as a consequence, severe and chronic microcytic anemia in the proband.- Published
- 2019
- Full Text
- View/download PDF
30. Integrated proteomic analysis of tumor necrosis factor α and interleukin 1β-induced endothelial inflammation.
- Author
-
Béguin EP, van den Eshof BL, Hoogendijk AJ, Nota B, Mertens K, Meijer AB, and van den Biggelaar M
- Subjects
- Cells, Cultured, Endothelial Cells pathology, Humans, Inflammation chemically induced, Inflammation metabolism, Inflammation pathology, Proteomics, Cell Adhesion Molecules biosynthesis, Endothelial Cells metabolism, Interleukin-1beta pharmacology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
The vascular endothelium provides a unique interaction plane for plasma proteins and leukocytes in inflammation. The pro-inflammatory cytokines Tumor Necrosis Factor α (TNFα) and interleukin 1β (IL-1β) have a profound effect on endothelial cells, which includes increased levels of adhesion molecules and a disrupted barrier function. To assess the endothelial response to these cytokines at the protein level, we evaluated changes in the whole proteome, cell surface proteome and phosphoproteome after 24 h of cytokine treatment. The effects of TNFα and IL-1β on endothelial cells were strikingly similar and included changes in proteins not previously associated with endothelial inflammation. Temporal profiling revealed time-dependent proteomic changes, including a limited number of early responsive proteins such as adhesion receptors ICAM1 and SELE. In addition, this approach uncovered a greater number of late responsive proteins, including proteins related to self-antigen peptide presentation, and a transient increase in ferritin. Peptide-based cell surface proteomics revealed extensive changes at the cell surface, which were in agreement with the whole proteome. In addition, site-specific changes within ITGA5 and ICAM1 were detected. Combined, our integrated proteomic data provide detailed information on endothelial inflammation, emphasize the role of the extracellular matrix therein, and include potential targets for therapeutic intervention. SIGNIFICANCE: Pro-inflammatory cytokines induce the expression of cell adhesion molecules in vascular endothelial cells. These molecules mediate the adhesion and migration of immune cells across the vessel wall, which is a key process to resolve infections in the underlying tissue. Dysregulation of endothelial inflammation can contribute to vascular diseases and the vascular endothelium is therefore an attractive target to control inflammation. Current strategies targeting endothelial adhesion molecules, including PECAM, CD99, ICAM1 and VCAM1 do not completely prevent transmigration. To identify additional therapeutic targets, we mapped the endothelial proteome after pro-inflammatory cytokine treatment. In addition to the whole proteome, we assessed the surface proteome to focus on cell adhesion molecules, and the phosphoproteome to uncover protein activation states. Here, we present an integrated overview of affected processes which further improves our understanding of endothelial inflammation and may eventually aid in therapeutic intervention of imbalanced inflammation., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
31. Loss-of-function nuclear factor κB subunit 1 (NFKB1) variants are the most common monogenic cause of common variable immunodeficiency in Europeans.
- Author
-
Tuijnenburg P, Lango Allen H, Burns SO, Greene D, Jansen MH, Staples E, Stephens J, Carss KJ, Biasci D, Baxendale H, Thomas M, Chandra A, Kiani-Alikhan S, Longhurst HJ, Seneviratne SL, Oksenhendler E, Simeoni I, de Bree GJ, Tool ATJ, van Leeuwen EMM, Ebberink EHTM, Meijer AB, Tuna S, Whitehorn D, Brown M, Turro E, Thrasher AJ, Smith KGC, Thaventhiran JE, and Kuijpers TW
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Europe, Female, Humans, Infant, Infant, Newborn, Loss of Function Mutation, Male, Middle Aged, Phenotype, T-Lymphocytes immunology, Young Adult, B-Lymphocytes immunology, Common Variable Immunodeficiency genetics, NF-kappa B p50 Subunit genetics
- Abstract
Background: The genetic cause of primary immunodeficiency disease (PID) carries prognostic information., Objective: We conducted a whole-genome sequencing study assessing a large proportion of the NIHR BioResource-Rare Diseases cohort., Methods: In the predominantly European study population of principally sporadic unrelated PID cases (n = 846), a novel Bayesian method identified nuclear factor κB subunit 1 (NFKB1) as one of the genes most strongly associated with PID, and the association was explained by 16 novel heterozygous truncating, missense, and gene deletion variants. This accounted for 4% of common variable immunodeficiency (CVID) cases (n = 390) in the cohort. Amino acid substitutions predicted to be pathogenic were assessed by means of analysis of structural protein data. Immunophenotyping, immunoblotting, and ex vivo stimulation of lymphocytes determined the functional effects of these variants. Detailed clinical and pedigree information was collected for genotype-phenotype cosegregation analyses., Results: Both sporadic and familial cases demonstrated evidence of the noninfective complications of CVID, including massive lymphadenopathy (24%), unexplained splenomegaly (48%), and autoimmune disease (48%), features prior studies correlated with worse clinical prognosis. Although partial penetrance of clinical symptoms was noted in certain pedigrees, all carriers have a deficiency in B-lymphocyte differentiation. Detailed assessment of B-lymphocyte numbers, phenotype, and function identifies the presence of an increased CD21
low B-cell population. Combined with identification of the disease-causing variant, this distinguishes between healthy subjects, asymptomatic carriers, and clinically affected cases., Conclusion: We show that heterozygous loss-of-function variants in NFKB1 are the most common known monogenic cause of CVID, which results in a temporally progressive defect in the formation of immunoglobulin-producing B cells., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
32. The D' domain of von Willebrand factor requires the presence of the D3 domain for optimal factor VIII binding.
- Author
-
Przeradzka MA, Meems H, van der Zwaan C, Ebberink EHTM, van den Biggelaar M, Mertens K, and Meijer AB
- Subjects
- Amino Acid Substitution, Factor VIII genetics, Factor VIII metabolism, Humans, Protein Binding, Protein Domains, von Willebrand Factor genetics, von Willebrand Factor metabolism, Factor VIII chemistry, Mutation, Missense, Surface Plasmon Resonance, von Willebrand Factor chemistry
- 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., (© 2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2018
- Full Text
- View/download PDF
33. Strap associates with Csde1 and affects expression of select Csde1-bound transcripts.
- Author
-
Moore KS, Yagci N, van Alphen F, Meijer AB, 't Hoen PAC, and von Lindern M
- Subjects
- Animals, Carrier Proteins genetics, Cell Cycle, Cell Differentiation, Erythroblasts cytology, Erythroblasts metabolism, Gene Knockdown Techniques, HEK293 Cells, Humans, Mice, Protein Binding, RNA, Messenger genetics, RNA, Messenger metabolism, Ribosomes metabolism, Carrier Proteins metabolism, DNA-Binding Proteins metabolism, Gene Expression Regulation, RNA-Binding Proteins metabolism
- Abstract
Erythropoiesis is regulated at many levels, including control of mRNA translation. Changing environmental conditions, such as hypoxia or the availability of nutrients and growth factors, require a rapid response enacted by the enhanced or repressed translation of existing transcripts. Cold shock domain protein e1 (Csde1/Unr) is an RNA-binding protein required for erythropoiesis and strongly upregulated in erythroblasts relative to other hematopoietic progenitors. The aim of this study is to identify the Csde1-containing protein complexes and investigate their role in post-transcriptional expression control of Csde1-bound transcripts. We show that Serine/Threonine kinase receptor-associated protein (Strap/Unrip), was the protein most strongly associated with Csde1 in erythroblasts. Strap is a WD40 protein involved in signaling and RNA splicing, but its role when associated with Csde1 is unknown. Reduced expression of Strap did not alter the pool of transcripts bound by Csde1. Instead, it altered the mRNA and/or protein expression of several Csde1-bound transcripts that encode for proteins essential for translational regulation during hypoxia, such as Hmbs, eIF4g3 and Pabpc4. Also affected by Strap knockdown were Vim, a Gata-1 target crucial for erythrocyte enucleation, and Elavl1, which stabilizes Gata-1 mRNA. The major cellular processes affected by both Csde1 and Strap were ribosome function and cell cycle control., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
34. Mass spectrometry-assisted identification of ADAMTS13-derived peptides presented on HLA-DR and HLA-DQ.
- Author
-
Hrdinová J, Verbij FC, Kaijen PHP, Hartholt RB, van Alphen F, Lardy N, Ten Brinke A, Vanhoorelbeke K, Hindocha PJ, De Groot AS, Meijer AB, Voorberg J, and Peyron I
- Subjects
- ADAMTS13 Protein metabolism, Animals, Antigen Presentation, Dendritic Cells, Epitope Mapping methods, Genotype, HEK293 Cells, HLA-DQ Antigens genetics, HLA-DQ Antigens metabolism, HLA-DR Antigens genetics, HLA-DR Antigens metabolism, High-Throughput Nucleotide Sequencing, Humans, Mice, Peptides metabolism, Protein Binding, ADAMTS13 Protein chemistry, ADAMTS13 Protein immunology, HLA-DQ Antigens immunology, HLA-DR Antigens immunology, Mass Spectrometry methods, Peptides chemistry, Peptides immunology
- Abstract
Formation of microthrombi is a hallmark of acquired thrombotic thrombocytopenic purpura. These microthrombi originate from insufficient processing of ultra large von Willebrand factor multimers by ADAMTS13 due to the development of anti-ADAMTS13 autoantibodies. Several studies have identified the major histocompatibility complex class II alleles HLA-DRB1*11, HLA-DQB1*03 and HLA-DQB1*02:02 as risk factors for acquired thrombotic thrombocytopenic purpura development. Previous research in our department indicated that ADAMTS13 CUB2 domain-derived peptides FINVAPHAR and LIRDTHSLR are presented on HLA-DRB1*11 and HLA-DRB1*03, respectively. Here, we describe the repertoire of ADAMTS13 peptides presented on HLA-DQ. In parallel, the repertoire of ADAMTS13-derived peptides presented on HLA-DR was monitored. Using HLA-DR- and HLA-DQ-specific antibodies, we purified HLA/peptide complexes from ADAMTS13-pulsed monocyte-derived dendritic cells. Using this approach, we identified ADAMTS13-derived peptides presented on HLA-DR for all 9 samples analyzed; ADAMTS13-derived peptides presented on HLA-DQ were identified in 4 out of 9 samples. We were able to confirm the presentation of the CUB2 domain-derived peptides FINVAPHAR and LIRDTHSLR on HLA-DR. In total, 12 different core-peptide sequences were identified on HLA-DR and 8 on HLA-DQ. For HLA-DR11, several potential new core-peptides were found; 4 novel core-peptides were exclusively identified on HLA-DQ. Furthermore, an in silico analysis was performed using the EpiMatrix and JanusMatrix tools to evaluate the eluted peptides, in the context of HLA-DR, for putative effector or regulatory T-cell responses at the population level. The results from this study provide a basis for the identification of immuno-dominant epitopes on ADAMTS13 involved in the onset of acquired thrombotic thrombocytopenic purpura., (Copyright © 2018 Ferrata Storti Foundation.)
- Published
- 2018
- Full Text
- View/download PDF
35. Identifying Children with HEreditary Coagulation disorders (iCHEC): a protocol for a prospective cohort study.
- Author
-
Stokhuijzen E, Rand ML, Cnossen MH, Biss TT, James PD, Suijker MH, Peters M, van der Lee JH, Peters B, Meijer AB, Blanchette VS, and Fijnvandraat K
- Subjects
- Adolescent, Canada, Child, Child, Preschool, Female, Humans, Infant, Infant, Newborn, Male, Netherlands, Prospective Studies, ROC Curve, Research Design, Self Report, Severity of Illness Index, United Kingdom, Blood Coagulation Disorders, Inherited diagnosis, Hemorrhage diagnosis, Mass Screening methods
- Abstract
Introduction: It is challenging to obtain a reliable bleeding history in children who are referred for a suspected inherited bleeding disorder. Bleeding symptoms may be subtle as children face fewer haemostatic challenges compared with adults. In order to standardise bleeding histories, questionnaires have been developed, called bleeding assessment tools (BATs). Although it has been shown that high bleeding scores are associated with the presence of a mucocutaneous bleeding disorder, these BATs lack sensitivity, efficiency and flexibility in the paediatric setting. We developed a new BAT (the iCHEC (identifying Children with HEreditary Coagulation disorders) BAT) to improve on these characteristics. We aim to evaluate the diagnostic accuracy of the iCHEC BAT as a screening tool for children who are suspected for having a bleeding disorder., Methods and Analysis: This is a prospective cohort study. Children (age 0-18 years) suspected for a bleeding disorder who present at tertiary haematology clinics, and/or their parents/guardians, will be asked to complete the iCHEC BAT. Sensitivity was increased by inclusion of paediatric-specific bleeding symptoms and novel qualitative questions per bleeding symptom. Efficiency was improved by developing a self-administered (online) version of the questionnaire. Flexibility for changes in the bleeding phenotype of developing children was improved by including questions that define when the bleeding symptoms occurred in the past. The diagnostic accuracy of the specific bleeding items will be evaluated by receiver operator characteristic curves, using classification based on the results from laboratory assessment as the reference standard. Analysis of the discriminative power of individual bleeding symptoms will be assessed., Ethics and Dissemination: The study has been approved by the medical ethics committees of all participating centres in the Netherlands, Canada and the UK. All paediatric subjects and/or their parents/guardians will provide written informed consent. Study results will be submitted for publication in peer-reviewed journals., Competing Interests: Competing interests: None declared., (© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.)
- Published
- 2018
- Full Text
- View/download PDF
36. Csde1 binds transcripts involved in protein homeostasis and controls their expression in an erythroid cell line.
- Author
-
Moore KS, Yagci N, van Alphen F, Paolini NA, Horos R, Held NM, Houtkooper RH, van den Akker E, Meijer AB, 't Hoen PAC, and von Lindern M
- Subjects
- Animals, CRISPR-Cas Systems genetics, DNA-Binding Proteins genetics, Erythropoiesis, HEK293 Cells, Humans, Poly(A)-Binding Proteins metabolism, RNA, Messenger genetics, RNA-Binding Proteins genetics, Tumor Cells, Cultured, DNA-Binding Proteins metabolism, Erythroid Cells metabolism, Gene Expression Regulation, Proteostasis, RNA-Binding Proteins metabolism
- Abstract
Expression of the RNA-binding protein Csde1 (Cold shock domain protein e1) is strongly upregulated during erythropoiesis compared to other hematopoietic lineages. Csde1 expression is impaired in the severe congenital anemia Diamond Blackfan Anemia (DBA), and reduced expression of Csde1 in healthy erythroblasts impaired their proliferation and differentiation. To investigate the cellular pathways controlled by Csde1 in erythropoiesis, we identified the transcripts that physically associate with Csde1 in erythroid cells. These mainly encoded proteins involved in ribogenesis, mRNA translation and protein degradation, but also proteins associated with the mitochondrial respiratory chain and mitosis. Crispr/Cas9-mediated deletion of the first cold shock domain of Csde1 affected RNA expression and/or protein expression of Csde1-bound transcripts. For instance, protein expression of Pabpc1 was enhanced while Pabpc1 mRNA expression was reduced indicating more efficient translation of Pabpc1 followed by negative feedback on mRNA stability. Overall, the effect of reduced Csde1 function on mRNA stability and translation of Csde1-bound transcripts was modest. Clones with complete loss of Csde1, however, could not be generated. We suggest that Csde1 is involved in feed-back control in protein homeostasis and that it dampens stochastic changes in mRNA expression.
- Published
- 2018
- Full Text
- View/download PDF
37. Chemical Footprinting Reveals Conformational Changes Following Activation of Factor XI.
- Author
-
Stroo I, Marquart JA, Bakhtiari K, Plug T, Meijer AB, and Meijers JCM
- Subjects
- Arginine chemistry, Binding Sites, Blood Coagulation, Blood Coagulation Tests, Catalytic Domain, Cloning, Molecular, Crystallography, X-Ray, Humans, Isoleucine chemistry, Mass Spectrometry, Peptides chemistry, Protein Conformation, Recombinant Proteins chemistry, Factor XI chemistry, Factor XIa chemistry, Lysine chemistry
- Abstract
Coagulation factor XI is activated by thrombin or factor XIIa resulting in a conformational change that converts the catalytic domain into its active form and exposing exosites for factor IX on the apple domains. Although crystal structures of the zymogen factor XI and the catalytic domain of the protease are available, the structure of the apple domains and hence the interactions with the catalytic domain in factor XIa are unknown. We now used chemical footprinting to identify lysine residue containing regions that undergo a conformational change following activation of factor XI. To this end, we employed tandem mass tag in conjunction with mass spectrometry. Fifty-two unique peptides were identified, covering 37 of the 41 lysine residues present in factor XI. Two identified lysine residues that showed altered flexibility upon activation were mutated to study their contribution in factor XI stability or enzymatic activity. Lys357, part of the connecting loop between A4 and the catalytic domain, was more reactive in factor XIa but mutation of this lysine residue did not impact on factor XIa activity. Lys516 and its possible interactor Glu380 are located in the catalytic domain and are covered by the activation loop of factor XIa. Mutating Glu380 enhanced Arg369 cleavage and thrombin generation in plasma. In conclusion, we have identified novel regions that undergo a conformational change following activation. This information improves knowledge about factor XI and will contribute to development of novel inhibitors or activators for this coagulation protein., Competing Interests: None declared., (Schattauer GmbH Stuttgart.)
- Published
- 2018
- Full Text
- View/download PDF
38. Comparative profiling of HLA-DR and HLA-DQ associated factor VIII peptides presented by monocyte-derived dendritic cells.
- Author
-
Peyron I, Hartholt RB, Pedró-Cos L, van Alphen F, Brinke AT, Lardy N, Meijer AB, and Voorberg J
- Subjects
- Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, Factor VIII chemistry, Gene Expression Profiling, HLA-DQ Antigens genetics, HLA-DR Antigens genetics, Hemophilia A genetics, Hemophilia A immunology, Humans, Proteome, Proteomics methods, Antigen Presentation immunology, Dendritic Cells immunology, Dendritic Cells metabolism, Factor VIII immunology, HLA-DQ Antigens immunology, HLA-DR Antigens immunology, Peptides immunology
- Abstract
The development of anti-factor VIII antibodies is a major complication of the treatment of patients with hemophilia A. Generation of high affinity anti-factor VIII antibodies is dependent on help provided by CD4
+ T cells that recognize factor VIII-derived peptides presented on class II major histocompatibility complex on the surface of antigen-presenting cells. In order to identify the immune-dominant epitopes that can be presented to CD4+ T cells, we previously developed a mass spectrometry-based method to identify factor VIII-derived peptides that are presented on human leukocyte antigen (HLA)-DR. In the present work, we compared the repertoire of FVIII-derived peptide presented on HLA-DR and HLA-DQ. Monocyte-derived dendritic cells from nine HLA-typed healthy donors were pulsed with recombinant factor VIII. HLA-DR and HLA-DQ molecules were purified using monoclonal antibodies. Our data show that HLA-DQ and HLA-DR present a similar repertoire of factor VIII-derived peptides. However, the number of peptides associated with HLA-DQ was lower than that with HLA-DR. We also identified a peptide, within the acidic a3 domains of factor VIII, which is presented with higher frequency on HLA-DQ. Interestingly, this peptide was found to have a higher predicted affinity for HLA-DQ than for HLA-DR. Taken together, our data suggest that HLA-DQ participates in the presentation of factor VIII peptides, thereby contributing to the development of inhibitory antibodies in a proportion of patients with severe hemophilia A., (Copyright© 2018 Ferrata Storti Foundation.)- Published
- 2018
- Full Text
- View/download PDF
39. Differences between Platelets Derived from Neonatal Cord Blood and Adult Peripheral Blood Assessed by Mass Spectrometry.
- Author
-
Stokhuijzen E, Koornneef JM, Nota B, van den Eshof BL, van Alphen FPJ, van den Biggelaar M, van der Zwaan C, Kuijk C, Mertens K, Fijnvandraat K, and Meijer AB
- Subjects
- Adult, Collagen metabolism, Female, Humans, Infant, Newborn, Male, Mass Spectrometry, Middle Aged, Platelet Activation genetics, Receptor, PAR-1 metabolism, Receptors, Thrombin metabolism, Transcriptome genetics, Blood Platelets metabolism, Fetal Blood metabolism, Platelet Aggregation genetics, Proteomics
- Abstract
It has been proposed that differences may exist between umbilical cord blood (CB) platelets and adult peripheral blood (APB) platelets, including altered protein levels of the main platelet integrins. We have now compared the protein expression profiles of CB and APB platelets employing a label-free comparative proteomics approach. Aggregation studies showed that CB platelets effectively aggregate in the presence of thromboxane A2 analogue, collagen, and peptide agonists of the proteinase-activated receptors 1 and 4. In agreement with previous studies, higher concentrations of the agonists were required to initiate aggregation in the CB platelets. Mass spectrometry analysis revealed no significant difference in the expression levels of critical platelet receptors like glycoprotein (GP)Ib, GPV, GPIX, and integrin αIIbβ3. This was confirmed using flow cytometry-based approaches. Gene ontology enrichment analysis revealed that elevated proteins in CB platelets were in particular enriched in proteins contributing to mitochondrial energy metabolism processes. The reduced proteins were enriched in proteins involved in, among others, platelet degranulation and activation. In conclusion, this study reveals that the CB and APB platelets are distinct. In particular, changes were observed for proteins that belong to metabolic and energy generation processes and not for the critical adhesive platelet integrins and glycoproteins.
- Published
- 2017
- Full Text
- View/download PDF
40. Paradigm of Biased PAR1 (Protease-Activated Receptor-1) Activation and Inhibition in Endothelial Cells Dissected by Phosphoproteomics.
- Author
-
van den Eshof BL, Hoogendijk AJ, Simpson PJ, van Alphen FPJ, Zanivan S, Mertens K, Meijer AB, and van den Biggelaar M
- Subjects
- Humans, Lactones pharmacology, Phosphorylation, Proteomics, Pyridines pharmacology, Signal Transduction, Endothelial Cells physiology, Receptor, PAR-1 antagonists & inhibitors, Receptor, PAR-1 physiology
- Abstract
Objective: Thrombin is the key serine protease of the coagulation cascade and mediates cellular responses by activation of PARs (protease-activated receptors). The predominant thrombin receptor is PAR1, and in endothelial cells (ECs), thrombin dynamically regulates a plethora of phosphorylation events. However, it has remained unclear whether thrombin signaling is exclusively mediated through PAR1. Furthermore, mechanistic insight into activation and inhibition of PAR1-mediated EC signaling is lacking. In addition, signaling networks of biased PAR1 activation after differential cleavage of the PAR1 N terminus have remained an unresolved issue., Approach and Results: Here, we used a quantitative phosphoproteomics approach to show that classical and peptide activation of PAR1 induce highly similar signaling, that low thrombin concentrations initiate only limited phosphoregulation, and that the PAR1 inhibitors vorapaxar and parmodulin-2 demonstrate distinct antagonistic properties. Subsequent analysis of the thrombin-regulated phosphosites in the presence of PAR1 inhibitors revealed that biased activation of PAR1 is not solely linked to a specific G-protein downstream of PAR1. In addition, we showed that only the canonical thrombin PAR1 tethered ligand induces extensive early phosphoregulation in ECs., Conclusions: Our study provides detailed insight in the signaling mechanisms downstream of PAR1. Our data demonstrate that thrombin-induced EC phosphoregulation is mediated exclusively through PAR1, that thrombin and thrombin-tethered ligand peptide induce similar phosphoregulation, and that only canonical PAR1 cleavage by thrombin generates a tethered ligand that potently induces early signaling. Furthermore, platelet PAR1 inhibitors directly affect EC signaling, indicating that it will be a challenge to design a PAR1 antagonist that will target only those pathways responsible for tissue pathology., (© 2017 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
41. Hermansky-Pudlak syndrome type 2: Aberrant pre-mRNA splicing and mislocalization of granule proteins in neutrophils.
- Author
-
de Boer M, van Leeuwen K, Geissler J, van Alphen F, de Vries E, van der Kuip M, Terheggen SWJ, Janssen H, van den Berg TK, Meijer AB, Roos D, and Kuijpers TW
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Codon, Nonsense genetics, Exons genetics, Female, Hermanski-Pudlak Syndrome physiopathology, Humans, Infant, Male, Middle Aged, Neutrophils metabolism, Neutrophils pathology, Phenotype, Point Mutation, RNA Splice Sites genetics, Sequence Deletion genetics, Adaptor Protein Complex 3 genetics, Adaptor Protein Complex beta Subunits genetics, Hermanski-Pudlak Syndrome genetics, RNA Precursors genetics, RNA Splicing genetics
- Abstract
Hermansky-Pudlak syndrome type 2 (HPS2) is a syndrome caused by mutations in the beta-3A subunit of the adaptor protein (AP)-3 complex (AP3B1 gene). We describe five unreported cases with four novel mutations, one of which caused aberrant pre-mRNA splicing. A point mutation c.2702C>G in exon 23 of the AP3B1 gene caused deletion of 112 bp in the mRNA in two siblings. This mutation activates a cryptic donor splice site that overrules the wild-type donor splice site of this exon. Three other novel mutations in AP3B1 were identified, that is, a nonsense mutation c.716G>A (p.Trp239Ter), a 1-bp and a 4-bp deletion c.177delA and c.1839_1842delTAGA, respectively, both causing frameshift and premature termination of translation. Mass spectrometry in four of these HPS2 patients demonstrated the (near) absence of all AP-3 complex subunits. Immunoelectron microscopy on the neutrophils of two of these patients showed abnormal granule formation. We found clear mislocalization of myeloperoxidase in the neutrophils even though the content of this protein but not the activity seemed to be present at normal levels. In sum, HPS2 is the result of the absence of the entire AP-3 complex, which results in severe neutropenia with a defect in granule formation as the major hematological finding., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
42. Monitoring storage induced changes in the platelet proteome employing label free quantitative mass spectrometry.
- Author
-
Rijkers M, van den Eshof BL, van der Meer PF, van Alphen FPJ, de Korte D, Leebeek FWG, Meijer AB, Voorberg J, and Jansen AJG
- Subjects
- Gene Ontology, Humans, Proteomics methods, Biological Products chemistry, Blood Platelets chemistry, Drug Storage, Mass Spectrometry, Proteome analysis
- Abstract
Shelf life of platelet concentrates is limited to 5-7 days due to loss of platelet function during storage, commonly referred to as the platelet storage lesion (PSL). To get more insight into the development of the PSL, we used label free quantitative mass spectrometry to identify changes in the platelet proteome during storage. In total 2501 proteins were accurately quantified in 3 biological replicates on at least 1 of the 7 different time-points analyzed. Significant changes in levels of 21 proteins were observed over time. Gene ontology enrichment analysis of these proteins revealed that the majority of this set was involved in platelet degranulation, secretion and regulated exocytosis. Twelve of these proteins have been shown to reside in α-granules. Upon prolonged storage (13-16 days) elevated levels of α-2-macroglobulin, glycogenin and Ig μ chain C region were identified. Taken together this study identifies novel markers for monitoring of the PSL that may potentially also be used for the detection of "young" and "old" platelets in the circulation.
- Published
- 2017
- Full Text
- View/download PDF
43. Cellular uptake of coagulation factor VIII: Elusive role of the membrane-binding spikes in the C1 domain.
- Author
-
Castro-Núñez L, Koornneef JM, Rondaij MG, Bloem E, van der Zwaan C, Mertens K, Meijer AB, and Meems H
- Subjects
- Cell Line, Humans, Low Density Lipoprotein Receptor-Related Protein-1 metabolism, Models, Molecular, Protein Binding, Protein Domains, Cell Membrane metabolism, Endocytosis, Factor VIII chemistry, Factor VIII metabolism
- Abstract
Low density lipoprotein receptor-related protein 1 (LRP1) is involved in the catabolism of many ligands, including factor VIII (FVIII) and alpha-2-macroglobulin (α2M). Transfer of FVIII to LRP1 is currently believed to be preceded by pre-concentration on the cell surface, by interacting with a so far unidentified component. In the present study, we used confocal microscopy and flow cytometry to compare endocytosis of FVIII and α2M using U87MG cells. The results show that α2M is rapidly internalized and does not compete for LRP1 mediated internalization of FVIII. FVIII endocytosis did not occur in the presence of receptor-associated-protein (RAP), but FVIII remained visible as a striated fluorescent pattern at the cell borders. In the presence of Von Willebrand Factor (VWF), no FVIII was observed on or within the cells, suggesting that VWF blocks interaction with both cell surface and LRP1. The same dual inhibition has previously been observed for FVIII C1 domain directed monoclonal antibody KM33. Elimination of the KM33 epitope by replacing FVIII C1 residues 2091-2095 and 2155-2160 for the homologues from factor V (FV), however, did not impair FVIII endocytosis. These membrane spikes alone were insufficient for cellular uptake, because FV was neither internalized by U87MG cells nor capable of effectively competing for FVIII endocytosis. These results show that FVIII endocytosis is driven by interaction with LRP1, but at the same time involves the spikes in the C1 domain that have been implicated in lipid binding., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
44. Combined immunodeficiency with severe inflammation and allergy caused by ARPC1B deficiency.
- Author
-
Kuijpers TW, Tool ATJ, van der Bijl I, de Boer M, van Houdt M, de Cuyper IM, Roos D, van Alphen F, van Leeuwen K, Cambridge EL, Arends MJ, Dougan G, Clare S, Ramirez-Solis R, Pals ST, Adams DJ, Meijer AB, and van den Berg TK
- Subjects
- Animals, Biomarkers, Biopsy, DNA Mutational Analysis, Disease Models, Animal, Disease Susceptibility, Humans, Hypersensitivity metabolism, Inflammation metabolism, Mice, Phenotype, Severe Combined Immunodeficiency metabolism, Severity of Illness Index, Skin immunology, Skin pathology, Actin-Related Protein 2-3 Complex deficiency, Hypersensitivity complications, Hypersensitivity genetics, Inflammation complications, Inflammation genetics, Severe Combined Immunodeficiency diagnosis, Severe Combined Immunodeficiency etiology
- Published
- 2017
- Full Text
- View/download PDF
45. Label-free Analysis of CD8 + T Cell Subset Proteomes Supports a Progressive Differentiation Model of Human-Virus-Specific T Cells.
- Author
-
van Aalderen MC, van den Biggelaar M, Remmerswaal EBM, van Alphen FPJ, Meijer AB, Ten Berge IJM, and van Lier RAW
- Subjects
- CD8-Positive T-Lymphocytes cytology, Humans, Proteome chemistry, Proteome genetics, T-Lymphocyte Subsets cytology, CD8-Positive T-Lymphocytes metabolism, Cell Differentiation, Proteome metabolism, T-Lymphocyte Subsets metabolism
- Abstract
Pathogens trigger T cells to express distinct sets of effector proteins. To better understand the molecular mechanisms that drive functional specification, we used high-resolution mass spectrometry and label-free protein quantification to measure proteomic differences between the seven largest circulating human CD8
+ T cell subsets. Hierarchical clustering of the proteomes placed naive and CD45RA-expressing effector-type T cells at the extremes of the spectrum, with central memory and other effector memory stages located in between. Prominent differences between the subsets included expression of specific granzymes, signaling proteins, and molecules involved in metabolic regulation and cell adhesion. Remarkably, whereas most of the proteomic relationships between the subsets occurred in linear variations, a small proportion of proteins was regulated only in discrete subsets. The data obtained from this proteome analysis correspond best to a progressive differentiation model in which specific stable traits are gradually acquired during development., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
46. Role of glycine 221 in catalytic activity of hyaluronan-binding protein 2.
- Author
-
Stavenuiter F, Ebberink EHTM, Mertens K, and Meijer AB
- Subjects
- Amino Acid Substitution, Calcium chemistry, Catalysis, Glycine chemistry, Glycine genetics, Mutation, Missense, Protein Domains, Serine Endopeptidases genetics, Urokinase-Type Plasminogen Activator chemistry, Urokinase-Type Plasminogen Activator genetics, Serine Endopeptidases chemistry
- Abstract
HABP2 (hyaluronan-binding protein 2) is a Ca
2+ -dependent serine protease with putative roles in blood coagulation and fibrinolysis. A G221E substitution, known as the Marburg I polymorphism, reportedly affects HABP2 function and has been associated with increased risk for cardiovascular disease. However, the importance of Gly-221 for HABP2 activity is unclear. Here, we used G221E, G221A, and G221S mutants to assess the role of Gly-221 in HABP2 catalysis. The G221E variant failed to activate the single-chain urokinase-type plasminogen activator, and the G221A and G221S variants displayed moderately reduced single-chain urokinase-type plasminogen activator activation. Activity toward the peptide substrate S-2288 was markedly decreased in all HABP2 variants, with G221E being the most defective and G221A being the least defective. In the absence of Ca2+ , S-2288 cleavage by wild-type HABP2 was Na+ -dependent, with Km decreasing from 3.0 to 0.6 mm upon titration from 0 to 0.3 m Na+ In the presence of 5 mm Ca2+ , Km was further reduced to 0.05 mm, but without an appreciable contribution of Na+ At physiological concentrations of Na+ and Ca2+ , the three HABP2 variants, and particularly G221E, displayed a major Km increase for S-2288. Chemical footprinting revealed that Ile-16 is significantly less protected from chemical modification in G221E than in wild-type HABP2, suggesting impaired insertion of the N terminus into the G221E protease domain, with a concomitant impact on catalytic activity. Homology modeling suggested that the Glu-221 side chain could sterically hinder insertion of the N terminus into the HABP2 protease domain, helping to explain the detrimental effects of Glu-221 substitution on HABP2 activity., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
47. Factor VIII/V C-domain swaps reveal discrete C-domain roles in factor VIII function and intracellular trafficking.
- Author
-
Ebberink EH, Bouwens EA, Bloem E, Boon-Spijker M, van den Biggelaar M, Voorberg J, Meijer AB, and Mertens K
- Subjects
- Endothelial Cells metabolism, Factor V chemistry, Factor V genetics, Factor VIII chemistry, Factor VIII genetics, Gene Expression, Humans, Intracellular Space metabolism, Models, Molecular, Protein Binding, Protein Conformation, Protein Transport, Structure-Activity Relationship, von Willebrand Factor metabolism, Factor V metabolism, Factor VIII metabolism, Protein Interaction Domains and Motifs
- 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., (Copyright© Ferrata Storti Foundation.)
- Published
- 2017
- Full Text
- View/download PDF
48. The class I scavenger receptor CD163 promotes internalization of ADAMTS13 by macrophages.
- Author
-
Verbij FC, Sorvillo N, Kaijen PHP, Hrdinova J, Peyron I, Fijnheer R, Ten Brinke A, Meijer AB, van Alphen FPJ, van den Berg TK, Graversen JJH, Moestrup SK, and Voorberg J
- Abstract
Internalization of ADAMTS13 by macrophages may contribute to its clearance from the circulation. Here we investigated endocytic mechanisms that contribute to the uptake of ADAMTS13 by macrophages. Human monocyte-derived macrophages were used to monitor the uptake of fluorescently labeled recombinant ADAMTS13 by flow cytometry. Internalization of ADAMTS13 was blocked upon addition of the cell-permeable dynamin inhibitor dynasore. Partial blocking of ADAMTS13 uptake was observed by using mannan; however, uptake was not affected by an antibody that blocked binding to the macrophage mannose receptor CD206, which suggests that other endocytic receptors contribute to the internalization of ADAMTS13 by macrophages. A pull-down with ADAMTS13 and subsequent mass spectrometric analysis identified the class I scavenger receptor CD163 as a candidate receptor for ADAMTS13. Blocking experiments with monoclonal anti-CD163 antibody EDHu-1 resulted in decreased ADAMTS13 internalization by macrophages. Pronounced inhibition of ADAMTS13 uptake by EDHu-1 was observed in CD163 high-expressing macrophages. In agreement with these findings, CD163-expressing Chinese hamster ovary cells were capable of rapidly internalizing ADAMTS13. Surface plasmon resonance revealed binding of ADAMTS13 to scavenger receptor cysteine-rich domains 1-9 and 1-5 of CD163. Taken together, our data identify CD163 as a major endocytic receptor for ADAMTS13 on macrophages., Competing Interests: Conflict-of-interest disclosure: The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
49. Analysis of the HLA-DR peptidome from human dendritic cells reveals high affinity repertoires and nonconventional pathways of peptide generation.
- Author
-
Ciudad MT, Sorvillo N, van Alphen FP, Catalán D, Meijer AB, Voorberg J, and Jaraquemada D
- Subjects
- Alleles, Amino Acid Motifs, Amino Acid Sequence, Cell Differentiation, Cytosol metabolism, Databases, Protein, Endosomes metabolism, Humans, Lysosomes metabolism, Monocytes cytology, Nuclear Proteins metabolism, Peptide Hydrolases metabolism, Peptides chemistry, Phenotype, Dendritic Cells metabolism, HLA-DR Antigens metabolism, Peptides metabolism, Proteome metabolism
- Abstract
Dendritic cells (DCs) are the major professional APCs of the immune system; however, their MHC-II-associated peptide repertoires have been hard to analyze, mostly because of their scarce presence in blood and tissues. In vitro matured human monocyte-derived DCs (MoDCs) are widely used as professional APCs in experimental systems. In this work, we have applied mass spectrometry to identify the HLA-DR-associated self-peptide repertoires from small numbers of mature MoDCs (∼5 × 10
6 cells), derived from 7 different donors. Repertoires of 9 different HLA-DR alleles were defined from analysis of 1319 peptides, showing the expected characteristics of MHC-II-associated peptides. Most peptides identified were predicted high binders for their respective allele, formed nested sets, and belonged to endo-lysosomal pathway-degraded proteins. Approximately 20% of the peptides were derived from cytosolic and nuclear proteins, a recurrent finding in HLA-DR peptide repertoires. Of interest, most of these peptides corresponded to single sequences, did not form nested sets, and were located at the C terminus of the parental protein, which suggested alternative processing. Analysis of cleavage patterns for terminal peptides predominantly showed aspartic acid before the cleavage site of both C- and N-terminal peptides and proline immediately after the cleavage site in C-terminal peptides. Proline was also frequent next to the cut sites of internal peptides. These data provide new insights into the Ag processing capabilities of DCs. The relevance of these processing pathways and their contribution to response to infection, tolerance induction, or autoimmunity deserve further analysis., (© Society for Leukocyte Biology.)- Published
- 2017
- Full Text
- View/download PDF
50. Identification of glycans on plasma-derived ADAMTS13.
- Author
-
Verbij FC, Stokhuijzen E, Kaijen PH, van Alphen F, Meijer AB, and Voorberg J
- Subjects
- ADAMTS13 Protein isolation & purification, Carbohydrate Conformation, Glycosylation, Humans, ADAMTS13 Protein chemistry, Polysaccharides chemistry
- Abstract
Patients suffering from acquired thrombotic thrombocytopenic purpura develop autoantibodies directed toward the plasma glycoprotein ADAMTS13. Here, we studied the glycan composition of plasma-derived ADAMTS13. Purified ADAMTS13 was reduced, alkylated, and processed into peptides with either trypsin or chymotrypsin. Glycopeptides were enriched using zwitterionic HILIC zip-tips and analyzed by tandem mass spectrometry employing higher-energy collision dissociation fragmentation. Upon detection of a diagnostic ion of a glycan fragment, electron transfer dissociation fragmentation was performed on the same precursor ion. The majority of N-linked glycans were of the complex type containing terminal sialic acids and fucose residues. A high mannose-containing glycan was attached to Asn614 in the spacer domain. Six O-linked glycans mostly terminating in sialic acid were found dispersed over ADAMTS13. Five O-linked glycans were attached to a Ser and one to Thr. All 6 O-linked glycans contained a terminal sialic acid. O-fucosylation is a common posttranslational modification of thrombospondin type 1 repeats. We identified 7 O-fucosylation sites in the thrombospondin (TSP) type 1 repeats. Unexpectedly, one additional O-fucosylation site was found in the disintegrin domain. This O-fucosylation site did not meet the proposed consensus sequence CSX(S/T)CG. C-mannosylation sites were identified in TSP1, linker TSP4-TSP5, and TSP8. Overall, our findings highlight the complexity of glycan modifications on ADAMTS13, which may have implications for its interaction with immune- or clearance receptors containing carbohydrate recognition domains., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.