1
|
Deng S, Zhang Y, Shen S, Li C, Qin C. Immunometabolism of Liver Xenotransplantation and Prospective Solutions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2407610. [PMID: 39912334 DOI: 10.1002/advs.202407610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/26/2024] [Indexed: 02/07/2025]
Abstract
End-stage liver diseases, such as hepatocellular carcinoma or acute liver failure, critically necessitate liver transplantation. However, the shortage of available organ donors fails to meet the rapidly growing transplantation demand. Due to the high similarity of liver tissue structure and metabolism between miniature pigs and humans, xenotransplantation of pig livers is considered as a potentially viable solution to organ scarcity. In the 2024, teams from China first time have successfully transplanted a genetically modified Bama miniature pig liver into a clinically brain-dead man lasting for 10 days. This milestone in human xenotransplantation research not only confirms the feasibility of clinical application of xenotransplantation, but also underscores the daunting and protracted nature of this pathway. Despite advanced gene-editing technologies theoretically circumventing the occurrence of most transplant rejection reactions, patients still face challenges such as chronic immune rejection, coagulation disorders, and thrombotic microangiopathy after receiving xenografts. Moreover, prolonged use of immunosuppressive drugs may induce irreversible immune dysfunction, leading to opportunistic infections and metabolic disorders. This article compares the similarities and differences in livers between humans and pigs, summarizes the immunometabolism of xenotransplantation based on current findings, and provides research perspectives on pre-transplantation and post-transplantation strategies for prolonging the survival time of xenografts.
Collapse
Affiliation(s)
- Shoulong Deng
- National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, National Health Commission of China (NHC) Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| | - Yi Zhang
- Department of Medicine, Panzhihua University, Sichuan, 61700, China
| | - Shasha Shen
- Department of Medicine, Panzhihua University, Sichuan, 61700, China
| | - Chongyang Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Chuan Qin
- National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, National Health Commission of China (NHC) Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
2
|
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, van den Biggelaar M. Variant mapping using mass spectrometry-based proteotyping as a diagnostic tool in von Willebrand disease. J Thromb Haemost 2024; 22:1894-1908. [PMID: 38679335 DOI: 10.1016/j.jtha.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/20/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
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.
Collapse
Affiliation(s)
- Iris C Kreft
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands.
| | - Tirsa T van Duijl
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Calvin van Kwawegen
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ferdows Atiq
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Winny Phan
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Margo B P Schuller
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Mariëtte Boon-Spijker
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Carmen van der Zwaan
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Alexander B Meijer
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Arie J Hoogendijk
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeroen C J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maartje van den Biggelaar
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Karampini E, Doherty D, Bürgisser PE, Garre M, Schoen I, Elliott S, Bierings R, O’Donnell JS. O-glycan determinants regulate VWF trafficking to Weibel-Palade bodies. Blood Adv 2024; 8:3254-3266. [PMID: 38640438 PMCID: PMC11226974 DOI: 10.1182/bloodadvances.2023012499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT von Willebrand factor (VWF) undergoes complex posttranslational modification within endothelial cells (ECs) before secretion. This includes significant N- and O-linked glycosylation. Previous studies have demonstrated that changes in N-linked glycan structures significantly influence VWF biosynthesis. In contrast, although abnormalities in VWF O-linked glycans (OLGs) have been associated with enhanced VWF clearance, their effect on VWF biosynthesis remains poorly explored. Herein, we report a novel role for OLG determinants in regulating VWF biosynthesis and trafficking within ECs. We demonstrate that alterations in OLGs (notably reduced terminal sialylation) lead to activation of the A1 domain of VWF within EC. In the presence of altered OLG, VWF multimerization is reduced and Weibel-Palade body (WPB) formation significantly impaired. Consistently, the amount of VWF secreted from WPB after EC activation was significantly reduced in the context of O-glycosylation inhibition. Finally, altered OLG on VWF not only reduced the amount of VWF secreted after EC activation but also affected its hemostatic efficacy. Notably, VWF secreted after WPB exocytosis consisted predominantly of low molecular weight multimers, and the length of tethered VWF string formation on the surface of activated ECs was significantly reduced. In conclusion, our data therefore support the hypothesis that alterations in O-glycosylation pathways directly affect VWF trafficking within human EC. These findings are interesting given that previous studies have reported altered OLG on plasma VWF (notably increased T-antigen expression) in patients with von Willebrand disease.
Collapse
Affiliation(s)
- Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Petra E. Bürgisser
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Massimiliano Garre
- Super-Resolution Imaging Consortium, Department of Chemistry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stephanie Elliott
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ruben Bierings
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Coagulation Centre, St James’s Hospital, Dublin, Ireland
| |
Collapse
|
4
|
Garapati K, Jain A, Madden BJ, Mun DG, Sharma J, Budhraja R, Pandey A. Defining albumin as a glycoprotein with multiple N-linked glycosylation sites. J Transl Med 2024; 22:454. [PMID: 38741158 PMCID: PMC11090807 DOI: 10.1186/s12967-024-05000-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/14/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Glycosylation is an enzyme-catalyzed post-translational modification that is distinct from glycation and is present on a majority of plasma proteins. N-glycosylation occurs on asparagine residues predominantly within canonical N-glycosylation motifs (Asn-X-Ser/Thr) although non-canonical N-glycosylation motifs Asn-X-Cys/Val have also been reported. Albumin is the most abundant protein in plasma whose glycation is well-studied in diabetes mellitus. However, albumin has long been considered a non-glycosylated protein due to absence of canonical motifs. Albumin contains two non-canonical N-glycosylation motifs, of which one was recently reported to be glycosylated. METHODS We enriched abundant serum proteins to investigate their N-linked glycosylation followed by trypsin digestion and glycopeptide enrichment by size-exclusion or mixed-mode anion-exchange chromatography. Glycosylation at canonical as well as non-canonical sites was evaluated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) of enriched glycopeptides. Deglycosylation analysis was performed to confirm N-linked glycosylation at non-canonical sites. Albumin-derived glycopeptides were fragmented by MS3 to confirm attached glycans. Parallel reaction monitoring was carried out on twenty additional samples to validate these findings. Bovine and rabbit albumin-derived glycopeptides were similarly analyzed by LC-MS/MS. RESULTS Human albumin is N-glycosylated at two non-canonical sites, Asn68 and Asn123. N-glycopeptides were detected at both sites bearing four complex sialylated glycans and validated by MS3-based fragmentation and deglycosylation studies. Targeted mass spectrometry confirmed glycosylation in twenty additional donor samples. Finally, the highly conserved Asn123 in bovine and rabbit serum albumin was also found to be glycosylated. CONCLUSIONS Albumin is a glycoprotein with conserved N-linked glycosylation sites that could have potential clinical applications.
Collapse
Affiliation(s)
- Kishore Garapati
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Anu Jain
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Dong-Gi Mun
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jyoti Sharma
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India
| | - Rohit Budhraja
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
5
|
Sadler B. Novel risk loci for thrombotic factors. Blood 2024; 143:1790-1792. [PMID: 38696197 DOI: 10.1182/blood.2024023891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
|
6
|
de Vries PS, Reventun P, Brown MR, Heath AS, Huffman JE, Le NQ, Bebo A, Brody JA, Temprano-Sagrera G, Raffield LM, Ozel AB, Thibord F, Jain D, Lewis JP, Rodriguez BAT, Pankratz N, Taylor KD, Polasek O, Chen MH, Yanek LR, Carrasquilla GD, Marioni RE, Kleber ME, Trégouët DA, Yao J, Li-Gao R, Joshi PK, Trompet S, Martinez-Perez A, Ghanbari M, Howard TE, Reiner AP, Arvanitis M, Ryan KA, Bartz TM, Rudan I, Faraday N, Linneberg A, Ekunwe L, Davies G, Delgado GE, Suchon P, Guo X, Rosendaal FR, Klaric L, Noordam R, van Rooij F, Curran JE, Wheeler MM, Osburn WO, O'Connell JR, Boerwinkle E, Beswick A, Psaty BM, Kolcic I, Souto JC, Becker LC, Hansen T, Doyle MF, Harris SE, Moissl AP, Deleuze JF, Rich SS, van Hylckama Vlieg A, Campbell H, Stott DJ, Soria JM, de Maat MPM, Almasy L, Brody LC, Auer PL, Mitchell BD, Ben-Shlomo Y, Fornage M, Hayward C, Mathias RA, Kilpeläinen TO, Lange LA, Cox SR, März W, Morange PE, Rotter JI, Mook-Kanamori DO, Wilson JF, van der Harst P, Jukema JW, Ikram MA, Blangero J, Kooperberg C, Desch KC, Johnson AD, Sabater-Lleal M, Lowenstein CJ, Smith NL, Morrison AC. A genetic association study of circulating coagulation factor VIII and von Willebrand factor levels. Blood 2024; 143:1845-1855. [PMID: 38320121 PMCID: PMC11443575 DOI: 10.1182/blood.2023021452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
ABSTRACT Coagulation factor VIII (FVIII) and its carrier protein von Willebrand factor (VWF) are critical to coagulation and platelet aggregation. We leveraged whole-genome sequence data from the Trans-Omics for Precision Medicine (TOPMed) program along with TOPMed-based imputation of genotypes in additional samples to identify genetic associations with circulating FVIII and VWF levels in a single-variant meta-analysis, including up to 45 289 participants. Gene-based aggregate tests were implemented in TOPMed. We identified 3 candidate causal genes and tested their functional effect on FVIII release from human liver endothelial cells (HLECs) and VWF release from human umbilical vein endothelial cells. Mendelian randomization was also performed to provide evidence for causal associations of FVIII and VWF with thrombotic outcomes. We identified associations (P < 5 × 10-9) at 7 new loci for FVIII (ST3GAL4, CLEC4M, B3GNT2, ASGR1, F12, KNG1, and TREM1/NCR2) and 1 for VWF (B3GNT2). VWF, ABO, and STAB2 were associated with FVIII and VWF in gene-based analyses. Multiphenotype analysis of FVIII and VWF identified another 3 new loci, including PDIA3. Silencing of B3GNT2 and the previously reported CD36 gene decreased release of FVIII by HLECs, whereas silencing of B3GNT2, CD36, and PDIA3 decreased release of VWF by HVECs. Mendelian randomization supports causal association of higher FVIII and VWF with increased risk of thrombotic outcomes. Seven new loci were identified for FVIII and 1 for VWF, with evidence supporting causal associations of FVIII and VWF with thrombotic outcomes. B3GNT2, CD36, and PDIA3 modulate the release of FVIII and/or VWF in vitro.
Collapse
Affiliation(s)
- Paul S. de Vries
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Paula Reventun
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michael R. Brown
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Adam S. Heath
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jennifer E. Huffman
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA
| | - Ngoc-Quynh Le
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Pau, Barcelona, Spain
| | - Allison Bebo
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | | | - Laura M. Raffield
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | - Florian Thibord
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, MA
| | - Deepti Jain
- Department of Biostatistics, Genetic Analysis Center, School of Public Health, University of Washington, Seattle, WA
| | - Joshua P. Lewis
- Department of Medicine, University of Maryland, Baltimore, MD
| | - Benjamin A. T. Rodriguez
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, MA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kent D. Taylor
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - Ming-Huei Chen
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, MA
| | - Lisa R. Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - German D. Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Marcus E. Kleber
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Jie Yao
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter K. Joshi
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Angel Martinez-Perez
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Pau, Barcelona, Spain
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Tom E. Howard
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
| | - Alex P. Reiner
- Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA
| | - Marios Arvanitis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Traci M. Bartz
- Departments of Biostatistics and Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Nauder Faraday
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lynette Ekunwe
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Gail Davies
- Department of Psychology, Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland
| | - Graciela E. Delgado
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Pierre Suchon
- C2VN, INSERM, INRAE, Aix Marseille University, Marseille, France
- Laboratory of Haematology, La Timone Hospital, Marseille, France
| | - Xiuqing Guo
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Frits R. Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lucija Klaric
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank van Rooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Joanne E. Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
| | - Marsha M. Wheeler
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - William O. Osburn
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Andrew Beswick
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Departments of Epidemiology and Health Systems and Population Health, Seattle, WA
| | - Ivana Kolcic
- Faculty of Medicine, University of Split, Split, Croatia
| | - Juan Carlos Souto
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Pau, Barcelona, Spain
- Unit of Thrombosis and Hemostasis, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Lewis C. Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Margaret F. Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Colchester, VT
| | - Sarah E. Harris
- Department of Psychology, Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland
| | - Angela P. Moissl
- Institute of Nutritional Sciences, Friedrich-Schiller-University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health Halle-Jena-Leipzig, Jena, Germany
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, CEA, Evry, France
- Centre d'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Stephen S. Rich
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | | | - Harry Campbell
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland
| | - Jose Manuel Soria
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Pau, Barcelona, Spain
| | - Moniek P. M. de Maat
- Department of Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Laura Almasy
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Lawrence C. Brody
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Paul L. Auer
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, WI
| | - Braxton D. Mitchell
- Department of Medicine, University of Maryland, Baltimore, MD
- Geriatric Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD
| | - Yoav Ben-Shlomo
- Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Myriam Fornage
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Rasika A. Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Leslie A. Lange
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Simon R. Cox
- Department of Psychology, Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland
| | - Winfried März
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Pierre-Emmanuel Morange
- C2VN, INSERM, INRAE, Aix Marseille University, Marseille, France
- Laboratory of Haematology, La Timone Hospital, Marseille, France
| | - Jerome I. Rotter
- Department of Pediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Dennis O. Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - James F. Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Pim van der Harst
- Division of Heart and Lungs, Department of Cardiology, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
| | | | - Karl C. Desch
- Department of Pediatrics, University of Michigan, C.S. Mott Children's Hospital, Ann Arbor, MI
| | - Andrew D. Johnson
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham Heart Study, Framingham, MA
| | - Maria Sabater-Lleal
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Pau, Barcelona, Spain
- Department of Medicine, Cardiovascular Medicine Unit, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | | | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Department of Veterans Affairs Office of Research and Development, Seattle Epidemiologic and Information Center, Seattle, WA
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
7
|
Shi H, Gao L, Kirby N, Shao B, Shan X, Kudo M, Silasi R, McDaniel JM, Zhou M, McGee S, Jing W, Lupu F, Cleuren A, George JN, Xia L. Clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in sickle cell anemia mice. Blood 2024; 143:1293-1309. [PMID: 38142410 PMCID: PMC10997916 DOI: 10.1182/blood.2023021583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT Although it is caused by a single-nucleotide mutation in the β-globin gene, sickle cell anemia (SCA) is a systemic disease with complex, incompletely elucidated pathologies. The mononuclear phagocyte system plays critical roles in SCA pathophysiology. However, how heterogeneous populations of hepatic macrophages contribute to SCA remains unclear. Using a combination of single-cell RNA sequencing and spatial transcriptomics via multiplexed error-robust fluorescence in situ hybridization, we identified distinct macrophage populations with diversified origins and biological functions in SCA mouse liver. We previously found that administering the von Willebrand factor (VWF)-cleaving protease ADAMTS13 alleviated vaso-occlusive episode in mice with SCA. Here, we discovered that the ADAMTS13-cleaved VWF was cleared from the circulation by a Clec4f+Marcohigh macrophage subset in a desialylation-dependent manner in the liver. In addition, sickle erythrocytes were phagocytized predominantly by Clec4f+Marcohigh macrophages. Depletion of macrophages not only abolished the protective effect of ADAMTS13 but exacerbated vaso-occlusive episode in mice with SCA. Furthermore, promoting macrophage-mediated VWF clearance reduced vaso-occlusion in SCA mice. Our study demonstrates that hepatic macrophages are important in the pathogenesis of SCA, and efficient clearance of VWF by hepatic macrophages is critical for the protective effect of ADAMTS13 in SCA mice.
Collapse
Affiliation(s)
- Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Liang Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Nicole Kirby
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Xindi Shan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Mariko Kudo
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - John Michael McDaniel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Meixiang Zhou
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Samuel McGee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Wei Jing
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Audrey Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - James N. George
- Hematology-Oncology Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
8
|
Swystun LL, Michels A, Lillicrap D. The contribution of the sinusoidal endothelial cell receptors CLEC4M, stabilin-2, and SCARA5 to VWF-FVIII clearance in thrombosis and hemostasis. J Thromb Haemost 2023; 21:2007-2019. [PMID: 37085036 PMCID: PMC11539076 DOI: 10.1016/j.jtha.2023.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Quantitative abnormalities in factor VIII (FVIII) and its binding partner, von Willebrand factor (VWF), are associated with an increased risk of bleeding or thrombosis, and pathways that regulate the clearance of VWF-FVIII can strongly influence their plasma levels. In 2010, the Cohorts for Heart and Aging Research in Genome Epidemiology (CHARGE) on genome-wide association study meta-analysis identified variants in the genes for the sinusoidal endothelial receptors C-type lectin domain family 4 member M (CLEC4M), stabilin-2, and scavenger receptor class A member 5 (SCARA5) as being associated with plasma levels of VWF and/or FVIII in normal individuals. The ability of these receptors to bind, internalize, and clear the VWF-FVIII complex from the circulation has now been reported in a series of studies using in vitro and in vivo models. The receptor stabilin-2 has also been shown to modulate the immune response to infused VWF-FVIII concentrates in a murine model. In addition, the influence of genetic variants in CLEC4M, STAB2, and SCARA5 on type 1 von Willebrand disease/low VWF phenotype, FVIII pharmacokinetics, and the risk of venous thromboembolism has been described in a number of patient-based studies. Understanding the role of these receptors in the regulation of VWF-FVIII clearance has led to significant insights into the genomic architecture that modulates plasma VWF and FVIII levels, improving the understanding of pathways that regulate VWF-FVIII clearance and the mechanistic basis of quantitative VWF-FVIII pathologies.
Collapse
Affiliation(s)
- Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Alison Michels
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada. https://twitter.com/michels_alison
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
9
|
Sarafanov AG. Plasma Clearance of Coagulation Factor VIII and Extension of Its Half-Life for the Therapy of Hemophilia A: A Critical Review of the Current State of Research and Practice. Int J Mol Sci 2023; 24:ijms24108584. [PMID: 37239930 DOI: 10.3390/ijms24108584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Factor VIII (FVIII) is an important component of blood coagulation as its congenital deficiency results in life-threatening bleeding. Current prophylactic therapy of the disease (hemophilia A) is based on 3-4 intravenous infusions of therapeutic FVIII per week. This poses a burden on patients, demanding reduction of infusion frequency by using FVIII with extended plasma half-life (EHL). Development of these products requires understanding FVIII plasma clearance mechanisms. This paper overviews (i) an up-to-date state of the research in this field and (ii) current EHL FVIII products, including recently approved efanesoctocog alfa, for which the plasma half-life exceeds a biochemical barrier posed by von Willebrand factor, complexed with FVIII in plasma, which results in ~1 per week infusion frequency. We focus on the EHL FVIII products' structure and function, in particular related to the known discrepancy in results of one-stage clotting (OC) and chromogenic substrate (CS) assays used to assign the products' potency, dosing, and for clinical monitoring in plasma. We suggest a possible root cause of these assays' discrepancy that is also pertinent to EHL factor IX variants used to treat hemophilia B. Finally, we discuss approaches in designing future EHL FVIII variants, including those to be used for hemophilia A gene therapy.
Collapse
Affiliation(s)
- Andrey G Sarafanov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
10
|
Lunghi B, Morfini M, Martinelli N, Branchini A, Linari S, Castaman G, Bernardi F. Modulation of factor VIII pharmacokinetics by genetic components in factor VIII receptors. Haemophilia 2023; 29:479-487. [PMID: 36533781 DOI: 10.1111/hae.14722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Gene variation in receptors for circulating factor VIII (FVIII) is candidate to explain the large inter-patient variability of infused FVIII pharmacokinetics (PK) in haemophilia A (HA). AIM To compare in an Italian HA cohort (n = 26) the influence on FVIII PK of genetic components in four von Willebrand factor (VWF)/FVIII receptors. METHODS Genotypes of low-density lipoprotein receptor (LDLR), asialoglycoprotein receptor minor subunit (ASGR2), family 4 member M (CLEC4M), stabilin2 (STAB2) and ABO blood-group, and VWF:Ag levels were included as independent variables in linear regression analyses of two-compartment model (TCM) - standard half-life (SHL) FVIII PK parameters. RESULTS In the initial FVIII distribution phase, the STAB2 rs4981022 AA, ASGR2 rs2289645 TT and LDLR rs688 TT genotypes may contribute to increase Cmax , and prolong or shorten AlphaHL. In the elimination phase, a shorter BetaHL was associated with the CLEC4M rs868875 GG (beta-coefficient .366, p = .025) and ASGR2 rs2289645 TC (beta-coefficient .456, p = .006) genotypes, which also showed shorter mean residence time (MRT) than TT genotypes (p = .021). The alpha and beta phase effects were independent of ABO and VWF:Ag levels at baseline. The association of the LDLR rs2228671 genotypes with clearance was independent of ABO (beta-coefficient -.363, p = .035) but not of other receptors or VWF:Ag, which may point out multiple and competing interactions. CONCLUSIONS With the limitation of the small number of HA patients, these observations highlight multiple genetic components acting in distinct phases of FVIII PK and contributing to explain FVIII PK variability. This analysis provides candidates for genotype-based, individual tailoring of FVIII substitutive treatment.
Collapse
Affiliation(s)
- Barbara Lunghi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Massimo Morfini
- Italian Association of Hemophilia Centers (AICE), Naples, Italy
| | | | - Alessio Branchini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Silvia Linari
- Center for Bleeding Disorders, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - Giancarlo Castaman
- Center for Bleeding Disorders, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
11
|
Ward SE, O'Sullivan JM, Moran AB, Spencer DIR, Gardner RA, Sharma J, Fazavana J, Monopoli M, McKinnon TAJ, Chion A, Haberichter S, O'Donnell JS. Sialylation on O-linked glycans protects von Willebrand factor from macrophage galactose lectin-mediated clearance. Haematologica 2022; 107:668-679. [PMID: 33763999 PMCID: PMC8883566 DOI: 10.3324/haematol.2020.274720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Terminal sialylation determines the plasma half-life of von Willebrand factor (VWF). A role for macrophage galactose lectin (MGL) in regulating hyposialylated VWF clearance has recently been proposed. In this study, we showed that MGL influences physiological plasma VWF clearance. MGL inhibition was associated with a significantly extended mean residence time and 3-fold increase in endogenous plasma VWF antigen levels (P<0.05). Using a series of VWF truncations, we further demonstrated that the A1 domain of VWF is predominantly responsible for enabling the MGL interaction. Binding of both full-length and VWF-A1-A2-A3 to MGL was significantly enhanced in the presence of ristocetin (P<0.05), suggesting that the MGL-binding site in A1 is not fully accessible in globular VWF. Additional studies using different VWF glycoforms demonstrated that VWF O-linked glycans, clustered at either end of the A1 domain, play a key role in protecting VWF against MGLmediated clearance. Reduced sialylation has been associated with pathological, increased clearance of VWF in patients with von Willebrand disease. Herein, we demonstrate that specific loss of α2-3 linked sialylation from O-glycans results in markedly increased MGL-binding in vitro, and markedly enhanced MGL-mediated clearance of VWF in vivo. Our data further show that the asialoglycoprotein receptor (ASGPR) does not have a significant role in mediating the increased clearance of VWF following loss of O-sialylation. Conversely however, we observed that loss of N-linked sialylation from VWF drives enhanced circulatory clearance predominantly via the ASGPR. Collectively, our data support the hypothesis that in addition to regulating physiological VWF clearance, the MGL receptor works in tandem with ASGPR to modulate enhanced clearance of aberrantly sialylated VWF in the pathogenesis of von Willebrand disease.
Collapse
Affiliation(s)
- Soracha E Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Alan B Moran
- Ludger, Ltd., Culham Science Centre, Abingdon, Oxfordshire OX14 3EB, United Kingdom; Leiden University Medical Centre, Centre for Proteomics and Metabolomics, 2300 RC Leiden
| | | | | | - Jyotika Sharma
- Department of Basic Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota
| | - Judicael Fazavana
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Marco Monopoli
- Department of Chemistry, RCSI, 123 St. Stephen's Green, Dublin 2
| | - Thomas A J McKinnon
- Faculty of Medicine, Imperial College, Hammersmith Hospital, Ducane Road, London
| | - Alain Chion
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | | | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; National Coagulation Centre, St James's Hospital, Dublin.
| |
Collapse
|
12
|
Combination of CLEC4M rs868875 G-Carriership and ABO O Genotypes May Predict Faster Decay of FVIII Infused in Hemophilia A Patients. J Clin Med 2022; 11:jcm11030733. [PMID: 35160186 PMCID: PMC8837058 DOI: 10.3390/jcm11030733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 11/26/2022] Open
Abstract
The C-type lectin CLEC4M binds and internalizes factor VIII (FVIII). Common CLEC4M variants have been associated with FVIII pharmacokinetic (PK) profiles in hemophilia A (HA) patients. The two-compartment PK analysis of plasma-derived (pd-) and full length recombinant FVIII concentrates was conducted in twenty-six patients (FVIII:C ≤ 2 IU/dL). F8, ABO blood-groups, and the CLEC4M rs868875A/G polymorphism were genotyped. CLEC4M genotype groups differed for the elimination rate constant K 1-0 (p < 0.001), half-life (K 1-0 HL), and the Beta rate constant. Patients treated with pd-FVIII also differed in the Alpha phase. In linear regression models, the contribution of the CLEC4M genotypes to FVIII PK parameters remained significant after correction for ABO, age, and VWF antigen levels at PK. Combined CLEC4M rs868875A/G and ABO genotypes displayed significant interaction (K 1-0, p = 0.014). Compared to other combined genotypes, the G-carriers/O genotypes showed half-reduced K 1-0 HL (p = 0.008), and faster FVIII clearance (mean 7.1 ± 2.2 mL/h/kg SE) than in the G-carriers/non-O (mean 2.4 ± 0.3 mL/h/kg SE), (p = 0.038). Comparison in HA patients recruited in several countries suggests that CLEC4M genotypes coherently influence infused FVIII half-life and clearance. Our analysis supports substantially faster FVIII decay associated with the rs868875 G-carrier/ABO O genotypes, which has potential implications for genetically tailored substitutive HA treatment.
Collapse
|
13
|
Voos KM, Cao W, Arce NA, Legan ER, Wang Y, Shajahan A, Azadi P, Lollar P, Zhang XF, Li R. Desialylation of O-glycans activates von Willebrand factor by destabilizing its autoinhibitory module. J Thromb Haemost 2022; 20:196-207. [PMID: 34529349 PMCID: PMC9134874 DOI: 10.1111/jth.15528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/15/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The binding of the A1 domain of von Willebrand factor (VWF) to platelet receptor glycoprotein (GP)Ibα defines the VWF activity in hemostasis. Recent studies suggest that sequences flanking A1 form cooperatively an autoinhibitory module (AIM) that reduces the accessibility of the GPIbα binding site on A1. Application of a tensile force induces unfolding of the AIM. Desialylation induces spontaneous binding of plasma VWF to platelets. Most O-glycans in VWF are located around the A1 domain. Removing certain O-glycans in the flanking sequences by site-directed mutagenesis enhances A1 binding to GPIbα and produces an effect similar to type 2B von Willebrand disease in animals. OBJECTIVES To understand if and how desialylation of O-glycans in the flanking sequences increases A1 activity. METHODS A recombinant AIM-A1 fragment encompassing VWF residues 1238-1493 and only O-glycans was treated with neuraminidase to produce desialylated protein. The glycan structure, dynamics, stability, and function of the desialylated protein was characterized by biochemical and biophysical methods and compared to the sialylated fragment. RESULTS Asialo-AIM-A1 exhibited increased binding activity and induced more apparent platelet aggregation than its sialylated counterpart. It exhibited a lower melting temperature, and increased hydrogen-deuterium exchange rates at residues near the secondary GPIbα binding site and the N-terminal flanking sequence. Asialo-AIM-A1 is less mechanically stable than sialo-AIM-A1, with its unstressed unfolding rate approximately 3-fold greater than the latter. CONCLUSIONS Desialylation of O-glycans around A1 increases its activity by destabilizing the AIM.
Collapse
Affiliation(s)
- Kayleigh M. Voos
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Wenpeng Cao
- Department of Bioengineering, Lehigh University, Bethlehem, PA
| | - Nicholas A. Arce
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Emily R. Legan
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Yingchun Wang
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Pete Lollar
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - X. Frank Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
14
|
Avdonin PP, Tsvetaeva NV, Goncharov NV, Rybakova EY, Trufanov SK, Tsitrina AA, Avdonin PV. Von Willebrand Factor in Health and Disease. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2021. [DOI: 10.1134/s1990747821040036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Abstract—
Von Willebrand factor (vWF), the key component of hemostasis, is synthesized in endothelial cells and megakaryocytes and released into the blood as high molecular weight multimeric glycoproteins weighing up to 20 million Daltons. Blood plasma metalloprotease ADAMTS13 cleaves ultra-large vWF multimers to smaller multimeric and oligomeric molecules. The vWF molecules attach to the sites of damage at the surface of arterioles and capillaries and unfold under conditions of shear stress. On the unfolded vWF molecule, the regions interacting with receptors on the platelet membrane are exposed. After binding to the vWF filaments, platelets are activated; platelets circulating in the vessels are additionally attached to them, leading to thrombus formation, blocking of microvessels, and cessation of bleeding. This review describes the history of the discovery of vWF, presents data on the mechanisms of vWF secretion and its structure, and characterizes the processes of vWF metabolism in the body under normal and pathological conditions.
Collapse
|
15
|
Ward S, O'Sullivan JM, O'Donnell JS. The Biological Significance of von Willebrand Factor O-Linked Glycosylation. Semin Thromb Hemost 2021; 47:855-861. [PMID: 34130346 DOI: 10.1055/s-0041-1726373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glycosylation is a key posttranslational modification, known to occur on more than half of all secreted proteins in man. As such, the role of N- and O-linked glycan structures in modulating various aspects of protein biology is an area of much research. Given their prevalence, it is perhaps unsurprising that variations in glycan structures have been demonstrated to play critical roles in modulating protein function and have been implicated in the pathophysiology of human diseases. von Willebrand factor (VWF), a plasma glycoprotein that is essential for normal hemostasis, is heavily glycosylated, containing 13 N-linked and 10 O-linked glycans. Together, these carbohydrate chains account for 20% of VWF monomeric mass, and have been shown to modulate VWF structure, function, and half-life. In this review, we focus on the specific role played by O-linked glycans in modulating VWF biology. Specifically, VWF O-linked glycans have been shown to modulate tertiary protein structure, susceptibility to ADAMTS13 proteolysis, platelet tethering, and VWF circulatory half-life.
Collapse
Affiliation(s)
- Soracha Ward
- Haemostasis Research Group, Irish Centre for Vascular Biology, School of Pharmacy and Bimolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jamie M O'Sullivan
- Haemostasis Research Group, Irish Centre for Vascular Biology, School of Pharmacy and Bimolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - James S O'Donnell
- Haemostasis Research Group, Irish Centre for Vascular Biology, School of Pharmacy and Bimolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Coagulation Centre, St James's Hospital, Dublin, Ireland
| |
Collapse
|
16
|
The relationship between ABO blood group, von Willebrand factor, and primary hemostasis. Blood 2021; 136:2864-2874. [PMID: 32785650 DOI: 10.1182/blood.2020005843] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
Numerous studies have reported significant associations between ABO blood group and risk of cardiovascular disease. These studies have consistently demonstrated that thrombotic risk is significantly reduced in individuals in blood group O. Nevertheless, the biological mechanisms through which ABO influences hemostasis have remained poorly understood. Exciting recent data have provided novel insights into how these ABO effects are modulated and have highlighted that ABO group significantly influences platelet plug formation at sites of vascular injury (primary hemostasis). In particular, ABO affects multiple aspects of von Willebrand factor (VWF) biology. In keeping with their reduced thrombotic risk, plasma VWF levels are ∼25% lower in healthy group O compared with healthy group non-O individuals. In addition, blood group O VWF demonstrates enhanced susceptibility to ADAMTS13 proteolysis. Finally, preliminary findings suggest that the interaction of group O VWF with platelets may also be reduced. Although the molecular mechanisms underlying these ABO effects on VWF have not been fully elucidated, it seems likely that they are mediated in large part by the ABO(H) carbohydrate structures that are carried on both the N- and O-linked glycans of VWF. Interestingly, ABO(H) determinants are also expressed on several different platelet surface glycoprotein receptors. Recent studies support the hypothesis that ABO group not only exerts major quantitative and qualitative effects on VWF, but also affect specific aspects of platelet function. Given the severe morbidity and the mortality associated with thrombotic disorders, defining the mechanisms underlying these ABO effects is not only of scientific interest, but also of direct clinical importance.
Collapse
|
17
|
Rawley O, Lillicrap D. Functional Roles of the von Willebrand Factor Propeptide. Hamostaseologie 2021; 41:63-68. [PMID: 33588457 DOI: 10.1055/a-1334-8002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The primary polypeptide sequence of von Willebrand factor (VWF) includes an N-terminal 741-amino acid VWF propeptide (VWFpp). In cells expressing VWF, the VWFpp performs two critical functions. In the Golgi, VWFpp mediates the intermolecular disulfide linkages that generate high-molecular-weight VWF multimers. Subsequently, the VWFpp, which is proteolytically cleaved from mature VWF by furin, functions to generate the endothelial storage organelles (Weibel-Palade bodies) in which VWF and a distinct collection of proteins are stored, and from where they undergo regulated secretion from the endothelium. The VWFpp is secreted from endothelial cells as dimers and circulates in plasma with at least some of the dimers associating with a noncovalent manner with the D'D3 domain of mature VWF. The VWFpp has a half-life of 2 to 3 hours in plasma, but to date no extracellular function has been determined for the molecule. Nevertheless, its large size and several biologically interesting structural features (two sets of vicinal cysteines and an RGD sequence) suggest that there may be roles that the VWFpp plays in hemostasis or associated physiological processes such as angiogenesis or wound repair.
Collapse
Affiliation(s)
- Orla Rawley
- Department of Pathology and Molecular Medicine, Richardson Laboratory, Queen's University, Kingston, Ontario, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Richardson Laboratory, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
18
|
O'Donghaile D, Jenkins PV, McGrath RT, Preston L, Field SP, Ward SE, O'Sullivan JM, O'Donnell JS. Expresser phenotype determines ABO(H) blood group antigen loading on platelets and von Willebrand factor. Sci Rep 2020; 10:18366. [PMID: 33110150 PMCID: PMC7591562 DOI: 10.1038/s41598-020-75462-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/15/2020] [Indexed: 01/22/2023] Open
Abstract
ABO blood group is associated with cardiovascular disease, with significantly lower risk in blood group O individuals. ABO(H) blood group determinants are expressed on different glycoproteins on platelet surfaces. In addition, ABO(H) structures are also present on VWF glycans. These ABO(H) carbohydrates influence both platelet and VWF function. Previous studies have reported that approximately 5-10% of normal blood donors express abnormally high or low levels of A or B blood group antigens on their platelet surfaces (high expresser phenotype, HXP or low expresser phenotype, LXP respectively). In this study, the biological effects of the ABO Expresser phenotype were investigated. ABO(H) expression on platelets and plasma VWF was studied in a series of 541 healthy blood donors. Overall, 5.6% of our study cohort were classified as HXP, whilst 4.4% satisfied criteria for LXP. We demonstrate that genotype at the ABO blood group locus plays a critical role in modulating the platelet HXP phenotype. In particular, A1A1 genotype is a major determinant of ABO high-expresser trait. Our data further show that ABH loading on VWF is also affected by ABO expresser phenotype. Consequently, A antigen expression on VWF was significantly elevated in HXP individuals and moderately reduced in LXP subjects (P < 0.05). Collectively, these findings suggest that ABO expresser phenotype influences primary hemostasis though several different pathways. Further studies will be required to define whether inter-individual variations in ABO(H) expression on platelets and/or VWF (particularly HXP and LXP) impact upon risk for cardiovascular disease.
Collapse
Affiliation(s)
- Diarmaid O'Donghaile
- Irish Blood Transfusion Service, Dublin, Ireland
- Department of Haematology, Trinity College Dublin, Dublin, Ireland
| | - P Vincent Jenkins
- Department of Haematology, University Hospital of Wales, Cardiff, Wales, UK
| | - Rachel T McGrath
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| | - Lisa Preston
- Cancer Molecular Diagnostics, Trinity Centre for Health Sciences, St James's Hospital, Dublin, Ireland
| | | | - Soracha E Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Ardilaun House 111 St Stephen's Green, Dublin 2, Ireland
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Ardilaun House 111 St Stephen's Green, Dublin 2, Ireland
| | - James S O'Donnell
- Department of Haematology, University Hospital of Wales, Cardiff, Wales, UK.
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Ardilaun House 111 St Stephen's Green, Dublin 2, Ireland.
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland.
| |
Collapse
|
19
|
Platelets and Defective N-Glycosylation. Int J Mol Sci 2020; 21:ijms21165630. [PMID: 32781578 PMCID: PMC7460655 DOI: 10.3390/ijms21165630] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
N-glycans are covalently linked to an asparagine residue in a simple acceptor sequence of proteins, called a sequon. This modification is important for protein folding, enhancing thermodynamic stability, and decreasing abnormal protein aggregation within the endoplasmic reticulum (ER), for the lifetime and for the subcellular localization of proteins besides other functions. Hypoglycosylation is the hallmark of a group of rare genetic diseases called congenital disorders of glycosylation (CDG). These diseases are due to defects in glycan synthesis, processing, and attachment to proteins and lipids, thereby modifying signaling functions and metabolic pathways. Defects in N-glycosylation and O-glycosylation constitute the largest CDG groups. Clotting and anticlotting factor defects as well as a tendency to thrombosis or bleeding have been described in CDG patients. However, N-glycosylation of platelet proteins has been poorly investigated in CDG. In this review, we highlight normal and deficient N-glycosylation of platelet-derived molecules and discuss the involvement of platelets in the congenital disorders of N-glycosylation.
Collapse
|
20
|
Identification of carbohydrate peripheral epitopes important for recognition by positive-ion MALDI multistage mass spectrometry. Carbohydr Polym 2020; 229:115528. [DOI: 10.1016/j.carbpol.2019.115528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/08/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
|
21
|
Tischer A, Machha VR, Moon-Tasson L, Benson LM, Auton M. Glycosylation sterically inhibits platelet adhesion to von Willebrand factor without altering intrinsic conformational dynamics. J Thromb Haemost 2020; 18:79-90. [PMID: 31479573 PMCID: PMC6940534 DOI: 10.1111/jth.14628] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND A molecular basis for von Willebrand factor (VWF) self-inhibition has been proposed by which the N-terminal and C-terminal flanking sequences of the globular A1 domain disulfide loop bind to and suppress the conformational dynamics of A1. These flanking sequences are rich in O-linked glycosylation (OLG), which is known to suppress platelet adhesion to VWF, presumably by steric hindrance. The inhibitory mechanism remains unresolved as to whether inhibition is due to steric exclusion by OLGs or a direct self-association interaction that stabilizes the domain. OBJECTIVES The platelet adhesive function, thermodynamic stability, and conformational dynamics of the wild-type and type 2M G1324S A1 domain lacking glycosylation (Escherichia coli) are compared with the wild-type glycosylated A1 domain (HEK293 cell culture) to decipher the self-inhibitory mechanism. METHODS Surface plasmon resonance and analytical rheology are utilized to assess Glycoprotein Ibα (GPIbα) binding at equilibrium and platelet adhesion under shear flow. The conformational stability is assessed through a combination of protein unfolding thermodynamics and hydrogen-deuterium exchange mass spectrometry (HXMS). RESULTS A1 glycosylation inhibits both GPIbα binding and platelet adhesion. Glycosylation increases the hydrodynamic size of A1 and stabilizes the thermal unfolding of A1 without changing its equilibrium stability. Glycosylation does not alter the intrinsic conformational dynamics of the A1 domain. CONCLUSIONS These studies invalidate the proposed inhibition through conformational suppression since glycosylation within these flanking sequences does not alter the native state stability or the conformational dynamics of A1. Rather, they confirm a mechanism by which glycosylation sterically hinders platelet adhesion to the A1 domain at equilibrium and under rheological shear stress.
Collapse
Affiliation(s)
- Alexander Tischer
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Venkata R. Machha
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Laurie Moon-Tasson
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Linda M. Benson
- Proteomics Core, Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, Minnesota, 55905, USA
| | - Matthew Auton
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, 55905, USA
| |
Collapse
|
22
|
Abstract
Von Willebrand factor (VWF) and coagulation factor VIII (FVIII) circulate as a complex in plasma and have a major role in the hemostatic system. VWF has a dual role in hemostasis. It promotes platelet adhesion by anchoring the platelets to the subendothelial matrix of damaged vessels and it protects FVIII from proteolytic degradation. Moreover, VWF is an acute phase protein that has multiple roles in vascular inflammation and is massively secreted from Weibel-Palade bodies upon endothelial cell activation. Activated FVIII on the other hand, together with coagulation factor IX forms the tenase complex, an essential feature of the propagation phase of coagulation on the surface of activated platelets. VWF deficiency, either quantitative or qualitative, results in von Willebrand disease (VWD), the most common bleeding disorder. The deficiency of FVIII is responsible for Hemophilia A, an X-linked bleeding disorder. Here, we provide an overview on the role of the VWF-FVIII interaction in vascular physiology.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany.
| |
Collapse
|
23
|
Genetic determinants of VWF clearance and FVIII binding modify FVIII pharmacokinetics in pediatric hemophilia A patients. Blood 2019; 134:880-891. [DOI: 10.1182/blood.2019000190] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Factor VIII (FVIII) pharmacokinetic (PK) properties show high interpatient variability in hemophilia A patients. Although previous studies have determined that age, body mass index, von Willebrand factor antigen (VWF:Ag) levels, and ABO blood group status can influence FVIII PK, they do not account for all observed variability. In this study, we aim to describe the genetic determinants that modify the FVIII PK profile in a population of 43 pediatric hemophilia A patients. We observed that VWF:Ag and VWF propeptide (VWFpp)/VWF:Ag, but not VWFpp, were associated with FVIII half-life. VWFpp/VWF:Ag negatively correlated with FVIII half-life in patients with non-O blood type, but no correlation was observed for type O patients, suggesting that von Willebrand factor (VWF) half-life, as modified by the ABO blood group, is a strong regulator of FVIII PK. The FVIII-binding activity of VWF positively correlated with FVIII half-life, and the rare or low-frequency nonsynonymous VWF variants p.(Arg826Lys) and p.(Arg852Glu) were identified in patients with reduced VWF:FVIIIB but not VWF:Ag. Common variants at the VWF, CLEC4M, and STAB2 loci, which have been previously associated with plasma levels of VWF and FVIII, were associated with the FVIII PK profile. Together, these studies characterize the mechanistic basis by which VWF clearance and ABO glycosylation modify FVIII PK in a pediatric population. Moreover, this study is the first to identify non-VWF and non-ABO variants that modify FVIII PK in pediatric hemophilia A patients.
Collapse
|
24
|
Ward S, O'Sullivan JM, O'Donnell JS. von Willebrand factor sialylation-A critical regulator of biological function. J Thromb Haemost 2019; 17:1018-1029. [PMID: 31055873 DOI: 10.1111/jth.14471] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/24/2019] [Indexed: 12/29/2022]
Abstract
Essentials Von Willebrand Factor (VWF) is extensively glycosylated with serial studies demonstrating that these carbohydrate determinants play critical roles in regulating multiple aspects of VWF biology. Terminal sialic acid residues, expressed on both the N- and O-linked glycans of VWF, regulate VWF functional activity, susceptibility to proteolysis and plasma clearance in vivo. Quantitative and qualitative variations in VWF sialylation have been reported in patients with von Willebrand Disease, as well as in a number of other physiological and pathological states. Further studies are warranted to define the molecular mechanisms through which N- and O-linked sialylation impacts upon the multiple biological activities of VWF. von Willebrand factor (VWF) undergoes complex post-translational modification prior to its secretion into the plasma. Consequently, VWF monomers contain complex N-glycan and O-glycan structures that, together, account for approximately 20% of the final monomeric mass. An increasing body of evidence has confirmed that these carbohydrate determinants play critical roles in regulating multiple aspects of VWF biology. In particular, studies have demonstrated that terminal ABO blood group has an important effect on plasma VWF levels. This effect is interesting, given that only 15% of the N-glycans and 1% of the O-glycans of VWF actually express terminal ABO(H) determinants. In contrast, the vast majority of the N-glycans and O-glycans on human VWF are capped by terminal negatively charged sialic acid residues. Recent data suggest that sialylation significantly regulates VWF functional activity, susceptibility to proteolysis, and clearance, through a number of independent pathways. These findings are of direct clinical relevence, in that quantitative and qualitative variations in VWF sialylation have been described in patients with VWD, as well as in patients with a number of other physiologic and pathologic conditions. Moreover, platelet-derived VWF is significantly hyposialylated as compared with plasma-derived VWF, whereas the recently licensed recombinant VWF therapeutic is hypersialylated. In this review, we examine the evidence supporting the hypothesis that VWF sialylation plays multiple biological roles. In addition, we consider data suggesting that quantitative and qualitative variations in VWF sialylation may play specific roles in the pathogenesis of VWD, and that sialic acid expression on VWF may also differ across a number of other physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Soracha Ward
- Haemostasis Research Group, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jamie M O'Sullivan
- Haemostasis Research Group, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - James S O'Donnell
- Haemostasis Research Group, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Coagulation Centre, St James's Hospital, Dublin, Ireland
| |
Collapse
|
25
|
De novo glycan structural identification from mass spectra using tree merging strategy. Comput Biol Chem 2019; 80:217-224. [DOI: 10.1016/j.compbiolchem.2019.03.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 03/23/2019] [Indexed: 11/19/2022]
|
26
|
Stowell SR, Stowell CP. Biologic roles of the ABH and Lewis histo-blood group antigens part II: thrombosis, cardiovascular disease and metabolism. Vox Sang 2019; 114:535-552. [PMID: 31090093 DOI: 10.1111/vox.12786] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
The ABH and Lewis antigens were among the first of the human red blood cell polymorphisms to be identified and, in the case of the former, play a dominant role in transfusion and transplantation. But these two therapies are largely twentieth-century innovations, and the ABH and related carbohydrate antigens are not only expressed on a very wide range of human tissues, but were present in primates long before modern humans evolved. Although we have learned a great deal about the biochemistry and genetics of these structures, the biological roles that they play in human health and disease are incompletely understood. This review and its companion, which appeared in a previous issue of Vox Sanguinis, will focus on a few of the biologic and pathologic processes which appear to be affected by histo-blood group phenotype. The first of the two reviews explored the interactions of two bacteria with the ABH and Lewis glycoconjugates of their human host cells, and described the possible connections between the immune response of the human host to infection and the development of the AB-isoagglutinins. This second review will describe the relationship between ABO phenotype and thromboembolic disease, cardiovascular disease states, and general metabolism.
Collapse
Affiliation(s)
- Sean R Stowell
- Center for Apheresis, Center for Transfusion and Cellular Therapies, Emory Hospital, Emory University School of Medicine, Atlanta, GA, USA.,Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher P Stowell
- Blood Transfusion Service, Massachusetts General Hospital, Boston, MA, USA.,Department of Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Increased galactose expression and enhanced clearance in patients with low von Willebrand factor. Blood 2019; 133:1585-1596. [DOI: 10.1182/blood-2018-09-874636] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/06/2019] [Indexed: 11/20/2022] Open
Abstract
Abstract
Glycan determinants on von Willebrand factor (VWF) play critical roles in regulating its susceptibility to proteolysis and clearance. Abnormal glycosylation has been shown to cause von Willebrand disease (VWD) in a number of different mouse models. However, because of the significant technical challenges associated with accurate assessment of VWF glycan composition, the importance of carbohydrates in human VWD pathogenesis remains largely unexplored. To address this, we developed a novel lectin-binding panel to enable human VWF glycan characterization. This methodology was then used to study glycan expression in a cohort of 110 patients with low VWF compared with O blood group-matched healthy controls. Interestingly, significant interindividual heterogeneity in VWF glycan expression was seen in the healthy control population. This variation included terminal sialylation and ABO(H) blood group expression on VWF. Importantly, we also observed evidence of aberrant glycosylation in a subgroup of patients with low VWF. In particular, terminal α(2-6)-linked sialylation was reduced in patients with low VWF, with a secondary increase in galactose (Gal) exposure. Furthermore, an inverse correlation between Gal exposure and estimated VWF half-life was observed in those patients with enhanced VWF clearance. Together, these findings support the hypothesis that loss of terminal sialylation contributes to the pathophysiology underpinning low VWF in at least a subgroup of patients by promoting enhanced clearance. In addition, alterations in VWF carbohydrate expression are likely to contribute to quantitative and qualitative variations in VWF levels in the normal population. This trial was registered at www.clinicaltrials.gov as #NCT03167320.
Collapse
|
28
|
Swystun LL, Notley C, Georgescu I, Lai JD, Nesbitt K, James PD, Lillicrap D. The endothelial lectin clearance receptor CLEC4M binds and internalizes factor VIII in a VWF-dependent and independent manner. J Thromb Haemost 2019; 17:681-694. [PMID: 30740857 PMCID: PMC7083068 DOI: 10.1111/jth.14404] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Indexed: 01/23/2023]
Abstract
Essentials CLEC4M is an endocytic receptor for factor FVIII. CLEC4M interacts with FVIII in a VWF-dependent and independent manner. CLEC4M binds to mannose-containing glycans on FVIII. CLEC4M internalization of FVIII involves clathrin coated pits. SUMMARY: Background von Willebrand factor (VWF) and factor VIII (FVIII) circulate in the plasma as a non-covalent complex, and the majority of FVIII is likely to be cleared by VWF-dependent pathways. Clearance of VWF-free FVIII is rapid and underlies the pathological basis of some quantitative FVIII deficiencies. The receptor pathways that regulate the clearance of VWF-bound and VWF-free FVIII are incompletely uncharacterized. The human liver-expressed endothelial lectin CLEC4M has been previously characterized as a clearance receptor for VWF, although its influence on FVIII is unknown. Objective The interaction between FVIII and CLEC4M was characterized in the presence or absence of VWF. Methods FVIII interactions with CLEC4M were evaluated by in vitro cell-based and solid phase binding assays. Interactions between FVIII and CLEC4M or liver sinusoidal endothelial cells were evaluated in vivo by immunohistochemistry. Results CLEC4M-expressing HEK 293 cells bound and internalized recombinant and plasma-derived FVIII through VWF-dependent and independent mechanisms. CLEC4M binding to recombinant FVIII was dependent on mannose-exposed N-linked glycans. CLEC4M mediated FVIII internalization via a clathrin-coated pit-dependent mechanism, resulting in transport of FVIII from early and late endosomes for catabolism by lysosomes. In vivo hepatic expression of CLEC4M after hydrodynamic liver transfer was associated with a decrease in plasma levels of endogenous murine FVIII:C in normal mice, whereas infused recombinant human FVIII was associated with sinusoidal endothelial cells in the presence or absence of VWF. Conclusions These findings suggest that CLEC4M is a novel clearance receptor that interacts with mannose-exposed glycans on FVIII in the presence or absence of VWF.
Collapse
Affiliation(s)
- Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Colleen Notley
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Ilinca Georgescu
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Jesse D Lai
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Kate Nesbitt
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Paula D James
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
29
|
Sun D, Hu F, Gao H, Song Z, Xie W, Wang P, Shi L, Wang K, Li Y, Huang C, Li Z. Distribution of abnormal IgG glycosylation patterns from rheumatoid arthritis and osteoarthritis patients by MALDI-TOF-MSn. Analyst 2019; 144:2042-2051. [PMID: 30714583 DOI: 10.1039/c8an02014k] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
IgG glycosylation differs in rheumatoid arthritis (RA) and osteoarthritis (OA), which should contribute to their pathogenesis research and diagnosis.
Collapse
|
30
|
Swystun LL, Lillicrap D. Genetic regulation of plasma von Willebrand factor levels in health and disease. J Thromb Haemost 2018; 16:2375-2390. [PMID: 30246494 PMCID: PMC7147242 DOI: 10.1111/jth.14304] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Indexed: 02/06/2023]
Abstract
Plasma levels of the multimeric glycoprotein von Willebrand factor (VWF) constitute a complex quantitative trait with a continuous distribution and wide range in the normal population (50-200%). Quantitative deficiencies of VWF (< 50%) are associated with an increased risk of bleeding, whereas high plasma levels of VWF (> 150%) influence the risk of arterial and venous thromboembolism. Although environmental factors can strongly influence plasma VWF levels, it is estimated that approximately 65% of this variability is heritable. Interestingly, although variability in VWF can account for ~ 5% of the genetic influence on plasma VWF levels, other genetic loci also strongly modify plasma VWF levels. The identification of the additional sources of VWF heritability has been the focus of recent observational trait-mapping studies, including genome-wide association studies or linkage analyses, as well as hypothesis-driven research studies. Quantitative trait loci influencing VWF glycosylation, secretion and clearance have been associated with plasma VWF antigen levels in normal individuals, and may contribute to quantitative VWF abnormalities in patients with a thrombotic tendency or type 1 von Willebrand disease (VWD). The identification of genetic modifiers of plasma VWF levels may allow for better molecular diagnosis of type 1 VWD, and enable the identification of individuals at increased risk for thrombosis. Validation of trait-mapping studies with in vitro and in vivo methodologies has led to novel insights into the life cycle of VWF and the pathogenesis of quantitative VWF abnormalities.
Collapse
Affiliation(s)
- L L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - D Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
31
|
Richards E, Bouché L, Panico M, Arbeloa A, Vinogradov E, Morris H, Wren B, Logan SM, Dell A, Fairweather NF. The S-layer protein of a Clostridium difficile SLCT-11 strain displays a complex glycan required for normal cell growth and morphology. J Biol Chem 2018; 293:18123-18137. [PMID: 30275012 PMCID: PMC6254364 DOI: 10.1074/jbc.ra118.004530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/27/2018] [Indexed: 12/16/2022] Open
Abstract
Clostridium difficile is a bacterial pathogen that causes major health challenges worldwide. It has a well-characterized surface (S)-layer, a para-crystalline proteinaceous layer surrounding the cell wall. In many bacterial and archaeal species, the S-layer is glycosylated, but no such modifications have been demonstrated in C. difficile. Here, we show that a C. difficile strain of S-layer cassette type 11, Ox247, has a complex glycan attached via an O-linkage to Thr-38 of the S-layer low-molecular-weight subunit. Using MS and NMR, we fully characterized this glycan. We present evidence that it is composed of three domains: (i) a core peptide-linked tetrasaccharide with the sequence -4-α-Rha-3-α-Rha-3-α-Rha-3-β-Gal-peptide; (ii) a repeating pentasaccharide with the sequence -4-β-Rha-4-α-Glc-3-β-Rha-4-(α-Rib-3-)β-Rha-; and (iii) a nonreducing end-terminal 2,3 cyclophosphoryl-rhamnose attached to a ribose-branched sub-terminal rhamnose residue. The Ox247 genome contains a 24-kb locus containing genes for synthesis and protein attachment of this glycan. Mutations in genes within this locus altered or completely abrogated formation of this glycan, and their phenotypes suggested that this S-layer modification may affect sporulation, cell length, and biofilm formation of C. difficile In summary, our findings indicate that the S-layer protein of SLCT-11 strains displays a complex glycan and suggest that this glycan is required for C. difficile sporulation and control of cell shape, a discovery with implications for the development of antimicrobials targeting the S-layer.
Collapse
Affiliation(s)
- Emma Richards
- From the Department of Life Sciences, Imperial College London, SW7 2AZ London, United Kingdom
| | - Laura Bouché
- From the Department of Life Sciences, Imperial College London, SW7 2AZ London, United Kingdom
| | - Maria Panico
- From the Department of Life Sciences, Imperial College London, SW7 2AZ London, United Kingdom
| | - Ana Arbeloa
- From the Department of Life Sciences, Imperial College London, SW7 2AZ London, United Kingdom
| | - Evgeny Vinogradov
- the Vaccine Program, Human Health Therapeutics Research Centre, National Research Council, Ottawa, Ontario K1A 0R6, Canada
| | - Howard Morris
- From the Department of Life Sciences, Imperial College London, SW7 2AZ London, United Kingdom,; Biopharmaspec, Suite 3.1, Lido Medical Centre, St. Saviours Road, JE2 7LA Jersey, United Kingdom, and
| | - Brendan Wren
- the London School of Hygiene and Tropical Medicine, WC1E 7HT, London, United Kingdom
| | - Susan M Logan
- the Vaccine Program, Human Health Therapeutics Research Centre, National Research Council, Ottawa, Ontario K1A 0R6, Canada
| | - Anne Dell
- From the Department of Life Sciences, Imperial College London, SW7 2AZ London, United Kingdom,.
| | - Neil F Fairweather
- From the Department of Life Sciences, Imperial College London, SW7 2AZ London, United Kingdom,.
| |
Collapse
|
32
|
O'Sullivan JM, Ward S, Lavin M, O'Donnell JS. von Willebrand factor clearance - biological mechanisms and clinical significance. Br J Haematol 2018; 183:185-195. [DOI: 10.1111/bjh.15565] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jamie M. O'Sullivan
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Soracha Ward
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Michelle Lavin
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
- National Coagulation Centre; St James's Hospital; Dublin Ireland
| | - James S. O'Donnell
- Haemostasis Research Group; Irish Centre for Vascular Biology; Royal College of Surgeons in Ireland; Dublin Ireland
- National Coagulation Centre; St James's Hospital; Dublin Ireland
| |
Collapse
|
33
|
Dong C, Lee J, Kim S, Lai W, Webb EB, Oztekin A, Zhang XF, Im W. Long-ranged Protein-glycan Interactions Stabilize von Willebrand Factor A2 Domain from Mechanical Unfolding. Sci Rep 2018; 8:16017. [PMID: 30375453 PMCID: PMC6207679 DOI: 10.1038/s41598-018-34374-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022] Open
Abstract
von Willebrand Factor (vWF) is a large multimeric protein that binds to platelets and collagen in blood clotting. vWF A2 domain hosts a proteolytic site for ADAMTS13 (A Disintegrin and Metalloprotease with a ThromboSpondin type 1 motif, member 13) to regulate the size of vWF multimers. This regulation process is highly sensitive to force conditions and protein-glycan interactions as the process occurs in flowing blood. There are two sites on A2 domain (N1515 and N1574) bearing various N-linked glycan structures. In this study, we used molecular dynamics (MD) simulation to study the force-induced unfolding of A2 domain with and without a single N-linked glycan type on each site. The sequential pullout of β-strands was used to represent a characteristic unfolding sequence of A2. This unfolding sequence varied due to protein-glycan interactions. The force-extension and total energy-extension profiles also show differences in magnitude but similar characteristic shapes between the systems with and without glycans. Systems with N-linked glycans encountered higher energy barriers for full unfolding and even for unfolding up to the point of ADAMTS13 cleavage site exposure. Interestingly, there is not much difference observed for A2 domain structure itself with and without glycans from standard MD simulations, suggesting roles of N-glycans in A2 unfolding through long-ranged protein-glycan interactions.
Collapse
Affiliation(s)
- Chuqiao Dong
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA, 18015, United States
| | - Jumin Lee
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, 18015, United States
| | - Seonghoon Kim
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, 18015, United States
| | - Whitney Lai
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, United States
| | - Edmund B Webb
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA, 18015, United States
| | - Alparslan Oztekin
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA, 18015, United States
| | - X Frank Zhang
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, PA, 18015, United States
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, United States
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, 18015, United States.
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, United States.
| |
Collapse
|
34
|
Canis K, Anzengruber J, Garenaux E, Feichtinger M, Benamara K, Scheiflinger F, Savoy LA, Reipert BM, Malisauskas M. In-depth comparison of N-glycosylation of human plasma-derived factor VIII and different recombinant products: from structure to clinical implications. J Thromb Haemost 2018; 16:S1538-7836(22)02223-1. [PMID: 29888865 DOI: 10.1111/jth.14204] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Indexed: 12/21/2022]
Abstract
Essentials Glycosylation heterogeneity of recombinant proteins affects pharmacokinetics and immunogenicity. N-glycomics/glycoproteomics of plasma-derived Factor VIII and 6 recombinant FVIIIs were compared. Depending on cell line, significant differences to plasma-derived FVIII were observed. Recombinant FVIIIs expressed distinct and immunologically relevant epitopes. SUMMARY Background/Objective Human factor VIII (FVIII) is a plasma glycoprotein, defects of which result in hemophilia A. Current substitution therapy uses FVIII products purified from human plasma or from various cell lines (recombinant FVIII) with different levels of B-domain deletion. Glycosylation is a post-translational protein modification in FVIII that has a substantial influence on its physical, functional and antigenic properties. Variation in glycosylation is likely to be the reason that FVIII products differ in their pharmacokinetics, pharmacodynamics and immunogenicity. However, the literature on FVIII glycosylation is inconsistent, preventing assembly into a coherent model. Seeking to better understand the glycosylation mechanisms underlying FVIII biology, we studied the N-glycosylation of human plasma-derived (pd)FVIII and six rFVIII products expressed in CHO, BHK or HEK cell lines. Methods FVIII samples were subjected to head-to-head detailed glycomic and glycoproteomic characterization using a combination of MALDI-MS and MS/MS, GC-MS and UPLC-UV-MSE technologies. Results/Conclusion The results of our study detail the N-glycan repertoire of pdFVIII to an unprecedented level, and for the first time, provide evidence of N-glycolylneuraminic acid (NeuGc) found on pdFVIII. Although site-specific glycosylation of rFVIII proved consistent with pdFVIII regardless of the expression system, the entire N-glycan content of each sample appeared significantly different. Although the proportion of biologically important epitopes common to all samples (i.e. sialylation and high-mannose) varied between samples, some recombinant products expressed distinct and immunologically relevant epitopes, such as LacdiNAc (LDN), fucosylated LacdiNAc (FucLDN), NeuGc, LewisX/Y and Galα1,3 Gal epitopes. rFVIII expressed in HEK cells showed the greatest glycomic differences to human pdFVIII.
Collapse
Affiliation(s)
- K Canis
- SGS M-Scan SA, Plan-le-Ouates, Switzerland
| | | | - E Garenaux
- SGS M-Scan SA, Plan-le-Ouates, Switzerland
| | | | - K Benamara
- Research & Development, Shire, Vienna, Austria
| | | | - L-A Savoy
- SGS M-Scan SA, Plan-le-Ouates, Switzerland
| | - B M Reipert
- Research & Development, Shire, Vienna, Austria
| | | |
Collapse
|
35
|
Harvey DJ, Struwe WB. Structural Studies of Fucosylated N-Glycans by Ion Mobility Mass Spectrometry and Collision-Induced Fragmentation of Negative Ions. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2018; 29:1179-1193. [PMID: 29790113 PMCID: PMC6003995 DOI: 10.1007/s13361-018-1950-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/16/2018] [Accepted: 03/16/2018] [Indexed: 05/03/2023]
Abstract
There is considerable potential for the use of ion mobility mass spectrometry in structural glycobiology due in large part to the gas-phase separation attributes not typically observed by orthogonal methods. Here, we evaluate the capability of traveling wave ion mobility combined with negative ion collision-induced dissociation to provide structural information on N-linked glycans containing multiple fucose residues forming the Lewisx and Lewisy epitopes. These epitopes are involved in processes such as cell-cell recognition and are important as cancer biomarkers. Specific information that could be obtained from the intact N-glycans by negative ion CID included the general topology of the glycan such as the presence or absence of a bisecting GlcNAc residue and the branching pattern of the triantennary glycans. Information on the location of the fucose residues was also readily obtainable from ions specific to each antenna. Some isobaric fragment ions produced prior to ion mobility could subsequently be separated and, in some cases, provided additional valuable structural information that was missing from the CID spectra alone. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- David J Harvey
- Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK.
| | - Weston B Struwe
- Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
| |
Collapse
|
36
|
Effects of plasma glycosyltransferase on the ABO(H) blood group antigens of human von Willebrand factor. Int J Hematol 2018; 108:139-144. [PMID: 29619625 DOI: 10.1007/s12185-018-2452-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/28/2018] [Accepted: 03/28/2018] [Indexed: 10/17/2022]
Abstract
Von Willebrand factor (VWF) is one of the plasma protein carrying ABO(H) blood group antigens, but the combining process of these antigens is not clear. In the present study, we examined whether plasma glycosyltransferase affects the blood group antigens on VWF. VWF expressing H-antigen (H-VWF) from blood group O and bovine serum albumin conjugated with H-antigen (H-BSA) were incubated with recombinant α1-3-N-acetylgalactosaminyltransferase (rA-transferase) and A-plasma with or without an additional UDP-GalNAc. Transformed antigens were detected by western blotting and ELISA, using an anti-A antibody. Both H-VWF and H-BSA acquired the A-antigen after incubation with rA-transferase and UDP-GalNAc. Incubation with A-plasma very weakly converted the H-antigen on BSA and VWF to A-antigen only in the presence of supplemented UDP-GalNAc. This conversion was enhanced on desialylation of H-VWF. These results indicate that sugar chains of plasma VWF can be modified by the external glycosyltransferase, but that plasma glycosyltransferase has no effect on the blood group antigens of VWF due to its low activity and the lack of donor sugars. Further, sialic acid residues of VWF may exert a protective effect against post-translational glycosylation. Our results clearly exclude the possibility that blood group antigens of VWF are constructed extracellularly in plasma.
Collapse
|
37
|
Sheng L, He Z, Liu Y, Ma M, Cai Z. Mass spectrometry characterization for N-glycosylation of immunoglobulin Y from hen egg yolk. Int J Biol Macromol 2018; 108:277-283. [DOI: 10.1016/j.ijbiomac.2017.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 01/26/2023]
|
38
|
Zhao C, Cooper DKC, Dai Y, Hara H, Cai Z, Mou L. The Sda and Cad glycan antigens and their glycosyltransferase, β1,4GalNAcT-II, in xenotransplantation. Xenotransplantation 2018; 25:e12386. [PMID: 29430727 DOI: 10.1111/xen.12386] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/19/2017] [Accepted: 01/12/2018] [Indexed: 12/21/2022]
Abstract
Antibody-mediated rejection is a barrier to the clinical application of xenotransplantation, and xenoantigens play an important role in this process. Early research suggested that N-acetyl-D-galactosamine (GalNAc) could serve as a potential xenoantigen. GalNAc is the immunodominant glycan of the Sda antigen. Recently, knockout of β1,4-N-acetylgalactosaminyltransferase 2 (β1,4GalNAcT-II) from the pig results in a decrease in binding of human serum antibodies to pig cells. It is believed that this is the result of the elimination of the GalNAc on the Sda antigen, which is catalyzed by the enzyme, β1,4GalNAcT-II. However, research into human blood group antigens suggests that only a small percentage (1%-2%) of people express anti-Sda antibodies directed to Sda antigen, and yet a majority appear to have antibodies directed to the products of pig B4GALNT2. Questions can therefore be asked as to (i) whether the comprehensive structure of the Sda antigen in humans, that is, the underlying sugar structure, is identical to the Sda antigen in pigs, (ii) whether the human anti-Sda antibody binds ubiquitously to pig cells, but not to human cells, and (iii) what role the Sda++ (also called Cad) antigen is playing in this discrepancy. We review what is known about these antigens and discuss the discrepancies that have been noted above.
Collapse
Affiliation(s)
- Chengjiang Zhao
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hidetaka Hara
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhiming Cai
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
39
|
Differential N-glycan patterns identified in lung adenocarcinoma by N-glycan profiling of formalin-fixed paraffin-embedded (FFPE) tissue sections. J Proteomics 2018; 172:1-10. [DOI: 10.1016/j.jprot.2017.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/08/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
|
40
|
A novel role for the macrophage galactose-type lectin receptor in mediating von Willebrand factor clearance. Blood 2017; 131:911-916. [PMID: 29282218 DOI: 10.1182/blood-2017-06-787853] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/12/2017] [Indexed: 01/22/2023] Open
Abstract
Previous studies have shown that loss of terminal sialic acid causes enhanced von Willebrand factor (VWF) clearance through the Ashwell-Morrell receptor (AMR). In this study, we investigated (1) the specific importance of N- vs O-linked sialic acid in protecting against VWF clearance and (2) whether additional receptors contribute to the reduced half-life of hyposialylated VWF. α2-3-linked sialic acid accounts for <20% of total sialic acid and is predominantly expressed on VWF O-glycans. Nevertheless, specific digestion with α2-3 neuraminidase (α2-3Neu-VWF) was sufficient to cause markedly enhanced VWF clearance. Interestingly, in vivo clearance experiments in dual VWF-/-/Asgr1-/- mice demonstrated enhanced clearance of α2-3Neu-VWF even in the absence of the AMR. The macrophage galactose-type lectin (MGL) is a C-type lectin that binds to glycoproteins expressing terminal N-acetylgalactosamine or galactose residues. Importantly, the markedly enhanced clearance of hyposialylated VWF in VWF-/-/Asgr1-/- mice was significantly attenuated in the presence of an anti-MGL inhibitory antibody. Furthermore, dose-dependent binding of human VWF to purified recombinant human MGL was confirmed using surface plasmon resonance. Additionally, plasma VWF:Ag levels were significantly elevated in MGL1-/- mice compared with controls. Collectively, these findings identify MGL as a novel macrophage receptor for VWF that significantly contributes to the clearance of both wild-type and hyposialylated VWF.
Collapse
|
41
|
Liu G, Cheng K, Lo CY, Li J, Qu J, Neelamegham S. A Comprehensive, Open-source Platform for Mass Spectrometry-based Glycoproteomics Data Analysis. Mol Cell Proteomics 2017; 16:2032-2047. [PMID: 28887379 DOI: 10.1074/mcp.m117.068239] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/23/2017] [Indexed: 12/12/2022] Open
Abstract
Glycosylation is among the most abundant and diverse protein post-translational modifications (PTMs) identified to date. The structural analysis of this PTM is challenging because of the diverse monosaccharides which are not conserved among organisms, the branched nature of glycans, their isomeric structures, and heterogeneity in the glycan distribution at a given site. Glycoproteomics experiments have adopted the traditional high-throughput LC-MSn proteomics workflow to analyze site-specific glycosylation. However, comprehensive computational platforms for data analyses are scarce. To address this limitation, we present a comprehensive, open-source, modular software for glycoproteomics data analysis called GlycoPAT (GlycoProteomics Analysis Toolbox; freely available from www.VirtualGlycome.org/glycopat). The program includes three major advances: (1) "SmallGlyPep," a minimal linear representation of glycopeptides for MSn data analysis. This format allows facile serial fragmentation of both the peptide backbone and PTM at one or more locations. (2) A novel scoring scheme based on calculation of the "Ensemble Score (ES)," a measure that scores and rank-orders MS/MS spectrum for N- and O-linked glycopeptides using cross-correlation and probability based analyses. (3) A false discovery rate (FDR) calculation scheme where decoy glycopeptides are created by simultaneously scrambling the amino acid sequence and by introducing artificial monosaccharides by perturbing the original sugar mass. Parallel computing facilities and user-friendly GUIs (Graphical User Interfaces) are also provided. GlycoPAT is used to catalogue site-specific glycosylation on simple glycoproteins, standard protein mixtures and human plasma cryoprecipitate samples in three common MS/MS fragmentation modes: CID, HCD and ETD. It is also used to identify 960 unique glycopeptides in cell lysates from prostate cancer cells. The results show that the simultaneous consideration of peptide and glycan fragmentation is necessary for high quality MSn spectrum annotation in CID and HCD fragmentation modes. Additionally, they confirm the suitability of GlycoPAT to analyze shotgun glycoproteomics data.
Collapse
Affiliation(s)
- Gang Liu
- From the ‡Chemical and Biological Engineering
| | - Kai Cheng
- From the ‡Chemical and Biological Engineering.,§Clinical & Translational Research Center
| | - Chi Y Lo
- From the ‡Chemical and Biological Engineering
| | - Jun Li
- ¶Pharmaceutical Sciences; and.,‖New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York
| | - Jun Qu
- ¶Pharmaceutical Sciences; and.,‖New York State Center for Excellence in Bioinformatics and Life Sciences, Buffalo, New York
| | - Sriram Neelamegham
- From the ‡Chemical and Biological Engineering; .,§Clinical & Translational Research Center
| |
Collapse
|
42
|
Gashash EA, Aloor A, Li D, Zhu H, Xu XQ, Xiao C, Zhang J, Parameswaran A, Song J, Ma C, Xiao W, Wang PG. An Insight into Glyco-Microheterogeneity of Plasma von Willebrand Factor by Mass Spectrometry. J Proteome Res 2017; 16:3348-3362. [PMID: 28696719 PMCID: PMC6309539 DOI: 10.1021/acs.jproteome.7b00359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human plasma von Willebrand Factor (VWF) plays essential roles in primary hemostasis in cooperation with other coagulations factors. There is ample indication that glycosylation affects many biological phases during the protein life cycle. However, comprehensive characterization of all probable N-glycosites simultaneous with O-glycosites is still not fully revealed. Thus, the intention of this exploration was to estimate the occupancy of all canonical N-glycosites besides simultaneous characterization of N- and O-glycoforms. An RP-LC-MS/MS system functionalized with CID and HCD tandem mass was utilized to analyze VWF. N-Glycosite occupancy varied along the protein backbone chain. Out of 257 HCD spectra, 181 characterized glycoforms were specified as either N- or O-glycosites. Sequential cleavage of glycosidic bonds along with Human Database mass matching have confirmed the glycoform structures. A total of 173 glycoforms represented most commonly biantennary and infrequently tri- and tetra-antennary N-glycans beside high mannose, hybrid, ABH antigen-terminated, and sulfated N-glycans. Many glycoforms were common across all N-sites. Noteworthy, previously unreported N-glycosites within domain D'(TIL'-E') showed glycosylation. Moreover, sialylated core 1 and core 2 O-glycans were detected on 2298T. Given subtle characterization of site-specific glycoforms, we can attain a profound understanding of the biological roles of VWF as well as facilitate the production of VWF-based therapeutics.
Collapse
Affiliation(s)
- Ebtesam A. Gashash
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
- Department of Chemistry, College of Science, Albaha University, Baljurashi, Albaha 65635, Saudi Arabia
| | - Arya Aloor
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Dong Li
- Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, 200065 Shanghai, China
| | - He Zhu
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Xiao-Qian Xu
- Department of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Cong Xiao
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Junping Zhang
- Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Aishwarya Parameswaran
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Jing Song
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Cheng Ma
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| | - Weidong Xiao
- Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Peng George Wang
- Center for Diagnostics & Therapeutics and Department of Chemistry, Georgia State University, Atlanta, Georgia 30302, United States
| |
Collapse
|
43
|
Hartholt RB, van Velzen AS, Peyron I, Ten Brinke A, Fijnvandraat K, Voorberg J. To serve and protect: The modulatory role of von Willebrand factor on factor VIII immunogenicity. Blood Rev 2017; 31:339-347. [PMID: 28716211 DOI: 10.1016/j.blre.2017.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/26/2017] [Accepted: 07/03/2017] [Indexed: 12/23/2022]
Abstract
Hemophilia A is a bleeding disorder characterized by the absence or dysfunction of blood coagulation factor VIII (FVIII). Patients are treated with regular infusions of FVIII concentrate. In response to treatment, approximately 30% of patients with severe hemophilia A develop inhibitory antibodies targeting FVIII. Both patient and treatment related risk factors for inhibitor development have been described. Multiple studies comparing the immunogenicity of recombinant and plasma-derived FVIII have yielded conflicting results. The randomized controlled SIPPET (Survey of Inhibitors in Plasma-Product Exposed Toddlers) trial demonstrated an increased risk of inhibitor development of recombinant FVIII when compared to von Willebrand factor (VWF)-containing plasma-derived FVIII. Presently, it is unclear which mechanism underlies the reduced immunogenicity of plasma-derived FVIII. In this review we address the potential role of VWF on FVIII immunogenicity and we discuss how VWF affects the immune recognition, processing and presentation of FVIII. We also briefly discuss the potential impact of glycan-composition on FVIII immunogenicity. It is well established that VWF shields the uptake of FVIII by antigen presenting cells. We have recently shown that VWF binds to the surface of dendritic cells. Here, we present a novel model in which surface bound FVIII-VWF complexes regulate the internalization of FVIII. Binding of FVIII to VWF is critically dependent on sulfation of Tyr1699 (HVGS numbering) in the light chain of FVIII. Incomplete sulfation of Tyr1699 has been suggested to occur in several recombinant FVIII products resulting in a loss of VWF binding. We hypothesize that this results in alternative pathways of FVIII internalization by antigen presenting cells which are not regulated by VWF. This hypothetical mechanism may explain the reduced immunogenicity of VWF containing plasma-derived FVIII concentrates as found in the SIPPET study.
Collapse
Affiliation(s)
- Robin B Hartholt
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Alice S van Velzen
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | - Ivan Peyron
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Anja Ten Brinke
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | - Karin Fijnvandraat
- Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands.
| | - Jan Voorberg
- Department of Plasma Proteins, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
44
|
Biological considerations of plasma-derived and recombinant factor VIII immunogenicity. Blood 2017; 129:3147-3154. [DOI: 10.1182/blood-2016-11-750885] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 04/19/2017] [Indexed: 02/06/2023] Open
Abstract
Abstract
In hemophilia A, the most severe complication of factor VIII (FVIII) replacement therapy involves the formation of FVIII neutralizing antibodies, also known as inhibitors, in 25% to 30% of patients. This adverse event is associated with a significant increase in morbidity and economic burden, thus highlighting the need to identify methods to limit FVIII immunogenicity. Inhibitor development is regulated by a complex balance of genetic factors, such as FVIII genotype, and environmental variables, such as coexistent inflammation. One of the hypothesized risk factors of inhibitor development is the source of the FVIII concentrate, which could be either recombinant or plasma derived. Differential immunogenicity of these concentrates has been documented in several recent epidemiologic studies, thus generating significant debate within the hemophilia treatment community. To date, these discussions have been unable to reach a consensus regarding how these outcomes might be integrated into enhancing clinical care. Moreover, the biological mechanistic explanations for the observed differences are poorly understood. In this article, we complement the existing epidemiologic investigations with an overview of the range of possible biochemical and immunologic mechanisms that may contribute to the different immune outcomes observed with plasma-derived and recombinant FVIII products.
Collapse
|
45
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2011-2012. MASS SPECTROMETRY REVIEWS 2017; 36:255-422. [PMID: 26270629 DOI: 10.1002/mas.21471] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 01/15/2015] [Indexed: 06/04/2023]
Abstract
This review is the seventh update of the original article published in 1999 on the application of MALDI mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2012. General aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, and fragmentation are covered in the first part of the review and applications to various structural types constitute the remainder. The main groups of compound are oligo- and poly-saccharides, glycoproteins, glycolipids, glycosides, and biopharmaceuticals. Much of this material is presented in tabular form. Also discussed are medical and industrial applications of the technique, studies of enzyme reactions, and applications to chemical synthesis. © 2015 Wiley Periodicals, Inc. Mass Spec Rev 36:255-422, 2017.
Collapse
Affiliation(s)
- David J Harvey
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, OX1 3QU, UK
| |
Collapse
|
46
|
Brophy TM, Ward SE, McGimsey TR, Schneppenheim S, Drakeford C, O’Sullivan JM, Chion A, Budde U, O’Donnell JS. Plasmin Cleaves Von Willebrand Factor at K1491-R1492 in the A1–A2 Linker Region in a Shear- and Glycan-Dependent Manner In Vitro. Arterioscler Thromb Vasc Biol 2017; 37:845-855. [DOI: 10.1161/atvbaha.116.308524] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022]
Abstract
Objective—
Previous studies have demonstrated a role for plasmin in regulating plasma von Willebrand factor (VWF) multimer composition. Moreover, emerging data have shown that plasmin-induced cleavage of VWF is of particular importance in specific pathological states. Interestingly, plasmin has been successfully used as an alternative to ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif) in a mouse model of thrombotic thrombocytopenic purpura. Consequently, elucidating the molecular mechanisms through which plasmin binds and cleaves VWF is not only of basic scientific interest but also of direct clinical importance. Our aim was to investigate factors that modulate the susceptibility of human VWF to proteolysis by plasmin.
Approach and Results—
We have adapted the VWF vortex proteolysis assay to allow for time-dependent shear exposure studies. We show that globular VWF is resistant to plasmin cleavage under static conditions, but is readily cleaved by plasmin under shear. Although both plasmin and ADAMTS13 cleave VWF in a shear-dependent manner, plasmin does not cleave at the Tyr1605-Met1606 ADAMTS13 proteolytic site in the A2 domain. Rather under shear stress conditions, or in the presence of denaturants, such as urea or ristocetin, plasmin cleaves the K1491-R1492 peptide bond within the VWF A1–A2 linker region. Finally, we demonstrate that VWF susceptibility to plasmin proteolysis at K1491-R1492 is modulated by local N-linked glycan expression within A1A2A3, and specifically inhibited by heparin binding to the A1 domain.
Conclusions—
Improved understanding of the plasmin–VWF interaction offers exciting opportunities to develop novel adjunctive therapies for the treatment of refractory thrombotic thrombocytopenic purpura.
Collapse
Affiliation(s)
- Teresa M. Brophy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Soracha E. Ward
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Thomas R. McGimsey
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Sonja Schneppenheim
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Clive Drakeford
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Jamie M. O’Sullivan
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Alain Chion
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - Ulrich Budde
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| | - James S. O’Donnell
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, Trinity College Dublin, Ireland (T.M.B., S.E.W., T.R.M.G., C.D., J.M.O., A.C., J.S.O.); Medilys Laborgesellschaft mbH, Department of Hämostaseology, Hamburg, Germany (S.S., U.B.); National Centre for Hereditary Coagulation Disorders, St. James’s Hospital, Dublin, Ireland (J.S.O.); and Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin (J.S.O.)
| |
Collapse
|
47
|
Huang C, Liu Y, Wu H, Sun D, Li Y. Characterization of IgG glycosylation in rheumatoid arthritis patients by MALDI-TOF-MS n and capillary electrophoresis. Anal Bioanal Chem 2017; 409:3731-3739. [PMID: 28397166 DOI: 10.1007/s00216-017-0302-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/27/2017] [Accepted: 03/08/2017] [Indexed: 11/28/2022]
Abstract
An analytical method based on the combination of multistage mass spectrometry (MSn) and capillary electrophoresis (CE) was developed for the analysis of immunoglobulin G (IgG) glycosylation in rheumatoid arthritis (RA) patients. It has been recently suggested that IgG glycosylation defect may be involved in RA immunopathogenesis. Complete characterization of glycans, including both qualitative and quantitative analysis, requires a combination of different techniques, and accurate, robust, sensitive, and high-throughput methodologies are important for analysis of clinical samples. In the present study, N-glycosylation of IgG in RA patients and in healthy people was characterized through identification of the released glycans using multistage matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF-MSn), and quantitation by CE. Assignment of the IgG N-glycan structures was made through branching pattern analysis by MSn with high-throughput. Further accurate quantitation indicated that galactosylation and sialylation of IgG N-glycans in RA cases were significantly lower than in healthy subjects. The results indicate that CE coupled with MSn can identify abnormal glycosylation of IgG in RA patients compared with healthy people, and that the present work is useful for RA mechanism studies and RA diagnosis. Graphical Abstract Qualitative and quantitative analysis of IgG glycosylation in rheumatoid arthritis patients by MALDI-TOF-MSn and capillary electrophoresis.
Collapse
Affiliation(s)
- Chuncui Huang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yaming Liu
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Hongmei Wu
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China.,GuangDong Bio-Healtech Advanced Co., Ltd, Foshan, Guangdong, 528000, China
| | - Dehui Sun
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yan Li
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China. .,University of Chinese Academy of Sciences, 19 A Yuquan Rd, Shijingshan District, Beijing, 100049, China.
| |
Collapse
|
48
|
Löf A, Müller JP, Benoit M, Brehm MA. Biophysical approaches promote advances in the understanding of von Willebrand factor processing and function. Adv Biol Regul 2017; 63:81-91. [PMID: 27717713 DOI: 10.1016/j.jbior.2016.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/19/2016] [Indexed: 06/06/2023]
Abstract
The large multimeric plasma glycoprotein von Willebrand factor (VWF) is essential for primary hemostasis by recruiting platelets to sites of vascular injury. VWF multimers respond to elevated hydrodynamic forces by elongation, thereby increasing their adhesiveness to platelets. Thus, the activation of VWF is force-induced, as is its inactivation. Due to these attributes, VWF is a highly interesting system from a biophysical point of view, and is well suited for investigation using biophysical approaches. Here, we give an overview on recent studies that predominantly employed biophysical methods to gain novel insights into multiple aspects of VWF: Electron microscopy was used to shed light on the domain structure of VWF and the mechanism of VWF secretion. High-resolution stochastic optical reconstruction microscopy, atomic force microscopy (AFM), microscale thermophoresis and fluorescence correlation spectroscopy allowed identification of protein disulfide isomerase isoform A1 as the VWF dimerizing enzyme and, together with molecular dynamics simulations, postulation of the dimerization mechanism. Advanced mass spectrometry led to detailed identification of the glycan structures carried by VWF. Microfluidics was used to illustrate the interplay of force and VWF function. Results from optical tweezers measurements explained mechanisms of the force-dependent functions of VWF's domains A1 and A2 and, together with thermodynamic approaches, increased our understanding of mutation-induced dysfunctions of platelet-binding. AFM-based force measurements and AFM imaging enabled exploration of intermonomer interactions and their dependence on pH and divalent cations. These advances would not have been possible by the use of biochemical methods alone and show the benefit of interdisciplinary research approaches.
Collapse
Affiliation(s)
- Achim Löf
- Department of Physics and Center for NanoScience, LMU Munich, Munich, Germany
| | - Jochen P Müller
- Department of Physics and Center for NanoScience, LMU Munich, Munich, Germany
| | - Martin Benoit
- Department of Physics and Center for NanoScience, LMU Munich, Munich, Germany
| | - Maria A Brehm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
49
|
Reiding KR, Ruhaak LR, Uh HW, El Bouhaddani S, van den Akker EB, Plomp R, McDonnell LA, Houwing-Duistermaat JJ, Slagboom PE, Beekman M, Wuhrer M. Human Plasma N-glycosylation as Analyzed by Matrix-Assisted Laser Desorption/Ionization-Fourier Transform Ion Cyclotron Resonance-MS Associates with Markers of Inflammation and Metabolic Health. Mol Cell Proteomics 2016; 16:228-242. [PMID: 27932526 DOI: 10.1074/mcp.m116.065250] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/01/2016] [Indexed: 12/22/2022] Open
Abstract
Glycosylation is an abundant co- and post-translational protein modification of importance to protein processing and activity. Although not template-defined, glycosylation does reflect the biological state of an organism and is a high-potential biomarker for disease and patient stratification. However, to interpret a complex but informative sample like the total plasma N-glycome, it is important to establish its baseline association with plasma protein levels and systemic processes. Thus far, large-scale studies (n >200) of the total plasma N-glycome have been performed with methods of chromatographic and electrophoretic separation, which, although being informative, are limited in resolving the structural complexity of plasma N-glycans. MS has the opportunity to contribute additional information on, among others, antennarity, sialylation, and the identity of high-mannose type species.Here, we have used matrix-assisted laser desorption/ionization (MALDI)-Fourier transform ion cyclotron resonance (FTICR)-MS to study the total plasma N-glycome of 2144 healthy middle-aged individuals from the Leiden Longevity Study, to allow association analysis with markers of metabolic health and inflammation. To achieve this, N-glycans were enzymatically released from their protein backbones, labeled at the reducing end with 2-aminobenzoic acid, and following purification analyzed by negative ion mode intermediate pressure MALDI-FTICR-MS. In doing so, we achieved the relative quantification of 61 glycan compositions, ranging from Hex4HexNAc2 to Hex7HexNAc6dHex1Neu5Ac4, as well as that of 39 glycosylation traits derived thereof. Next to confirming known associations of glycosylation with age and sex by MALDI-FTICR-MS, we report novel associations with C-reactive protein (CRP), interleukin 6 (IL-6), body mass index (BMI), leptin, adiponectin, HDL cholesterol, triglycerides (TG), insulin, gamma-glutamyl transferase (GGT), alanine aminotransferase (ALT), and smoking. Overall, the bisection, galactosylation, and sialylation of diantennary species, the sialylation of tetraantennary species, and the size of high-mannose species proved to be important plasma characteristics associated with inflammation and metabolic health.
Collapse
Affiliation(s)
- Karli R Reiding
- From the ‡Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - L Renee Ruhaak
- §Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Hae-Won Uh
- ¶Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Said El Bouhaddani
- ¶Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Erik B van den Akker
- ¶Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,**Pattern Recognition & Bioinformatics, Delft University of Technology, 2600 GA Delft, The Netherlands
| | - Rosina Plomp
- From the ‡Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Liam A McDonnell
- From the ‡Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jeanine J Houwing-Duistermaat
- ¶Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,‡‡Department of Statistics, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - P Eline Slagboom
- ‖Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Marian Beekman
- ‖Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Manfred Wuhrer
- From the ‡Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| |
Collapse
|
50
|
O'Sullivan JM, Aguila S, McRae E, Ward SE, Rawley O, Fallon PG, Brophy TM, Preston RJS, Brady L, Sheils O, Chion A, O'Donnell JS. N-linked glycan truncation causes enhanced clearance of plasma-derived von Willebrand factor. J Thromb Haemost 2016; 14:2446-2457. [PMID: 27732771 DOI: 10.1111/jth.13537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 12/31/2022]
Abstract
Essentials von Willebrands factor (VWF) glycosylation plays a key role in modulating in vivo clearance. VWF glycoforms were used to examine the role of specific glycan moieties in regulating clearance. Reduction in sialylation resulted in enhanced VWF clearance through asialoglycoprotein receptor. Progressive VWF N-linked glycan trimming resulted in increased macrophage-mediated clearance. Click to hear Dr Denis discuss clearance of von Willebrand factor in a free presentation from the ISTH Academy SUMMARY: Background Enhanced von Willebrand factor (VWF) clearance is important in the etiology of both type 1 and type 2 von Willebrand disease (VWD). In addition, previous studies have demonstrated that VWF glycans play a key role in regulating in vivo clearance. However, the molecular mechanisms underlying VWF clearance remain poorly understood. Objective To define the molecular mechanisms through which VWF N-linked glycan structures influence in vivo clearance. Methods By use of a series of exoglycosidases, different plasma-derived VWF (pd-VWF) glycoforms were generated. In vivo clearance of these glycoforms was then assessed in VWF-/- mice in the presence or absence of inhibitors of asialoglycoprotein receptor (ASGPR), or following clodronate-induced macrophage depletion. Results Reduced amounts of N-linked and O-linked sialylation resulted in enhanced pd-VWF clearance modulated via ASGPR. In addition to this role of terminal sialylation, we further observed that progressive N-linked glycan trimming also resulted in markedly enhanced VWF clearance. Furthermore, these additional N-linked glycan effects on clearance were ASGPR-independent, and instead involved enhanced macrophage clearance that was mediated, at least in part, through LDL receptor-related protein 1. Conclusion The carbohydrate determinants expressed on VWF regulate susceptibility to proteolysis by ADAMTS-13. In addition, our findings now further demonstrate that non-sialic acid carbohydrate determinants expressed on VWF also play an unexpectedly important role in modulating in vivo clearance through both hepatic ASGPR-dependent and macrophage-dependent pathways. In addition, these data further support the hypothesis that variation in VWF glycosylation may be important in the pathophysiology underlying type 1C VWD.
Collapse
Affiliation(s)
- J M O'Sullivan
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - S Aguila
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - E McRae
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - S E Ward
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - O Rawley
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - P G Fallon
- Inflammation and Immunity Research Group, Institute of Molecular Medicine, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - T M Brophy
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - R J S Preston
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - L Brady
- Department of Histopathology, Sir Patrick Dun Research Laboratory, Trinity College Dublin, St James's Hospital Dublin, Dublin, Ireland
| | - O Sheils
- Department of Histopathology, Sir Patrick Dun Research Laboratory, Trinity College Dublin, St James's Hospital Dublin, Dublin, Ireland
| | - A Chion
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - J S O'Donnell
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St James's Hospital, Trinity College Dublin, Dublin, Ireland
- National Centre for Hereditary Coagulation Disorders, St James's Hospital, Dublin, Ireland
| |
Collapse
|