1
|
Bär I, Barraclough A, Bürgisser PE, van Kwawegen C, Fijnvandraat K, Eikenboom JCJ, Leebeek FWG, Voorberg J, Bierings R. The severe von Willebrand disease variant p.M771V leads to impaired anterograde trafficking of von Willebrand factor in patient-derived and base-edited endothelial colony-forming cells. J Thromb Haemost 2025; 23:466-479. [PMID: 39510415 DOI: 10.1016/j.jtha.2024.10.023] [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: 07/09/2024] [Revised: 10/03/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND von Willebrand disease (VWD) is the most common inherited bleeding disorder caused by quantitative or qualitative defects in von Willebrand factor (VWF). The p.M771V VWF variant leads to a severe bleeding phenotype in homozygous patients. However, the exact molecular mechanism remains unclear, which prevents personalized treatment of those VWD patients. OBJECTIVES This study aimed to characterize the underlying molecular mechanisms of the p.M771V variant in multiple representative ex vivo cell models. METHODS Endothelial colony-forming cells (ECFCs) were isolated from venous blood of VWD patients from the Willebrand in the Netherlands cohort carrying homozygous and heterozygous p.M771V VWF variants. The p.M771V variant was also introduced in cord blood-derived ECFCs (CB-ECFCs) through adenine base editing and was overexpressed in HEK293 cells. Biosynthesis, storage, and secretion of VWF was studied using biochemical methods and confocal microscopy. RESULTS Two unrelated homozygous p.M771V patients presented with very low VWF activity and antigen levels in plasma. Patient ECFCs showed impaired uncleaved VWF processing into mature VWF, with secreted VWF being severely reduced when compared to ECFCs of healthy donors. Multimer analysis of p.M771V ECFCs showed a deficiency of high molecular weight VWF multimers. Immunofluorescent staining revealed VWF retention in the endoplasmic reticulum; this was confirmed in various populations of base-edited CB-ECFCs harboring the p.M771V variant. CONCLUSION The severe endothelial phenotype observed in patient-derived p.M771V ECFCs, HEK293 cells, and an original base-edited CB-ECFC modeling system show that endoplasmic reticulum retention of VWF and failure to undergo subsequent proteolytic processing underpins the severe bleeding phenotype of patients with homozygous variants at M771.
Collapse
Affiliation(s)
- Isabel Bär
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, The Netherlands. https://twitter.com/IsabelBr12
| | - Alastair Barraclough
- Department of Pediatric Hematology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Petra E Bürgisser
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Calvin van Kwawegen
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Karin Fijnvandraat
- Department of Pediatric Hematology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeroen C J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, The Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, The Netherlands. https://twitter.com/FLeebeek
| | - Jan Voorberg
- Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam, The Netherlands. https://twitter.com/VoorbergJ
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Paschalaki K, Pericleous C. Isolation and Characterization of Endothelial-Colony Forming Cells (ECFC): Studying Endothelial Senescence for Translational Studies and for Personalized Medicine. Methods Mol Biol 2025; 2906:255-270. [PMID: 40082361 DOI: 10.1007/978-1-0716-4426-3_15] [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] [Indexed: 03/16/2025]
Abstract
Endothelium regulates vascular homeostasis maintaining a healthy cardiovascular system but also plays a key role in tumor development, progression, and metastasis. Endothelial senescence can be driven by aging, DNA damage, oxidative stress, oncogenes and chemotherapy, and contributes to vascular dysfunction. Endothelial colony-forming cells (ECFC) are endothelial-committed progenitors with clonogenic potential, de novo angiogenic capacity and endothelial regenerative abilities. Studying ECFC senescence provides a novel approach to investigate the molecular mechanisms of endothelial dysfunction and response to treatment, in a noninvasive and personalized manner.
Collapse
Affiliation(s)
| | - Charis Pericleous
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
3
|
Laan SNJ, Lenderink BG, Eikenboom JCJ, Bierings R. Endothelial colony-forming cells in the spotlight: insights into the pathophysiology of von Willebrand disease and rare bleeding disorders. J Thromb Haemost 2024; 22:3355-3365. [PMID: 39243860 DOI: 10.1016/j.jtha.2024.08.011] [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: 03/20/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Endothelial cells deliver a vital contribution to the maintenance of hemostasis by constituting an anatomical as well as functional barrier between the blood and the rest of the body. Apart from the physical barrier function, endothelial cells maintain the hemostatic equilibrium by their pro- and anticoagulant functions. An important part of their procoagulant contribution is the production of von Willebrand factor (VWF), which is a carrier protein for coagulation factor VIII and facilitates the formation of a platelet plug. Thus, VWF is indispensable for both primary and secondary hemostasis, which is exemplified by the bleeding disorder von Willebrand disease that results from qualitative or quantitative deficiencies in VWF. A cellular model that was found to accurately reflect the endothelium and its secretory organelles are endothelial colony-forming cells, which can be readily isolated from peripheral blood and constitute a robust ex vivo model to investigate the donor's endothelial cell function. This review summarizes some of the valuable insights on biology of VWF and pathogenic mechanisms of von Willebrand disease that have been made possible using studies with endothelial colony-forming cells derived from patients with bleeding disorders.
Collapse
Affiliation(s)
- Sebastiaan N J Laan
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands; Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands. https://twitter.com/laan_bas
| | - Britte G Lenderink
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jeroen C J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
4
|
Seidizadeh O, Baronciani L, Lillicrap D, Peyvandi F. Application of genetic testing for the diagnosis of von Willebrand disease. J Thromb Haemost 2024; 22:2115-2128. [PMID: 38762018 PMCID: PMC11548015 DOI: 10.1016/j.jtha.2024.05.006] [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] [Revised: 04/13/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
von Willebrand disease (VWD) is the most frequent inherited bleeding disorder, with an estimated symptomatic prevalence of 1 per 1000 in the general population. VWD is characterized by defects in the quantity, quality, or multimeric structure of von Willebrand factor (VWF), a glycoprotein being hemostatically essential in circulation. VWD is classified into 3 principal types: low VWF/type 1 with partial quantitative deficiency of VWF, type 3 with virtual absence of VWF, and type 2 with functional abnormalities of VWF, being classified as 2A, 2B, 2M, and 2N. A new VWD type has been officially recognized by the ISTH SSC on von Willebrand factor which has also been discussed by the joint ASH/ISTH/NHF/WFH 2021 guidelines (ie, type 1C), indicating patients with quantitative deficiency due to an enhanced VWF clearance. With the advent of next-generation sequencing technologies, the process of genetic diagnosis has substantially changed and improved accuracy. Therefore, nowadays, patients with type 3 and severe type 1 VWD can benefit from genetic testing as much as type 2 VWD. Specifically, genetic testing can be used to confirm or differentiate a VWD diagnosis, as well as to provide genetic counseling. The focus of this manuscript is to discuss the current knowledge on VWD molecular pathophysiology and the application of genetic testing for VWD diagnosis.
Collapse
Affiliation(s)
- Omid Seidizadeh
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy. https://twitter.com/OmidSeidi
| | - Luciano Baronciani
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada. https://twitter.com/DavidLillicrap
| | - Flora Peyvandi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
5
|
Laan SNJ, de Boer S, Dirven RJ, van Moort I, Kuipers TB, Mei H, Bierings R, Eikenboom J. Transcriptional and functional profiling identifies inflammation and endothelial-to-mesenchymal transition as potential drivers for phenotypic heterogeneity within a cohort of endothelial colony forming cells. J Thromb Haemost 2024; 22:2027-2038. [PMID: 38574861 DOI: 10.1016/j.jtha.2024.03.018] [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: 10/19/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Endothelial colony-forming cells (ECFCs) derived from patients can be used to investigate pathogenic mechanisms of vascular diseases like von Willebrand disease. Considerable phenotypic heterogeneity has been observed between ECFC clones derived from healthy donors. This heterogeneity needs to be well understood in order to use ECFCs as endothelial models for disease. OBJECTIVES Therefore, we aimed to determine phenotypic and gene expression differences between control ECFCs. METHODS A total of 34 ECFC clones derived from 16 healthy controls were analyzed. The transcriptome of a selection of ECFC clones (n = 15) was analyzed by bulk RNA sequencing and gene set enrichment analysis. Gene expression was measured in all ECFC clones by quantitative polymerase chain reaction. Phenotypic profiling was performed and migration speed of the ECFCs was measured using confocal microscopy, followed by automated quantification of cell morphometrics and migration speed. RESULTS Through hierarchical clustering of RNA expression profiles, we could distinguish 2 major clusters within the ECFC cohort. Major differences were associated with proliferation and migration in cluster 1 and inflammation and endothelial-to-mesenchymal transition in cluster 2. Phenotypic profiling showed significantly more and smaller ECFCs in cluster 1, which contained more and longer Weibel-Palade bodies. Migration speed in cluster 1 was also significantly higher. CONCLUSION We observed a range of different RNA expression patterns between ECFC clones, mostly associated with inflammation and clear differences in Weibel-Palade body count and structure. We developed a quantitative polymerase chain reaction panel that can be used for the characterization of ECFC clones, which is essential for the correct analysis of pathogenic mechanisms in vascular disorders.
Collapse
Affiliation(s)
- Sebastiaan N J Laan
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands; Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands. https://twitter.com/laan_bas
| | - Suzan de Boer
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Richard J Dirven
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Iris van Moort
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Thomas B Kuipers
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jeroen Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
6
|
Cox AA, Liu A, Ng CJ. Clusterin knockdown has effects on intracellular and secreted von Willebrand factor in human umbilical vein endothelial cells. PLoS One 2024; 19:e0298133. [PMID: 38363768 PMCID: PMC10871512 DOI: 10.1371/journal.pone.0298133] [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: 10/04/2023] [Accepted: 01/17/2024] [Indexed: 02/18/2024] Open
Abstract
Alterations in von Willebrand factor (VWF) have an important role in human health and disease. Deficiency of VWF is associated with symptoms of bleeding and excesses of VWF are associated with thrombotic outcomes. Understanding the mechanisms that drive VWF regulation can lead to a better understanding of modulation of VWF levels in humans. We identified clusterin (CLU) as a potential candidate regulator of VWF based on a single cell RNA sequencing (scRNA-seq) analysis in control endothelial cells (ECs) and von Willebrand disease (VWD) endothelial colony-forming-cells (ECFCs). We found that patients with deficiencies of VWF (von Willebrand disease, VWD) had decreased CLU expression and ECs with low VWF expression also had low CLU expression. Based on these findings, we sought to evaluate the role of CLU in the regulation of VWF, specifically as it relates to VWD. As CLU is primarily thought to be a golgi protein involved in protein chaperoning, we hypothesized that knockdown of CLU would lead to decreases in VWF and alterations in Weibel-Palade bodies (WPBs). We used both siRNA- and CRISPR-Cas9-based approaches to modulate CLU in human umbilical vein endothelial cells (HUVECs) and evaluated VWF protein levels, VWF mRNA copy number, and WPB quantity and size. We demonstrated that siRNA-based knockdown of CLU resulted in decreases in VWF content in cellular lysates and supernatants, but no significant change in WPB quantity or size. A CRISPR-Cas9-based knockdown of CLU demonstrated similar findings of decreases in intracellular VWF content but no significant change in WPB quantity or size. Our data suggests that CLU knockdown is associated with decreases in cellular VWF content but does not affect VWF mRNA levels or WPB quantity or size.
Collapse
Affiliation(s)
- Allaura A. Cox
- Department of Pediatrics, University of Colorado–Anschutz Medical Campus, Aurora, CO, United States of America
| | - Alice Liu
- Department of Bioengineering, Washington University, St. Louis, MO, United States of America
| | - Christopher J. Ng
- Department of Pediatrics, University of Colorado–Anschutz Medical Campus, Aurora, CO, United States of America
| |
Collapse
|
7
|
Thachil J, Lassila R. What can historical literature on von Willebrand disease teach us? Res Pract Thromb Haemost 2023; 7:102244. [PMID: 38193051 PMCID: PMC10772893 DOI: 10.1016/j.rpth.2023.102244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 01/10/2024] Open
Abstract
Dr Erik von Willebrand first described a family with bleeding symptoms in a Finnish publication in 1926. A closer look at this landmark publication sheds light on some pathophysiological aspects of von Willebrand disease that may be applicable even in the current era. We attempt to relay in this article how the teachings from this original description may provide a benchmark for further research in this condition.
Collapse
Affiliation(s)
- Jecko Thachil
- Department of Haematology, Manchester University Hospitals, Manchester, United Kingdom
- University of Manchester, Manchester, United Kingdom
| | - Riitta Lassila
- Research Program Unit in Systems Oncology, Oncosys, Medical Faculty, University of Helsinki, Helsinki, Finland
- Coagulation Disorders Unit, Department of Hematology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
8
|
Barrett L, Curry N, Abu-Hanna J. Experimental Models of Traumatic Injuries: Do They Capture the Coagulopathy and Underlying Endotheliopathy Induced by Human Trauma? Int J Mol Sci 2023; 24:11174. [PMID: 37446351 PMCID: PMC10343021 DOI: 10.3390/ijms241311174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Trauma-induced coagulopathy (TIC) is a major cause of morbidity and mortality in patients with traumatic injury. It describes the spectrum of coagulation abnormalities that occur because of the trauma itself and the body's response to the trauma. These coagulation abnormalities range from hypocoagulability and hyperfibrinolysis, resulting in potentially fatal bleeding, in the early stages of trauma to hypercoagulability, leading to widespread clot formation, in the later stages. Pathological changes in the vascular endothelium and its regulation of haemostasis, a phenomenon known as the endotheliopathy of trauma (EoT), are thought to underlie TIC. Our understanding of EoT and its contribution to TIC remains in its infancy largely due to the scarcity of experimental research. This review discusses the mechanisms employed by the vascular endothelium to regulate haemostasis and their dysregulation following traumatic injury before providing an overview of the available experimental in vitro and in vivo models of trauma and their applicability for the study of the EoT and its contribution to TIC.
Collapse
Affiliation(s)
- Liam Barrett
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK;
- Emergency Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Nicola Curry
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LD, UK
| | - Jeries Abu-Hanna
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
| |
Collapse
|
9
|
Mobayen G, Smith K, Ediriwickrema K, Starke RD, Solomonidis EG, Laffan MA, Randi AM, McKinnon TAJ. von Willebrand factor binds to angiopoietin-2 within endothelial cells and after release from Weibel-Palade bodies. J Thromb Haemost 2023; 21:1802-1812. [PMID: 37011710 DOI: 10.1016/j.jtha.2023.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND The von Willebrand factor (VWF) is a multimeric plasma glycoprotein essential for hemostasis, inflammation, and angiogenesis. The majority of VWF is synthesized by endothelial cells (ECs) and stored in Weibel-Palade bodies (WPB). Among the range of proteins shown to co-localize to WPB is angiopoietin-2 (Angpt-2), a ligand of the receptor tyrosine kinase Tie-2. We have previously shown that VWF itself regulates angiogenesis, raising the hypothesis that some of the angiogenic activity of VWF may be mediated by its interaction with Angpt-2. METHODS Static-binding assays were used to probe the interaction between Angpt-2 and VWF. Binding in media from cultured human umbilical vein ECs s and in plasma was determined by immunoprecipitation experiments. Immunofluorescence was used to detect the presence of Angpt-2 on VWF strings, and flow assays were used to investigate the effect on VWF function. RESULTS Static-binding assays revealed that Angpt-2 bound to VWF with high affinity (KD,app ∼3 nM) in a pH and calcium-dependent manner. The interaction was localized to the VWF A1 domain. Co-immunoprecipitation experiments demonstrated that the complex persisted following stimulated secretion from ECs and was present in plasma. Angpt-2 was also visible on VWF strings on stimulated ECs. The VWF-Angpt-2 complex did not inhibit the binding of Angpt-2 to Tie-2 and did not significantly interfere with VWF-platelet capture. CONCLUSIONS Together, these data demonstrate a direct binding interaction between Angpt-2 and VWF that persists after secretion. VWF may act to localize Angpt-2; further work is required to establish the functional consequences of this interaction.
Collapse
Affiliation(s)
- Golzar Mobayen
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Koval Smith
- National Heart and Lung Institute (NHLI) Cardiovascular Sciences, Unit Imperial College Academic Health Science Centre, Hammersmith Hospital, London, United Kingdom
| | - Kushani Ediriwickrema
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Richard D Starke
- National Heart and Lung Institute (NHLI) Cardiovascular Sciences, Unit Imperial College Academic Health Science Centre, Hammersmith Hospital, London, United Kingdom
| | - Emmanouil Georgios Solomonidis
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Michael A Laffan
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Anna M Randi
- National Heart and Lung Institute (NHLI) Cardiovascular Sciences, Unit Imperial College Academic Health Science Centre, Hammersmith Hospital, London, United Kingdom
| | - Thomas A J McKinnon
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom.
| |
Collapse
|
10
|
Ocran E, Chornenki NLJ, Bowman M, Sholzberg M, James P. Gastrointestinal bleeding in von Willebrand patients: special diagnostic and management considerations. Expert Rev Hematol 2023; 16:575-584. [PMID: 37278227 DOI: 10.1080/17474086.2023.2221846] [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: 03/20/2023] [Accepted: 06/01/2023] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Severe and recurrent gastrointestinal (GI) bleeding caused by angiodysplasia is a significant problem in patients with von Willebrand disease (VWD) and in those with acquired von Willebrand syndrome (AVWS). At present, angiodysplasia-related GI bleeding is often refractory to standard treatment including replacement therapy with von Willebrand factor (VWF) concentrates and continues to remain a major challenge and cause of significant morbidity in patients despite advances in diagnostics and therapeutics. AREAS COVERED This paper reviews the available literature on GI bleeding in VWD patients, examines the molecular mechanisms implicated in angiodysplasia-related GI bleeding, and summarizes existing strategies in the management of bleeding GI angiodysplasia in patients with VWF abnormalities. Suggestions are made for further research directions. EXPERT OPINION Bleeding from angiodysplasia poses a significant challenge for individuals with abnormal VWF. Diagnosis remains a challenge and may require multiple radiologic and endoscopic investigations. Additionally, there is a need for enhanced understanding at a molecular level to identify effective therapies. Future studies of VWF replacement therapies using newer formulations as well as other adjunctive treatments to prevent and treat bleeding will hopefully improve care.
Collapse
Affiliation(s)
- Edwin Ocran
- Department of Medicine, Queen's University, Kingston, Canada
| | | | | | - Michelle Sholzberg
- Division of Hematology-Oncology, St. Michael's Hospital, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, Canada
| | - Paula James
- Department of Medicine, Queen's University, Kingston, Canada
| |
Collapse
|
11
|
Stritt S, Nurden P, Nurden AT, Schved JF, Bordet JC, Roux M, Alessi MC, Trégouët DA, Mäkinen T, Giansily-Blaizot M. APOLD1 loss causes endothelial dysfunction involving cell junctions, cytoskeletal architecture, and Weibel-Palade bodies, while disrupting hemostasis. Haematologica 2023; 108:772-784. [PMID: 35638551 PMCID: PMC9973481 DOI: 10.3324/haematol.2022.280816] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Indexed: 11/09/2022] Open
Abstract
Vascular homeostasis is impaired in various diseases thereby contributing to the progression of their underlying pathologies. The endothelial immediate early gene Apolipoprotein L domain-containing 1 (APOLD1) helps to regulate endothelial function. However, its precise role in endothelial cell biology remains unclear. We have localized APOLD1 to endothelial cell contacts and to Weibel-Palade bodies (WPB) where it associates with von Willebrand factor (VWF) tubules. Silencing of APOLD1 in primary human endothelial cells disrupted the cell junction-cytoskeletal interface, thereby altering endothelial permeability accompanied by spontaneous release of WPB contents. This resulted in an increased presence of WPB cargoes, notably VWF and angiopoietin-2 in the extracellular medium. Autophagy flux, previously recognized as an essential mechanism for the regulated release of WPB, was impaired in the absence of APOLD1. In addition, we report APOLD1 as a candidate gene for a novel inherited bleeding disorder across three generations of a large family in which an atypical bleeding diathesis was associated with episodic impaired microcirculation. A dominant heterozygous nonsense APOLD1:p.R49* variant segregated to affected family members. Compromised vascular integrity resulting from an excess of plasma angiopoietin-2, and locally impaired availability of VWF may explain the unusual clinical profile of APOLD1:p.R49* patients. In summary, our findings identify APOLD1 as an important regulator of vascular homeostasis and raise the need to consider testing of endothelial cell function in patients with inherited bleeding disorders without apparent platelet or coagulation defects.
Collapse
Affiliation(s)
- Simon Stritt
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala
| | - Paquita Nurden
- Institut de Rythmologie et de Modélisation Cardiaque, Hôpital Xavier Arnozan, Pessac, France.
| | - Alan T Nurden
- Institut de Rythmologie et de Modélisation Cardiaque, Hôpital Xavier Arnozan, Pessac, France
| | - Jean-François Schved
- Department of Biological Hematology, CHU Montpellier, Université de Montpellier, Montpellier
| | - Jean-Claude Bordet
- Hematology, Hospices civils de Lyon, Bron biology center and Hemostasis- Thrombosis, Lyon-1 University, Lyon
| | | | | | - David-Alexandre Trégouët
- Laboratory of Excellence GENMED (Medical Genomics), Paris; University of Bordeaux, INSERM, Bordeaux Population Health Research Center, U1219, Bordeaux
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Muriel Giansily-Blaizot
- Department of Biological Hematology, CHU Montpellier, Université de Montpellier, Montpellier
| |
Collapse
|
12
|
Chornenki NLJ, Ocran E, James PD. Special considerations in GI bleeding in VWD patients. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:624-630. [PMID: 36485078 PMCID: PMC9820382 DOI: 10.1182/hematology.2022000390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Gastrointestinal (GI) bleeding is an important cause of morbidity and mortality in von Willebrand disease (VWD). It has been noted that GI bleeding caused by angiodysplasia is overrepresented in VWD patients compared to other causes. The bleeding from angiodysplasia is notoriously difficult to treat; recurrences and rebleeds are common. A growing body of basic science evidence demonstrates that von Willebrand factor negatively regulates angiogenesis through multiple pathways. VWD is clinically highly associated with angiodysplasia. The predisposition to angiodysplasia likely accounts for many of the clinical difficulties related to managing GI bleeding in VWD patients. Diagnosis and treatment are challenging with the current tools available, and much further research is needed to further optimize care for these patients with regard to acute treatment, prophylaxis, and adjunctive therapies. In this review we provide an overview of the available literature on GI bleeding in VWD and explore the molecular underpinnings of angiodysplasia-related GI bleeding. Considerations for diagnostic effectiveness are discussed, as well as the natural history and recurrence of these lesions and which therapeutic options are available for acute and prophylactic management.
Collapse
Affiliation(s)
| | - Edwin Ocran
- Department of Medicine, Queen's University, Kingston, Canada
| | - Paula D James
- Department of Medicine, Queen's University, Kingston, Canada
| |
Collapse
|
13
|
Abstract
Endothelial colony-forming cells (ECFCs) are progenitor cells that can give rise to colonies of highly proliferative vascular endothelial cells (ECs) with clonal expansion and in vivo blood vessel-forming potential. More than two decades ago, the identification of ECFCs in human peripheral blood created tremendous opportunities as having a clinically accessible source of autologous ECs could facilitate meaningful therapies with the potential to impact multiple vascular diseases. Nevertheless, until recently, the field of endothelial progenitor cells has been plagued with ambiguities and controversies, and reaching a consensus on the definition of ECFCs has not been straightforward. Moreover, although the basic phenotypical and functional characteristics of cultured ECFCs are now well established, some fundamental questions such as the origin of ECFCs and their physiological roles in health and disease remain incompletely understood. Here, I highlight some critical studies that have shaped our current understanding of ECFCs in humans. Insights into the biological attributes of ECFCs are essential for facilitating the clinical translation of their therapeutic potential.
Collapse
Affiliation(s)
- Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
14
|
Kat M, Margadant C, Voorberg J, Bierings R. Dispatch and delivery at the ER-Golgi interface: how endothelial cells tune their hemostatic response. FEBS J 2022; 289:6863-6870. [PMID: 35246944 PMCID: PMC9790534 DOI: 10.1111/febs.16421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 01/13/2023]
Abstract
Von Willebrand factor (VWF) is a glycoprotein that is secreted into the circulation and controls bleeding by promoting adhesion and aggregation of blood platelets at sites of vascular injury. Substantial inter-individual variation in VWF plasma levels exists among the healthy population. Prior to secretion, VWF polymers are assembled and condensed into helical tubules, which are packaged into Weibel-Palade bodies (WPBs), a highly specialized post-Golgi storage compartment in vascular endothelial cells. In the inherited bleeding disorder Von Willebrand disease (VWD), mutations in the VWF gene can cause qualitative or quantitative defects, limiting protein function, secretion, or plasma survival. However, pathogenic VWF mutations cannot be found in all VWD cases. Although an increasing number of genetic modifiers have been identified, even more rare genetic variants that impact VWF plasma levels likely remain to be discovered. Here, we summarize recent evidence that modulation of the early secretory pathway has great impact on the biogenesis and release of WPBs. Based on these findings, we propose that rare, as yet unidentified quantitative trait loci influencing intracellular VWF transport contribute to highly variable VWF levels in the population. These may underlie the thrombotic complications linked to high VWF levels, as well as the bleeding tendency in individuals with low VWF levels.
Collapse
Affiliation(s)
- Marije Kat
- Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdam University Medical CenterUniversity of AmsterdamThe Netherlands
| | - Coert Margadant
- Angiogenesis laboratoryCancer Center AmsterdamAmsterdam University Medical Center location VUmcThe Netherlands
| | - Jan Voorberg
- Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdam University Medical CenterUniversity of AmsterdamThe Netherlands,Experimental Vascular MedicineAmsterdam University Medical CenterUniversity of AmsterdamThe Netherlands
| | - Ruben Bierings
- Hematology, Erasmus University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
15
|
Bowman M, Casey L, Selvam SN, Lima PD, Rawley O, Hinds M, Tuttle A, Grabell J, Iorio A, Walker I, Lillicrap D, James P. von Willebrand factor propeptide variants lead to impaired storage and ER retention in patient-derived endothelial colony-forming cells. J Thromb Haemost 2022; 20:1599-1609. [PMID: 35466528 PMCID: PMC9246936 DOI: 10.1111/jth.15740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/29/2022] [Accepted: 04/21/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND von Willebrand factor (VWF) is synthesized by vascular endothelial cells and megakaryocytes. The VWF propeptide is critical for multimerization and acts as an intra-molecular chaperone for mature VWF in sorting to its storage organelles, Weibel-Palade bodies (WPBs). In the Canadian Type 3 VWD study, almost half of the identified variants were in the VWF propeptide and these were associated with an increased bleeding phenotype. OBJECTIVE To investigate VWF propeptide variants that cause quantitative von Willebrand disease (VWD) by utilizing patient-derived endothelial colony-forming cells (ECFCs). PATIENTS/METHODS Endothelial colony-forming cells were isolated from five Type 3 VWD patients from four families with the following variants: (1) homozygous p.Asp75_Gly178del (deletion of exons 4 and 5 deletion; Ex4-5del), (2) homozygous p.Cys633Arg, (3) homozygous p.Arg273Trp, and (4) p.Pro293Glnfs*164 and p.Gln419* inherited in the compound heterozygous state. Additionally, ECFCs were isolated from six family members (two Type 1 VWD, four unaffected). RESULTS Endothelial colony-forming cells from the Type 3 patient with the compound heterozygous genotype exhibited a true null VWF cellular phenotype, with negligible VWF detected. In contrast, the other three propeptide variants presented a similar expression pattern in homozygous ECFCs where VWF was synthesized but not packaged in WPBs, and variant VWF had an increased association with the endoplasmic reticulum (ER) marker, protein disulfide-isomerase (PDI), indicating an ER-retention phenotype. The biosynthetic phenotype was similar but to a lesser degree in heterozygous ECFCs expressing the non-null variants. CONCLUSION This study further elucidates the importance of the VWF propeptide in the VWD phenotype using patient-derived cells.
Collapse
Affiliation(s)
- Mackenzie Bowman
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Lara Casey
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Soundarya N. Selvam
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | | | - Orla Rawley
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Megan Hinds
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Angie Tuttle
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Julie Grabell
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Alfonso Iorio
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Irwin Walker
- Department of Medicine, Division of Hematology & Thromboembolism, McMaster University, Hamilton, ON, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Paula James
- Department of Medicine, Queen’s University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
16
|
Wang Y, Keshavarz M, Barhouse P, Smith Q. Strategies for Regenerative Vascular Tissue Engineering. Adv Biol (Weinh) 2022; 7:e2200050. [PMID: 35751461 DOI: 10.1002/adbi.202200050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/15/2022] [Indexed: 11/11/2022]
Abstract
Vascularization remains one of the key challenges in creating functional tissue-engineered constructs for therapeutic applications. This review aims to provide a developmental lens on the necessity of blood vessels in defining tissue function while exploring stem cells as a suitable source for vascular tissue engineering applications. The intersections of stem cell biology, material science, and engineering are explored as potential solutions for directing vascular assembly.
Collapse
Affiliation(s)
- Yao Wang
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| | - Mozhgan Keshavarz
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| | - Patrick Barhouse
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center University of California Irvine CA 92697 USA
| |
Collapse
|
17
|
Exploring Endothelial Colony-Forming Cells to Better Understand the Pathophysiology of Disease: An Updated Review. Stem Cells Int 2022; 2022:4460041. [PMID: 35615696 PMCID: PMC9126670 DOI: 10.1155/2022/4460041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/20/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Endothelial cell (EC) dysfunction has been implicated in a variety of pathological conditions. The collection of ECs from patients is typically conducted postmortem or through invasive procedures, such as surgery and interventional procedures, hampering efforts to clarify the role of ECs in disease onset and progression. In contrast, endothelial colony-forming cells (ECFCs), also termed late endothelial progenitor cells, late outgrowth endothelial cells, blood outgrowth endothelial cells, or endothelial outgrowth cells, are obtained in a minimally invasive manner, namely, by the culture of human peripheral blood mononuclear cells in endothelial growth medium. ECFCs resemble mature ECs phenotypically, genetically, and functionally, making them excellent surrogates for ECs. Numerous studies have been performed that examined ECFC function in conditions such as coronary artery disease, diabetes mellitus, hereditary hemorrhagic telangiectasia, congenital bicuspid aortic valve disease, pulmonary arterial hypertension, venous thromboembolic disease, and von Willebrand disease. Here, we provide an updated review of studies using ECFCs that were performed to better understand the pathophysiology of disease. We also discuss the potential of ECFCs as disease biomarkers and the standardized methods to culture, quantify, and evaluate ECFCs and suggest the future direction of research in this field.
Collapse
|
18
|
Page KM, McCormack JJ, Lopes-da-Silva M, Patella F, Harrison-Lavoie K, Burden JJ, Quah YYB, Scaglioni D, Ferraro F, Cutler DF. Structure modeling hints at a granular organization of the Golgi ribbon. BMC Biol 2022; 20:111. [PMID: 35549945 PMCID: PMC9102599 DOI: 10.1186/s12915-022-01305-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/21/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND In vertebrate cells, the Golgi functional subunits, mini-stacks, are linked into a tri-dimensional network. How this "ribbon" architecture relates to Golgi functions remains unclear. Are all connections between mini-stacks equal? Is the local structure of the ribbon of functional importance? These are difficult questions to address, without a quantifiable readout of the output of ribbon-embedded mini-stacks. Endothelial cells produce secretory granules, the Weibel-Palade bodies (WPB), whose von Willebrand Factor (VWF) cargo is central to hemostasis. The Golgi apparatus controls WPB size at both mini-stack and ribbon levels. Mini-stack dimensions delimit the size of VWF "boluses" whilst the ribbon architecture allows their linear co-packaging, thereby generating WPBs of different lengths. This Golgi/WPB size relationship suits mathematical analysis. RESULTS WPB lengths were quantized as multiples of the bolus size and mathematical modeling simulated the effects of different Golgi ribbon organizations on WPB size, to be compared with the ground truth of experimental data. An initial simple model, with the Golgi as a single long ribbon composed of linearly interlinked mini-stacks, was refined to a collection of mini-ribbons and then to a mixture of mini-stack dimers plus long ribbon segments. Complementing these models with cell culture experiments led to novel findings. Firstly, one-bolus sized WPBs are secreted faster than larger secretory granules. Secondly, microtubule depolymerization unlinks the Golgi into equal proportions of mini-stack monomers and dimers. Kinetics of binding/unbinding of mini-stack monomers underpinning the presence of stable dimers was then simulated. Assuming that stable mini-stack dimers and monomers persist within the ribbon resulted in a final model that predicts a "breathing" arrangement of the Golgi, where monomer and dimer mini-stacks within longer structures undergo continuous linking/unlinking, consistent with experimentally observed WPB size distributions. CONCLUSIONS Hypothetical Golgi organizations were validated against a quantifiable secretory output. The best-fitting Golgi model, accounting for stable mini-stack dimers, is consistent with a highly dynamic ribbon structure, capable of rapid rearrangement. Our modeling exercise therefore predicts that at the fine-grained level the Golgi ribbon is more complex than generally thought. Future experiments will confirm whether such a ribbon organization is endothelial-specific or a general feature of vertebrate cells.
Collapse
Affiliation(s)
- Karen M. Page
- Department of Mathematics, University College London, Gower Street, London, WC1E 6BT UK
| | - Jessica J. McCormack
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Mafalda Lopes-da-Silva
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
- Current address: iNOVA4Health, CEDOC-Chronic Diseases Research Center, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Francesca Patella
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
- Current address: Kinomica, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG UK
| | - Kimberly Harrison-Lavoie
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Jemima J. Burden
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Ying-Yi Bernadette Quah
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Dominic Scaglioni
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Francesco Ferraro
- Department of Biology and Evolution of Marine Organisms, BEOM, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy
| | - Daniel F. Cutler
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| |
Collapse
|
19
|
Ng CJ, Liu A, Venkataraman S, Ashworth KJ, Baker CD, O'Rourke R, Vibhakar R, Jones KL, Di Paola J. Single-cell transcriptional analysis of human endothelial colony-forming cells from patients with low VWF levels. Blood 2022; 139:2240-2251. [PMID: 35143643 PMCID: PMC8990376 DOI: 10.1182/blood.2021010683] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022] Open
Abstract
von Willebrand factor (VWF) plays a key role in normal hemostasis, and deficiencies of VWF lead to clinically significant bleeding. We sought to identify novel modifiers of VWF levels in endothelial colony-forming cells (ECFCs) using single-cell RNA sequencing (scRNA-seq). ECFCs were isolated from patients with low VWF levels (plasma VWF antigen levels between 30 and 50 IU/dL) and from healthy controls. Human umbilical vein endothelial cells were used as an additional control cell line. Cells were characterized for their Weibel Palade body (WPB) content and VWF release. scRNA-seq of all cell lines was performed to evaluate for gene expression heterogeneity and for candidate modifiers of VWF regulation. Candidate modifiers identified by scRNA-seq were further characterized with small-interfering RNA (siRNA) experiments to evaluate for effects on VWF. We observed that ECFCs derived from patients with low VWF demonstrated alterations in baseline WPB metrics and exhibit impaired VWF release. scRNA-seq analyses of these endothelial cells revealed overall decreased VWF transcription, mosaicism of VWF expression, and genes that are differentially expressed in low VWF ECFCs and control endothelial cells (control ECs). An siRNA screen of potential VWF modifiers provided further evidence of regulatory candidates, and 1 such candidate, FLI1, alters the transcriptional activity of VWF. In conclusion, ECFCs from individuals with low VWF demonstrate alterations in their baseline VWF packaging and release compared with control ECs. scRNA-seq revealed alterations in VWF transcription, and siRNA screening identified multiple candidate regulators of VWF.
Collapse
Affiliation(s)
- Christopher J Ng
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Alice Liu
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Sujatha Venkataraman
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Katrina J Ashworth
- Division of Hematology Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO; and
| | - Christopher D Baker
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Rebecca O'Rourke
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Kenneth L Jones
- Department of Cell Biology and
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Jorge Di Paola
- Division of Hematology Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO; and
| |
Collapse
|
20
|
Low von Willebrand factor phenotype: the enigma continues. Blood 2022; 139:2102-2103. [PMID: 35389442 DOI: 10.1182/blood.2021013541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/05/2021] [Indexed: 11/20/2022] Open
|
21
|
Multifaceted Pathomolecular Mechanism of a VWF Large Deletion Involved in the Pathogenesis of Severe VWD. Blood Adv 2021; 6:1038-1053. [PMID: 34861678 PMCID: PMC8945295 DOI: 10.1182/bloodadvances.2021005895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/21/2021] [Indexed: 11/20/2022] Open
Abstract
The present study demonstrates the dominant-negative impact of an in-frame large deletion on VWF biosynthesis and biogenesis of the WPBs. The malformed WPBs/altered trafficking of its inflammatory cargos cause distresses in endothelial cell signaling pathways and phenotype.
An in-frame heterozygous large deletion of exons 4 through 34 of the von Willebrand factor (VWF) gene was identified in a type 3 von Willebrand disease (VWD) index patient (IP), as the only VWF variant. The IP exhibited severe bleeding episodes despite prophylaxis treatment, with a short VWF half-life after infusion of VWF/factor VIII concentrates. Transcript analysis confirmed transcription of normal VWF messenger RNA besides an aberrant deleted transcript. The IP endothelial colony-forming cells (ECFCs) exhibited a defect in the VWF multimers and Weibel-Palade bodies (WPBs) biogenesis, although demonstrating normal VWF secretion compared with healthy cells. Immunostaining of IP-ECFCs revealed subcellular mislocalization of WPBs pro-inflammatory cargos angiopoietin-2 (Ang2, nuclear accumulation) and P-selectin. Besides, the RNA-sequencing (RNA-seq) analysis showed upregulation of pro-inflammatory and proangiogenic genes, P-selectin, interleukin 8 (IL-8), IL-6, and GROα, copackaged with VWF into WPBs. Further, whole-transcriptome RNA-seq and subsequent gene ontology (GO) enrichment analysis indicated the most enriched GO-biological process terms among the differentially expressed genes in IP-ECFCs were regulation of cell differentiation, cell adhesion, leukocyte adhesion to vascular endothelial, blood vessel morphogenesis, and angiogenesis, which resemble downstream signaling pathways associated with inflammatory stimuli and Ang2 priming. Accordingly, our functional experiments exhibited an increased endothelial cell adhesiveness and interruption in endothelial cell–cell junctions of the IP-ECFCs. In conclusion, the deleted VWF has a dominant-negative impact on multimer assembly and the biogenesis of WPBs, leading to altered trafficking of their pro-inflammatory cargos uniquely, which, in turn, causes changes in cellular signaling pathways, phenotype, and function of the endothelial cells.
Collapse
|
22
|
Mathur T, Tronolone JJ, Jain A. Comparative Analysis of Blood-Derived Endothelial Cells for Designing Next-Generation Personalized Organ-on-Chips. J Am Heart Assoc 2021; 10:e022795. [PMID: 34743553 PMCID: PMC8751908 DOI: 10.1161/jaha.121.022795] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Organ‐on‐chip technology has accelerated in vitro preclinical research of the vascular system, and a key strength of this platform is its promise to impact personalized medicine by providing a primary human cell–culture environment where endothelial cells are directly biopsied from individual tissue or differentiated through stem cell biotechniques. However, these methods are difficult to adopt in laboratories, and often result in impurity and heterogeneity of cells. This limits the power of organ‐chips in making accurate physiological predictions. In this study, we report the use of blood‐derived endothelial cells as alternatives to primary and induced pluripotent stem cell–derived endothelial cells. Methods and Results Here, the genotype, phenotype, and organ‐chip functional characteristics of blood‐derived outgrowth endothelial cells were compared against commercially available and most used primary endothelial cells and induced pluripotent stem cell–derived endothelial cells. The methods include RNA‐sequencing, as well as criterion standard assays of cell marker expression, growth kinetics, migration potential, and vasculogenesis. Finally, thromboinflammatory responses under shear using vessel‐chips engineered with blood‐derived endothelial cells were assessed. Blood‐derived endothelial cells exhibit the criterion standard hallmarks of typical endothelial cells. There are differences in gene expression profiles between different sources of endothelial cells, but blood‐derived cells are relatively closer to primary cells than induced pluripotent stem cell–derived. Furthermore, blood‐derived endothelial cells are much easier to obtain from individuals and yet, they serve as an equally effective cell source for functional studies and organ‐chips compared with primary cells or induced pluripotent stem cell–derived cells. Conclusions Blood‐derived endothelial cells may be used in preclinical research for developing more robust and personalized next‐generation disease models using organ‐on‐chips.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, College of Engineering Texas A&M University College Station TX
| | - James J Tronolone
- Department of Biomedical Engineering, College of Engineering Texas A&M University College Station TX
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering Texas A&M University College Station TX.,Department of Medical Physiology College of MedicineTexas A&M Health Science Center Bryan TX.,Department of Cardiovascular Sciences Houston Methodist Research Institute Houston TX
| |
Collapse
|
23
|
Sharda AV, Bogue T, Barr A, Mendez LM, Flaumenhaft R, Zwicker JI. Circulating Protein Disulfide Isomerase Is Associated with Increased Risk of Thrombosis in JAK2-Mutated Myeloproliferative Neoplasms. Clin Cancer Res 2021; 27:5708-5717. [PMID: 34400417 DOI: 10.1158/1078-0432.ccr-21-1140] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/07/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Thromboembolic events (TE) are the most common complications of myeloproliferative neoplasms (MPN). Clinical parameters, including patient age and mutation status, are used to risk-stratify patients with MPN, but a true biomarker of TE risk is lacking. Protein disulfide isomerase (PDI), an endoplasmic reticulum protein vital for protein folding, also possesses essential extracellular functions, including regulation of thrombus formation. Pharmacologic PDI inhibition prevents thrombus formation, but whether pathologic increases in PDI increase TE risk remains unknown. EXPERIMENTAL DESIGN We evaluated the association of plasma PDI levels and risk of TE in a cohort of patients with MPN with established diagnosis of polycythemia vera (PV) or essential thrombocythemia (ET), compared with healthy controls. Plasma PDI was measured at enrollment and subjects followed prospectively for development of TE. RESULTS A subset of patients, primarily those with JAK2-mutated MPN, had significantly elevated plasma PDI levels as compared with controls. Plasma PDI was functionally active. There was no association between PDI levels and clinical parameters typically used to risk-stratify patients with MPN. The risk of TE was 8-fold greater in those with PDI levels above 2.5 ng/mL. Circulating endothelial cells from JAK2-mutated MPN patients, but not platelets, demonstrated augmented PDI release, suggesting endothelial activation as a source of increased plasma PDI in MPN. CONCLUSIONS The observed association between plasma PDI levels and increased risk of TE in patients with JAK2-mutated MPN has both prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Anish V Sharda
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.,Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Thomas Bogue
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Alexandra Barr
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Lourdes M Mendez
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Jeffrey I Zwicker
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts. .,Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
Characterization of large in-frame von Willebrand factor deletions highlights differing pathogenic mechanisms. Blood Adv 2021; 4:2979-2990. [PMID: 32609846 DOI: 10.1182/bloodadvances.2018027813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/02/2020] [Indexed: 11/20/2022] Open
Abstract
Copy number variation (CNV) is known to cause all von Willebrand disease (VWD) types, although the associated pathogenic mechanisms involved have not been extensively studied. Notably, in-frame CNV provides a unique opportunity to investigate how specific von Willebrand factor (VWF) domains influence the processing and packaging of the protein. Using multiplex ligation-dependent probe amplification, this study determined the extent to which CNV contributed to VWD in the Molecular and Clinical Markers for the Diagnosis and Management of Type 1 von Willebrand Disease cohort, highlighting in-frame deletions of exons 3, 4-5, 32-34, and 33-34. Heterozygous in vitro recombinant VWF expression demonstrated that, although deletion of exons 3, 32-34, and 33-34 all resulted in significant reductions in total VWF (P < .0001, P < .001, and P < .01, respectively), only deletion of exons 3 and 32-34 had a significant impact on VWF secretion (P < .0001). High-resolution microscopy of heterozygous and homozygous deletions confirmed these observations, indicating that deletion of exons 3 and 32-34 severely impaired pseudo-Weibel-Palade body (WPB) formation, whereas deletion of exons 33-34 did not, with this variant still exhibiting pseudo-WPB formation similar to wild-type VWF. In-frame deletions in VWD, therefore, contribute to pathogenesis via moderate or severe defects in VWF biosynthesis and secretion.
Collapse
|
25
|
Rauch A, Paris C, Repesse Y, Branche J, D'Oiron R, Harroche A, Ternisien C, Castet SM, Lebreton A, Pan-Petesch B, Volot F, Claeyssens S, Chamouni P, Gay V, Berger C, Desprez D, Falaise C, Biron Andreani C, Marichez C, Pradines B, Zawadzki C, Itzhar Baikian N, Borel-Derlon A, Goudemand J, Gerard R, Susen S. Gastrointestinal bleeding from angiodysplasia in von Willebrand disease: Improved diagnosis and outcome prediction using videocapsule on top of conventional endoscopy. J Thromb Haemost 2021; 19:380-386. [PMID: 33113216 DOI: 10.1111/jth.15155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/09/2020] [Accepted: 10/21/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Despite a high prevalence of angiodysplasia, no specific guidelines are available for the modalities of endoscopic exploration of gastrointestinal (GI) bleeding in von Willebrand disease (VWD). Whether VWD patients could benefit from video capsule endoscopy (VCE) looking for angiodysplasia eligible to endoscopic treatment or at high risk of bleeding is unknown. OBJECTIVES To assess the diagnostic efficacy for angiodysplasia and the prognostic value of VCE on top of conventional endoscopy in VWD patients with GI bleeding. PATIENTS/METHODS A survey was sent to the 30 centers of the French-network on inherited bleeding disorders to identify VWD patients referred for endoscopic exploration of GI bleeding from January 2015 to December 2017. Data obtained included patient characteristics, VWD phenotype/genotype, GI bleeding pattern, results of endoscopic investigations, and medical management applied including endoscopic therapy. We assessed by Kaplan-Meier analysis the recurrence-free survival after the first GI bleeding event according to endoscopic categorization and, in patients with angiodysplasia, to the presence of small-bowel localizations on VCE exploration. RESULTS GI bleeding source localization was significantly improved when including VCE exploration (P < .01), even in patients without history of angiodysplasia (P < .05). Patients with angiodysplasia had more GI bleeding recurrences (P < .01). A lower recurrence-free survival was observed in patients with angiodysplasia (log-rank test, P = .02), and especially when lesions were located in the small bowel (log-rank test, P < .01), even after endoscopic treatment with argon plasma coagulation (log-rank test, P < .01). CONCLUSION VCE should be more systematically used in VWD patients with unexplained or recurrent GI bleeding looking for angiodysplasia eligible to endoscopic treatment or at high risk of relapse.
Collapse
Affiliation(s)
- Antoine Rauch
- Department of Hematology and Transfusion, CHU Lille, Institut d'Hématologie Transfusion, Lille, France
| | - Camille Paris
- Department of Hematology and Transfusion, CHU Lille, Institut d'Hématologie Transfusion, Lille, France
| | | | - Julien Branche
- Department of Gastroenterology, CHU Lille, Lille, France
| | - Roseline D'Oiron
- Center for Hemophilia and Rare Congenital Bleeding Disorders, University Hospital Paris-Sud, AP-HP, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | | - Valérie Gay
- CH Métropole Savoie - Site de Chambéry, Chambéry, France
| | | | | | - Céline Falaise
- Centre régional de traitement des hémophiles, Hôpital de La Timone, AP-HM, Marseille, France
| | | | - Catherine Marichez
- Department of Hematology and Transfusion, CHU Lille, Institut d'Hématologie Transfusion, Lille, France
| | - Benedicte Pradines
- Department of Hematology and Transfusion, CHU Lille, Institut d'Hématologie Transfusion, Lille, France
| | - Christophe Zawadzki
- Department of Hematology and Transfusion, CHU Lille, Institut d'Hématologie Transfusion, Lille, France
| | - Nathalie Itzhar Baikian
- Laboratoire d'Hématologie, GH St-Louis Lariboisière F.Widal - Hôpital Lariboisière APHP, Paris, France
| | | | - Jenny Goudemand
- Department of Hematology and Transfusion, CHU Lille, Institut d'Hématologie Transfusion, Lille, France
| | - Romain Gerard
- Department of Gastroenterology, CHU Lille, Lille, France
| | - Sophie Susen
- Department of Hematology and Transfusion, CHU Lille, Institut d'Hématologie Transfusion, Lille, France
| |
Collapse
|
26
|
de Boer S, Bowman M, Notley C, Mo A, Lima P, de Jong A, Dirven R, Weijers E, Lillicrap D, James P, Eikenboom J. Endothelial characteristics in healthy endothelial colony forming cells; generating a robust and valid ex vivo model for vascular disease. J Thromb Haemost 2020; 18:2721-2731. [PMID: 32654420 PMCID: PMC7590112 DOI: 10.1111/jth.14998] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 06/14/2020] [Accepted: 07/06/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Endothelial colony forming cells (ECFCs) derived from peripheral blood can be used to analyze the pathophysiology of vascular diseases ex vivo. However, heterogeneity is observed between ECFC clones and this variability needs to be understood and standardized for ECFCs to be used as a cell model for applications in vascular studies. OBJECTIVE Determine reference characteristics of healthy control ECFCs to generate a valid ex vivo model for vascular disease. METHODS Putative ECFCs (n = 47) derived from 21 individual healthy subjects were studied for cell morphology and specific cell characteristics. Clones were analyzed for the production and secretion of von Willebrand factor (VWF), cell proliferation, and the expression of endothelial cell markers. RESULTS Based on morphology, clones were categorized into three groups. Group 1 consisted of clones with classic endothelial cell morphology, whereas groups 2 and 3 contained less condensed cells with increasing cell sizes. All clones had comparable endothelial cell surface expression profiles, with low levels of non-endothelial markers. However, a decrease in CD31 and a group-related increase in CD309 and CD45 expression, combined with a decrease in cell proliferation and VWF production and secretion, was observed in clones in group 3 and to a lesser extent in group 2. CONCLUSIONS We observed group-related variations in endothelial cell characteristics when clones lacked the classic endothelial cell morphology. Despite this variation, clones in all groups expressed endothelial cell surface markers. Provided that clones with similar characteristics are compared, we believe ECFCs are a valid ex vivo model to study vascular disease.
Collapse
Affiliation(s)
- Suzan de Boer
- Division of Thrombosis and HemostasisDepartment of Internal MedicineEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | | | - Colleen Notley
- Department of Pathology and Molecular MedicineQueen’s UniversityKingstonONCanada
| | - Aomei Mo
- Department of Pathology and Molecular MedicineQueen’s UniversityKingstonONCanada
| | - Patricia Lima
- Department of MedicineQueen’s UniversityKingstonONCanada
| | - Annika de Jong
- Division of Thrombosis and HemostasisDepartment of Internal MedicineEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Richard Dirven
- Division of Thrombosis and HemostasisDepartment of Internal MedicineEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Ester Weijers
- Division of Thrombosis and HemostasisDepartment of Internal MedicineEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - David Lillicrap
- Department of Pathology and Molecular MedicineQueen’s UniversityKingstonONCanada
| | - Paula James
- Department of MedicineQueen’s UniversityKingstonONCanada
| | - Jeroen Eikenboom
- Division of Thrombosis and HemostasisDepartment of Internal MedicineEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
27
|
Liao G, Zheng K, Shorr R, Allan DS. Human endothelial colony-forming cells in regenerative therapy: A systematic review of controlled preclinical animal studies. Stem Cells Transl Med 2020; 9:1344-1352. [PMID: 32681814 PMCID: PMC7581447 DOI: 10.1002/sctm.20-0141] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/11/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Endothelial colony‐forming cells (ECFCs) hold significant promise as candidates for regenerative therapy of vascular injury. Existing studies remain largely preclinical and exhibit marked design heterogeneity. A systematic review of controlled preclinical trials of human ECFCs is needed to guide future study design and to accelerate clinical translation. A systematic search of Medline and EMBASE on 1 April 2019 returned 3131 unique entries of which 66 fulfilled the inclusion criteria. Most studies used ECFCs derived from umbilical cord or adult peripheral blood. Studies used genetically modified immunodeficient mice (n = 52) and/or rats (n = 16). ECFC phenotypes were inconsistently characterized. While >90% of studies used CD31+ and CD45−, CD14− was demonstrated in 73% of studies, CD146+ in 42%, and CD10+ in 35%. Most disease models invoked ischemia. Peripheral vascular ischemia (n = 29), central nervous system ischemia (n = 14), connective tissue injury (n = 10), and cardiovascular ischemia and reperfusion injury (n = 7) were studied most commonly. Studies showed predominantly positive results; only 13 studies reported ≥1 outcome with null results, three reported only null results, and one reported harm. Quality assessment with SYRCLE revealed potential sources of bias in most studies. Preclinical ECFC studies are associated with benefit across several ischemic conditions in animal models, although combining results is limited by marked heterogeneity in study design. In particular, characterization of ECFCs varied and aspects of reporting introduced risk of bias in most studies. More studies with greater focus on standardized cell characterization and consistency of the disease model are needed.
Collapse
Affiliation(s)
- Gary Liao
- Clinical Epidemiology and Regenerative Medicine Programs, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Katina Zheng
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Risa Shorr
- Information Services, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - David S Allan
- Clinical Epidemiology and Regenerative Medicine Programs, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
28
|
Von Willebrand Disease: From In Vivo to In Vitro Disease Models. Hemasphere 2020; 3:e297. [PMID: 31942548 PMCID: PMC6919471 DOI: 10.1097/hs9.0000000000000297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/04/2019] [Indexed: 01/28/2023] Open
Abstract
Von Willebrand factor (VWF) plays an essential role in primary hemostasis and is exclusively synthesized and stored in endothelial cells and megakaryocytes. Upon vascular injury, VWF is released into the circulation where this multimeric protein is required for platelet adhesion. Defects of VWF lead to the most common inherited bleeding disorder von Willebrand disease (VWD). Three different types of VWD exist, presenting with varying degrees of bleeding tendencies. The pathophysiology of VWD can be investigated by examining the synthesis, storage and secretion in VWF producing cells. These cells can either be primary VWF producing cells or transfected heterologous cell models. For many years transfected heterologous cells have been used successfully to elucidate many aspects of VWF synthesis. However, those cells do not fully reflect the characteristics of primary cells. Obtaining primary endothelial cells or megakaryocytes with a VWD phenotype, requires invasive procedures, such as vessel collection or a bone marrow biopsy. A more recent and promising development is the isolation of endothelial colony forming cells (ECFCs) from peripheral blood as a true-to-nature cell model. Alternatively, various animal models are available but limiting, therefore, new approaches are needed to study VWD and other bleeding disorders. A potential versatile source of endothelial cells and megakaryocytes could be induced pluripotent stem cells (iPSCs). This review gives an overview of models that are available to study VWD and VWF and will discuss novel approaches that can be considered to improve the understanding of the structural and functional mechanisms underlying this disease.
Collapse
|
29
|
Schillemans M, Kat M, Westeneng J, Gangaev A, Hofman M, Nota B, van Alphen FPJ, de Boer M, van den Biggelaar M, Margadant C, Voorberg J, Bierings R. Alternative trafficking of Weibel-Palade body proteins in CRISPR/Cas9-engineered von Willebrand factor-deficient blood outgrowth endothelial cells. Res Pract Thromb Haemost 2019; 3:718-732. [PMID: 31624792 PMCID: PMC6782018 DOI: 10.1002/rth2.12242] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/24/2019] [Accepted: 06/10/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Synthesis of the hemostatic protein von Willebrand factor (VWF) drives formation of endothelial storage organelles called Weibel-Palade bodies (WPBs). In the absence of VWF, angiogenic and inflammatory mediators that are costored in WPBs are subject to alternative trafficking routes. In patients with von Willebrand disease (VWD), partial or complete absence of VWF/WPBs may lead to additional bleeding complications, such as angiodysplasia. Studies addressing the role of VWF using VWD patient-derived blood outgrowth endothelial cells (BOECs) have reported conflicting results due to the intrinsic heterogeneity of patient-derived BOECs. OBJECTIVE To generate a VWF-deficient endothelial cell model using clustered regularly interspaced short palindromic repeats (CRISPR) genome engineering of blood outgrowth endothelial cells. METHODS We used CRISPR/CRISPR-associated protein 9 editing in single-donor cord blood-derived BOECs (cbBOECs) to generate clonal VWF -/- cbBOECs. Clones were selected using high-throughput screening, VWF mutations were validated by sequencing, and cells were phenotypically characterized. RESULTS Two VWF -/- BOEC clones were obtained and were entirely devoid of WPBs, while their overall cell morphology was unaltered. Several WPB proteins, including CD63, syntaxin-3 and the cargo proteins angiopoietin (Ang)-2, interleukin (IL)-6, and IL-8 showed alternative trafficking and secretion in the absence of VWF. Interestingly, Ang-2 was relocated to the cell periphery and colocalized with Tie-2. CONCLUSIONS CRISPR editing of VWF provides a robust method to create VWF- deficient BOECs that can be directly compared to their wild-type counterparts. Results obtained with our model system confirmed alternative trafficking of several WPB proteins in the absence of VWF and support the theory that increased Ang-2/Tie-2 interaction contributes to angiogenic abnormalities in VWD patients.
Collapse
Affiliation(s)
- Maaike Schillemans
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marije Kat
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jurjen Westeneng
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Anastasia Gangaev
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Menno Hofman
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Benjamin Nota
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Floris P. J. van Alphen
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martin de Boer
- Blood Cell ResearchSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Maartje van den Biggelaar
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Coert Margadant
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jan Voorberg
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Experimental Vascular MedicineAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ruben Bierings
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- HematologyErasmus University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
30
|
de Jong A, Weijers E, Dirven R, de Boer S, Streur J, Eikenboom J. Variability of von Willebrand factor-related parameters in endothelial colony forming cells. J Thromb Haemost 2019; 17:1544-1554. [PMID: 31265169 PMCID: PMC6852380 DOI: 10.1111/jth.14558] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/03/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023]
Abstract
Essentials Endothelial colony forming cells (ECFCs) are a powerful tool to study vascular diseases ex vivo. Separate ECFC lines show variations in morphology and von Willebrand factor-related parameters. Maximum cell density is correlated with von Willebrand factor expression in ECFCs. Variations in ECFC lines are dependent on the age and mesenchymal state of the cells. ABSTRACT: Background Endothelial colony forming cells (ECFCs) are cultured endothelial cells derived from peripheral blood. ECFCs are a powerful tool to study pathophysiological mechanisms underlying vascular diseases, including von Willebrand disease. In prior research, however, large variations between ECFC lines were observed in, among others, von Willebrand factor (VWF) expression. Objective Understand the relation between phenotypic characteristics and VWF-related parameters of healthy control ECFCs. Methods ECFC lines (n = 16) derived from six donors were studied at maximum cell density. Secreted and intracellular VWF antigen were measured by ELISA. The angiogenic capacity of ECFCs was investigated by the Matrigel tube formation assay. Differences in expression of genes involved in angiogenesis, aging, and endothelial to mesenchymal transition (EndoMT) were measured by quantitative PCR. Results Different ECFC lines show variable morphologies and cell density at maximum confluency and cell lines with a low maximum cell density show a mixed and more mesenchymal phenotype. We identified a significant positive correlation between maximum cell density and VWF production, both at protein and mRNA level. Also, significant correlations were observed between maximum cell density and several angiogenic, aging and EndoMT parameters. Conclusions We observed variations in morphology, maximum cell density, VWF production, and angiogenic potential between healthy control ECFCs. These variations seem to be attributable to differences in aging and EndoMT. Because variations correlate with cell density, we believe that ECFCs maintain a powerful tool to study vascular diseases. It is however important to compare cell lines with the same characteristics and perform experiments at maximum cell density.
Collapse
Affiliation(s)
- Annika de Jong
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Ester Weijers
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Richard Dirven
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Suzan de Boer
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Jasmin Streur
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| | - Jeroen Eikenboom
- Department of Internal medicinedivision of Thrombosis and HemostasisEinthoven Laboratory for Vascular and Regenerative MedicineLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
31
|
Mathur T, Singh KA, R Pandian NK, Tsai SH, Hein TW, Gaharwar AK, Flanagan JM, Jain A. Organ-on-chips made of blood: endothelial progenitor cells from blood reconstitute vascular thromboinflammation in vessel-chips. LAB ON A CHIP 2019; 19:2500-2511. [PMID: 31246211 PMCID: PMC6650325 DOI: 10.1039/c9lc00469f] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Development of therapeutic approaches to treat vascular dysfunction and thrombosis at disease- and patient-specific levels is an exciting proposed direction in biomedical research. However, this cannot be achieved with animal preclinical models alone, and new in vitro techniques, like human organ-on-chips, currently lack inclusion of easily obtainable and phenotypically-similar human cell sources. Therefore, there is an unmet need to identify sources of patient primary cells and apply them in organ-on-chips to increase personalized mechanistic understanding of diseases and to assess drugs. In this study, we provide a proof-of-feasibility of utilizing blood outgrowth endothelial cells (BOECs) as a disease-specific primary cell source to analyze vascular inflammation and thrombosis in vascular organ-chips or "vessel-chips". These blood-derived BOECs express several factors that confirm their endothelial identity. The vessel-chips are cultured with BOECs from healthy or diabetic patients and form an intact 3D endothelial lumen. Inflammation of the BOEC endothelium with exogenous cytokines reveals vascular dysfunction and thrombosis in vitro similar to in vivo observations. Interestingly, our study with vessel-chips also reveals that unstimulated BOECs of type 1 diabetic pigs show phenotypic behavior of the disease - high vascular dysfunction and thrombogenicity - when compared to control BOECs or normal primary endothelial cells. These results demonstrate the potential of organ-on-chips made from autologous endothelial cells obtained from blood in modeling vascular pathologies and therapeutic outcomes at a disease and patient-specific level.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| | - Kanwar Abhay Singh
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| | - Navaneeth K R Pandian
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| | - Shu-Huai Tsai
- Department of Medical Physiology, Texas A&M University System Health Science Center, Temple, USA
| | - Travis W Hein
- Department of Medical Physiology, Texas A&M University System Health Science Center, Temple, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA. and Center for Remote Health Technologies and Systems, Texas A&M University, College Station, USA and Department of Materials Science and Engineering, Texas A&M University, College Station, USA
| | - Jonathan M Flanagan
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St, College Station, TX 77843, USA.
| |
Collapse
|
32
|
Smadja DM, Melero-Martin JM, Eikenboom J, Bowman M, Sabatier F, Randi AM. Standardization of methods to quantify and culture endothelial colony-forming cells derived from peripheral blood: Position paper from the International Society on Thrombosis and Haemostasis SSC. J Thromb Haemost 2019; 17:1190-1194. [PMID: 31119878 PMCID: PMC7028216 DOI: 10.1111/jth.14462] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/18/2019] [Indexed: 12/27/2022]
Affiliation(s)
- David M. Smadja
- Université Paris Descartes, Paris, France
- Faculté de Pharmacie de Paris, INSERM UMR-S 1140,
Paris, France
- Hematology Department, AP-HP, Hôpital
Européen Georges Pompidou, Paris, France
- Laboratory of Biosurgical Research, Carpentier Foundation,
Hôpital Européen Georges Pompidou, Paris, France
| | - Juan M. Melero-Martin
- Department of Cardiac Surgery, Boston Children’s
Hospital, Boston, Massachusetts
- Department of Surgery, Harvard Medical School, Boston,
Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Jeroen Eikenboom
- Einthoven Laboratory for Vascular and Regenerative
Medicine, Department of Thrombosis and Hemostasis, Leiden University Medical Center,
Leiden, the Netherlands
| | - Mackenzie Bowman
- Department of Medicine, Queen’s University,
Kingston, Ontario, Canada
| | - Florence Sabatier
- C2VN Aix Marseille University, INSERM, INRA, Marseille,
France
- Laboratory of Cell Therapy, INSERM CBT-1409, CHU La
Conception, AP-HM, Marseille, France
| | - Anna M. Randi
- Imperial Centre for Translational and Experimental
Medicine, National Heart and Lung Institute, Imperial College London, London,
UK
| |
Collapse
|
33
|
|
34
|
Significant gynecological bleeding in women with low von Willebrand factor levels. Blood Adv 2019; 2:1784-1791. [PMID: 30042144 DOI: 10.1182/bloodadvances.2018017418] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/19/2018] [Indexed: 01/11/2023] Open
Abstract
Gynecological bleeding is frequently reported in women with von Willebrand disease (VWD). Low von Willebrand factor (VWF) may be associated with significant bleeding phenotype despite only mild plasma VWF reductions. The contribution of gynecological bleeding to this phenotype has yet to be described. The optimal clinical bleeding assessment tool (BAT) to evaluate bleeding remains unclear. Using a standardized approach to phenotypic assessment, we evaluated gynecological bleeding and directly compared the Condensed Molecular and Clinical Markers for the Diagnosis and Management of type 1 VWD (Condensed MCMDM-1 VWD) and International Society on Thrombosis and Haemostasis (ISTH) BAT scores in 120 women enrolled in the Low von Willebrand in Ireland Cohort study. Heavy menstrual bleeding (HMB) was reported in 89% of female participants; 45.8% developed iron deficiency. Using identical data, Condensed MCMDM-1 VWD menorrhagia domain scores were significantly lower than ISTH BAT scores (2 vs 3; P < .0001), the discrepant results related to 40% of women not seeking medical consultation for HMB, reducing the sensitivity of the Condensed score. For those who reported HMB to physicians, the low VWF diagnosis was not expedited (age at diagnosis 34.2 vs 33.4 years in women failing to present; P = .7). Postpartum hemorrhage (PPH) was self-reported in 63.5% of parous women (n = 74); 21.6% required transfusion, critical care, radiological, or surgical intervention. Our data demonstrate that gynecological bleeding is frequently reported in women with low VWF; despite pregnancy-related increases in plasma VWF levels, these women may experience PPH. Defining the optimal management approach for these patients requires further research. This trial was registered at www.clinicaltrials.gov as #NCT03167320.
Collapse
|
35
|
Vasculogenic Stem and Progenitor Cells in Human: Future Cell Therapy Product or Liquid Biopsy for Vascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:215-237. [PMID: 31898789 DOI: 10.1007/978-3-030-31206-0_11] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
New blood vessel formation in adults was considered to result exclusively from sprouting of preexisting endothelial cells, a process referred to angiogenesis. Vasculogenesis, the formation of new blood vessels from endothelial progenitor cells, was thought to occur only during embryonic life. Discovery of adult endothelial progenitor cells (EPCs) in 1997 opened the door for cell therapy in vascular disease. Endothelial progenitor cells contribute to vascular repair and are now well established as postnatal vasculogenic cells in humans. It is now admitted that endothelial colony-forming cells (ECFCs) are the vasculogenic subtype. ECFCs could be used as a cell therapy product and also as a liquid biopsy in several vascular diseases or as vector for gene therapy. However, despite a huge interest in these cells, their tissue and molecular origin is still unclear. We recently proposed that endothelial progenitor could come from very small embryonic-like stem cells (VSELs) isolated in human from CD133 positive cells. VSELs are small dormant stem cells related to migratory primordial germ cells. They have been described in bone marrow and other organs. This chapter discusses the reported findings from in vitro data and also preclinical studies that aimed to explore stem cells at the origin of vasculogenesis in human and then explore the potential use of ECFCs to promote newly formed vessels or serve as liquid biopsy to understand vascular pathophysiology and in particular pulmonary disease and haemostasis disorders.
Collapse
|
36
|
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: 48] [Impact Index Per Article: 6.9] [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
|
37
|
Leebeek FWG, Susen S. Von Willebrand disease: Clinical conundrums. Haemophilia 2018; 24 Suppl 6:37-43. [PMID: 29878657 DOI: 10.1111/hae.13508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2018] [Indexed: 12/22/2022]
Affiliation(s)
- F W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S Susen
- Inserm, U1011, Hemostasis and Transfusion Department, French Reference Center for Von Willebrand Disease, CHU Lille, Lille, France
| |
Collapse
|
38
|
Paschalaki KE, Randi AM. Recent Advances in Endothelial Colony Forming Cells Toward Their Use in Clinical Translation. Front Med (Lausanne) 2018; 5:295. [PMID: 30406106 PMCID: PMC6205967 DOI: 10.3389/fmed.2018.00295] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/28/2018] [Indexed: 12/17/2022] Open
Abstract
The term “Endothelial progenitor cell” (EPC) has been used to describe multiple cell populations that express endothelial surface makers and promote vascularisation. However, the only population that has all the characteristics of a real “EPC” is the Endothelial Colony Forming Cells (ECFC). ECFC possess clonal proliferative potential, display endothelial and not myeloid cell surface markers, and exhibit pronounced postnatal vascularisation ability in vivo. ECFC have been used to investigate endothelial molecular dysfunction in several diseases, as they give access to endothelial cells from patients in a non-invasive way. ECFC also represent a promising tool for revascularization of damaged tissue. Here we review the translational applications of ECFC research. We discuss studies which have used ECFC to investigate molecular endothelial abnormalities in several diseases and review the evidence supporting the use of ECFC for autologous cell therapy, gene therapy and tissue regeneration. Finally, we discuss ways to improve the therapeutic efficacy of ECFC in clinical applications, as well as the challenges that must be overcome to use ECFC in clinical trials for regenerative approaches.
Collapse
Affiliation(s)
- Koralia E Paschalaki
- Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Anna M Randi
- Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
39
|
Ecklu-Mensah G, Olsen RW, Bengtsson A, Ofori MF, Hviid L, Jensen ATR, Adams Y. Blood outgrowth endothelial cells (BOECs) as a novel tool for studying adhesion of Plasmodium falciparum-infected erythrocytes. PLoS One 2018; 13:e0204177. [PMID: 30300360 PMCID: PMC6177148 DOI: 10.1371/journal.pone.0204177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/03/2018] [Indexed: 11/19/2022] Open
Abstract
The lack of suitable animal models for the study of cytoadhesion of P. falciparum-infected erythrocytes (IEs) has necessitated in vitro studies employing a range of cell lines of either human tumour origin (e.g., BeWo and C32 cells) or non-human origin (e.g., CHO cells). Of the human cells available, many were isolated from adults, or derived from a pool of donors (e.g., HBEC-5i). Here we demonstrate, for the first time, the successful isolation of blood outgrowth endothelial cells (BOECs) from frozen stabilates of peripheral blood mononuclear cells obtained from small-volume peripheral blood samples from paediatric malaria patients. BOECs are a sub-population of human endothelial cells, found within the peripheral blood. We demonstrate that these cells express receptors such as Intercellular Adhesion Molecule 1 (ICAM-1/CD54), Endothelial Protein C Receptor (EPCR/CD201), platelet/endothelial cell adhesion molecule 1 (PECAM-1/CD31), Thrombomodulin (CD141), and support adhesion of P. falciparum IEs.
Collapse
Affiliation(s)
- Gertrude Ecklu-Mensah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
- Centre for Medical Parasitology at Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca W. Olsen
- Centre for Medical Parasitology at Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anja Bengtsson
- Centre for Medical Parasitology at Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael F. Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Lars Hviid
- Centre for Medical Parasitology at Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Anja T. R. Jensen
- Centre for Medical Parasitology at Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yvonne Adams
- Centre for Medical Parasitology at Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
40
|
Randi AM, Smith KE, Castaman G. von Willebrand factor regulation of blood vessel formation. Blood 2018; 132:132-140. [PMID: 29866817 PMCID: PMC6182264 DOI: 10.1182/blood-2018-01-769018] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023] Open
Abstract
Several important physiological processes, from permeability to inflammation to hemostasis, take place at the vessel wall and are regulated by endothelial cells (ECs). Thus, proteins that have been identified as regulators of one process are increasingly found to be involved in other vascular functions. Such is the case for von Willebrand factor (VWF), a large glycoprotein best known for its critical role in hemostasis. In vitro and in vivo studies have shown that lack of VWF causes enhanced vascularization, both constitutively and following ischemia. This evidence is supported by studies on blood outgrowth EC (BOEC) from patients with lack of VWF synthesis (type 3 von Willebrand disease [VWD]). The molecular pathways are likely to involve VWF binding partners, such as integrin αvβ3, and components of Weibel-Palade bodies, such as angiopoietin-2 and galectin-3, whose storage is regulated by VWF; these converge on the master regulator of angiogenesis and endothelial homeostasis, vascular endothelial growth factor signaling. Recent studies suggest that the roles of VWF may be tissue specific. The ability of VWF to regulate angiogenesis has clinical implications for a subset of VWD patients with severe, intractable gastrointestinal bleeding resulting from vascular malformations. In this article, we review the evidence showing that VWF is involved in blood vessel formation, discuss the role of VWF high-molecular-weight multimers in regulating angiogenesis, and review the value of studies on BOEC in developing a precision medicine approach to validate novel treatments for angiodysplasia in congenital VWD and acquired von Willebrand syndrome.
Collapse
Affiliation(s)
- Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Koval E Smith
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Giancarlo Castaman
- Center for Bleeding Disorders and Coagulation, Department of Oncology, Careggi University Hospital, Florence, Italy
| |
Collapse
|
41
|
de Jong A, Dirven RJ, Oud JA, Tio D, van Vlijmen BJM, Eikenboom J. Correction of a dominant-negative von Willebrand factor multimerization defect by small interfering RNA-mediated allele-specific inhibition of mutant von Willebrand factor. J Thromb Haemost 2018; 16:1357-1368. [PMID: 29734512 DOI: 10.1111/jth.14140] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Indexed: 01/30/2023]
Abstract
Essentials Substitution therapy for von Willebrand (VW) disease leaves mutant VW factor (VWF) unhindered. Presence of mutant VWF may negatively affect phenotypes despite treatment. Inhibition of VWF by allele-specific siRNAs targeting single-nucleotide polymorphisms is effective. Allele-specific inhibition of VWF p.Cys2773Ser improves multimerization. SUMMARY Background Treatment of the bleeding disorder von Willebrand disease (VWD) focuses on increasing von Willebrand factor (VWF) levels by administration of desmopressin or VWF-containing concentrates. Both therapies leave the production of mutant VWF unhindered, which may have additional consequences, such as thrombocytopenia in patients with VWD type 2B, competition between mutant and normal VWF for platelet receptors, and the potential development of intestinal angiodysplasia. Most cases of VWD are caused by dominant-negative mutations in VWF, and we hypothesize that diminishing expression of mutant VWF positively affects VWD phenotypes. Objectives To investigate allele-specific inhibition of VWF by applying small interfering RNAs (siRNAs) targeting common single-nucleotide polymorphisms (SNPs) in VWF. This approach allows allele-specific knockdown irrespective of the mutations causing VWD. Methods Four SNPs with a high predicted heterozygosity within VWF were selected, and siRNAs were designed against both alleles of the four SNPs. siRNA efficiency, allele specificity and siRNA-mediated phenotypic improvements were determined in VWF-expressing HEK293 cells. Results Twelve siRNAs were able to efficiently inhibit single VWF alleles in HEK293 cells that stably produce VWF. Transient cotransfections of these siRNAs with two VWF alleles resulted in a clear preference for the targeted allele over the untargeted allele for 11 siRNAs. We also demonstrated siRNA-mediated phenotypic improvement of the VWF multimerization pattern of the VWD type 2A mutation VWF p.Cys2773Ser. Conclusions Allele-specific siRNAs are able to distinguish VWF alleles on the basis of one nucleotide variation, and are able to improve a severe multimerization defect caused by VWF p.Cys2773Ser. This holds promise for the therapeutic application of allele-specific siRNAs in dominant-negative VWD.
Collapse
Affiliation(s)
- A de Jong
- Department of Internal Medicine (Thrombosis and Hemostasis), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - R J Dirven
- Department of Internal Medicine (Thrombosis and Hemostasis), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - J A Oud
- Department of Internal Medicine (Thrombosis and Hemostasis), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - D Tio
- Department of Internal Medicine (Thrombosis and Hemostasis), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - B J M van Vlijmen
- Department of Internal Medicine (Thrombosis and Hemostasis), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - J Eikenboom
- Department of Internal Medicine (Thrombosis and Hemostasis), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
42
|
Abnormal angiogenesis in blood outgrowth endothelial cells derived from von Willebrand disease patients. Blood Coagul Fibrinolysis 2018; 28:521-533. [PMID: 28362648 DOI: 10.1097/mbc.0000000000000635] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
: Bleeding associated with angiodysplasia is a common, often intractable complication in patients with von Willebrand disease (VWD). von Willebrand factor (VWF), the protein deficient or defective in VWD, is a negative regulator of angiogenesis, which may explain the pathologic blood vessel growth in VWD. This study explores the normal range of angiogenesis in blood outgrowth endothelial cells (BOECs) derived from healthy donors and compares this to angiogenesis in BOECs from VWD patients of all types and subtypes. BOECs were assessed for VWF and angiopoietin-2 (Ang-2) gene expression, secretion, and storage. To explore angiogenic potential, we characterized cellular proliferation, matrix protein adhesion, migration, and tubule formation. We found great angiogenic variability in VWD BOECs with respect to each of the angiogenesis parameters. However, type 1 and 3 VWD BOECs had higher Ang-2 secretion associated with impaired endothelial cell migration velocity and enhanced directionality. Type 2A and 2B BOECs were the most proliferative and multiple VWD BOECs had impaired tubule formation in Matrigel. This study highlights the angiogenic variability in BOECs derived from VWD patients. Abnormal cell proliferation, migration, and increased Ang-2 secretion are common features of VWD BOECs. Despite the many abnormalities of VWD BOECs, significant heterogeneity among individual VWD phenotypes precludes a simple description of relationship between VWD type and in vitro surrogates for angiodysplasia.
Collapse
|
43
|
Mitchell JA, Knowles RB, Kirkby NS, Reed DM, Edin ML, White WE, Chan MV, Longhurst H, Yaqoob MM, Milne GL, Zeldin DC, Warner TD. Kidney Transplantation in a Patient Lacking Cytosolic Phospholipase A 2 Proves Renal Origins of Urinary PGI-M and TX-M. Circ Res 2018; 122:555-559. [PMID: 29298774 PMCID: PMC5816977 DOI: 10.1161/circresaha.117.312144] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/14/2017] [Accepted: 12/20/2017] [Indexed: 01/31/2023]
Abstract
RATIONALE The balance between vascular prostacyclin, which is antithrombotic, and platelet thromboxane A2, which is prothrombotic, is fundamental to cardiovascular health. Prostacyclin and thromboxane A2 are formed after the concerted actions of cPLA2α (cytosolic phospholipase A2) and COX (cyclooxygenase). Urinary 2,3-dinor-6-keto-PGF1α (PGI-M) and 11-dehydro-TXB2 (TX-M) have been taken as biomarkers of prostacyclin and thromboxane A2 formation within the circulation and used to explain COX biology and patient phenotypes, despite concerns that urinary PGI-M and TX-M originate in the kidney. OBJECTIVE We report data from a remarkable patient carrying an extremely rare genetic mutation in cPLA2α, causing almost complete loss of prostacyclin and thromboxane A2, who was transplanted with a normal kidney resulting in an experimental scenario of whole-body cPLA2α knockout, kidney-specific knockin. By studying this patient, we can determine definitively the contribution of the kidney to the productions of PGI-M and TX-M and test their validity as markers of prostacyclin and thromboxane A2 in the circulation. METHODS AND RESULTS Metabolites were measured using liquid chromatography-tandem mass spectrometry. Endothelial cells were grown from blood progenitors. Before kidney transplantation, the patient's endothelial cells and platelets released negligible levels of prostacyclin (measured as 6-keto-prostaglandin F1α) and thromboxane A2 (measured as TXB2), respectively. Likewise, the urinary levels of PGI-M and TX-M were very low. After transplantation and the establishment of normal renal function, the levels of PGI-M and TX-M in the patient's urine rose to within normal ranges, whereas endothelial production of prostacyclin and platelet production of thromboxane A2 remained negligible. CONCLUSIONS These data show that PGI-M and TX-M can be derived exclusively from the kidney without contribution from prostacyclin made by endothelial cells or thromboxane A2 by platelets in the general circulation. Previous work relying on urinary metabolites of prostacyclin and thromboxane A2 as markers of whole-body endothelial and platelet function now requires reevaluation.
Collapse
Affiliation(s)
- Jane A Mitchell
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Rebecca B Knowles
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Nicholas S Kirkby
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Daniel M Reed
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Matthew L Edin
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - William E White
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Melissa V Chan
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Hilary Longhurst
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Magdi M Yaqoob
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Ginger L Milne
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Darryl C Zeldin
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.)
| | - Timothy D Warner
- From the National Heart and Lung Institute, Imperial College London, United Kingdom (J.A.M., N.S.K., D.M.R.); Blizard Institute, Queen Mary University of London, United Kingdom (R.B.K., W.E.W., M.V.C., M.M.Y., T.D.W.); National Institute for Environmental Health Sciences, Research Triangle, NC (M.L.E., D.C.Z.); Department of Nephrology (W.E.W., M.M.Y.) and Immunology Department (H.L.), Barts Health NHS Trust, London, United Kingdom; and Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, TN (G.L.M.).
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW In the last nine decades, large advances have been made toward the characterization of the pathogenic basis and clinical management of von Willebrand disease (VWD), the most prevalent inherited bleeding disorder. Pathological variations at the von Willebrand factor (VWF) locus present as a range of both quantitative and qualitative abnormalities that make up the complex clinical spectrum of VWD. This review describes the current understanding of the pathobiological basis of VWD. RECENT FINDINGS The molecular basis of type 2 (qualitative abnormalities) and type 3 VWD (total quantitative deficiency) have been well characterized in recent decades. However, knowledge of type 1 VWD (partial quantitative deficiency) remains incomplete because of the allelic and locus heterogeneity of this trait, and is complicated by genetic variability at the VWF gene, interactions between the VWF gene and the environment, and the involvement of external modifying loci. Recent genome wide association studies and linkage analyses have sought to identify additional genes that modify the type 1 VWD phenotype. SUMMARY Understanding the pathogenic basis of VWD will facilitate the development of novel treatment regimens for this disorder, and improve the ability to provide complementary molecular diagnostics for type 1 VWD.
Collapse
|
45
|
de Jong A, Eikenboom J. Von Willebrand disease mutation spectrum and associated mutation mechanisms. Thromb Res 2017; 159:65-75. [PMID: 28987708 DOI: 10.1016/j.thromres.2017.09.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/13/2017] [Accepted: 09/22/2017] [Indexed: 01/24/2023]
Abstract
Von Willebrand disease (VWD) is a bleeding disorder that is mainly caused by mutations in the multimeric protein von Willebrand factor (VWF). These mutations may lead to deficiencies in plasma VWF or dysfunctional VWF. VWF is a heterogeneous protein and over the past three decades, hundreds of VWF mutations have been identified. In this review we have organized all reported mutations, spanning a timeline from the late eighties until early 2017. This resulted in an overview of 750 unique mutations that are divided over the VWD types 1, 2A, 2B, 2M, 2N and 3. For many of these mutations the disease-causing effects have been characterized in vitro through expression studies, ex vivo by analysis of patient-derived endothelial cells, as well as in animal or (bio)physical models. Here we describe the mechanisms associated with the VWF mutations per VWD type.
Collapse
Affiliation(s)
- Annika de Jong
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
46
|
Michels A, Swystun LL, Mewburn J, Albánez S, Lillicrap D. Investigating von Willebrand Factor Pathophysiology Using a Flow Chamber Model of von Willebrand Factor-platelet String Formation. J Vis Exp 2017. [PMID: 28829426 DOI: 10.3791/55917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Von Willebrand factor (VWF) is a multimeric glycoprotein coagulation factor that mediates platelet adhesion and aggregation at sites of endothelial damage and that carries factor VIII in the circulation. VWF is synthesized by endothelial cells and is either released constitutively into the plasma or is stored in specialized organelles, called Weibel-Palade bodies (WPBs), for on-demand release in response to hemostatic challenge. Procoagulant and proinflammatory stimuli can rapidly induce WPB exocytosis and VWF release. The majority of VWF released by endothelial cells circulates in the plasma; however, a proportion of VWF is anchored to the endothelial cell surface. Under conditions of physiological shear, endothelial-anchored VWF can bind to platelets, forming a VWF-platelet string that may represent the nidus of thrombus formation. A flow chamber system can be used to visually observe the release of VWF from endothelial cells and the subsequent platelet capture in a manner that is reproducible and relevant to the pathophysiology of VWF-mediated thrombus formation. Using this methodology, endothelial cells are cultured in a flow chamber and are subsequently stimulated with secretagogues to induce WPB exocytosis. Washed platelets are then perfused over the activated endothelium. The platelets are activated and subsequently bind to elongated VWF strings in the direction of fluid flow. Using extracellular histones as a procoagulant and proinflammatory stimulus, we observed increased VWF-platelet string formation on histone-treated endothelial cells compared to untreated endothelial cells. This protocol describes a quantitative, visual, and real-time assessment of the activation of VWF-platelet interactions in models of thrombosis and hemostasis.
Collapse
Affiliation(s)
- Alison Michels
- Department of Pathology and Molecular Medicine, Queen's University
| | - Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen's University
| | | | - Silvia Albánez
- Department of Pathology and Molecular Medicine, Queen's University
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University;
| |
Collapse
|
47
|
Bowman ML, Pluthero F, Tuttle A, Casey L, Li L, Christensen H, Robinson K, Lillicrap D, Kahr WH, James P. Discrepant platelet and plasma von Willebrand factor in von Willebrand disease patients with p.Pro2808Leufs*24. J Thromb Haemost 2017; 15:1403-1411. [PMID: 28453889 PMCID: PMC5949882 DOI: 10.1111/jth.13722] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Indexed: 12/23/2022]
Abstract
Essentials von Willebrand factor (VWF) is synthesized in endothelial cells and platelet precursors. Type 3 patients with Pro2808Leufs*24 have lower bleeding scores than other type 3s. The Pro2808Leufs*24 variant was examined in patient platelets and endothelial cells. Type 3s with this variant contain releaseable VWF, possibly reducing bleeding. SUMMARY Background A novel variant, p.Pro2808Leufs*24, in the von Willebrand factor (VWF) gene was previously identified in the Canadian von Willebrand disease (VWD) patient population. Clinical observations of type 3 VWD patients with this variant indicate a milder bleeding phenotype compared with other type 3 patients. Objective To assess the effect of the Pro2808Leufs*24 variant on the molecular pathogenesis of VWD and correlate this with the phenotype observed in patients. Patients/Methods Phenotypic data from individuals in the Canadian type 3 VWD study were analyzed. VWF expression in platelets and plasma was assessed via immunoblotting. Cellular expression of VWF in platelets and blood outgrowth endothelial cells (BOEC) was examined via immunofluorescence microscopy and biochemical analysis in a type 3 index case and family member with Pro2808Leufs*24. Results Twenty-six individuals with the Pro2808Leufs*24 variant (16 type 3 VWD homozygous or compound heterozygous and 10 heterozygous family members) were studied. Bleeding scores were lower in type 3 patients with Pro2808Leufs*24 compared with type 3 patients with other variants, confirming a milder bleeding phenotype. Immunoblotting of platelet lysates detected VWF in the platelets of type 3 patients with Pro2808Leufs*24. Examination of an index case detected VWF within platelets via immunofluorescence microscopy, and in vitro experiments showed that this VWF was released upon platelet activation. Patient BOECs showed decreased VWF synthesis and secretion, although some VWF-containing granules were observed. Conclusion Type 3 VWD patients with the Pro2808Leufs*24 have bioavailable platelet-derived VWF that may produce a milder bleeding phenotype than other type 3s.
Collapse
Affiliation(s)
| | - Fred.G. Pluthero
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Angie Tuttle
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Lara Casey
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Ling Li
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hilary Christensen
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - K.Sue Robinson
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Walter H.A. Kahr
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
- Departments of Paediatrics & Biochemistry, Division of Haematology/Oncology, University of Toronto and The Hospital for Sick Children, Toronto, ON, Canada
| | - Paula James
- Department of Medicine, Queen’s University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
48
|
Mohamed NA, Davies RP, Lickiss PD, Ahmetaj-Shala B, Reed DM, Gashaw HH, Saleem H, Freeman GR, George PM, Wort SJ, Morales-Cano D, Barreira B, Tetley TD, Chester AH, Yacoub MH, Kirkby NS, Moreno L, Mitchell JA. Chemical and biological assessment of metal organic frameworks (MOFs) in pulmonary cells and in an acute in vivo model: relevance to pulmonary arterial hypertension therapy. Pulm Circ 2017; 7:643-653. [PMID: 28447910 PMCID: PMC5841901 DOI: 10.1177/2045893217710224] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and debilitating condition. Despite promoting vasodilation, current drugs have a therapeutic window within which they are limited by systemic side effects. Nanomedicine uses nanoparticles to improve drug delivery and/or reduce side effects. We hypothesize that this approach could be used to deliver PAH drugs avoiding the systemic circulation. Here we report the use of iron metal organic framework (MOF) MIL-89 and PEGylated MIL-89 (MIL-89 PEG) as suitable carriers for PAH drugs. We assessed their effects on viability and inflammatory responses in a wide range of lung cells including endothelial cells grown from blood of donors with/without PAH. Both MOFs conformed to the predicted structures with MIL-89 PEG being more stable at room temperature. At concentrations up to 10 or 30 µg/mL, toxicity was only seen in pulmonary artery smooth muscle cells where both MOFs reduced cell viability and CXCL8 release. In endothelial cells from both control donors and PAH patients, both preparations inhibited the release of CXCL8 and endothelin-1 and in macrophages inhibited inducible nitric oxide synthase activity. Finally, MIL-89 was well-tolerated and accumulated in the rat lungs when given in vivo. Thus, the prototypes MIL-89 and MIL-89 PEG with core capacity suitable to accommodate PAH drugs are relatively non-toxic and may have the added advantage of being anti-inflammatory and reducing the release of endothelin-1. These data are consistent with the idea that these materials may not only be useful as drug carriers in PAH but also offer some therapeutic benefit in their own right.
Collapse
Affiliation(s)
- Nura A Mohamed
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK.,2 Heart Science Centre at Harefield Hospital, Harefield, UK.,3 Qatar Foundation Research and Development Division, Doha, Qatar
| | - Robert P Davies
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Paul D Lickiss
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Blerina Ahmetaj-Shala
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Daniel M Reed
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Hime H Gashaw
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Hira Saleem
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Gemma R Freeman
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Peter M George
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Stephen J Wort
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Daniel Morales-Cano
- 5 Department of Pharmacology, Faculty of Medicine, Universidad Complutense de Madrid- Instituto de Investigacion Sanitaria Gregorio Marañón (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Bianca Barreira
- 5 Department of Pharmacology, Faculty of Medicine, Universidad Complutense de Madrid- Instituto de Investigacion Sanitaria Gregorio Marañón (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Teresa D Tetley
- 6 Lung Cell Biology Group, National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Magdi H Yacoub
- 2 Heart Science Centre at Harefield Hospital, Harefield, UK
| | - Nicholas S Kirkby
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Laura Moreno
- 5 Department of Pharmacology, Faculty of Medicine, Universidad Complutense de Madrid- Instituto de Investigacion Sanitaria Gregorio Marañón (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Jane A Mitchell
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
49
|
Selvam S, James P. Angiodysplasia in von Willebrand Disease: Understanding the Clinical and Basic Science. Semin Thromb Hemost 2017; 43:572-580. [PMID: 28476066 DOI: 10.1055/s-0037-1599145] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Severe and intractable gastrointestinal bleeding caused by angiodysplasia is a debilitating problem for up to 20% of patients with von Willebrand disease (VWD). Currently, the lack of an optimal treatment for this recurrent problem presents an ongoing challenge for many physicians in their management of affected patients. Over the past few years, studies have pointed to a regulatory role for the hemostatic protein, von Willebrand factor (VWF), in angiogenesis, providing a novel target for the modulation of vessel development. This article will review the clinical implications and molecular pathology of angiodysplasia in VWD.
Collapse
Affiliation(s)
- Soundarya Selvam
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Paula James
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada.,Department of Medicine, Queen's University, Kingston, Canada
| |
Collapse
|
50
|
Swystun LL, James PD. Genetic diagnosis in hemophilia and von Willebrand disease. Blood Rev 2017; 31:47-56. [DOI: 10.1016/j.blre.2016.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 11/24/2022]
|