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 2024:S1538-7836(24)00639-1. [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] [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
|
Lu CY, Wu JZ, Yao HHY, Liu RJY, Li L, Pluthero FG, Freeman SA, Kahr WHA. Acidification of α-granules in megakaryocytes by vacuolar-type adenosine triphosphatase is essential for organelle biogenesis. J Thromb Haemost 2024; 22:2294-2305. [PMID: 38718926 DOI: 10.1016/j.jtha.2024.04.021] [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: 02/17/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Platelets coordinate blood coagulation at sites of vascular injury and play fundamental roles in a wide variety of (patho)physiological processes. Key to many platelet functions is the transport and secretion of proteins packaged within α-granules, organelles produced by platelet precursor megakaryocytes. Prominent among α-granule cargo are fibrinogen endocytosed from plasma and endogenously synthesized von Willebrand factor. These and other proteins are known to require acidic pH for stable packaging. Luminal acidity has been confirmed for mature α-granules isolated from platelets, but direct measurement of megakaryocyte granule acidity has not been reported. OBJECTIVES To determine the luminal pH of α-granules and their precursors in megakaryocytes and assess the requirement of vacuolar-type adenosine triphosphatase (V-ATPase) activity to establish and maintain the luminal acidity and integrity of these organelles. METHODS Cresyl violet staining was used to detect acidic granules in megakaryocytes. Endocytosis of fibrinogen tagged with the pH-sensitive fluorescent dye fluorescein isothiocyanate was used to load a subset of these organelles. Ratiometric fluorescence analysis was used to determine their luminal pH. RESULTS We show that most of the acidic granules detected in megakaryocytes appear to be α-granules/precursors, for which we established a median luminal pH of 5.2 (IQR, 5.0-5.5). Inhibition of megakaryocyte V-ATPase activity led to enlargement of cargo-containing compartments detected by fluorescence microscopy and electron microscopy. CONCLUSION These observations reveal that V-ATPase activity is required to establish and maintain a luminal acidic pH in megakaryocyte α-granules/precursors, confirming its importance for stable packaging of cargo proteins such as von Willebrand factor.
Collapse
Affiliation(s)
- Chien-Yi Lu
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Jing Ze Wu
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Helen H Y Yao
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Richard J Y Liu
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Ling Li
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Fred G Pluthero
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Spencer A Freeman
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Walter H A Kahr
- Cell Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Division of Haematology/Oncology, Department of Paediatrics, University of Toronto and The Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Seidizadeh O, Eikenboom JCJ, Denis CV, Flood VH, James P, Lenting PJ, Baronciani L, O'Donnell JS, Lillicrap D, Peyvandi F. von Willebrand disease. Nat Rev Dis Primers 2024; 10:51. [PMID: 39054329 DOI: 10.1038/s41572-024-00536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
von Willebrand disease (VWD) is the most common inherited bleeding disorder. The disorder is characterized by excessive mucocutaneous bleeding. The most common bleeding manifestations of this condition include nosebleeds, bruising, bleeding from minor wounds, menorrhagia or postpartum bleeding in women as well as bleeding after surgery. Other less frequent symptoms include gastrointestinal bleeding, haematomas or haemarthroses. VWD pathophysiology is complex and results from defects in von Willebrand factor (VWF) glycoprotein. Quantitative deficiencies are responsible for type 1 VWD with a partial decrease of VWF and type 3 with the complete absence of VWF. Qualitative abnormalities cause type 2 VWD, being further divided into types 2A, 2B, 2M and 2N. Although common, VWD is at risk of misdiagnosis, overdiagnosis and underdiagnosis owing to several factors, including complex diagnosis, variability of bleeding symptoms, presence of external variables (blood groups and other physiological modifiers such as exercise, thyroid hormones, oestrogens, and ageing), and lack of disease awareness among non-specialist health-care providers. Establishing the correct VWD diagnosis requires an array of specialized phenotypic assays and/or molecular genetic testing of the VWF gene. The management of bleeding includes increasing endogenous VWF levels with desmopressin or infusion of exogenous VWF concentrates (plasma-derived or recombinant). Fibrinolytic inhibitors, topical haemostatic agents and hormonal therapies are used as effective adjunctive measures.
Collapse
Affiliation(s)
- Omid Seidizadeh
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Jeroen C J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Cécile V Denis
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Veronica H Flood
- Department of Paediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Paula James
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Peter J Lenting
- Laboratory for Hemostasis, Inflammation & Thrombosis, Unité Mixte de Recherche 1176, Institut National de la Santé et de la Recherche Médicale, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Luciano Baronciani
- Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Flora Peyvandi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy.
| |
Collapse
|
4
|
Duranova H, Kuzelova L, Borotova P, Simora V, Fialkova V. Human Umbilical Vein Endothelial Cells as a Versatile Cellular Model System in Diverse Experimental Paradigms: An Ultrastructural Perspective. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2024; 30:419-439. [PMID: 38817111 DOI: 10.1093/mam/ozae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024]
Abstract
Human umbilical vein endothelial cells (HUVECs) are primary cells isolated from the vein of an umbilical cord, extensively used in cardiovascular studies and medical research. These cells, retaining the characteristics of endothelial cells in vivo, serve as a valuable cellular model system for understanding vascular biology, endothelial dysfunction, pathophysiology of diseases such as atherosclerosis, and responses to different drugs or treatments. Transmission electron microscopy (TEM) has been a cornerstone in revealing the detailed architecture of multiple cellular model systems including HUVECs, allowing researchers to visualize subcellular organelles, membrane structures, and cytoskeletal elements. Among them, the endoplasmic reticulum, Golgi apparatus, mitochondria, and nucleus can be meticulously examined to recognize alterations indicative of cellular responses to various stimuli. Importantly, Weibel-Palade bodies are characteristic secretory organelles found in HUVECs, which can be easily distinguished in the TEM. These distinctive structures also dynamically react to different factors through regulated exocytosis, resulting in complete or selective release of their contents. This detailed review summarizes the ultrastructural features of HUVECs and highlights the utility of TEM as a pivotal tool for analyzing HUVECs in diverse research frameworks, contributing valuable insights into the comprehension of HUVEC behavior and enriching our knowledge into the complexity of vascular biology.
Collapse
Affiliation(s)
- Hana Duranova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Lenka Kuzelova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
- Faculty of Biotechnology and Food Sciences, Institute of Biotechnology, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Petra Borotova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Veronika Simora
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Veronika Fialkova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| |
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
|
Karampini E, Doherty D, Bürgisser PE, Garre M, Schoen I, Elliott S, Bierings R, O’Donnell JS. O-glycan determinants regulate VWF trafficking to Weibel-Palade bodies. Blood Adv 2024; 8:3254-3266. [PMID: 38640438 PMCID: PMC11226974 DOI: 10.1182/bloodadvances.2023012499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT von Willebrand factor (VWF) undergoes complex posttranslational modification within endothelial cells (ECs) before secretion. This includes significant N- and O-linked glycosylation. Previous studies have demonstrated that changes in N-linked glycan structures significantly influence VWF biosynthesis. In contrast, although abnormalities in VWF O-linked glycans (OLGs) have been associated with enhanced VWF clearance, their effect on VWF biosynthesis remains poorly explored. Herein, we report a novel role for OLG determinants in regulating VWF biosynthesis and trafficking within ECs. We demonstrate that alterations in OLGs (notably reduced terminal sialylation) lead to activation of the A1 domain of VWF within EC. In the presence of altered OLG, VWF multimerization is reduced and Weibel-Palade body (WPB) formation significantly impaired. Consistently, the amount of VWF secreted from WPB after EC activation was significantly reduced in the context of O-glycosylation inhibition. Finally, altered OLG on VWF not only reduced the amount of VWF secreted after EC activation but also affected its hemostatic efficacy. Notably, VWF secreted after WPB exocytosis consisted predominantly of low molecular weight multimers, and the length of tethered VWF string formation on the surface of activated ECs was significantly reduced. In conclusion, our data therefore support the hypothesis that alterations in O-glycosylation pathways directly affect VWF trafficking within human EC. These findings are interesting given that previous studies have reported altered OLG on plasma VWF (notably increased T-antigen expression) in patients with von Willebrand disease.
Collapse
Affiliation(s)
- Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Petra E. Bürgisser
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Massimiliano Garre
- Super-Resolution Imaging Consortium, Department of Chemistry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stephanie Elliott
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ruben Bierings
- Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Coagulation Centre, St James’s Hospital, Dublin, Ireland
| |
Collapse
|
7
|
Hordijk S, Carter T, Bierings R. A new look at an old body: molecular determinants of Weibel-Palade body composition and von Willebrand factor exocytosis. J Thromb Haemost 2024; 22:1290-1303. [PMID: 38307391 DOI: 10.1016/j.jtha.2024.01.015] [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: 11/10/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/04/2024]
Abstract
Endothelial cells, forming a monolayer along blood vessels, intricately regulate vascular hemostasis, inflammatory responses, and angiogenesis. A key determinant of these functions is the controlled secretion of Weibel-Palade bodies (WPBs), which are specialized endothelial storage organelles housing a presynthesized pool of the hemostatic protein von Willebrand factor and various other hemostatic, inflammatory, angiogenic, and vasoactive mediators. This review delves into recent mechanistic insights into WPB biology, including the biogenesis that results in their unique morphology, the acquisition of intraluminal vesicles and other cargo, and the contribution of proton pumps to organelle acidification. Additionally, in light of a number of proteomic approaches to unravel the regulatory networks that control WPB formation and secretion, we provide a comprehensive overview of the WPB exocytotic machinery, including their molecular and cellular mechanisms.
Collapse
Affiliation(s)
- Sophie Hordijk
- Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands. https://twitter.com/SophieHordijk
| | - Tom Carter
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Ruben Bierings
- Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
8
|
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
|
9
|
Ma J, Hao Z, Zhang Y, Li L, Huang X, Wang Y, Chen L, Yang G, Li W. Physical Contacts Between Mitochondria and WPBs Participate in WPB Maturation. Arterioscler Thromb Vasc Biol 2024; 44:108-123. [PMID: 37942609 DOI: 10.1161/atvbaha.123.319939] [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: 07/31/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Weibel-Palade bodies (WPBs) are endothelial cell-specific cigar-shaped secretory organelles containing various biologically active molecules. WPBs play crucial roles in thrombosis, hemostasis, angiogenesis, and inflammation. The main content of WPBs is the procoagulant protein vWF (von Willebrand factor). Physical contacts and functional cross talk between mitochondria and other organelles have been demonstrated. Whether an interorganellar connection exists between mitochondria and WPBs is unknown. METHODS We observed physical contacts between mitochondria and WPBs in human umbilical vein endothelial cells by electron microscopy and living cell confocal microscopy. We developed an artificial intelligence-assisted method to quantify the duration and length of organelle contact sites in live cells. RESULTS We found there existed physical contacts between mitochondria and WPBs. Disruption of mitochondrial function affected the morphology of WPBs. Furthermore, we found that Rab3b, a small GTPase on the WPBs, was enriched at the mitochondrion-WPB contact sites. Rab3b deficiency reduced interaction between the two organelles and impaired the maturation of WPBs and vWF multimer secretion. CONCLUSIONS Our results reveal that Rab3b plays a crucial role in mediating the mitochondrion-WPB contacts, and that mitochondrion-WPB coupling is critical for the maturation of WPBs in vascular endothelial cells.
Collapse
Affiliation(s)
- Jing Ma
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, China (J.M., Z.H., W.L.)
- MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Beijing, China (J.M., Z.H., W.L.)
- Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China (J.M., Z.H., W.L.)
| | - Zhenhua Hao
- MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Beijing, China (J.M., Z.H., W.L.)
- Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China (J.M., Z.H., W.L.)
| | - Yudong Zhang
- National Laboratory of Pattern Recognition, Institute of Automation (Y.Z., G.Y.), Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China (Y.Z., G.Y.)
| | - Liuju Li
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, National Biomedical Imaging Center, School of Future Technology (L.L., L.C.), Peking University, Beijing, China
| | - Xiaoshuai Huang
- Biomedical Engineering Department (X.H.), Peking University, Beijing, China
| | - Yu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology (Y.W.), Chinese Academy of Sciences, Beijing, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, National Biomedical Imaging Center, School of Future Technology (L.L., L.C.), Peking University, Beijing, China
| | - Ge Yang
- National Laboratory of Pattern Recognition, Institute of Automation (Y.Z., G.Y.), Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China (Y.Z., G.Y.)
| | - Wei Li
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, China (J.M., Z.H., W.L.)
- MOE Key Laboratory of Major Diseases in Children, Capital Medical University, Beijing, China (J.M., Z.H., W.L.)
- Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China (J.M., Z.H., W.L.)
| |
Collapse
|
10
|
Zhao M, Zhang Y, Qiang L, Lu Z, Zhao Z, Fu Y, Wu B, Chai Q, Ge P, Lei Z, Zhang X, Li B, Wang J, Zhang L, Liu CH. A Golgi-resident GPR108 cooperates with E3 ubiquitin ligase Smurf1 to suppress antiviral innate immunity. Cell Rep 2023; 42:112655. [PMID: 37330913 DOI: 10.1016/j.celrep.2023.112655] [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: 11/14/2022] [Revised: 04/10/2023] [Accepted: 06/01/2023] [Indexed: 06/20/2023] Open
Abstract
The regulation of antiviral immunity is crucial in maintaining host immune homeostasis, a process that involves dynamic modulations of host organelles. The Golgi apparatus is increasingly perceived as a host organelle functioning as a critical platform for innate immunity, but the detailed mechanism by which it regulates antiviral immunity remains elusive. Here, we identify the Golgi-localized G protein-coupled receptor 108 (GPR108) as a regulator of type Ι interferon responses by targeting interferon regulatory factor 3 (IRF3). Mechanistically, GPR108 enhances the ubiquitin ligase Smad ubiquitylation regulatory factor 1 (Smurf1)-mediated K63-linked polyubiquitination of phosphorylated IRF3 for nuclear dot 10 protein 52 (NDP52)-dependent autophagic degradation, leading to suppression of antiviral immune responses against DNA or RNA viruses. Taken together, our study provides insight into the crosstalk between the Golgi apparatus and antiviral immunity via a dynamic and spatiotemporal regulation of GPR108-Smurf1 axis, thereby indicating a potential target for treating viral infection.
Collapse
Affiliation(s)
- Mengyuan Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yong Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lihua Qiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zhe Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zhuo Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yesheng Fu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| | - Bo Wu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| | - Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Pupu Ge
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zehui Lei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xinwen Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Bingxi Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China.
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
11
|
Laan SNJ, Dirven RJ, Bürgisser PE, Eikenboom J, Bierings R. Automated segmentation and quantitative analysis of organelle morphology, localization and content using CellProfiler. PLoS One 2023; 18:e0278009. [PMID: 37315066 DOI: 10.1371/journal.pone.0278009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/03/2023] [Indexed: 06/16/2023] Open
Abstract
One of the most used and versatile methods to study number, dimensions, content and localization of secretory organelles is confocal microscopy analysis. However, considerable heterogeneity exists in the number, size and shape of secretory organelles that can be present in the cell. One thus needs to analyze large numbers of organelles for valid quantification. Properly evaluating these parameters requires an automated, unbiased method to process and quantitatively analyze microscopy data. Here, we describe two pipelines, run by CellProfiler software, called OrganelleProfiler and OrganelleContentProfiler. These pipelines were used on confocal images of endothelial colony forming cells (ECFCs), which contain unique secretory organelles called Weibel-Palade bodies (WPBs), and on early endosomes in ECFCs and human embryonic kidney 293T (HEK293T) cells. Results show that the pipelines can quantify the cell count, size, organelle count, organelle size, shape, relation to cells and nuclei, and distance to these objects in both endothelial and HEK293T cells. Additionally, the pipelines were used to measure the reduction in WPB size after disruption of the Golgi and to quantify the perinuclear clustering of WPBs after triggering of cAMP-mediated signaling pathways in ECFCs. Furthermore, the pipeline is able to quantify secondary signals located in or on the organelle or in the cytoplasm, such as the small WPB GTPase Rab27A. Cell profiler measurements were checked for validity using Fiji. To conclude, these pipelines provide a powerful, high-processing quantitative tool for the characterization of multiple cell and organelle types. These pipelines are freely available and easily editable for use on different cell types or organelles.
Collapse
Affiliation(s)
- Sebastiaan N J Laan
- Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
- Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Richard J Dirven
- Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Petra E Bürgisser
- Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen Eikenboom
- Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Ruben Bierings
- Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
12
|
Chen P, Levy DL. Regulation of organelle size and organization during development. Semin Cell Dev Biol 2023; 133:53-64. [PMID: 35148938 PMCID: PMC9357868 DOI: 10.1016/j.semcdb.2022.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/20/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
During early embryogenesis, as cells divide in the developing embryo, the size of intracellular organelles generally decreases to scale with the decrease in overall cell size. Organelle size scaling is thought to be important to establish and maintain proper cellular function, and defective scaling may lead to impaired development and disease. However, how the cell regulates organelle size and organization are largely unanswered questions. In this review, we summarize the process of size scaling at both the cell and organelle levels and discuss recently discovered mechanisms that regulate this process during early embryogenesis. In addition, we describe how some recently developed techniques and Xenopus as an animal model can be used to investigate the underlying mechanisms of size regulation and to uncover the significance of proper organelle size scaling and organization.
Collapse
Affiliation(s)
- Pan Chen
- Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Daniel L Levy
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA.
| |
Collapse
|
13
|
Amiri KP, Kalish A, Mukherji S. Robustness and Universality in Organelle Size Control. PHYSICAL REVIEW LETTERS 2023; 130:018401. [PMID: 36669211 PMCID: PMC10316456 DOI: 10.1103/physrevlett.130.018401] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/07/2022] [Indexed: 06/15/2023]
Abstract
One of the grand challenges in cellular biophysics is understanding the precision with which cells assemble and maintain subcellular structures. Organelle sizes, for example, must be flexible enough to allow cells to grow or shrink them as environments demand yet be maintained within homeostatic limits. Despite identification of molecular factors that regulate organelle sizes we lack insight into the quantitative principles underlying organelle size control. Here we show experimentally that cells can robustly control average fluctuations in organelle size. By demonstrating that organelle sizes obey a universal scaling relationship we predict theoretically, our framework suggests that organelles grow in random bursts from a limiting pool of building blocks. Burstlike growth provides a general biophysical mechanism by which cells can maintain on average reliable yet plastic organelle sizes.
Collapse
Affiliation(s)
| | - Asa Kalish
- Department of Physics, Washington University in St. Louis
| | | |
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: 4.5] [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
|
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: 1.5] [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
|
16
|
Nakano A. The Golgi Apparatus and its Next-Door Neighbors. Front Cell Dev Biol 2022; 10:884360. [PMID: 35573670 PMCID: PMC9096111 DOI: 10.3389/fcell.2022.884360] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/28/2022] [Indexed: 12/20/2022] Open
Abstract
The Golgi apparatus represents a central compartment of membrane traffic. Its apparent architecture, however, differs considerably among species, from unstacked and scattered cisternae in the budding yeast Saccharomyces cerevisiae to beautiful ministacks in plants and further to gigantic ribbon structures typically seen in mammals. Considering the well-conserved functions of the Golgi, its fundamental structure must have been optimized despite seemingly different architectures. In addition to the core layers of cisternae, the Golgi is usually accompanied by next-door compartments on its cis and trans sides. The trans-Golgi network (TGN) can be now considered as a compartment independent from the Golgi stack. On the cis side, the intermediate compartment between the ER and the Golgi (ERGIC) has been known in mammalian cells, and its functional equivalent is now suggested for yeast and plant cells. High-resolution live imaging is extremely powerful for elucidating the dynamics of these compartments and has revealed amazing similarities in their behaviors, indicating common mechanisms conserved along the long course of evolution. From these new findings, I would like to propose reconsideration of compartments and suggest a new concept to describe their roles comprehensively around the Golgi and in the post-Golgi trafficking.
Collapse
|
17
|
Helical self-assembly of a mucin segment suggests an evolutionary origin for von Willebrand factor tubules. Proc Natl Acad Sci U S A 2022; 119:e2116790119. [PMID: 35377815 PMCID: PMC9169620 DOI: 10.1073/pnas.2116790119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Extracellular proteins with mechanical functions often require specialized assembly processes to form covalent oligomers. Progress in tissue bioengineering and repair will benefit from an understanding of how to harness and manipulate these processes. Here, we show that a particular supramolecular assembly mode was pre-encoded in the ancient domain organization common to gel-forming mucins and von Willebrand factor, glycoproteins that are deceptively different due to their divergence for distinct mechanical tasks. This finding highlights symmetry principles and building blocks retooled in nature to construct polymers with wide-ranging properties. These building blocks and knowledge of their self-assembly can be used to design new polymeric structures. The glycoprotein von Willebrand factor (VWF) contributes to hemostasis by stanching injuries in blood vessel walls. A distinctive feature of VWF is its assembly into long, helical tubules in endothelial cells prior to secretion. When VWF is released into the bloodstream, these tubules unfurl to release linear polymers that bind subendothelial collagen at wound sites, recruit platelets, and initiate the clotting cascade. VWF evolved from gel-forming mucins, the polymeric glycoproteins that coat and protect exposed epithelia. Despite the divergent function of VWF in blood vessel repair, sequence conservation and shared domain organization imply that VWF retained key aspects of the mucin bioassembly mechanism. Here, we show using cryo-electron microscopy that the ability to form tubules, a property hitherto thought to have arisen as a VWF adaptation to the vasculature, is a feature of the amino-terminal region of mucin. This segment of the human intestinal gel-forming mucin (MUC2) was found to self-assemble into tubules with a striking resemblance to those of VWF itself. To facilitate a comparison, we determined the residue-resolution structure of tubules formed by the homologous segment of VWF. The structures of the MUC2 and VWF tubules revealed the flexible joints and the intermolecular interactions required for tubule formation. Steric constraints in full-length MUC2 suggest that linear filaments, a previously observed supramolecular assembly form, are more likely than tubules to be the physiological mucin storage intermediate. Nevertheless, MUC2 tubules indicate a possible evolutionary origin for VWF tubules and elucidate design principles present in mucins and VWF.
Collapse
|
18
|
Lu J, Ma J, Hao Z, Li W. HPS6 Regulates the Biogenesis of Weibel–Palade Body in Endothelial Cells Through Trafficking v-ATPase to Its Limiting Membrane. Front Cell Dev Biol 2022; 9:743124. [PMID: 35252216 PMCID: PMC8891752 DOI: 10.3389/fcell.2021.743124] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
The Weibel–Palade body (WPB) is one of the lysosome-related organelles (LROs) in endothelial cells, whose main content is von Willebrand factor (vWF). The biogenesis of LROs is regulated by the Hermansky–Pudlak syndrome (HPS) protein-associated complexes through transporting cargo proteins to WPBs. Our previous studies have shown that HPS6, a subunit of BLOC-2 complex, is likely involved in the maturation of WPBs. However, the underlying mechanism remains unknown. In this study, we found that the knockdown of HPS6 in human umbilical vein endothelial cells (HUVECs) resulted in misshaped WPBs, decreased WPB number, and impaired vWF tubulation, which are similar to the characteristics of HPS6-deficient mouse endothelial cells. We observed similar morphological changes of WPBs in HUVECs after the knockdown of ATP6V0D1 (a subunit of v-ATPase). Furthermore, we found that HPS6 interacted with ATP6V0D1, suggesting that HPS6 transports ATP6V0D1 to the WPB limiting membrane for the assembly of the v-ATPase complex to maintain its acidic luminal pH, which is critical for the formation of vWF tubules during WPB maturation. In conclusion, HPS6 likely regulates the biogenesis of WPBs by participating in the trafficking of v-ATPase to the WPB membrane.
Collapse
|
19
|
Naß J, Terglane J, Gerke V. Weibel Palade Bodies: Unique Secretory Organelles of Endothelial Cells that Control Blood Vessel Homeostasis. Front Cell Dev Biol 2022; 9:813995. [PMID: 34977047 PMCID: PMC8717947 DOI: 10.3389/fcell.2021.813995] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial cells produce and release compounds regulating vascular tone, blood vessel growth and differentiation, plasma composition, coagulation and fibrinolysis, and also engage in interactions with blood cells thereby controlling hemostasis and acute inflammatory reactions. These interactions have to be tightly regulated to guarantee smooth blood flow in normal physiology, but also allow specific and often local responses to blood vessel injury and infectious or inflammatory insults. To cope with these challenges, endothelial cells have the remarkable capability of rapidly changing their surface properties from non-adhesive (supporting unrestricted blood flow) to adhesive (capturing circulating blood cells). This is brought about by the evoked secretion of major adhesion receptors for platelets (von-Willebrand factor, VWF) and leukocytes (P-selectin) which are stored in a ready-to-be-used form in specialized secretory granules, the Weibel-Palade bodies (WPB). WPB are unique, lysosome related organelles that form at the trans-Golgi network and further mature by receiving material from the endolysosomal system. Failure to produce correctly matured VWF and release it through regulated WPB exocytosis results in pathologies, most importantly von-Willebrand disease, the most common inherited blood clotting disorder. The biogenesis of WPB, their intracellular motility and their fusion with the plasma membrane are regulated by a complex interplay of proteins and lipids, involving Rab proteins and their effectors, cytoskeletal components as well as membrane tethering and fusion machineries. This review will discuss aspects of WPB biogenesis, trafficking and exocytosis focussing on recent findings describing factors contributing to WPB maturation, WPB-actin interactions and WPB-plasma membrane tethering and fusion.
Collapse
Affiliation(s)
- Johannes Naß
- Centre for Molecular Biology of Inflammation, Institute of Medical Biochemistry, University of Muenster, Muenster, Germany
| | - Julian Terglane
- Centre for Molecular Biology of Inflammation, Institute of Medical Biochemistry, University of Muenster, Muenster, Germany
| | - Volker Gerke
- Centre for Molecular Biology of Inflammation, Institute of Medical Biochemistry, University of Muenster, Muenster, Germany
| |
Collapse
|
20
|
Harris NS, Pelletier JP, Marin MJ, Winter WE. Von Willebrand factor and disease: a review for laboratory professionals. Crit Rev Clin Lab Sci 2021; 59:241-256. [DOI: 10.1080/10408363.2021.2014781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Neil S. Harris
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - J. Peter Pelletier
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Maximo J. Marin
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - William E. Winter
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
21
|
Patella F, Vendramin C, Charles O, Scully MA, Cutler DF. Shrinking Weibel-Palade bodies prevents high platelet recruitment in assays using thrombotic thrombocytopenic purpura plasma. Res Pract Thromb Haemost 2021; 5:e12626. [PMID: 34934893 PMCID: PMC8652131 DOI: 10.1002/rth2.12626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/02/2021] [Accepted: 09/15/2021] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Thrombotic thrombocytopenic purpura (TTP), caused by a genetic or autoimmune-driven lack of ADAMTS-13 activity, leads to high levels of the ultra-large von Willebrand factor (VWF) multimers produced by endothelial cells, causing excess platelet recruitment into forming thrombi, often with mortal consequences. Treatments include plasma infusion or replacement to restore ADAMTS-13 activity, or prevention of platelet recruitment to VWF. OBJECTIVES We tested a different approach, exploiting the unique cell biology of the endothelium. Upon activation, the VWF released by exocytosis of Weibel-Palade bodies (WPBs), transiently anchored to the cell surface, unfurls as strings into flowing plasma, recruiting platelets. Using plasma from patients with TTP increases platelet recruitment to the surface of cultured endothelial cells under flow. WPBs are uniquely plastic, and shortening WPBs dramatically reduces VWF string lengths and the recruitment of platelets. We wished to test whether the TTP plasma-driven increase in platelet recruitment would be countered by reducing formation of the longest WPBs that release longer strings. METHODS Endothelial cells grown in flow chambers were treated with fluvastatin, one of 37 drugs shown to shorten WPBs, then activated under flow in the presence of platelets and plasma of either controls or patients with TTP. RESULT We found that the dramatic increase in platelet recruitment caused by TTP plasma is entirely countered by treatment with fluvastatin, shortening the WPBs. CONCLUSIONS This potential approach of ameliorating the endothelial contribution to thrombotic risk by intervening far upstream of hemostasis might prove a useful adjunct to more conventional and direct therapies.
Collapse
Affiliation(s)
- Francesca Patella
- MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
- KinomicaAlderley ParkAlderley EdgeMacclesfieldUK
| | | | - Oscar Charles
- MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | | | - Daniel F. Cutler
- MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| |
Collapse
|
22
|
Much CD, Sendtner BS, Schwefel K, Freund E, Bekeschus S, Otto O, Pagenstecher A, Felbor U, Rath M, Spiegler S. Inactivation of Cerebral Cavernous Malformation Genes Results in Accumulation of von Willebrand Factor and Redistribution of Weibel-Palade Bodies in Endothelial Cells. Front Mol Biosci 2021; 8:622547. [PMID: 34307446 PMCID: PMC8298835 DOI: 10.3389/fmolb.2021.622547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/21/2021] [Indexed: 01/06/2023] Open
Abstract
Cerebral cavernous malformations are slow-flow thrombi-containing vessels induced by two-step inactivation of the CCM1, CCM2 or CCM3 gene within endothelial cells. They predispose to intracerebral bleedings and focal neurological deficits. Our understanding of the cellular and molecular mechanisms that trigger endothelial dysfunction in cavernous malformations is still incomplete. To model both, hereditary and sporadic CCM disease, blood outgrowth endothelial cells (BOECs) with a heterozygous CCM1 germline mutation and immortalized wild-type human umbilical vein endothelial cells were subjected to CRISPR/Cas9-mediated CCM1 gene disruption. CCM1 -/- BOECs demonstrated alterations in cell morphology, actin cytoskeleton dynamics, tube formation, and expression of the transcription factors KLF2 and KLF4. Furthermore, high VWF immunoreactivity was observed in CCM1 -/- BOECs, in immortalized umbilical vein endothelial cells upon CRISPR/Cas9-induced inactivation of either CCM1, CCM2 or CCM3 as well as in CCM tissue samples of familial cases. Observer-independent high-content imaging revealed a striking reduction of perinuclear Weibel-Palade bodies in unstimulated CCM1 -/- BOECs which was observed in CCM1 +/- BOECs only after stimulation with PMA or histamine. Our results demonstrate that CRISPR/Cas9 genome editing is a powerful tool to model different aspects of CCM disease in vitro and that CCM1 inactivation induces high-level expression of VWF and redistribution of Weibel-Palade bodies within endothelial cells.
Collapse
Affiliation(s)
- Christiane D. Much
- Department of Human Genetics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Barbara S. Sendtner
- Department of Human Genetics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Konrad Schwefel
- Department of Human Genetics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Eric Freund
- Centre for Innovation Competence (ZIK) plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
| | - Sander Bekeschus
- Centre for Innovation Competence (ZIK) plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
| | - Oliver Otto
- Centre for Innovation Competence (ZIK) ‐ Humoral Immune Reactions in Cardiovascular Diseases, University of Greifswald, Greifswald, Germany
| | - Axel Pagenstecher
- Department of Neuropathology, Center for Mind, Brain and Behavior (CMBB), University Hospital Giessen and MarburgMarburg, Germany
| | - Ute Felbor
- Department of Human Genetics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Rath
- Department of Human Genetics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stefanie Spiegler
- Department of Human Genetics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
23
|
Bäck N, Mains RE, Eipper BA. PAM: diverse roles in neuroendocrine cells, cardiomyocytes, and green algae. FEBS J 2021; 289:4470-4496. [PMID: 34089560 DOI: 10.1111/febs.16049] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/28/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022]
Abstract
Our understanding of the ways in which peptides are used for communication in the nervous and endocrine systems began with the identification of oxytocin, vasopressin, and insulin, each of which is stored in electron-dense granules, ready for release in response to an appropriate stimulus. For each of these peptides, entry of its newly synthesized precursor into the ER lumen is followed by transport through the secretory pathway, exposing the precursor to a sequence of environments and enzymes that produce the bioactive products stored in mature granules. A final step in the biosynthesis of many peptides is C-terminal amidation by peptidylglycine α-amidating monooxygenase (PAM), an ascorbate- and copper-dependent membrane enzyme that enters secretory granules along with its soluble substrates. Biochemical and cell biological studies elucidated the highly conserved mechanism for amidated peptide production and raised many questions about PAM trafficking and the effects of PAM on cytoskeletal organization and gene expression. Phylogenetic studies and the discovery of active PAM in the ciliary membranes of Chlamydomonas reinhardtii, a green alga lacking secretory granules, suggested that a PAM-like enzyme was present in the last eukaryotic common ancestor. While the catalytic features of human and C. reinhardtii PAM are strikingly similar, the trafficking of PAM in C. reinhardtii and neuroendocrine cells and secretion of its amidated products differ. A comparison of PAM function in neuroendocrine cells, atrial myocytes, and C. reinhardtii reveals multiple ways in which altered trafficking allows PAM to accomplish different tasks in different species and cell types.
Collapse
Affiliation(s)
- Nils Bäck
- Department of Anatomy, University of Helsinki, Finland
| | - Richard E Mains
- Department of Neuroscience, UConn Health, Farmington, CT, USA
| | - Betty A Eipper
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, USA
| |
Collapse
|
24
|
Zhang Y, Seemann J. Rapid degradation of GRASP55 and GRASP65 reveals their immediate impact on the Golgi structure. J Cell Biol 2021; 220:211583. [PMID: 33301566 PMCID: PMC7735681 DOI: 10.1083/jcb.202007052] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 02/08/2023] Open
Abstract
GRASP55 and GRASP65 have been implicated in stacking of Golgi cisternae and lateral linking of stacks within the Golgi ribbon. However, RNAi or gene knockout approaches to dissect their respective roles have often resulted in conflicting conclusions. Here, we gene-edited GRASP55 and/or GRASP65 with a degron tag in human fibroblasts, allowing for induced rapid degradation by the proteasome. We show that acute depletion of either GRASP55 or GRASP65 does not affect the Golgi ribbon, while chronic degradation of GRASP55 disrupts lateral connectivity of the ribbon. Acute double depletion of both GRASPs coincides with the loss of the vesicle tethering proteins GM130, p115, and Golgin-45 from the Golgi and compromises ribbon linking. Furthermore, GRASP55 and/or GRASP65 is not required for maintaining stacks or de novo assembly of stacked cisternae at the end of mitosis. These results demonstrate that both GRASPs are dispensable for Golgi stacking but are involved in maintaining the integrity of the Golgi ribbon together with GM130 and Golgin-45.
Collapse
Affiliation(s)
- Yijun Zhang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Joachim Seemann
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
25
|
Karampini E, Bürgisser PE, Olins J, Mulder AA, Jost CR, Geerts D, Voorberg J, Bierings R. Sec22b determines Weibel-Palade body length by controlling anterograde ER-Golgi transport. Haematologica 2021; 106:1138-1147. [PMID: 32336681 PMCID: PMC8018124 DOI: 10.3324/haematol.2019.242727] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 01/07/2023] Open
Abstract
Von Willebrand factor (VWF) is a multimeric hemostatic protein that is synthesized in endothelial cells, where it is stored for secretion in elongated secretory organelles called Weibel-Palade bodies (WPB). The hemostatic activity of VWF is strongly related to the length of these bodies, but how endothelial cells control the dimensions of their WPB is unclear. In this study, using a targeted short hairpin RNA screen, we identified longin-SNARE Sec22b as a novel determinant of WPB size and VWF trafficking. We found that Sec22b depletion resulted in loss of the typically elongated WPB morphology together with disintegration of the Golgi and dilation of rough endoplasmic reticulum cisternae. This was accompanied by reduced proteolytic processing of VWF, accumulation of VWF in the dilated rough endoplasmic reticulum and reduced basal and stimulated VWF secretion. Our data demonstrate that the elongation of WPB, and thus adhesive activity of their cargo VWF, is determined by the rate of anterograde transport between endoplasmic reticulum and Golgi, which depends on Sec22b-containing SNARE complexes.
Collapse
Affiliation(s)
- Ellie Karampini
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, The Netherlands
| | - Petra E Bürgisser
- Dept. of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jenny Olins
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, The Netherlands
| | - Aat A Mulder
- Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carolina R Jost
- Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Geerts
- Medical Biology, Amsterdam University Medical Center, University of Amsterdam, The Netherlands
| | - Jan Voorberg
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, The Netherlands
| | - Ruben Bierings
- Dept. of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Emerging mechanisms to modulate VWF release from endothelial cells. Int J Biochem Cell Biol 2020; 131:105900. [PMID: 33301925 DOI: 10.1016/j.biocel.2020.105900] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/13/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
Agonist-mediated exocytosis of Weibel-Palade bodies underpins the endothelium's ability to respond to injury or infection. Much of this important response is mediated by the major constituent of Weibel-Palade bodies: the ultra-large glycoprotein von Willebrand factor. Upon regulated WPB exocytosis, von Willebrand factor multimers unfurl into long, platelet-catching 'strings' which instigate the pro-haemostatic response. Accordingly, excessive levels of VWF are associated with thrombotic pathologies, including myocardial infarction and ischaemic stroke. Failure to appropriately cleave von Willebrand Factor strings results in thrombotic thrombocytopenic purpura, a life-threatening pathology characterised by tissue ischaemia and multiple microvascular occlusions. Historically, treatment of thrombotic thrombocytopenic purpura has relied heavily on plasma exchange therapy. However, the demonstrated efficacy of Rituximab and Caplacizumab in the treatment of acquired thrombotic thrombocytopenic purpura highlights how insights into pathophysiology can improve treatment options for von Willebrand factor-related disease. Directly limiting von Willebrand factor release from Weibel-Palade bodies has the potential as a therapeutic for cardiovascular disease. Cell biologists aim to map the WPB biogenesis and secretory pathways in order to find novel ways to control von Willebrand factor release. Emerging paradigms include the modulation of Weibel-Palade body size, trafficking and mechanism of fusion. This review focuses on the promise, progress and challenges of targeting Weibel-Palade bodies as a means to inhibit von Willebrand factor release from endothelial cells.
Collapse
|
27
|
Ferraro F, Patella F, Costa JR, Ketteler R, Kriston‐Vizi J, Cutler DF. Modulation of endothelial organelle size as an antithrombotic strategy. J Thromb Haemost 2020; 18:3296-3308. [PMID: 32881285 PMCID: PMC8436738 DOI: 10.1111/jth.15084] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND It is long established that von Willebrand factor (VWF) is central to hemostasis and thrombosis. Endothelial VWF is stored in cell-specific secretory granules, Weibel-Palade bodies (WPBs), organelles generated in a wide range of lengths (0.5-5.0 µm). WPB size responds to physiological cues and pharmacological treatment, and VWF secretion from shortened WPBs dramatically reduces platelet and plasma VWF adhesion to an endothelial surface. OBJECTIVE We hypothesized that WPB-shortening represented a novel target for antithrombotic therapy. Our objective was to determine whether compounds exhibiting this activity do exist. METHODS Using a microscopy approach coupled to automated image analysis, we measured the size of WPB bodies in primary human endothelial cells treated with licensed compounds for 24 hours. RESULTS AND CONCLUSIONS A novel approach to identification of antithrombotic compounds generated a significant number of candidates with the ability to shorten WPBs. In vitro assays of two selected compounds confirm that they inhibit the pro-hemostatic activity of secreted VWF. This set of compounds acting at a very early stage of the hemostatic process could well prove to be a useful adjunct to current antithrombotic therapeutics. Further, in the current SARS-CoV-2 pandemic, with a considerable fraction of critically ill COVID-19 patients affected by hypercoagulability, these WPB size-reducing drugs might also provide welcome therapeutic leads for frontline clinicians and researchers.
Collapse
Affiliation(s)
- Francesco Ferraro
- Endothelial Cell Biology Group, MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
- Present address:
Department of Biology and Evolution of Marine Organisms (BEOM)Stazione Zoologica Anton DohrnVilla ComunaleNaplesItaly
| | - Francesca Patella
- Endothelial Cell Biology Group, MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - Joana R. Costa
- Cell Signalling and Autophagy GroupMRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
- Present address:
Leukaemia Biology Research GroupDepartment of Haematology, Cancer InstituteUniversity College LondonLondonUK
| | - Robin Ketteler
- Cell Signalling and Autophagy GroupMRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - Janos Kriston‐Vizi
- Bioinformatics Image Core (BIONIC)MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - Daniel F. Cutler
- Endothelial Cell Biology Group, MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| |
Collapse
|
28
|
Rab6 is required for rapid, cisternal-specific, intra-Golgi cargo transport. Sci Rep 2020; 10:16604. [PMID: 33024151 PMCID: PMC7538953 DOI: 10.1038/s41598-020-73276-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/28/2020] [Indexed: 11/08/2022] Open
Abstract
Rab6, the most abundant Golgi associated small GTPase, consists of 2 equally common isoforms, Rab6A and Rab6A′, that differ in 3 amino acids and localize to trans Golgi cisternae. The two isoforms are largely redundant in function and hence are often referred to generically as Rab6. Rab6 loss-of-function inhibits retrograde Golgi trafficking, induces an increase in Golgi cisternal number in HeLa cells and delays the cell surface appearance of the anterograde cargo protein, VSVG. We hypothesized that these effects are linked and might be explained by a cisternal-specific delay in cargo transport. In pulse chase experiments using a deconvolved, confocal line scanning approach to score the distribution of the tsO45 mutant of VSVG protein in Rab6 depleted cells, we found that anterograde transport at 32 °C, permissive conditions, through the Golgi apparatus was locally delayed, almost tenfold, between medial and trans Golgi cisterna. Cis to medial transport was nearly normal as was trans Golgi to TGN transport. TGN exit was unaffected by Rab6 depletion. These effects were the same with either of two siRNAs. Similar intra-Golgi transport delays were seen at 37 °C with RUSH VSVG or a RUSH GPI-anchored construct using a biotin pulse to release the marker proteins from the ER. Using 3D-SIM, a super resolution approach, we found that RUSH VSVG transport was delayed pre-trans Golgi. These visual approaches suggest a selective slowing of anterograde transport relative to 3 different marker proteins downstream of the trans Golgi. Using a biochemical approach, we found that the onset of VSVG endoglycosidase H resistance in Rab6 depleted cells was delayed. Depletion of neither Rab6A or Rab6A′ isoforms alone had any effect on anterograde transport through the Golgi suggesting that Rab6A and Rab6A′ act coordinately. Delayed cargo transport conditions correlate strongly with a proliferation of Golgi cisternae observed in earlier electron microscopy. Our results strongly indicate that Rab6 is selectively required for rapid anterograde transport from the medial to trans Golgi. We suggest that the observed correlation with localized cisternal proliferation fits best with a cisternal progression model of Golgi function.
Collapse
|
29
|
de Jong A, Dirven RJ, Boender J, Atiq F, Anvar SY, Leebeek FWG, van Vlijmen BJM, Eikenboom J. Ex vivo Improvement of a von Willebrand Disease Type 2A Phenotype Using an Allele-Specific Small-Interfering RNA. Thromb Haemost 2020; 120:1569-1579. [PMID: 32803740 PMCID: PMC7649061 DOI: 10.1055/s-0040-1715442] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Von Willebrand disease (VWD) is the most common inherited bleeding disorder and is mainly caused by dominant-negative mutations in the multimeric protein von Willebrand factor (VWF). These mutations may either result in quantitative or qualitative defects in VWF. VWF is an endothelial protein that is secreted to the circulation upon endothelial activation. Once secreted, VWF multimers bind platelets and chaperone coagulation factor VIII in the circulation. Treatment of VWD focuses on increasing VWF plasma levels, but production and secretion of mutant VWF remain uninterrupted. Presence of circulating mutant VWF might, however, still affect normal hemostasis or functionalities of VWF beyond hemostasis. We hypothesized that inhibition of the production of mutant VWF improves the function of VWF overall and ameliorates VWD phenotypes. We previously proposed the use of allele-specific small-interfering RNAs (siRNAs) that target frequent
VWF
single nucleotide polymorphisms to inhibit mutant
VWF
. The aim of this study is to prove the functionality of these allele-specific siRNAs in endothelial colony-forming cells (ECFCs). We isolated ECFCs from a VWD type 2A patient with an intracellular multimerization defect, reduced VWF collagen binding, and a defective processing of proVWF to VWF. After transfection of an allele-specific siRNA that specifically inhibited expression of mutant VWF, we showed amelioration of the laboratory phenotype, with normalization of the VWF collagen binding, improvement in VWF multimers, and enhanced VWF processing. Altogether, we prove that allele-specific inhibition of the production of mutant VWF by siRNAs is a promising therapeutic strategy to improve VWD phenotypes.
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
| | - Richard J Dirven
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan Boender
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ferdows Atiq
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Seyed Yahya Anvar
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bart J M van Vlijmen
- 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
|
30
|
Patella F, Cutler DF. RGS4 controls secretion of von Willebrand factor to the subendothelial matrix. J Cell Sci 2020; 133:jcs247312. [PMID: 32576664 DOI: 10.1242/jcs.247312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/11/2020] [Indexed: 11/20/2022] Open
Abstract
The haemostatic protein von Willebrand factor (VWF) exists in plasma and subendothelial pools. The plasma pools are secreted from endothelial storage granules, Weibel-Palade bodies (WPBs), by basal secretion with a contribution from agonist-stimulated secretion, and the subendothelial pool is secreted into the subendothelial matrix by a constitutive pathway not involving WPBs. We set out to determine whether the constitutive release of subendothelial VWF is actually regulated and, if so, what functional consequences this might have. Constitutive VWF secretion can be increased by a range of factors, including changes in VWF expression, levels of TNF and other environmental cues. An RNA-seq analysis revealed that expression of regulator of G protein signalling 4 (RGS4) was reduced in endothelial cells (HUVECs) grown under these conditions. siRNA RGS4 treatment of HUVECs increased constitutive basolateral secretion of VWF, probably by affecting the anterograde secretory pathway. In a simple model of endothelial damage, we show that RGS4-silenced cells increased platelet recruitment onto the subendothelial matrix under flow. These results show that changes in RGS4 expression alter levels of subendothelial VWF, affecting platelet recruitment. This introduces a novel control over VWF function.
Collapse
Affiliation(s)
- Francesca Patella
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Daniel F Cutler
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
31
|
Bowman SL, Bi-Karchin J, Le L, Marks MS. The road to lysosome-related organelles: Insights from Hermansky-Pudlak syndrome and other rare diseases. Traffic 2020; 20:404-435. [PMID: 30945407 DOI: 10.1111/tra.12646] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022]
Abstract
Lysosome-related organelles (LROs) comprise a diverse group of cell type-specific, membrane-bound subcellular organelles that derive at least in part from the endolysosomal system but that have unique contents, morphologies and functions to support specific physiological roles. They include: melanosomes that provide pigment to our eyes and skin; alpha and dense granules in platelets, and lytic granules in cytotoxic T cells and natural killer cells, which release effectors to regulate hemostasis and immunity; and distinct classes of lamellar bodies in lung epithelial cells and keratinocytes that support lung plasticity and skin lubrication. The formation, maturation and/or secretion of subsets of LROs are dysfunctional or entirely absent in a number of hereditary syndromic disorders, including in particular the Hermansky-Pudlak syndromes. This review provides a comprehensive overview of LROs in humans and model organisms and presents our current understanding of how the products of genes that are defective in heritable diseases impact their formation, motility and ultimate secretion.
Collapse
Affiliation(s)
- Shanna L Bowman
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jing Bi-Karchin
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Linh Le
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael S Marks
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Karampini E, Bierings R, Voorberg J. Orchestration of Primary Hemostasis by Platelet and Endothelial Lysosome-Related Organelles. Arterioscler Thromb Vasc Biol 2020; 40:1441-1453. [PMID: 32375545 DOI: 10.1161/atvbaha.120.314245] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Megakaryocyte-derived platelets and endothelial cells store their hemostatic cargo in α- and δ-granules and Weibel-Palade bodies, respectively. These storage granules belong to the lysosome-related organelles (LROs), a heterogeneous group of organelles that are rapidly released following agonist-induced triggering of intracellular signaling pathways. Following vascular injury, endothelial Weibel-Palade bodies release their content into the vascular lumen and promote the formation of long VWF (von Willebrand factor) strings that form an adhesive platform for platelets. Binding to VWF strings as well as exposed subendothelial collagen activates platelets resulting in the release of α- and δ-granules, which are crucial events in formation of a primary hemostatic plug. Biogenesis and secretion of these LROs are pivotal for the maintenance of proper hemostasis. Several bleeding disorders have been linked to abnormal generation of LROs in megakaryocytes and endothelial cells. Recent reviews have emphasized common pathways in the biogenesis and biological properties of LROs, focusing mainly on melanosomes. Despite many similarities, LROs in platelet and endothelial cells clearly possess distinct properties that allow them to provide a highly coordinated and synergistic contribution to primary hemostasis by sequentially releasing hemostatic cargo. In this brief review, we discuss in depth the known regulators of α- and δ-granules in megakaryocytes/platelets and Weibel-Palade bodies in endothelial cells, starting from transcription factors that have been associated with granule formation to protein complexes that promote granule maturation. In addition, we provide a detailed view on the interplay between platelet and endothelial LROs in controlling hemostasis as well as their dysfunction in LRO related bleeding disorders.
Collapse
Affiliation(s)
- Ellie Karampini
- From the Department of Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory (E.K., R.B., J.V.), Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - Ruben Bierings
- From the Department of Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory (E.K., R.B., J.V.), Amsterdam University Medical Center, University of Amsterdam, the Netherlands.,Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands (R.B.)
| | - Jan Voorberg
- From the Department of Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory (E.K., R.B., J.V.), Amsterdam University Medical Center, University of Amsterdam, the Netherlands.,Experimental Vascular Medicine (J.V.), Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
33
|
Tiemeier GL, de Koning R, Wang G, Kostidis S, Rietjens RGJ, Sol WMPJ, Dumas SJ, Giera M, van den Berg CW, Eikenboom JCJ, van den Berg BM, Carmeliet P, Rabelink TJ. Lowering the increased intracellular pH of human-induced pluripotent stem cell-derived endothelial cells induces formation of mature Weibel-Palade bodies. Stem Cells Transl Med 2020; 9:758-772. [PMID: 32163224 PMCID: PMC7308639 DOI: 10.1002/sctm.19-0392] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/17/2020] [Indexed: 12/11/2022] Open
Abstract
Differentiation of human‐induced pluripotent stem cells (hiPSCs) into vascular endothelium is of great importance to tissue engineering, disease modeling, and use in regenerative medicine. Although differentiation of hiPSCs into endothelial‐like cells (hiPSC‐derived endothelial cells [hiPSC‐ECs]) has been demonstrated before, controversy exists as to what extent these cells faithfully reflect mature endothelium. To address this issue, we investigate hiPSC‐ECs maturation by their ability to express von Willebrand factor (VWF) and formation of Weibel‐Palade bodies (WPBs). Using multiple hiPSCs lines, hiPSC‐ECs failed to form proper VWF and WPBs, essential for angiogenesis, primary and secondary homeostasis. Lowering the increased intracellular pH (pHi) of hiPSC‐ECs with acetic acid did result in the formation of elongated WPBs. Nuclear magnetic resonance data showed that the higher pHi in hiPSC‐ECs occurred in association with decreased intracellular lactate concentrations. This was explained by decreased glycolytic flux toward pyruvate and lactate in hiPSC‐ECs. In addition, decreased expression of monocarboxylate transporter member 1, a member of the solute carrier family (SLC16A1), which regulates lactate and H+ uptake, contributed to the high pHi of hiPSC‐EC. Mechanistically, pro‐VWF dimers require the lower pH environment of the trans‐Golgi network for maturation and tubulation. These data show that while hiPSC‐ECs may share many features with mature EC, they are characterized by metabolic immaturity hampering proper EC function.
Collapse
Affiliation(s)
- Gesa L Tiemeier
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rozemarijn de Koning
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Gangqi Wang
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Rosalie G J Rietjens
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Wendy M P J Sol
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sébastien J Dumas
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Cathelijne W van den Berg
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen C J Eikenboom
- The Einthoven Laboratory for Experimental Vascular Medicine, Department of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernard M van den Berg
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ton J Rabelink
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
34
|
Wesley CC, Mishra S, Levy DL. Organelle size scaling over embryonic development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 9:e376. [PMID: 32003549 DOI: 10.1002/wdev.376] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Abstract
Cell division without growth results in progressive cell size reductions during early embryonic development. How do the sizes of intracellular structures and organelles scale with cell size and what are the functional implications of such scaling relationships? Model organisms, in particular Caenorhabditis elegans worms, Drosophila melanogaster flies, Xenopus laevis frogs, and Mus musculus mice, have provided insights into developmental size scaling of the nucleus, mitotic spindle, and chromosomes. Nuclear size is regulated by nucleocytoplasmic transport, nuclear envelope proteins, and the cytoskeleton. Regulators of microtubule dynamics and chromatin compaction modulate spindle and mitotic chromosome size scaling, respectively. Developmental scaling relationships for membrane-bound organelles, like the endoplasmic reticulum, Golgi, mitochondria, and lysosomes, have been less studied, although new imaging approaches promise to rectify this deficiency. While models that invoke limiting components and dynamic regulation of assembly and disassembly can account for some size scaling relationships in early embryos, it will be exciting to investigate the contribution of newer concepts in cell biology such as phase separation and interorganellar contacts. With a growing understanding of the underlying mechanisms of organelle size scaling, future studies promise to uncover the significance of proper scaling for cell function and embryonic development, as well as how aberrant scaling contributes to disease. This article is categorized under: Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Early Embryonic Development > Fertilization to Gastrulation Comparative Development and Evolution > Model Systems.
Collapse
Affiliation(s)
- Chase C Wesley
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming
| | - Sampada Mishra
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming
| | - Daniel L Levy
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming
| |
Collapse
|
35
|
Abstract
Von Willebrand factor (VWF) and coagulation factor VIII (FVIII) circulate as a complex in plasma and have a major role in the hemostatic system. VWF has a dual role in hemostasis. It promotes platelet adhesion by anchoring the platelets to the subendothelial matrix of damaged vessels and it protects FVIII from proteolytic degradation. Moreover, VWF is an acute phase protein that has multiple roles in vascular inflammation and is massively secreted from Weibel-Palade bodies upon endothelial cell activation. Activated FVIII on the other hand, together with coagulation factor IX forms the tenase complex, an essential feature of the propagation phase of coagulation on the surface of activated platelets. VWF deficiency, either quantitative or qualitative, results in von Willebrand disease (VWD), the most common bleeding disorder. The deficiency of FVIII is responsible for Hemophilia A, an X-linked bleeding disorder. Here, we provide an overview on the role of the VWF-FVIII interaction in vascular physiology.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany.
| |
Collapse
|
36
|
McCormack JJ, Harrison‐Lavoie KJ, Cutler DF. Human endothelial cells size-select their secretory granules for exocytosis to modulate their functional output. J Thromb Haemost 2020; 18:243-254. [PMID: 31519030 PMCID: PMC7155122 DOI: 10.1111/jth.14634] [Citation(s) in RCA: 12] [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: 03/21/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The secretory granules of endothelial cells, Weibel-Palade bodies, are released in response to numerous extracellular signals. Their cargo is critical to many vascular functions including hemostasis and inflammation. This presents a fundamental problem: how can these cells initiate tailor-made responses from the release of a single type of organelle, each with similar cargo? Each cell contains Weibel-Palade bodies in a wide range of sizes, and we have shown that experimentally shortening these organelles disproportionately reduces their ability to initiate hemostasis in vitro, leaving leukocyte recruitment unaffected. Could the production of this range of sizes underpin differential responses? OBJECTIVES To determine whether different agonists drive the exocytosis of different sizes of Weibel-Palade bodies. METHODS We used a high-throughput automated unbiased imaging workflow to analyze the sizes of Weibel-Palade bodies within human umbilical vein endothelial cells (HUVECs) before and after agonist activation to determine changes in organelle size distributions. RESULTS We found that a subset of agonists differentially evoke the release of the longest, most pro-hemostatic organelles. Inhibiting the release of these longest organelles by just 15% gives a fall of 60% in an assay of secreted von Willebrand factor (vWF) function. CONCLUSIONS The size-selection of granules for exocytosis represents a novel layer of control, allowing endothelial cells to provide diverse responses to different signals via the release of a single type of organelle.
Collapse
Affiliation(s)
| | | | - Daniel F. Cutler
- MRC Laboratory of Molecular Cell BiologyUniversity College LondonLondonUK
| |
Collapse
|
37
|
Atakpa P, Thillaiappan NB, Mataragka S, Prole DL, Taylor CW. IP 3 Receptors Preferentially Associate with ER-Lysosome Contact Sites and Selectively Deliver Ca 2+ to Lysosomes. Cell Rep 2019; 25:3180-3193.e7. [PMID: 30540949 PMCID: PMC6302550 DOI: 10.1016/j.celrep.2018.11.064] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/30/2018] [Accepted: 11/15/2018] [Indexed: 12/22/2022] Open
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) allow extracellular stimuli to redistribute Ca2+ from the ER to cytosol or other organelles. We show, using small interfering RNA (siRNA) and vacuolar H+-ATPase (V-ATPase) inhibitors, that lysosomes sequester Ca2+ released by all IP3R subtypes, but not Ca2+ entering cells through store-operated Ca2+ entry (SOCE). A low-affinity Ca2+ sensor targeted to lysosomal membranes reports large, local increases in cytosolic [Ca2+] during IP3-evoked Ca2+ release, but not during SOCE. Most lysosomes associate with endoplasmic reticulum (ER) and dwell at regions populated by IP3R clusters, but IP3Rs do not assemble ER-lysosome contacts. Increasing lysosomal pH does not immediately prevent Ca2+ uptake, but it causes lysosomes to slowly redistribute and enlarge, reduces their association with IP3Rs, and disrupts Ca2+ exchange with ER. In a "piston-like" fashion, ER concentrates cytosolic Ca2+ and delivers it, through large-conductance IP3Rs, to a low-affinity lysosomal uptake system. The involvement of IP3Rs allows extracellular stimuli to regulate Ca2+ exchange between the ER and lysosomes.
Collapse
Affiliation(s)
- Peace Atakpa
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | | | - Stefania Mataragka
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - David L Prole
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Colin W Taylor
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| |
Collapse
|
38
|
Effect of Autophagy Regulated by Sirt1/FoxO1 Pathway on the Release of Factors Promoting Thrombosis from Vascular Endothelial Cells. Int J Mol Sci 2019; 20:ijms20174132. [PMID: 31450612 PMCID: PMC6747322 DOI: 10.3390/ijms20174132] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 01/08/2023] Open
Abstract
Factors promoting thrombosis such as von Willebrand factor (vWF) and P-selectin are essential for the development of atherosclerosis (AS) and arterial thrombosis. The processing, maturation and release of vWF are regulated by autophagy of vascular endothelial cells. The Sirt1/FoxO1 pathway is an important pathway to regulate autophagy of endothelial cells, therefore the Sirt1/FoxO1 pathway may be an important target for the prevention of thrombosis. We investigated the role of ox-LDL in the release of vWF and P-selectin and the expression of Sirt1 and FoxO1 by Western Blot, Flow Cytometry, ELISA, and tandem fluorescent mRFP-GFP-LC3. We found that vWF and P-selectin secretion increased and Sirt1/FoxO1 pathway was depressed in human umbilical vein endothelial cells (HUVEC) when treated with ox-LDL. Moreover, the expression of autophagy-related protein LC3-II/I and p62 increased. Then, we explored the relationship between autophagy regulated by the Sirt1/FoxO1 pathway and the secretion of vWF and P-selectin. We found that Sirt1/FoxO1, activated by the Sirt1 activators resveratrol (RSV) and SRT1720, decreased the secretion of vWF and P-selectin, which can be abolished by the autophagy inhibitor 3-MA. The expression of Rab7 increased when Sirt1/FoxO1 pathway was activated, and the accumulation of p62 was decreased. Autophagy flux was inhibited by ox-LDL and Sirt1/FoxO1 pathway might enhance autophagy flux through the promotion of the Rab7 expression. Taken together, our data suggest that by enhancing autophagy flux and decreasing the release of vWF and P-selectin, the Sirt1/FoxO1 pathway may be a promising target to prevent AS and arterial thrombosis.
Collapse
|
39
|
Saraste J, Prydz K. A New Look at the Functional Organization of the Golgi Ribbon. Front Cell Dev Biol 2019; 7:171. [PMID: 31497600 PMCID: PMC6713163 DOI: 10.3389/fcell.2019.00171] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
A characteristic feature of vertebrate cells is a Golgi ribbon consisting of multiple cisternal stacks connected into a single-copy organelle next to the centrosome. Despite numerous studies, the mechanisms that link the stacks together and the functional significance of ribbon formation remain poorly understood. Nevertheless, these questions are of considerable interest, since there is increasing evidence that Golgi fragmentation – the unlinking of the stacks in the ribbon – is intimately connected not only to normal physiological processes, such as cell division and migration, but also to pathological states, including neurodegeneration and cancer. Challenging a commonly held view that ribbon architecture involves the formation of homotypic tubular bridges between the Golgi stacks, we present an alternative model, based on direct interaction between the biosynthetic (pre-Golgi) and endocytic (post-Golgi) membrane networks and their connection with the centrosome. We propose that the central domains of these permanent pre- and post-Golgi networks function together in the biogenesis and maintenance of the more transient Golgi stacks, and thereby establish “linker compartments” that dynamically join the stacks together. This model provides insight into the reversible fragmentation of the Golgi ribbon that takes place in dividing and migrating cells and its regulation along a cell surface – Golgi – centrosome axis. Moreover, it helps to understand transport pathways that either traverse or bypass the Golgi stacks and the positioning of the Golgi apparatus in differentiated neuronal, epithelial, and muscle cells.
Collapse
Affiliation(s)
- Jaakko Saraste
- Department of Biomedicine and Molecular Imaging Center, University of Bergen, Bergen, Norway
| | - Kristian Prydz
- Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
40
|
Delevoye C, Marks MS, Raposo G. Lysosome-related organelles as functional adaptations of the endolysosomal system. Curr Opin Cell Biol 2019; 59:147-158. [PMID: 31234051 PMCID: PMC6726539 DOI: 10.1016/j.ceb.2019.05.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
Unique functions of specialised cells such as those of the immune and haemostasis systems, skin, blood vessels, lung, and bone require specialised compartments, collectively referred to as lysosome-related organelles (LROs), that share features of endosomes and lysosomes. LROs harbour unique morphological features and cell type-specific contents, and most if not all undergo regulated secretion for diverse functions. Ongoing research, largely driven by analyses of inherited diseases and their model systems, is unravelling the mechanisms involved in LRO generation, maturation, transport and secretion. A molecular understanding of these features will provide targets and markers that can be exploited for diagnosis and therapy of a myriad of diseases.
Collapse
Affiliation(s)
- Cédric Delevoye
- Structure and Membrane Compartments, Institut Curie, Paris Sciences and Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Michael S Marks
- Dept. of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA; Dept. of Pathology and Laboratory Medicine and Dept. of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Graça Raposo
- Structure and Membrane Compartments, Institut Curie, Paris Sciences and Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France.
| |
Collapse
|
41
|
Makhoul C, Gosavi P, Gleeson PA. Golgi Dynamics: The Morphology of the Mammalian Golgi Apparatus in Health and Disease. Front Cell Dev Biol 2019; 7:112. [PMID: 31334231 PMCID: PMC6616279 DOI: 10.3389/fcell.2019.00112] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
In vertebrate cells the Golgi consists of individual stacks fused together into a compact ribbon structure. The function of the ribbon structure of the Golgi has only begun to be appreciated (De Matteis et al., 2008; Gosavi and Gleeson, 2017; Wei and Seemann, 2017). Recent advances have identified a role for the Golgi in the regulation of a broad range of cellular processes and of particular interest is that the modulation of the Golgi ribbon is associated with regulation of a number of signaling pathways (Makhoul et al., 2018). Various cell responses, such as inflammation, and various disorders and diseases, including neurodegeneration and cancer, are associated with the loss of the Golgi ribbon and the appearance of a dispersed or semi-dispersed Golgi. Often the dispersed Golgi is referred to as a “fragmented” morphology. However, the description of a dispersed Golgi ribbon as “fragmented” is inadequate as it does not accurately define the morphological state of the Golgi. This issue is particularly relevant as there are an increasing number of reports describing Golgi fragmentation under physiological and pathological conditions. Knowledge of the precise Golgi architecture is relevant to an appreciation of the functional status of the Golgi apparatus and the underlying molecular mechanism for the contribution of the Golgi to different cellular processes. Here we propose a classification to define the various morphological states of the non-ribbon architecture of the Golgi in mammalian cells as a guide to more precisely define the relationship between the morphological and functional status of this organelle.
Collapse
Affiliation(s)
- Christian Makhoul
- The Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Prajakta Gosavi
- The Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
42
|
Jevtić P, Schibler AC, Wesley CC, Pegoraro G, Misteli T, Levy DL. The nucleoporin ELYS regulates nuclear size by controlling NPC number and nuclear import capacity. EMBO Rep 2019; 20:embr.201847283. [PMID: 31085625 DOI: 10.15252/embr.201847283] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022] Open
Abstract
How intracellular organelles acquire their characteristic sizes is a fundamental question in cell biology. Given stereotypical changes in nuclear size in cancer, it is important to understand the mechanisms that control nuclear size in human cells. Using a high-throughput imaging RNAi screen, we identify and mechanistically characterize ELYS, a nucleoporin required for post-mitotic nuclear pore complex (NPC) assembly, as a determinant of nuclear size in mammalian cells. ELYS knockdown results in small nuclei, reduced nuclear lamin B2 localization, lower NPC density, and decreased nuclear import. Increasing nuclear import by importin α overexpression rescues nuclear size and lamin B2 import, while inhibiting importin α/β-mediated nuclear import decreases nuclear size. Conversely, ELYS overexpression increases nuclear size, enriches nuclear lamin B2 at the nuclear periphery, and elevates NPC density and nuclear import. Consistent with these observations, knockdown or inhibition of exportin 1 increases nuclear size. Thus, we identify ELYS as a novel positive effector of mammalian nuclear size and propose that nuclear size is sensitive to NPC density and nuclear import capacity.
Collapse
Affiliation(s)
- Predrag Jevtić
- Department of Molecular Biology, University of Wyoming, Laramie, WY, USA
| | | | - Chase C Wesley
- Department of Molecular Biology, University of Wyoming, Laramie, WY, USA
| | - Gianluca Pegoraro
- High Throughput Imaging Facility (HiTIF), National Cancer Institute, NIH, Bethesda, MD, USA
| | - Tom Misteli
- National Cancer Institute, NIH, Bethesda, MD, USA
| | - Daniel L Levy
- Department of Molecular Biology, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
43
|
Lopes-da-Silva M, McCormack JJ, Burden JJ, Harrison-Lavoie KJ, Ferraro F, Cutler DF. A GBF1-Dependent Mechanism for Environmentally Responsive Regulation of ER-Golgi Transport. Dev Cell 2019; 49:786-801.e6. [PMID: 31056345 PMCID: PMC6764485 DOI: 10.1016/j.devcel.2019.04.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 02/19/2019] [Accepted: 04/04/2019] [Indexed: 12/17/2022]
Abstract
How can anterograde membrane trafficking be modulated by physiological cues? A screen of Golgi-associated proteins revealed that the ARF-GEF GBF1 can selectively modulate the ER-Golgi trafficking of prohaemostatic von Willebrand factor (VWF) and extracellular matrix (ECM) proteins in human endothelial cells and in mouse fibroblasts. The relationship between levels of GBF1 and the trafficking of VWF into forming secretory granules confirmed GBF1 is a limiting factor in this process. Further, GBF1 activation by AMPK couples its control of anterograde trafficking to physiological cues; levels of glucose control GBF1 activation in turn modulating VWF trafficking into secretory granules. GBF1 modulates both ER and TGN exit, the latter dramatically affecting the size of the VWF storage organelles, thereby influencing the hemostatic capacity of the endothelium. The role of AMPK as a central integrating element of cellular pathways with intra- and extra-cellular cues can now be extended to modulation of the anterograde secretory pathway. The Arf-GEF GBF1 modulates anterograde trafficking of VWF and ECM proteins Loss of GBF1 slows ER and TGN exit, producing swollen ER and giant WPBs Activation of GBF1 via AMPK reduces endothelial WPB size and secretion Metabolic change alters anterograde trafficking and cargo secretion via AMPK-GBF1
Collapse
Affiliation(s)
- Mafalda Lopes-da-Silva
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK.
| | - Jessica J McCormack
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Jemima J Burden
- Electron Microscopy Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Kimberly J Harrison-Lavoie
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Francesco Ferraro
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Daniel F Cutler
- Endothelial Cell Biology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, UK.
| |
Collapse
|
44
|
Abstract
The Golgi apparatus is a central sorting station in the cell. It receives newly synthesized molecules from the endoplasmic reticulum and directs them to different subcellular destinations, such as the plasma membrane or the endocytic pathway. Importantly, in the last few years, it has emerged that the maintenance of Golgi structure is connected to the proper regulation of membrane trafficking. Rab proteins are small GTPases that are considered to be the master regulators of the intracellular membrane trafficking. Several of the over 60 human Rabs are involved in the regulation of transport pathways at the Golgi as well as in the maintenance of its architecture. This chapter will summarize the different roles of Rab GTPases at the Golgi, both as regulators of membrane transport, scaffold, and tethering proteins and in preserving the structure and function of this organelle.
Collapse
|
45
|
Mietkowska M, Schuberth C, Wedlich-Söldner R, Gerke V. Actin dynamics during Ca 2+-dependent exocytosis of endothelial Weibel-Palade bodies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:1218-1229. [PMID: 30465794 DOI: 10.1016/j.bbamcr.2018.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 01/24/2023]
Abstract
Weibel-Palade bodies (WPBs) are specialized secretory organelles of endothelial cells that serve important functions in the response to inflammation and vascular injury. WPBs actively respond to different stimuli by regulated exocytosis leading to full or selective release of their contents. Cellular conditions and mechanisms that distinguish between these possibilities are only beginning to emerge. To address this we analyzed dynamic rearrangements of the actin cytoskeleton during histamine-stimulated, Ca2+-dependent WPB exocytosis. We show that most WPB fusion events are followed by a rapid release of von-Willebrand factor (VWF), the large WPB cargo, and that this occurs concomitant with a softening of the actin cortex by the recently described Ca2+-dependent actin reset (CaAR). However, a considerable fraction of WPB fusion events is characterized by a delayed release of VWF and observed after the CaAR reaction peak. These delayed VWF secretions are accompanied by an assembly of actin rings or coats around the WPB post-fusion structures and are also seen following direct elevation of intracellular Ca2+ by plasma membrane wounding. Actin ring/coat assembly at WPB post-fusion structures requires Rho GTPase activity and is significantly reduced upon expression of a dominant-active mutant of the formin INF2 that triggers a permanent CaAR peak-like sequestration of actin to the endoplasmic reticulum. These findings suggest that a rigid actin cortex correlates with a higher proportion of fused WPB which assemble actin rings/coats most likely required for efficient VWF expulsion and/or stabilization of a WPB post-fusion structure. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Magdalena Mietkowska
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation and Cells-in-Motion Cluster of Excellence, University of Münster, Germany
| | - Christian Schuberth
- Institute of Cell Dynamics and Imaging, Centre for Molecular Biology of Inflammation and Cells-in-Motion Cluster of Excellence, University of Münster, Germany
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging, Centre for Molecular Biology of Inflammation and Cells-in-Motion Cluster of Excellence, University of Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation and Cells-in-Motion Cluster of Excellence, University of Münster, Germany.
| |
Collapse
|
46
|
Holthenrich A, Gerke V. Regulation of von-Willebrand Factor Secretion from Endothelial Cells by the Annexin A2-S100A10 Complex. Int J Mol Sci 2018; 19:ijms19061752. [PMID: 29899263 PMCID: PMC6032327 DOI: 10.3390/ijms19061752] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/08/2018] [Accepted: 06/09/2018] [Indexed: 12/17/2022] Open
Abstract
Endothelial cells serve as gatekeepers of vascular hemostasis and local inflammatory reactions. They can rapidly respond to changes in the environment, caused, for example, by blood vessel injury, tissue damage or infection, by secreting in a strictly regulated manner factors regulating these processes. These factors include adhesion receptors for circulating leukocytes and platelets, P-selectin and von-Willebrand factor (VWF) that are stored in specialized secretory granules of endothelial cells, the Weibel-Palade bodies (WPB). Acute exposure of these adhesion molecules converts the endothelial cell surface from an anti-adhesive state enabling unrestricted flow of circulating blood cells to an adhesive one capable of capturing leukocytes (through P-selectin) and platelets (through VWF). While these are important (patho)physiological responses, compromised or dysregulated WPB secretion can cause pathologies such as excessive bleeding or vascular occlusion. Several factors are involved in regulating the exocytosis of WPB and thus represent potential targets for therapeutic interventions in these pathologies. Among them, the annexin A2 (AnxA2)-S100A10 complex has been shown to participate in the tethering/docking of secretion-competent WPB at the plasma membrane, and interference with AnxA2/S100A10 expression or complex formation significantly reduces acute WPB exocytosis and VWF release. Thus, developing specific means to efficiently block AnxA2-S100A10 complex formation in endothelial cells could lead to novel avenues towards interfering with acute vascular thrombosis.
Collapse
Affiliation(s)
- Anna Holthenrich
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Strasse 56, 48149 Münster, Germany.
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Münster, Von-Esmarch-Strasse 56, 48149 Münster, Germany.
| |
Collapse
|
47
|
Schillemans M, Karampini E, van den Eshof BL, Gangaev A, Hofman M, van Breevoort D, Meems H, Janssen H, Mulder AA, Jost CR, Escher JC, Adam R, Carter T, Koster AJ, van den Biggelaar M, Voorberg J, Bierings R. Weibel-Palade Body Localized Syntaxin-3 Modulates Von Willebrand Factor Secretion From Endothelial Cells. Arterioscler Thromb Vasc Biol 2018; 38:1549-1561. [PMID: 29880488 PMCID: PMC6039413 DOI: 10.1161/atvbaha.117.310701] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/17/2018] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— Endothelial cells store VWF (von Willebrand factor) in rod-shaped secretory organelles, called Weibel-Palade bodies (WPBs). WPB exocytosis is coordinated by a complex network of Rab GTPases, Rab effectors, and SNARE (soluble NSF attachment protein receptor) proteins. We have previously identified STXBP1 as the link between the Rab27A-Slp4-a complex on WPBs and the SNARE proteins syntaxin-2 and -3. In this study, we investigate the function of syntaxin-3 in VWF secretion. Approach and Results— In human umbilical vein endothelial cells and in blood outgrowth endothelial cells (BOECs) from healthy controls, endogenous syntaxin-3 immunolocalized to WPBs. A detailed analysis of BOECs isolated from a patient with variant microvillus inclusion disease, carrying a homozygous mutation in STX3(STX3−/−), showed a loss of syntaxin-3 protein and absence of WPB-associated syntaxin-3 immunoreactivity. Ultrastructural analysis revealed no detectable differences in morphology or prevalence of immature or mature WPBs in control versus STX3−/− BOECs. VWF multimer analysis showed normal patterns in plasma of the microvillus inclusion disease patient, and media from STX3−/− BOECs, together indicating WPB formation and maturation are unaffected by absence of syntaxin-3. However, a defect in basal as well as Ca2+- and cAMP-mediated VWF secretion was found in the STX3−/− BOECs. We also show that syntaxin-3 interacts with the WPB-associated SNARE protein VAMP8 (vesicle-associated membrane protein-8). Conclusions— Our data reveal syntaxin-3 as a novel WPB-associated SNARE protein that controls WPB exocytosis.
Collapse
Affiliation(s)
- Maaike Schillemans
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Ellie Karampini
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Bart L van den Eshof
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Anastasia Gangaev
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Menno Hofman
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Dorothee van Breevoort
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Henriët Meems
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Hans Janssen
- Cell Biology, The Netherlands Cancer Institute, Amsterdam (H.J.)
| | - Aat A Mulder
- Molecular Cell Biology, Section Electron Microscopy, Leiden University Medical Center, The Netherlands (A.A.M., C.R.J., A.J.K.)
| | - Carolina R Jost
- Molecular Cell Biology, Section Electron Microscopy, Leiden University Medical Center, The Netherlands (A.A.M., C.R.J., A.J.K.)
| | - Johanna C Escher
- Pediatric Gastroenterology, Sophia Children's Hospital, Erasmus MC, Rotterdam, The Netherlands (J.C.E.)
| | - Rüdiger Adam
- Pediatric Gastroenterology, University Medical Centre, Mannheim, Germany (R.A.)
| | - Tom Carter
- St George's, University of London, United Kingdom (T.C.)
| | - Abraham J Koster
- Molecular Cell Biology, Section Electron Microscopy, Leiden University Medical Center, The Netherlands (A.A.M., C.R.J., A.J.K.)
| | - Maartje van den Biggelaar
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Jan Voorberg
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.).,Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (J.V.)
| | - Ruben Bierings
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| |
Collapse
|
48
|
Berntorp E, Ågren A, Aledort L, Blombäck M, Cnossen MH, Croteau SE, von Depka M, Federici AB, Goodeve A, Goudemand J, Mannucci PM, Mourik M, Önundarson PT, Rodeghiero F, Szántó T, Windyga J. Fifth Åland Island conference on von Willebrand disease. Haemophilia 2018; 24 Suppl 4:5-19. [DOI: 10.1111/hae.13475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2018] [Indexed: 12/11/2022]
Affiliation(s)
- E. Berntorp
- Centre for Thrombosis and Haemostasis; Skåne University Hospital; Lund University; Malmö Sweden
| | - A. Ågren
- Coagulation Unit; Department of Medicine; Division of Haematology; Karolinska University Hospital; Stockholm Sweden
| | - L. Aledort
- Department of Hematology and Medical Oncology; Mount Sinai Hospital; New York NY USA
| | - M. Blombäck
- Department of Molecular Medicine and Surgery; Karolinska University Hospital; Stockholm Sweden
| | - M. H. Cnossen
- Erasmus University Medical Center - Sophia Children's Hospital Rotterdam; Rotterdam The Netherlands
| | - S. E. Croteau
- Boston Children's Hospital; Boston Hemophilia Center; Harvard Medical School; Boston MA USA
| | | | - A. B. Federici
- Department of Oncology and Oncologic Hematology, Haematology and Transfusion Medicine; L. Sacco University Hospital; University of Milan; Milan Italy
| | - A. Goodeve
- Haemostasis Research Group; University of Sheffield; Sheffield UK
- Sheffield Diagnostic Genetics Service; Sheffield Children's NHS Foundation Trust; Sheffield UK
| | - J. Goudemand
- Department of Haematology; Lille University Hospital; Lille France
| | - P. M. Mannucci
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center; IRCCS Cà Grande Maggiore Policlinico Hospital Foundation; Milan Italy
| | - M. Mourik
- Department of Molecular Cell Biology; Leiden University Medical Center; Leiden The Netherlands
| | - P. T. Önundarson
- Landspitali University Hospital; University of Iceland School of Medicine; Reykjavik Iceland
| | - F. Rodeghiero
- Department of Cell Therapy and Hematology; San Bortolo Hospital; Vicenza Italy
| | - T. Szántó
- Coagulation Disorders Unit; Departments of Hematology and Clinical Chemistry (HUSLAB Laboratory Services); Helsinki University Central Hospital; Helsinki Finland
| | - J. Windyga
- Department of Disorders of Hemostasis and Internal Medicine; Institute of Hematology and Transfusion Medicine; Warsaw Poland
| |
Collapse
|
49
|
McCormack JJ, Lopes da Silva M, Ferraro F, Patella F, Cutler DF. Weibel-Palade bodies at a glance. J Cell Sci 2017; 130:3611-3617. [PMID: 29093059 DOI: 10.1242/jcs.208033] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The vascular environment can rapidly alter, and the speed with which responses to both physiological and pathological changes are required necessitates the existence of a highly responsive system. The endothelium can quickly deliver bioactive molecules by regulated exocytosis of its secretory granules, the Weibel-Palade bodies (WPBs). WPBs include proteins that initiate both haemostasis and inflammation, as well those that modulate blood pressure and angiogenesis. WPB formation is driven by von Willebrand factor, their most abundant protein, which controls both shape and size of WPBs. WPB are generated in a range of sizes, with the largest granules over ten times the size of the smallest. In this Cell Science at a Glance and the accompanying poster, we discuss the emerging mechanisms by which WPB size is controlled and how this affects the ability of this organelle to modulate haemostasis. We will also outline the different modes of exocytosis and their polarity that are currently being explored, and illustrate that these large secretory organelles provide a model for how elements of secretory granule biogenesis and exocytosis cooperate to support a complex and diverse set of functions.
Collapse
Affiliation(s)
- Jessica J McCormack
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Mafalda Lopes da Silva
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Francesco Ferraro
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Francesca Patella
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Daniel F Cutler
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| |
Collapse
|
50
|
Deng S, Liu H, Qiu K, You H, Lei Q, Lu W. Role of the Golgi Apparatus in the Blood-Brain Barrier: Golgi Protection May Be a Targeted Therapy for Neurological Diseases. Mol Neurobiol 2017; 55:4788-4801. [PMID: 28730529 DOI: 10.1007/s12035-017-0691-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) protects the brain from toxic material in the blood, provides nutrients for brain tissues, and screens harmful substances from the brain. The specific brain microvascular endothelial cells (BMVECs), tight junction between endothelial cells, and astrocytes ensure proper function of the central nervous system (CNS). Pathological factors disrupt the integrity of the BBB by destroying the normal function of endothelial cells and decreasing the production of tight junction proteins or the expression of proteins specifically localized on astrocytes. Interestingly, fragmentation of the Golgi apparatus is observed in neurological diseases and is involved in the destruction of the BBB function. The Golgi acts as a processing center in which proteins are transported after being processed in the endoplasmic reticulum. Besides reprocessing, classifying, and packaging proteins, the Golgi apparatus (GA) also acts as a signaling platform and calcium pool. In this review, we summarized the current literature on the potential relationship between the Golgi and endothelial cells, tight junction, and astrocytes. The normal function of the BBB is maintained as long as the normal function and morphology of the GA are not disturbed. Furthermore, we speculate that protecting the Golgi may be a novel therapeutic approach to protect the BBB and treat neurological diseases due to BBB dysfunction.
Collapse
Affiliation(s)
- Shuwen Deng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Hui Liu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Ke Qiu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Hong You
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Qiang Lei
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.
| |
Collapse
|