Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Blood Adv ; 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669344

ABSTRACT

Blood endothelial cells control the hemostatic and inflammatory response by secreting von Willebrand factor (VWF) and P-selectin from storage organelles called Weibel-Palade bodies (WPB). Actin-associated motor proteins regulate this secretory pathway at multiple points. Prior to fusion, myosin Va forms a complex that anchors WPBs to peripheral actin structures allowing maturation of content. Post-fusion, an actomyosin ring/coat is recruited and compresses the WPB to forcibly expel the largest VWF multimers. Here we provide the first evidence for the involvement of class I myosins during regulated VWF secretion. We show that the unconventional myosin-1C (Myo1c) is recruited post-fusion via its pleckstrin homology domain in an actin-independent process. This provides a link between the actin ring and phosphatidylinositol 4,5-bisphosphate (PIP2) at the membrane of the fused organelle and is necessary to ensure maximal VWF secretion. This is an active process requiring Myo1c ATPase activity as inhibition of class I myosins using the inhibitor Pentachloropseudilin or expression of an ATPase deficient Myo1c rigor mutant perturbs the expulsion of VWF and alters the kinetics of the exocytic actin ring. These data offer a novel insight into the control of an essential physiological process and provide a new way in which it can be regulated.

2.
Sci Signal ; 16(786): eabo4863, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37220183

ABSTRACT

Linear and disturbed flow differentially regulate gene expression, with disturbed flow priming endothelial cells (ECs) for a proinflammatory, atheroprone expression profile and phenotype. Here, we investigated the role of the transmembrane protein neuropilin-1 (NRP1) in ECs exposed to flow using cultured ECs, mice with an endothelium-specific knockout of NRP1, and a mouse model of atherosclerosis. We demonstrated that NRP1 was a constituent of adherens junctions that interacted with VE-cadherin and promoted its association with p120 catenin, stabilizing adherens junctions and inducing cytoskeletal remodeling in alignment with the direction of flow. We also showed that NRP1 interacted with transforming growth factor-ß (TGF-ß) receptor II (TGFBR2) and reduced the plasma membrane localization of TGFBR2 and TGF-ß signaling. NRP1 knockdown increased the abundance of proinflammatory cytokines and adhesion molecules, resulting in increased leukocyte rolling and atherosclerotic plaque size. These findings describe a role for NRP1 in promoting endothelial function and reveal a mechanism by which NRP1 reduction in ECs may contribute to vascular disease by modulating adherens junction signaling and promoting TGF-ß signaling and inflammation.


Subject(s)
Endothelial Cells , Neuropilin-1 , Receptor, Transforming Growth Factor-beta Type II , Animals , Mice , Adherens Junctions , Endothelium , Cadherins
3.
Blood ; 141(8): 930-944, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36564030

ABSTRACT

In response to tissue injury, within seconds the ultra-large glycoprotein von Willebrand factor (VWF) is released from endothelial storage organelles (Weibel-Palade bodies) into the lumen of the blood vasculature, where it leads to the recruitment of platelets. The marked size of VWF multimers represents an unprecedented burden on the secretory machinery of endothelial cells (ECs). ECs have evolved mechanisms to overcome this, most notably an actomyosin ring that forms, contracts, and squeezes out its unwieldy cargo. Inhibiting the formation or function of these structures represents a novel therapeutic target for thrombotic pathologies, although characterizing proteins associated with such a dynamic process has been challenging. We have combined APEX2 proximity labeling with an innovative dual loss-of-function screen to identify proteins associated with actomyosin ring function. We show that p21 activated kinase 2 (PAK2) recruits septin hetero-oligomers, a molecular interaction that forms a ring around exocytic sites. This cascade of events controls actomyosin ring function, aiding efficient exocytic release. Genetic or pharmacological inhibition of PAK2 or septins led to inefficient release of VWF and a failure to form platelet-catching strings. This new molecular mechanism offers additional therapeutic targets for the control of thrombotic disease and is highly relevant to other secretory systems that employ exocytic actomyosin machinery.


Subject(s)
Actomyosin , von Willebrand Factor , von Willebrand Factor/metabolism , Actomyosin/metabolism , Septins/metabolism , p21-Activated Kinases/metabolism , Endothelial Cells/metabolism , Proteomics , Exocytosis/physiology , Cytokinesis , Weibel-Palade Bodies/metabolism
4.
Cell Death Dis ; 13(2): 163, 2022 02 19.
Article in English | MEDLINE | ID: mdl-35184131

ABSTRACT

During this last decade, the development of prosenescence therapies has become an attractive strategy as cellular senescence acts as a barrier against tumour progression. In this context, CDK4/6 inhibitors induce senescence and reduce tumour growth in breast cancer patients. However, even though cancer cells are arrested after CDK4/6 inhibitor treatment, genes regulating senescence in this context are still unknown limiting their antitumour activity. Here, using a functional genome-wide CRISPR/Cas9 genetic screen we found several genes that participate in the proliferation arrest induced by CDK4/6 inhibitors. We find that downregulation of the coagulation factor IX (F9) using sgRNA and shRNA prevents the cell cycle arrest and senescent-like phenotype induced in MCF7 breast tumour cells upon Palbociclib treatment. These results were confirmed using another breast cancer cell line, T47D, and with an alternative CDK4/6 inhibitor, Abemaciclib, and further tested in a panel of 22 cancer cells. While F9 knockout prevents the induction of senescence, treatment with a recombinant F9 protein was sufficient to induce a cell cycle arrest and senescence-like state in MCF7 tumour cells. Besides, endogenous F9 is upregulated in different human primary cells cultures undergoing senescence. Importantly, bioinformatics analysis of cancer datasets suggest a role for F9 in human tumours. Altogether, these data collectively propose key genes involved in CDK4/6 inhibitor response that will be useful to design new therapeutic strategies in personalised medicine in order to increase their efficiency, stratify patients and avoid drug resistance.


Subject(s)
Breast Neoplasms , Cyclin-Dependent Kinase 6 , Factor IX , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Factor IX/genetics , Female , Humans , MCF-7 Cells
5.
Immunity ; 54(9): 1989-2004.e9, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34363750

ABSTRACT

The migration of neutrophils from the blood circulation to sites of infection or injury is a key immune response and requires the breaching of endothelial cells (ECs) that line the inner aspect of blood vessels. Unregulated neutrophil transendothelial cell migration (TEM) is pathogenic, but the molecular basis of its physiological termination remains unknown. Here, we demonstrated that ECs of venules in inflamed tissues exhibited a robust autophagic response that was aligned temporally with the peak of neutrophil trafficking and was strictly localized to EC contacts. Genetic ablation of EC autophagy led to excessive neutrophil TEM and uncontrolled leukocyte migration in murine inflammatory models, while pharmacological induction of autophagy suppressed neutrophil infiltration into tissues. Mechanistically, autophagy regulated the remodeling of EC junctions and expression of key EC adhesion molecules, facilitating their intracellular trafficking and degradation. Collectively, we have identified autophagy as a modulator of EC leukocyte trafficking machinery aimed at terminating physiological inflammation.


Subject(s)
Autophagy/physiology , Endothelial Cells/physiology , Neutrophil Infiltration/physiology , Transendothelial and Transepithelial Migration/physiology , Animals , Chemotaxis, Leukocyte/physiology , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Intercellular Junctions/physiology , Mice , Mice, Inbred C57BL , Neutrophils/physiology
6.
Int J Biochem Cell Biol ; 131: 105900, 2021 02.
Article in English | MEDLINE | ID: mdl-33301925

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.


Subject(s)
Brain Ischemia/drug therapy , Fibrinolytic Agents/therapeutic use , Myocardial Infarction/drug therapy , Purpura, Thrombotic Thrombocytopenic/drug therapy , Weibel-Palade Bodies/drug effects , von Willebrand Factor/antagonists & inhibitors , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Communication/drug effects , Cell Communication/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Hemostasis/drug effects , Hemostasis/genetics , Humans , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Organelle Size/drug effects , Purpura, Thrombotic Thrombocytopenic/genetics , Purpura, Thrombotic Thrombocytopenic/metabolism , Purpura, Thrombotic Thrombocytopenic/pathology , Rituximab/therapeutic use , Secretory Pathway/drug effects , Secretory Pathway/genetics , Single-Domain Antibodies/therapeutic use , Weibel-Palade Bodies/genetics , Weibel-Palade Bodies/metabolism , Weibel-Palade Bodies/pathology , von Willebrand Factor/biosynthesis , von Willebrand Factor/genetics
7.
PLoS Biol ; 17(12): e3000554, 2019 12.
Article in English | MEDLINE | ID: mdl-31790392

ABSTRACT

Junctional complexes between endothelial cells form a dynamic barrier that hinders passive diffusion of blood constituents into interstitial tissues. Remodelling of junctions is an essential process during leukocyte trafficking, vascular permeability, and angiogenesis. However, for many junctional proteins, the mechanisms of junctional remodelling have yet to be determined. Here, we used receptor mutagenesis, horseradish peroxidase (HRP), and ascorbate peroxidase 2 (APEX-2) proximity labelling, alongside light and electron microscopy (EM), to map the intracellular trafficking routes of junctional adhesion molecule-C (JAM-C). We found that JAM-C cotraffics with receptors associated with changes in permeability such as vascular endothelial cadherin (VE-Cadherin) and neuropilin (NRP)-1 and 2, but not with junctional proteins associated with the transmigration of leukocytes. Dynamic JAM-C trafficking and degradation are necessary for junctional remodelling during cell migration and angiogenesis. By identifying new potential trafficking machinery, we show that a key point of regulation is the ubiquitylation of JAM-C by the E3 ligase Casitas B-lineage lymphoma (CBL), which controls the rate of trafficking versus lysosomal degradation.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Endothelial Cells/physiology , Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability , Cell Adhesion , Cell Adhesion Molecules/physiology , Endothelium, Vascular/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/physiology , Junctional Adhesion Molecule C , Leukocytes/physiology , Neuropilins/metabolism , Protein Transport/physiology , Proto-Oncogene Proteins c-cbl/metabolism
8.
J Cell Sci ; 130(15): 2591-2605, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28674075

ABSTRACT

Weibel-Palade bodies (WPBs), the storage organelles of endothelial cells, are essential to normal haemostatic and inflammatory responses. Their major constituent protein is von Willebrand factor (VWF) which, following stimulation with secretagogues, is released into the blood vessel lumen as large platelet-catching strings. This exocytosis changes the protein composition of the cell surface and also results in a net increase in the amount of plasma membrane. Compensatory endocytosis is thought to limit changes in cell size and retrieve fusion machinery and other misplaced integral membrane proteins following exocytosis; however, little is known about the extent, timing, mechanism and precise function of compensatory endocytosis in endothelial cells. Using biochemical assays, live-cell imaging and correlative spinning-disk microscopy and transmission electron microscopy assays we provide the first in-depth high-resolution characterisation of this process. We provide a model of compensatory endocytosis based on rapid clathrin- and dynamin-mediated retrieval. Inhibition of this process results in a change of exocytic mode: WPBs then fuse with previously fused WPBs rather than the plasma membrane, leading, in turn, to the formation of structurally impaired tangled VWF strings.This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Clathrin/metabolism , Exocytosis/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Membrane Fusion/physiology , Weibel-Palade Bodies/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Weibel-Palade Bodies/genetics
9.
Trends Cell Biol ; 22(6): 329-37, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22543050

ABSTRACT

It is well known that actin can associate with intracellular membranes to drive endocytosis and the rocketing motion of bacteria, virions and some organelles and to regulate synaptic vesicle plasticity. Actin also has been extensively reported to be involved at several steps of exocytosis; however, it has typically been described as functioning either within the actin cortex or by providing actin tracks for organelle transport. Increasingly, actin filament coats or rings have been directly localized on the surface of the exocytic organelle. Here, we suggest a common mechanism for actin-based regulation of large secretory granules whereby organelle-associated actomyosin II contraction either directly expels secretory content or stabilizes the exocytosing organelle.


Subject(s)
Actins/metabolism , Exocytosis , Animals , Humans , Protein Binding , Transport Vesicles/metabolism
10.
J Cell Biol ; 194(4): 613-29, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21844207

ABSTRACT

The study of actin in regulated exocytosis has a long history with many different results in numerous systems. A major limitation on identifying precise mechanisms has been the paucity of experimental systems in which actin function has been directly assessed alongside granule content release at distinct steps of exocytosis of a single secretory organelle with sufficient spatiotemporal resolution. Using dual-color confocal microscopy and correlative electron microscopy in human endothelial cells, we visually distinguished two sequential steps of secretagogue-stimulated exocytosis: fusion of individual secretory granules (Weibel-Palade bodies [WPBs]) and subsequent expulsion of von Willebrand factor (VWF) content. Based on our observations, we conclude that for fusion, WPBs are released from cellular sites of actin anchorage. However, once fused, a dynamic ring of actin filaments and myosin II forms around the granule, and actomyosin II contractility squeezes VWF content out into the extracellular environment. This study therefore demonstrates how discrete actin cytoskeleton functions within a single cellular system explain actin filament-based prevention and promotion of specific exocytic steps during regulated secretion.


Subject(s)
Actomyosin/metabolism , Endothelial Cells/metabolism , Exocytosis , Weibel-Palade Bodies/metabolism , von Willebrand Factor/metabolism , Actin Cytoskeleton/metabolism , Cells, Cultured , Cytochalasins/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Exocytosis/drug effects , Humans , Membrane Fusion , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Video , Myosin Type II/metabolism , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , Weibel-Palade Bodies/drug effects , Weibel-Palade Bodies/ultrastructure
11.
Traffic ; 12(10): 1371-82, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21740491

ABSTRACT

Von-Willebrand factor (vWF) is a highly multimerized hemostatic glycoprotein that is stored in endothelial Weibel-Palade bodies (WPB) and secreted upon cell stimulation to act in recruiting platelets to sites of vessel injury. Only fully matured multimeric vWF represents an efficient anchor for platelets, and endothelial cells have developed mechanisms to prevent release of immature vWF. Full maturation of vWF occurs within WPB following their translocation from a perinuclear site of emergence at the trans-Golgi network (TGN) to the cell periphery. The WPB-associated small GTPase Rab27a is involved in restricting immature WPB exocytosis and we searched for links between Rab27a and the actin cytoskeleton that could anchor WPB inside endothelial cells until they are fully matured. We here identify myosin Va as such link. Myosin Va forms a tripartite complex with Rab27a and its effector MyRIP and depletion of or dominant-negative interference with myosin Va leads to an increase in the ratio of perinuclear to more peripheral WPB. Concomitantly, myosin Va depletion results in an elevated secretion of less-oligomeric vWF from histamine-stimulated endothelial cells. These results indicate that a Rab27a/MyRIP/myosin Va complex is involved in linking WPB to the peripheral actin cytoskeleton of endothelial cells to allow full maturation and prevent premature secretion of vWF.


Subject(s)
Endothelial Cells/metabolism , Myosin Heavy Chains/physiology , Myosin Type V/physiology , Vesicular Transport Proteins/physiology , rab GTP-Binding Proteins/physiology , von Willebrand Factor/metabolism , Cell Culture Techniques , Cell Line , Endothelial Cells/physiology , Humans , Immunoprecipitation , Microscopy, Confocal , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Plasmids , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
12.
Blood ; 113(20): 5010-8, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19270261

ABSTRACT

Endothelial cells contain cigar-shaped secretory organelles called Weibel-Palade bodies (WPBs) that play a crucial role in both hemostasis and the initiation of inflammation. The major cargo protein of WPBs is von Willebrand factor (VWF). In unstimulated cells, this protein is stored in a highly multimerized state coiled into protein tubules, but after secretagogue stimulation and exocytosis it unfurls, under shear force, as long platelet-binding strings. Small GTPases of the Rab family play a key role in organelle function. Using siRNA depletion in primary endothelial cells, we have identified a role for the WPB-associated Rab27a and its effector MyRIP. Both these proteins are present on only mature WPBs, and this rab/effector complex appears to anchor these WPBs to peripheral actin. Depletion of either the Rab or its effector results in a loss of peripheral WPB localization, and this destabilization is coupled with an increase in both basal and stimulated secretion. The VWF released from Rab27a-depleted cells is less multimerized, and the VWF strings seen under flow are shorter. Our results indicate that this Rab/effector complex controls peripheral distribution and prevents release of incompletely processed WPB content.


Subject(s)
Endothelial Cells/metabolism , Protein Multimerization , Vesicular Transport Proteins/physiology , rab GTP-Binding Proteins/physiology , von Willebrand Factor/metabolism , Cells, Cultured , Exocytosis/genetics , Gene Knockdown Techniques , Humans , Molecular Weight , Protein Multimerization/genetics , Protein Transport/genetics , Tissue Distribution , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics , Weibel-Palade Bodies/metabolism , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
13.
J Biol Chem ; 284(6): 3935-45, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19033446

ABSTRACT

The major lymphatic endothelial hyaluronan receptor LYVE-1, a Link superfamily glycoprotein similar to the hyaluronan-binding/inflammatory leukocyte homing receptor CD44, was initially implicated in hyaluronan (HA)-mediated cell adhesion and lymph-borne hyaluronan metabolism. However, the apparently normal phenotype of Lyve-1 knock-out mice and the recent demonstration that the receptor undergoes cytokine-induced endocytosis independent of HA uptake have cast doubt on such functions. Here we present new data that reconcile these anomalies by showing that LYVE-1 is functionally "silenced" in a cell-specific fashion by autoinhibitory glycosylation. We demonstrate that LYVE-1 transfected in HEK 293T fibroblasts and Jurkat T cells is competent to bind HA, whereas the endogenous receptor in cultured lymphatic endothelial cells or the receptor transfected in Chinese hamster ovary and HeLa cells is not. Moreover, through a combination of mutagenesis and functional analysis in HEK 293T fibroblasts and glycosylation-defective Chinese hamster ovary cell lines, we reveal that the inhibitory mechanism is reversible and is exerted by terminal sialylation, most likely through alpha2-3 or alpha2-6 linkage to O-glycans. Finally, we provide evidence that the mechanism operates in vivo by showing that native LYVE-1 in primary lymphatic endothelial cells is extensively sialylated and that HA binding can be reactivated by neuraminidase treatment of the soluble ectodomain. These results reveal unexpected complexity in the regulation of LYVE-1 function and raise the possibility that this receptor, like CD44, may become active after appropriate unmasking in vivo.


Subject(s)
Endothelium, Lymphatic/metabolism , Glycoproteins/metabolism , Hyaluronic Acid/metabolism , N-Acetylneuraminic Acid/metabolism , Vesicular Transport Proteins/metabolism , Animals , CHO Cells , Carbohydrate Conformation , Cricetinae , Cricetulus , Endocytosis/physiology , Endothelium, Lymphatic/cytology , Glycoproteins/genetics , HeLa Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Hyaluronic Acid/genetics , Jurkat Cells , Membrane Transport Proteins , Mice , Mice, Knockout , N-Acetylneuraminic Acid/genetics , Protein Binding/physiology , Vesicular Transport Proteins/genetics
14.
Mol Biol Cell ; 19(12): 5072-81, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18815278

ABSTRACT

Formation of secretory organelles requires the coupling of cargo selection to targeting into the correct exocytic pathway. Although the assembly of regulated secretory granules is driven in part by selective aggregation and retention of content, we recently reported that adaptor protein-1 (AP-1) recruitment of clathrin is essential to the initial formation of Weibel-Palade bodies (WPBs) at the trans-Golgi network. A selective co-aggregation process might include recruitment of components required for targeting to the regulated secretory pathway. However, we find that acquisition of the regulated secretory phenotype by WPBs in endothelial cells is coupled to but can be separated from formation of the distinctive granule core by ablation of the AP-1 effectors aftiphilin and gamma-synergin. Their depletion by small interfering RNA leads to WPBs that fail to respond to secretagogue and release their content in an unregulated manner. We find that these non-responsive WPBs have density, markers of maturation, and highly multimerized von Willebrand factor similar to those of wild-type granules. Thus, by also recruiting aftiphilin/gamma-synergin in addition to clathrin, AP-1 coordinates formation of WPBs with their acquisition of a regulated secretory phenotype.


Subject(s)
Adaptor Protein Complex 1/metabolism , Carrier Proteins/metabolism , Endothelial Cells/metabolism , Nerve Tissue Proteins/metabolism , Secretory Pathway/physiology , Weibel-Palade Bodies/metabolism , Adaptor Protein Complex 1/genetics , Animals , Biomarkers/metabolism , Carrier Proteins/genetics , Cell Line , Clathrin/metabolism , Endothelial Cells/cytology , Exocytosis/physiology , Female , Humans , Nerve Tissue Proteins/genetics , Phenotype , Pregnancy , Protein Conformation , RNA Interference , Transcription Factor AP-1/metabolism , von Willebrand Factor/chemistry , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
15.
J Cell Sci ; 121(Pt 1): 19-27, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18096688

ABSTRACT

Weibel-Palade bodies (WPBs) are secretory organelles used for post-synthesis storage in endothelial cells that can, very rapidly, be triggered to release their contents. They carry a variety of bioactive molecules that are needed to mount a rapid response to the complex environment of cells that line blood vessels. They store factors that are essential to haemostasis and inflammation, as well as factors that modulate vascular tonicity and angiogenesis. The number of WPBs and their precise content vary between endothelial tissues, reflecting their differing physiological circumstances. The particular functional demands of the highly multimerised haemostatic protein von Willebrand Factor (VWF), which is stored in WPBs as tubules until release, are responsible for the cigar shape of these granules. How VWF tubules drive the formation of these uniquely shaped organelles, and how WPB density increases during maturation, has recently been revealed by EM analysis using high-pressure freezing and freeze substitution. In addition, an AP1/clathrin coat has been found to be essential to WPB formation. Following recruitment of cargo at the TGN, there is a second wave of recruitment that delivers integral and peripheral membrane proteins to WPBs, some of which is AP3 dependent.


Subject(s)
Weibel-Palade Bodies/physiology , Adaptor Protein Complex 1/metabolism , Animals , Clathrin/metabolism , Exocytosis , Humans , Weibel-Palade Bodies/ultrastructure , trans-Golgi Network/physiology , von Willebrand Factor/chemistry , von Willebrand Factor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...