Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
2.
Inflammation ; 42(1): 290-305, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30218321

ABSTRACT

We tested the ability of platelet-derived extracellular vesicles (PEV) to promote adhesion of flowing neutrophils to endothelial cells (EC). PEV were collected from platelets stimulated with collagen-related peptide, and differential centrifugation was used to collect larger vesicles enriched for platelet membrane microvesicles (PMV) or smaller vesicles enriched for platelet exosomes (Pexo). Vesicle binding and resultant activation of neutrophils and EC were assessed by flow cytometry. Flow-based adhesion assays assessed binding of neutrophils directly to deposited vesicles or to EC, after neutrophils or EC had been treated with vesicles. PEV bound efficiently to neutrophils or EC, with resultant upregulation of activation markers. Binding was Ca++-dependent and dominantly mediated by CD62P for neutrophils or by integrins for EC. Deposited PEV supported mainly transient attachments of flowing neutrophils through CD62P and some stable adhesion through CXC-chemokines. Neutrophil adhesion to EC was promoted when either cell was pre-treated with PEV, although the effect was less prominent when EC were pre-activated with tumor necrosis factor-α. The pro-adhesive effects on neutrophils could largely be attributed to the larger PMV rather than Pexo. Thus, surface-bound PEV can capture flowing neutrophils, while PEV also activate neutrophils and EC to promote interactions. PEV may potentiate inflammatory responses after tissue injury.


Subject(s)
Blood Platelets/ultrastructure , Cell Communication , Endothelial Cells/cytology , Extracellular Vesicles/physiology , Neutrophils/cytology , Blood Specimen Collection , Calcium , Cell Adhesion , Cells, Cultured , Endothelial Cells/chemistry , Healthy Volunteers , Humans , Integrins/metabolism , Neutrophils/chemistry , P-Selectin/metabolism
3.
J Immunol ; 193(1): 234-43, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24872191

ABSTRACT

Stromal cells actively modulate the inflammatory process, in part by influencing the ability of neighboring endothelial cells to support the recruitment of circulating leukocytes. We hypothesized that podocytes influence the ability of glomerular endothelial cells (GEnCs) to recruit neutrophils during inflammation. To address this, human podocytes and human GEnCs were cultured on opposite sides of porous inserts and then treated with or without increasing concentrations of TNF-α prior to addition of neutrophils. The presence of podocytes significantly reduced neutrophil recruitment to GEnCs by up to 50% when cultures were treated with high-dose TNF-α (100 U/ml), when compared with GEnC monocultures. Importantly, this phenomenon was dependent on paracrine actions of soluble IL-6, predominantly released by podocytes. A similar response was absent when HUVECs were cocultured with podocytes, indicating a tissue-specific phenomenon. Suppressor of cytokine signaling 3 elicited the immunosuppressive actions of IL-6 in a process that disrupted the presentation of chemokines on GEnCs by altering the expression of the duffy Ag receptor for chemokines. Interestingly, suppressor of cytokine signaling 3 knockdown in GEnCs upregulated duffy Ag receptor for chemokines and CXCL5 expression, thereby restoring the neutrophil recruitment. In summary, these studies reveal that podocytes can negatively regulate neutrophil recruitment to inflamed GEnCs by modulating IL-6 signaling, identifying a potential novel anti-inflammatory role of IL-6 in renal glomeruli.


Subject(s)
Cell Communication/immunology , Endothelial Cells/immunology , Interleukin-6/immunology , Neutrophil Infiltration , Neutrophils/immunology , Podocytes/immunology , Cell Communication/genetics , Cell Line, Transformed , Endothelial Cells/cytology , Female , Gene Knockdown Techniques , Humans , Interleukin-6/genetics , Male , Neutrophils/cytology , Podocytes/cytology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/immunology
4.
PLoS One ; 7(8): e43916, 2012.
Article in English | MEDLINE | ID: mdl-22952809

ABSTRACT

Neutrophil proteases, proteinase-3 (PR3) and elastase play key roles in glomerular endothelial cell (GEC) injury during glomerulonephritis. Endothelial protease-activated receptors (PARs) are potential serine protease targets in glomerulonephritis. We investigated whether PAR1/2 are required for alterations in GEC phenotype that are mediated by PR3 or elastase during active glomerulonephritis. Endothelial PARs were assessed by flow cytometry. Thrombin, trypsin and agonist peptides for PAR1 and PAR2, TFLLR-NH(2) and SLIGKV-NH(2,) respectively, were used to assess alterations in PAR activation induced by PR3 or elastase. Endothelial von Willebrand Factor (vWF)release and calcium signaling were used as PAR activation markers. Both PR3 and elastase induced endothelial vWF release, with elastase inducing the highest response. PAR1 peptide induced GEC vWF release to the same extent as PR3. However, knockdown of PARs by small interfering RNA showed that neither PAR1 nor PAR2 activation caused PR3 or elastase-mediated vWF release. Both proteases interacted with and disarmed surface GEC PAR1, but there was no detectable interaction with cellular PAR2. Neither protease induced a calcium response in GEC. Therefore, PAR signaling and serine protease-induced alterations in endothelial function modulate glomerular inflammation via parallel but independent pathways.


Subject(s)
Endothelial Cells/cytology , Kidney Glomerulus/cytology , Myeloblastin/metabolism , Pancreatic Elastase/metabolism , Receptor, PAR-1/metabolism , Signal Transduction , von Willebrand Factor/metabolism , Calcium/metabolism , Endothelial Cells/metabolism , HEK293 Cells , Humans , Proteolysis
5.
Cardiovasc Res ; 91(1): 134-41, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21285294

ABSTRACT

AIMS: Cells of the monocyte lineage are the most abundant inflammatory cells found in atherosclerotic lesions. Dominance of the inflammatory infiltrate by monocytes indicates that there is a disease-driven mechanism supporting their selective recruitment. Previous studies have demonstrated that interactions between endothelial cells (ECs) and platelets may promote monocyte recruitment. In this study, we sought to expand on this knowledge using a complex coculture model of the diseased vessel wall. METHODS AND RESULTS: Using primary human cells in an in vitro flow-based adhesion assay, we found that secretory arterial smooth muscle cells (SMCs), cocultured with ECs, promote preferential recruitment of monocytes from blood in a TGF-ß1-dependent manner. Approximately 85% of leucocytes recruited to the endothelium were CD14(+). Formation of adhesive platelet bridges on ECs was essential for monocyte recruitment as platelet removal or inhibition of adhesion to the ECs abolished monocyte recruitment. Monocytes were recruited from flow by platelet P-selectin and activated by EC-derived CC chemokine ligand 2 (CCL2), although the presentation of CCL2 to adherent monocytes was dependent upon platelet activation and release of CXC chemokine ligand 4 (CXCL4). In an intravital model of TGF-ß1-driven vascular inflammation in mice, platelets were also necessary for efficient leucocyte recruitment to vessels of the microcirculation in the cremaster muscle. CONCLUSIONS: In this study, we have demonstrated that stromal cells found within the diseased artery wall may promote the preferential recruitment of monocytes and this is achieved by establishing a cascade of interactions between SMCs, ECs, platelets, and monocytes.


Subject(s)
Atherosclerosis/immunology , Blood Platelets/immunology , Cell Adhesion , Endothelial Cells/immunology , Monocytes/immunology , Myocytes, Smooth Muscle/immunology , Platelet Activation , Animals , Atherosclerosis/blood , Blood Platelets/metabolism , Cell Communication , Cells, Cultured , Chemokine CCL2/metabolism , Coculture Techniques , Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Mice , Monocytes/metabolism , Myocytes, Smooth Muscle/metabolism , P-Selectin/metabolism , Platelet Adhesiveness , Platelet Factor 4/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...