Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Liver Int ; 42(5): 1185-1203, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35129269

RESUMEN

BACKGROUND AND AIMS: Leukocyte infiltration is a hallmark of hepatic inflammation. The Junctional Adhesion Molecule A (JAM-A) is a crucial regulator of leukocyte extravasation and is upregulated in human viral fibrosis. Reduced shear stress within hepatic sinusoids and the specific phenotype of liver sinusoidal endothelial cells (LSEC) cumulate in differing adhesion characteristics during liver fibrosis. The aim of this study was to define the functional role of cell-specific adhesion molecule JAM-A during hepatic fibrogenesis. METHODS: Complete, conditional (intestinal epithelial; endothelial) and bone marrow chimeric Jam-a knockout animals and corresponding C57Bl/6 wild-type animals were treated with carbon tetrachloride (CCl4 , 6 weeks). For functional analyses of JAM-A, comprehensive in vivo studies, co-culture models and flow-based adhesion assays were performed. RESULTS: Complete and bone marrow-derived Jam-a-/- animals showed aggravated fibrosis with increased non-sinusoidal, perivascular accumulation of CD11b+ F4/80+ monocyte-derived macrophages in contrast to wild-type mice. Despite being associated with disturbed epithelial barrier function, an intestinal epithelial Jam-a knockout did not affect fibrogenesis. In endothelial-specific Jam-a-/- animals, liver fibrosis was aggravated alongside sinusoid capillarization and hepatic stellate cell (HSC) activation. HSC activation is induced via Jam-a-/- LSEC-derived secretion of soluble factors. Sinusoid CD31 expression and hedgehog gene signalling were increased, but leukocyte infiltration and adhesion to LSECs remained unaffected. CONCLUSIONS: Our models decipher cell-specific JAM-A to exert crucial functions during hepatic fibrogenesis. JAM-A on bone marrow-derived cells regulates non-sinusoidal vascular immune cell recruitment, while endothelial JAM-A controls liver sinusoid capillarization and HSC quiescence.


Asunto(s)
Molécula A de Adhesión de Unión , Animales , Células Endoteliales/metabolismo , Fibrosis , Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/metabolismo , Humanos , Molécula A de Adhesión de Unión/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos C57BL
2.
J Biol Chem ; 295(42): 14367-14378, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-32817341

RESUMEN

Ticks, as blood-sucking parasites, have developed a complex strategy to evade and suppress host immune responses during feeding. The crucial part of this strategy is expression of a broad family of salivary proteins, called Evasins, to neutralize chemokines responsible for cell trafficking and recruitment. However, structural information about Evasins is still scarce, and little is known about the structural determinants of their binding mechanism to chemokines. Here, we studied the structurally uncharacterized Evasin-4, which neutralizes a broad range of CC-motif chemokines, including the chemokine CC-motif ligand 5 (CCL5) involved in atherogenesis. Crystal structures of Evasin-4 and E66S CCL5, an obligatory dimeric variant of CCL5, were determined to a resolution of 1.3-1.8 Å. The Evasin-4 crystal structure revealed an L-shaped architecture formed by an N- and C-terminal subdomain consisting of eight ß-strands and an α-helix that adopts a substantially different position compared with closely related Evasin-1. Further investigation into E66S CCL5-Evasin-4 complex formation with NMR spectroscopy showed that residues of the N terminus are involved in binding to CCL5. The peptide derived from the N-terminal region of Evasin-4 possessed nanomolar affinity to CCL5 and inhibited CCL5 activity in monocyte migration assays. This suggests that Evasin-4 derivatives could be used as a starting point for the development of anti-inflammatory drugs.


Asunto(s)
Quimiocina CCL5/antagonistas & inhibidores , Proteínas y Péptidos Salivales/química , Garrapatas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CCL5/metabolismo , Cristalografía por Rayos X , Humanos , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Proteínas y Péptidos Salivales/genética , Proteínas y Péptidos Salivales/metabolismo
3.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298951

RESUMEN

The chemokines CCL5 and CXCL4 are deposited by platelets onto endothelial cells, inducing monocyte arrest. Here, the fate of CCL5 and CXCL4 after endothelial deposition was investigated. Human umbilical vein endothelial cells (HUVECs) and EA.hy926 cells were incubated with CCL5 or CXCL4 for up to 120 min, and chemokine uptake was analyzed by microscopy and by ELISA. Intracellular calcium signaling was visualized upon chemokine treatment, and monocyte arrest was evaluated under laminar flow. Whereas CXCL4 remained partly on the cell surface, all of the CCL5 was internalized into endothelial cells. Endocytosis of CCL5 and CXCL4 was shown as a rapid and active process that primarily depended on dynamin, clathrin, and G protein-coupled receptors (GPCRs), but not on surface proteoglycans. Intracellular calcium signals were increased after chemokine treatment. Confocal microscopy and ELISA measurements in cell organelle fractions indicated that both chemokines accumulated in the nucleus. Internalization did not affect leukocyte arrest, as pretreatment of chemokines and subsequent washing did not alter monocyte adhesion to endothelial cells. Endothelial cells rapidly and actively internalize CCL5 and CXCL4 by clathrin and dynamin-dependent endocytosis, where the chemokines appear to be directed to the nucleus. These findings expand our knowledge of how chemokines attract leukocytes to sites of inflammation.


Asunto(s)
Núcleo Celular/metabolismo , Quimiocina CCL5/metabolismo , Células Endoteliales/metabolismo , Factor Plaquetario 4/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular , Humanos
4.
J Biol Chem ; 294(33): 12370-12379, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31235521

RESUMEN

Chemokines are a group of chemotaxis proteins that regulate cell trafficking and play important roles in immune responses and inflammation. Ticks are blood-sucking parasites that secrete numerous immune-modulatory agents in their saliva to evade host immune responses. Evasin-3 is a small salivary protein that belongs to a class of chemokine-binding proteins isolated from the brown dog tick, Rhipicephalus sanguineus Evasin-3 has been shown to have a high affinity for chemokines CXCL1 and CXCL8 and to diminish inflammation in mice. In the present study, solution NMR spectroscopy was used to investigate the structure of Evasin-3 and its CXCL8-Evasin-3 complex. Evasin-3 is found to disrupt the glycosaminoglycan-binding site of CXCL8 and inhibit the interaction of CXCL8 with CXCR2. Structural data were used to design two novel CXCL8-binding peptides. The linear tEv3 17-56 and cyclic tcEv3 16-56 dPG Evasin-3 variants were chemically synthesized by solid-phase peptide synthesis. The affinity of these newly synthesized variants to CXCL8 was measured by surface plasmon resonance biosensor analysis. The Kd values of tEv3 17-56 and tcEv3 16-56 dPG were 27 and 13 nm, respectively. Both compounds effectively inhibited CXCL8-induced migration of polymorphonuclear neutrophils. The present results suggest utility of synthetic Evasin-3 variants as scaffolds for designing and fine-tuning new chemokine-binding agents that suppress immune responses and inflammation.


Asunto(s)
Proteínas de Artrópodos , Glicosaminoglicanos , Neutrófilos/metabolismo , Receptores de Interleucina-8B , Rhipicephalus sanguineus/química , Proteínas y Péptidos Salivales , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/metabolismo , Movimiento Celular , Perros , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Estructura Cuaternaria de Proteína , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/metabolismo , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/metabolismo
5.
Bioconjug Chem ; 31(3): 948-955, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32077689

RESUMEN

Atherosclerosis is one of the leading causes of mortality in developed and developing countries. The onset of atherosclerosis development is accompanied by overexpression of several inflammatory chemokines. Neutralization of these chemokines by chemokine-binding agents attenuates atherosclerosis progression. Here, we studied structural binding features of the tick protein Evasin-3 to chemokine (C-X-C motif) ligand 1 (CXCL1). We showed that Evasin-3-bound CXCL1 is unable to activate the CXCR2 receptor, but retains affinity to glycosaminoglycans. This observation was exploited to detect inflammation by visualizing a group of closely related CXC-type chemokines deposited on cell walls in human endothelial cells and murine carotid arteries by a fluorescent Evasin-3 conjugate. This work highlights the applicability of tick-derived chemokine-binding conjugates as a platform for the development of new agents for inflammation imaging.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Quimiocinas CXC/metabolismo , Endotelio Vascular/metabolismo , Garrapatas , Animales , Enfermedades de las Arterias Carótidas/metabolismo , Glicosaminoglicanos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/diagnóstico por imagen , Inflamación/metabolismo , Ratones
6.
Respir Res ; 17(1): 68, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27259950

RESUMEN

BACKGROUND: Colonization of the airways with potential pathogenic bacteria is observed in a number of chronic respiratory diseases, such as COPD or cystic fibrosis. Infections with respiratory viruses are known triggers of exacerbations of these diseases. We here investigated if pre-exposure to bacteria alters the response of lung epithelial cells to subsequent viral infection. METHODS: Bronchial epithelial cells (BEAS-2B cells and primary bronchial epithelial cells) were exposed to heat-inactivated Haemophilus influenzae, Pseudomonas aeruginosa or Streptococcus pneumoniae and subsequently infected with respiratory syncytial virus (RSV), type 2 human adenovirus or influenza B. Levels of pro-inflammatory cytokines, viral replication and expression of pattern recognition receptors were determined in culture supernatants and/or cell lysates. RESULTS: Exposure of BEAS-2B cells to H. influenzae before and during RSV-infection synergistically increased the release of IL-6 (increase above calculated additive effect at 72 h: 56 % ± 3 %, mean ± SEM) and IL-8 (53 % ± 12 %). This effect was sustained even when bacteria were washed away before viral infection and was neither associated with enhanced viral replication, nor linked to increased expression of key pattern recognition receptors. P. aeruginosa enhanced the release of inflammatory cytokines to a similar extent, yet only if bacteria were also present during viral infection. S. pneumoniae did not enhance RSV-induced cytokine release. Surprisingly, adenovirus infection significantly reduced IL-6 release in cells exposed to either of the three tested bacterial strains by on average more than 50 %. Infection with influenza B on the other hand did not affect cytokine production in BEAS-2B cells exposed to the different bacterial strains. CONCLUSION: Pre-exposure of epithelial cells to bacteria alters the response to subsequent viral infection depending on the types of pathogen involved. These findings highlight the complexity of microbiome interactions in the airways, possibly contributing to the susceptibility to exacerbations and the natural course of airway diseases.


Asunto(s)
Bacterias/patogenicidad , Coinfección , Células Epiteliales/microbiología , Células Epiteliales/virología , Pulmón/microbiología , Pulmón/virología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Virus/patogenicidad , Adenoviridae/patogenicidad , Animales , Bacterias/inmunología , Chlorocebus aethiops , Citocinas/metabolismo , Perros , Células Epiteliales/metabolismo , Haemophilus influenzae/patogenicidad , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Virus de la Influenza B/patogenicidad , Pulmón/metabolismo , Células de Riñón Canino Madin Darby , Cultivo Primario de Células , Pseudomonas aeruginosa/patogenicidad , Virus Sincitiales Respiratorios/patogenicidad , Infecciones del Sistema Respiratorio/metabolismo , Streptococcus pneumoniae/patogenicidad , Factores de Tiempo , Células Vero , Virus/inmunología
7.
Atherosclerosis ; 319: 132-141, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33468314

RESUMEN

Atherosclerosis is an underlying cause of a broad array of cardiovascular diseases characterized by plaques, arterial wall thickening initiated by hyperlipidemia, pro-inflammatory signals, endothelial dysfunction and the influx of inflammatory cells. By still incompletely characterized mechanisms, these plaques can destabilize or erode, leading to thrombosis and blood vessel occlusion and becomes clinically manifest as angina pectoris, myocardial infarction (MI) or stroke. Among the several blood cell types that are involved in the development of atherosclerosis, the role of platelets during the thrombotic occlusion of ruptured or eroded plaques is well established and clinically exploited as evident by the extensive use of platelet inhibitors. However, there is increasing evidence that platelets are also involved in the earlier stages of atheroma development by exhibiting pro-inflammatory activities. The scope of this review is to describe the role of platelets in the initiation and propagation stages of atherosclerosis and beyond; in atherothrombotic complications.


Asunto(s)
Aterosclerosis , Infarto del Miocardio , Trombosis , Plaquetas , Humanos , Inhibidores de Agregación Plaquetaria
8.
TH Open ; 5(4): e533-e542, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34901735

RESUMEN

The cause of atherothrombosis is rupture or erosion of atherosclerotic lesions, leading to an increased risk of myocardial infarction or stroke. Here, platelet activation plays a major role, leading to the release of bioactive molecules, for example, chemokines and coagulation factors, and to platelet clot formation. Several antiplatelet therapies have been developed for secondary prevention of cardiovascular events, in which anticoagulant drugs are often combined. Besides playing a role in hemostasis, platelets are also involved in inflammation. However, it is unclear whether current antiplatelet therapies also affect platelet immune functions. In this study, the possible anti-inflammatory effects of antiplatelet medications on chemokine release were investigated using enzyme-linked immunosorbent assay and on the chemotaxis of THP-1 cells toward platelet releasates. We found that antiplatelet medication acetylsalicylic acid (ASA) led to reduced chemokine (CC motif) ligand 5 (CCL5) and chemokine (CXC motif) ligand 4 (CXCL4) release from platelets, while leukocyte chemotaxis was not affected. Depending on the agonist, α IIb ß 3 and P2Y 12 inhibitors also affected CCL5 or CXCL4 release. The combination of ASA with a P2Y 12 inhibitor or a phosphodiesterase (PDE) inhibitor did not lead to an additive reduction in CCL5 or CXCL4 release. Interestingly, these combinations did reduce leukocyte chemotaxis. This study provides evidence that combined therapy of ASA and a P2Y 12 or PDE3 inhibitor can decrease the inflammatory leukocyte recruiting potential of the releasate of activated platelets.

9.
Cells ; 10(8)2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34440764

RESUMEN

OBJECTIVE: platelets possess not only haemostatic but also inflammatory properties, which combined are thought to play a detrimental role in thromboinflammatory diseases such as acute coronary syndromes and stroke. Phosphodiesterase (PDE) 3 and -5 inhibitors have demonstrated efficacy in secondary prevention of arterial thrombosis, partially mediated by their antiplatelet action. Yet it is unclear whether such inhibitors also affect platelets' inflammatory functions. Here, we aimed to examine the effect of the PDE3A inhibitor cilostazol and the PDE5 inhibitor tadalafil on platelet function in various aspects of thromboinflammation. Approach and results: cilostazol, but not tadalafil, delayed ex vivo platelet-dependent fibrin formation under whole blood flow over type I collagen at 1000 s-1. Similar results were obtained with blood from Pde3a deficient mice, indicating that cilostazol effects are mediated via PDE3A. Interestingly, cilostazol specifically reduced the release of phosphatidylserine-positive extracellular vesicles (EVs) from human platelets while not affecting total EV release. Both cilostazol and tadalafil reduced the interaction of human platelets with inflamed endothelium under arterial flow and the release of the chemokines CCL5 and CXCL4 from platelets. Moreover, cilostazol, but not tadalafil, reduced monocyte recruitment and platelet-monocyte interaction in vitro. CONCLUSIONS: this study demonstrated yet unrecognised roles for platelet PDE3A and platelet PDE5 in platelet procoagulant and proinflammatory responses.


Asunto(s)
Antiinflamatorios/farmacología , Plaquetas/efectos de los fármacos , Cilostazol/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fibrinolíticos/farmacología , Inhibidores de Fosfodiesterasa 3/farmacología , Activación Plaquetaria/efectos de los fármacos , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/enzimología , Plaquetas/inmunología , Células Cultivadas , Quimiocinas/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fibrina/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de Fosfodiesterasa 5/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Transducción de Señal , Tadalafilo/farmacología
10.
Atherosclerosis ; 310: 17-25, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32877806

RESUMEN

BACKGROUND AND AIMS: Platelets can release extracellular vesicles (EVs) upon stimulation with various agonists. Interestingly, platelets from patients with Glanzmann thrombasthenia have reduced EV release. These platelets lack functional αIIbß3 integrins, indicating that αIIbß3 integrin is critical in vesicle release. Integrin activation is central in platelet function and is associated with e.g. adhesion, aggregation and cytoskeletal rearrangement. However, while platelet activation pathways are widely known, the mechanisms underlying EV release remain uncharacterized. We investigated the role of integrin αIIbß3, phosphatidyl serine (PS) exposure, cytoskeletal rearrangement and their associated signalling pathways in EV release. METHODS: EVs were isolated from activated platelets. Platelet activation status was measured by multicolour flow cytometry. A panel of pharmacologic inhibitors was used to interfere in specific signalling pathways. EV release was quantified enzymatically based on membrane PS content and nanoparticle tracking analysis. In addition, real-time visualization of EV shedding with confocal microscopy and EVs with Cryo-TEM imaging was performed. RESULTS: Platelet activation with convulxin resulted in higher EV release than with activation by thrombin. Kinetic measurements indicated that EV release followed the pattern of αIIbß3 integrin activation and subsequent closure paralleled by PS exposure. Prevention of αIIbß3 activation with the inhibitor tirofiban dramatically suppressed EV release. Similar results were obtained using αIIbß3-deficient platelets from patients with Glanzmann thrombasthenia. Inhibition of actin cytoskeleton rearrangement decreased EV release, whereas inhibition of individual signalling targets upstream of cytoskeletal rearrangement showed no such effects. CONCLUSION: Platelet EV release requires three main events: integrin activation and closure, PS exposure, and cytoskeletal rearrangement.


Asunto(s)
Vesículas Extracelulares , Fosfatidilserinas , Plaquetas , Humanos , Integrina beta3 , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria
11.
J Extracell Vesicles ; 8(1): 1585163, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30863515

RESUMEN

Airway epithelial cells secrete extracellular vesicles (EVs) under basal conditions and when exposed to cigarette smoke extract (CSE). Getting insights into the composition of these EVs will help unravel their functions in homeostasis and smoking-induced pathology. Here, we characterized the proteomic composition of basal and CSE-induced airway epithelial EVs. BEAS-2B cells were left unexposed or exposed to 1% CSE for 24 h, followed by EV isolation using ultrafiltration and size exclusion chromatography. Isolated EVs were labelled with tandem mass tags and their proteomic composition was determined using nano-LC-MS/MS. Tissue factor (TF) activity was determined by a factor Xa generation assay, phosphatidylserine (PS) content by prothrombinase assay and thrombin generation using calibrated automated thrombogram (CAT). Nano-LC-MS/MS identified 585 EV-associated proteins with high confidence. Of these, 201 were differentially expressed in the CSE-EVs according to the moderated t-test, followed by false discovery rate (FDR) adjustment with the FDR threshold set to 0.1. Functional enrichment analysis revealed that 24 proteins of the pathway haemostasis were significantly up-regulated in CSE-EVs, including TF. Increased TF expression on CSE-EVs was confirmed by bead-based flow cytometry and was associated with increased TF activity. CSE-EVs caused faster and more thrombin generation in normal human plasma than control-EVs, which was partly TF-, but also PS-dependent. In conclusion, proteomic analysis allowed us to predict procoagulant properties of CSE-EVs which were confirmed in vitro. Cigarette smoke-induced EVs may contribute to the increased cardiovascular and respiratory risk observed in smokers.

12.
J Vis Exp ; (134)2018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29683452

RESUMEN

The recruitment of leukocytes upon injury or inflammation to sites of injury or tissue damage has been investigated during recent decades and has resulted in the concept of the leukocyte adhesion cascade. However, the exact molecular mechanisms involved in leukocyte recruitment have not yet been fully identified. Since leukocyte recruitment remains an important subject in the field of infection, inflammation, and (auto-) immune research, we present a straightforward laminar flow-based assay to study underlying mechanisms of the adhesion, de-adhesion, and transmigration of leukocytes under venous and arterial flow regimes. The in vitro assay can be used to study the molecular mechanisms that underlie the interactions between leukocytes and their cellular partners in different models of vascular inflammation. This protocol describes a laminar flow-based assay using a parallel-flow chamber and an inverted phase contrast microscope connected to a camera to study the interactions of leukocytes and endothelial cells or platelets, which can be visualized and recorded then analyzed offline. Endothelial cells, platelets, or leukocytes can be pretreated with inhibitors or antibodies to determine the role of specific molecules during this process. Shear conditions, i.e. arterial or venous shear stress, can be easily adapted by the viscosity and flow rate of the perfused fluids and the height of the channel.


Asunto(s)
Endotelio Vascular/citología , Ambiente Controlado , Leucocitos/citología , Plaquetas/metabolismo , Técnicas de Cultivo de Célula/métodos , Endotelio Vascular/metabolismo , Humanos , Leucocitos/metabolismo
13.
J Extracell Vesicles ; 6(1): 1322454, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28717419

RESUMEN

Extracellular vesicles (EVs) are mediators of cell communication during health and disease, and abundantly released by platelets upon activation or during ageing. Platelet EVs exert modulatory effects on immune and vascular cells. Platelet EVs may modulate the function of vascular smooth muscle cells (SMC). Platelet EVs were isolated from platelet-rich plasma and incubated with SMC in order to assess binding, proliferation, migration and pro-inflammatory phenotype of the cells. Platelet EVs firmly bound to resting SMC through the platelet integrin αIIbß3, while binding also occurred in a CX3CL1-CX3CR1-dependent manner after cytokine stimulation. Platelet EVs increased SMC migration comparable to platelet derived growth factor or platelet factor 4 and induced SMC proliferation, which relied on CD40- and P-selectin interactions. Flow-resistant monocyte adhesion to platelet EV-treated SMC was increased compared with resting SMC. Again, this adhesion depended on integrin αIIbß3 and P-selectin, and to a lesser extent on CD40 and CX3CR1. Treatment of SMC with platelet EVs induced interleukin 6 secretion. Finally, platelet EVs induced a synthetic SMC morphology and decreased calponin expression. Collectively, these data indicate that platelet EVs exert a strong immunomodulatory activity on SMC. In particular, platelet EVs induce a switch towards a pro-inflammatory phenotype, stimulating vascular remodelling.

14.
Sci Rep ; 7(1): 15297, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29127410

RESUMEN

Appropriate isolation methods are essential for unravelling the relative contribution of extracellular vesicles (EVs) and the EV-free secretome to homeostasis and disease. We hypothesized that ultrafiltration followed by size exclusion chromatography (UF-SEC) provides well-matched concentrates of EVs and free secreted molecules for proteomic and functional studies. Conditioned media of BEAS-2B bronchial epithelial cells were concentrated on 10 kDa centrifuge filters, followed by separation of EVs and free protein using sepharose CL-4B SEC. Alternatively, EVs were isolated by ultracentrifugation. EV recovery was estimated by bead-coupled flow cytometry and tuneable resistive pulse sensing. The proteomic composition of EV isolates and SEC protein fractions was characterized by nano LC-MS/MS. UF-SEC EVs tended to have a higher yield and EV-to-protein rate of purity than ultracentrifugation EVs. UF-SEC EVs and ultracentrifugation EVs showed similar fold-enrichments for biological pathways that were distinct from those of UF-SEC protein. Treatment of BEAS-2B cells with UF-SEC protein, but not with either type of EV isolate increased the IL-8 concentration in the media whereas EVs, but not protein induced monocyte adhesion to endothelial cells. Thus, UF-SEC is a useful alternative for ultracentrifugation and allows comparing the proteomic composition and functional effects of EVs and free secreted molecules.


Asunto(s)
Cromatografía en Gel , Células Epiteliales/química , Células Epiteliales/metabolismo , Vesículas Extracelulares/química , Medios de Cultivo/química , Humanos , Sefarosa/análogos & derivados , Sefarosa/química , Células THP-1 , Ultrafiltración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA