Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Haematologica ; 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38572559

RESUMEN

Innate myeloid cells especially neutrophils and their extracellular traps are known to promote intravascular coagulation and thrombosis formation in infections and various other conditions. Innate myeloid cell dependent fibrin formation can support systemic immunity while its dysregulation enhances the severity of infectious diseases. Less is known about the immune mechanisms preventing dysregulation of fibrin homeostasis in infection. During experimental systemic infections local fibrin deposits in the liver microcirculation cause rapid arrest of CD4+ T cells. Arrested T helper cells mostly represent Th17 cells that partially originate from the small intestine. Intravascular fibrin deposits activate mouse and human CD4+ T cells which can be mediated by direct fibrin - CD4+ T cell interactions. Activated CD4+ T cells suppress fibrin deposition and microvascular thrombosis by directly counteracting coagulation activation by neutrophils and classical monocytes. T cell activation, which is initially triggered by IL- 12p40- and MHC-II dependent mechanisms, enhances intravascular fibrinolysis via LFA-1. Moreover, CD4+ T cells disfavor the association of the fibrinolysis inhibitor TAFI with fibrin whereby fibrin deposition is increased by TAFI in the absence but not presence of T cells. In human infections thrombosis development is inversely related to microvascular levels of CD4+ T cells. Thus, fibrin promotes LFA-1 dependent T helper cell activation in infections which drives a negative feedback cycle that rapidly restricts intravascular fibrin and thrombosis development.

2.
Cell Mol Life Sci ; 79(10): 512, 2022 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-36094626

RESUMEN

To fulfil its orchestration of immune cell trafficking, a network of chemokines and receptors developed that capitalizes on specificity, redundancy, and functional selectivity. The discovery of heteromeric interactions in the chemokine interactome has expanded the complexity within this network. Moreover, some inflammatory mediators, not structurally linked to classical chemokines, bind to chemokine receptors and behave as atypical chemokines (ACKs). We identified macrophage migration inhibitory factor (MIF) as an ACK that binds to chemokine receptors CXCR2 and CXCR4 to promote atherogenic leukocyte recruitment. Here, we hypothesized that chemokine-chemokine interactions extend to ACKs and that MIF forms heterocomplexes with classical chemokines. We tested this hypothesis by using an unbiased chemokine protein array. Platelet chemokine CXCL4L1 (but not its variant CXCL4 or the CXCR2/CXCR4 ligands CXCL8 or CXCL12) was identified as a candidate interactor. MIF/CXCL4L1 complexation was verified by co-immunoprecipitation, surface plasmon-resonance analysis, and microscale thermophoresis, also establishing high-affinity binding. We next determined whether heterocomplex formation modulates inflammatory/atherogenic activities of MIF. Complex formation was observed to inhibit MIF-elicited T-cell chemotaxis as assessed by transwell migration assay and in a 3D-matrix-based live cell-imaging set-up. Heterocomplexation also blocked MIF-triggered migration of microglia in cortical cultures in situ, as well as MIF-mediated monocyte adhesion on aortic endothelial cell monolayers under flow stress conditions. Of note, CXCL4L1 blocked binding of Alexa-MIF to a soluble surrogate of CXCR4 and co-incubation with CXCL4L1 attenuated MIF responses in HEK293-CXCR4 transfectants, indicating that complex formation interferes with MIF/CXCR4 pathways. Because MIF and CXCL4L1 are platelet-derived products, we finally tested their role in platelet activation. Multi-photon microscopy, FLIM-FRET, and proximity-ligation assay visualized heterocomplexes in platelet aggregates and in clinical human thrombus sections obtained from peripheral artery disease (PAD) in patients undergoing thrombectomy. Moreover, heterocomplexes inhibited MIF-stimulated thrombus formation under flow and skewed the lamellipodia phenotype of adhering platelets. Our study establishes a novel molecular interaction that adds to the complexity of the chemokine interactome and chemokine/receptor-network. MIF/CXCL4L1, or more generally, ACK/CXC-motif chemokine heterocomplexes may be target structures that can be exploited to modulate inflammation and thrombosis.


Asunto(s)
Aterosclerosis , Factores Inhibidores de la Migración de Macrófagos , Trombosis , Aterosclerosis/metabolismo , Células HEK293 , Humanos , Inflamación/metabolismo , Oxidorreductasas Intramoleculares , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factor Plaquetario 4 , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo
3.
FASEB J ; 33(2): 1860-1872, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30207797

RESUMEN

Tumor microvesicles are a peculiar type of extracellular vesicles that circulate in the blood of patients with metastatic cancer. The itineraries and immune cell interactions of tumor microvesicles during the intravascular and extravascular stages of metastasis are largely unknown. We found that the lipid receptor CD36 is a major mediator of the engulfment of pancreatic tumor microvesicles by myeloid immune cells in vitro and critically samples circulating tumor microvesicles by resident liver macrophages in mice in vivo. Direct nanoscopic imaging of individual tumor microvesicles shows that the microvesicles rapidly decay during engulfment whereby their cargo is targeted concomitantly to the plasma membrane and the cytoplasm excluding lysosomal compartments. CD36 also promotes internalization of blood cell (nontumor) microvesicles, which involves endolysosomal pathways. A portion of tumor microvesicles circulating in the liver microcirculation traverses the vessel wall in a CD36-dependent way. Extravasated microvesicles colonize distinct perivascular Ly6C- macrophages for at least 2 wk. Thus, the microvesicles are increasingly integrated into CD36-induced premetastatic cell clusters and enhance development of liver metastasis. Hence, promotion of metastasis by pancreatic tumor microvesicles is associated with CD36-regulated immune cell invasion and extravasation of microvesicles and persistent infiltration of specific tissue macrophages by microvesicle cargo.-Pfeiler, S., Thakur, M., Grünauer, P., Megens, R. T. A., Joshi, U., Coletti, R., Samara, V., Müller-Stoy, G., Ishikawa-Ankerhold, H., Stark, K., Klingl, A., Fröhlich, T., Arnold, G. J., Wörmann, S., Bruns, C. J., Algül, H., Weber, C., Massberg, S., Engelmann, B. CD36-triggered cell invasion and persistent tissue colonization by tumor microvesicles during metastasis.


Asunto(s)
Antígenos CD36/inmunología , Micropartículas Derivadas de Células/inmunología , Lisosomas/inmunología , Macrófagos/inmunología , Neoplasias Pancreáticas/inmunología , Micropartículas Derivadas de Células/patología , Humanos , Lisosomas/patología , Macrófagos/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Células THP-1
4.
Arterioscler Thromb Vasc Biol ; 38(4): 772-786, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29419408

RESUMEN

OBJECTIVE: Cancer patients are at high risk of developing deep venous thrombosis (DVT) and venous thromboembolism, a leading cause of mortality in this population. However, it is largely unclear how malignant tumors drive the prothrombotic cascade culminating in DVT. APPROACH AND RESULTS: Here, we addressed the pathophysiology of malignant DVT compared with nonmalignant DVT and focused on the role of tumor microvesicles as potential targets to prevent cancer-associated DVT. We show that microvesicles released by pancreatic adenocarcinoma cells (pancreatic tumor-derived microvesicles [pcMV]) boost thrombus formation in a model of flow restriction of the mouse vena cava. This depends on the synergistic activation of coagulation by pcMV and host tissue factor. Unlike nonmalignant DVT, which is initiated and propagated by innate immune cells, thrombosis triggered by pcMV was largely independent of myeloid leukocytes or platelets. Instead, we identified externalization of the phospholipid phosphatidylethanolamine as a major mechanism controlling the prothrombotic activity of pcMV. Disrupting phosphatidylethanolamine-dependent activation of factor X suppressed pcMV-induced DVT without causing changes in hemostasis. CONCLUSIONS: Together, we show here that the pathophysiology of pcMV-associated experimental DVT differs markedly from innate immune cell-promoted nonmalignant DVT and is therefore amenable to distinct antithrombotic strategies. Targeting phosphatidylethanolamine on tumor microvesicles could be a new strategy for prevention of cancer-associated DVT without causing bleeding complications.


Asunto(s)
Adenocarcinoma/complicaciones , Coagulación Sanguínea , Micropartículas Derivadas de Células/metabolismo , Neoplasias Pancreáticas/complicaciones , Vena Cava Inferior/metabolismo , Trombosis de la Vena/etiología , Adenocarcinoma/sangre , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Bacteriocinas/farmacología , Coagulación Sanguínea/efectos de los fármacos , Línea Celular Tumoral , Micropartículas Derivadas de Células/efectos de los fármacos , Micropartículas Derivadas de Células/patología , Modelos Animales de Enfermedad , Diseño de Fármacos , Factor Xa/metabolismo , Fibrinolíticos/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Péptidos/farmacología , Fosfatidiletanolaminas/antagonistas & inhibidores , Fosfatidiletanolaminas/sangre , Transducción de Señal , Tromboplastina/metabolismo , Vena Cava Inferior/efectos de los fármacos , Vena Cava Inferior/patología , Trombosis de la Vena/sangre , Trombosis de la Vena/patología , Trombosis de la Vena/prevención & control
5.
Haematologica ; 102(2): 206-213, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27927771

RESUMEN

Neutrophils, early mediators of the innate immune defense, are recruited to developing thrombi in different types of thrombosis. They amplify intravascular coagulation by stimulating the tissue factor-dependent extrinsic pathway via inactivation of endogenous anticoagulants, enhancing factor XII activation or decreasing plasmin generation. Neutrophil-dependent prothrombotic mechanisms are supported by the externalization of decondensed nucleosomes and granule proteins that together form neutrophil extracellular traps. These traps, either in intact or fragmented form, are causally involved in various forms of experimental thrombosis as first indicated by their role in the enhancement of both microvascular thrombosis during bacterial infection and carotid artery thrombosis. Neutrophil extracellular traps can be induced by interactions of neutrophils with activated platelets; vice versa, these traps enhance adhesion of platelets via von Willebrand factor. Neutrophil-induced microvascular thrombus formation can restrict the dissemination and survival of blood-borne bacteria and thereby sustain intravascular immunity. Dysregulation of this innate immune pathway may support sepsis-associated coagulopathies. Notably, neutrophils and extracellular nucleosomes, together with platelets, critically promote fibrin formation during flow restriction-induced deep vein thrombosis. Neutrophil extracellular traps/extracellular nucleosomes are increased in thrombi and in the blood of patients with different vaso-occlusive pathologies and could be therapeutically targeted for the prevention of thrombosis. Thus, during infections and in response to blood vessel damage, neutrophils and externalized nucleosomes are major promoters of intravascular blood coagulation and thrombosis.


Asunto(s)
Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , Nucleosomas/metabolismo , Trombosis/etiología , Trombosis/metabolismo , Animales , Biomarcadores , Coagulación Sanguínea , Plaquetas/inmunología , Plaquetas/metabolismo , Cromatina/metabolismo , Fibrina/metabolismo , Humanos , Inmunidad Innata , Neutrófilos/inmunología , Activación Plaquetaria , Trombosis/sangre , Trombosis/patología
6.
Basic Res Cardiol ; 111(6): 69, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27743118

RESUMEN

In this meeting report, particularly addressing the topic of protection of the cardiovascular system from ischemia/reperfusion injury, highlights are presented that relate to conditioning strategies of the heart with respect to molecular mechanisms and outcome in patients' cohorts, the influence of co-morbidities and medications, as well as the contribution of innate immune reactions in cardioprotection. Moreover, developmental or systems biology approaches bear great potential in systematically uncovering unexpected components involved in ischemia-reperfusion injury or heart regeneration. Based on the characterization of particular platelet integrins, mitochondrial redox-linked proteins, or lipid-diol compounds in cardiovascular diseases, their targeting by newly developed theranostics and technologies opens new avenues for diagnosis and therapy of myocardial infarction to improve the patients' outcome.


Asunto(s)
Cardiología/tendencias , Enfermedades Cardiovasculares , Nanomedicina Teranóstica/tendencias , Animales , Cardiología/métodos , Humanos
7.
Sci Adv ; 10(12): eadl1710, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38517968

RESUMEN

Neutrophils rapidly respond to inflammation and infection, but to which degree their functional trajectories after mobilization from the bone marrow are shaped within the circulation remains vague. Experimental limitations have so far hampered neutrophil research in human disease. Here, using innovative fixation and single-cell-based toolsets, we profile human and murine neutrophil transcriptomes and proteomes during steady state and bacterial infection. We find that peripheral priming of circulating neutrophils leads to dynamic shifts dominated by conserved up-regulation of antimicrobial genes across neutrophil substates, facilitating pathogen containment. We show the TLR4/NF-κB signaling-dependent up-regulation of canonical neutrophil activation markers like CD177/NB-1 during acute inflammation, resulting in functional shifts in vivo. Blocking de novo RNA synthesis in circulating neutrophils abrogates these plastic shifts and prevents the adaptation of antibacterial neutrophil programs by up-regulation of distinct effector molecules upon infection. These data underline transcriptional plasticity as a relevant mechanism of functional neutrophil reprogramming during acute infection to foster bacterial containment within the circulation.


Asunto(s)
Neutrófilos , Transcriptoma , Ratones , Humanos , Animales , Neutrófilos/metabolismo , Proteómica , Inflamación/genética , Inflamación/metabolismo , Perfilación de la Expresión Génica
8.
Blood ; 115(20): 4102-10, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20107233

RESUMEN

Platelets play a key role in hemostasis and various diseases including arterial thrombosis. Glycoprotein VI (GPVI) mediates adhesion to collagen structures exposed at sites of vascular injury and subsequent platelet activation. We determined the effects of specific activation of GPVI on the human platelet proteome. Isolated human platelets were stimulated with an activating monoclonal antibody specific for GPVI. Platelet proteins were analyzed by 2-dimensional difference gel electrophoresis (2D-DIGE) and mass spectrometry. We identified 8 differentially abundant proteins associated with cell signaling, metabolism, organization and rearrangement of the cytoskeleton, and membrane trafficking. Differentially abundant proteins included aldose reductase (AR), beta-centractin, charged multivesicular body protein 3, Src substrate cortactin, ERp57, and pleckstrin. Importantly, GPVI-modulated protein abundance was functionally relevant. Correspondingly, AR enzyme activity significantly increased upon GPVI activation and inhibition of AR resulted in reduced platelet aggregation. Furthermore, ERp57 was released upon ligation of platelet GPVI and increased the activity of tissue factor, a major initiator of blood coagulation. In summary, GPVI activation results in differential changes in abundance of platelet proteins, including AR and ERp57, which support platelet aggregation and platelet-dependent coagulation. These results provide further insight into the mechanisms that underlie platelet activation through the GPVI receptor and may help to identify novel pharmacologic targets.


Asunto(s)
Biomarcadores/metabolismo , Plaquetas/metabolismo , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteoma/análisis , Trombosis/metabolismo , Adulto , Anticuerpos Monoclonales , Coagulación Sanguínea , Western Blotting , Electroforesis en Gel Bidimensional , Femenino , Citometría de Flujo , Humanos , Masculino , Activación Plaquetaria , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Adulto Joven
9.
Biochim Biophys Acta ; 1801(6): 609-16, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20226876

RESUMEN

Mammalian and arthropod cells acquire phospholipids by protein-mediated pathways that comprise selective and whole particle uptake routes. Phospholipid uptake critically supports cellular incorporation of nutrition-derived polyunsaturated fatty acids. It can occur jointly with cholesterol uptake, but intracellular processing of phospholipids is distinctively different from sterol processing. The newly imported phospholipids are utilized for production of bioactive lipids, such as thromboxane A(2) and lyso phosphatidic acid, and for synthesis of triacylglycerol. Class B scavenger receptor BI (SR-BI) represents a major mediator of the uptake of various phospholipids. The related scavenger receptor CD36, as shown here, also facilitates cellular phospholipid uptake. CD36 supports import of the choline phospholipids phosphatidylcholine (PC) and sphingomyelin (SM), but not of phosphatidylethanolamine (PE). Other transferases trigger cellular uptake of selective phospholipids, such as phosphatidic acid (PA) phosphatases that facilitate PA import and thereby modify cell survival and synaptic transmission. Phospholipid uptake depends on the activation status of cells. Activation of blood platelets indeed increases PE uptake. This is mediated by the serpin protein C inhibitor (PCI) and enhances thrombin formation. Exchange of phospholipids between blood cells and lipoproteins partially adjusts the lipid distribution pattern of blood cells to the one of lipoprotein particles. This in turn modifies the activities of cell membrane sodium transporters and could thereby contribute to sodium flux alterations in the metabolic syndrome. The in vivo relevance of phospholipid uptake in humans is indicated by comparable and reversible changes in the same phospholipid species in both lipoproteins and cells after rapid removal of low-density lipoproteins. Finally, cells also incorporate oxidized (pathogenic) phospholipids using partially overlapping entry pathways as native phospholipids which might support the ability of oxidized lipids to promote atherothrombosis.


Asunto(s)
Fosfolípidos/metabolismo , Transporte Biológico , Ácidos Grasos Insaturados/sangre , Transducción de Señal , Transmisión Sináptica
10.
J Clin Invest ; 118(3): 1110-22, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18274674

RESUMEN

The activation of initiator protein tissue factor (TF) is likely to be a crucial step in the blood coagulation process, which leads to fibrin formation. The stimuli responsible for inducing TF activation are largely undefined. Here we show that the oxidoreductase protein disulfide isomerase (PDI) directly promotes TF-dependent fibrin production during thrombus formation in vivo. After endothelial denudation of mouse carotid arteries, PDI was released at the injury site from adherent platelets and disrupted vessel wall cells. Inhibition of PDI decreased TF-triggered fibrin formation in different in vivo murine models of thrombus formation, as determined by intravital fluorescence microscopy. PDI infusion increased - and, under conditions of decreased platelet adhesion, PDI inhibition reduced - fibrin generation at the injury site, indicating that PDI can directly initiate blood coagulation. In vitro, human platelet-secreted PDI contributed to the activation of cryptic TF on microvesicles (microparticles). Mass spectrometry analyses indicated that part of the extracellular cysteine 209 of TF was constitutively glutathionylated. Mixed disulfide formation contributed to maintaining TF in a state of low functionality. We propose that reduced PDI activates TF by isomerization of a mixed disulfide and a free thiol to an intramolecular disulfide. Our findings suggest that disulfide isomerases can act as injury response signals that trigger the activation of fibrin formation following vessel injury.


Asunto(s)
Coagulación Sanguínea , Fibrina/biosíntesis , Proteína Disulfuro Isomerasas/fisiología , Transducción de Señal/fisiología , Tromboplastina/fisiología , Animales , Células Cultivadas , Disulfuros/química , Glutatión/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Tromboplastina/química
11.
J Cachexia Sarcopenia Muscle ; 12(5): 1333-1351, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34427055

RESUMEN

BACKGROUND: Cancer cachexia (CCx) is a multifactorial wasting disorder characterized by involuntary loss of body weight that affects many cancer patients and implies a poor prognosis, reducing both tolerance to and efficiency of anticancer therapies. Actual challenges in management of CCx remain in the identification of tumour-derived and host-derived mediators involved in systemic inflammation and tissue wasting and in the discovery of biomarkers that would allow for an earlier and personalized care of cancer patients. The aim of this study was to identify new markers of CCx across different species and tumour entities. METHODS: Quantitative secretome analysis was performed to identify specific factors characteristic of cachexia-inducing cancer cell lines. To establish the subsequently identified phospholipase PLA2G7 as a marker of CCx, plasma PLA2G7 activity and/or protein levels were measured in well-established mouse models of CCx and in different cohorts of weight-stable and weight-losing cancer patients with different tumour entities. Genetic PLA2G7 knock-down in tumours and pharmacological treatment using the well-studied PLA2G7 inhibitor darapladib were performed to assess its implication in the pathogenesis of CCx in C26 tumour-bearing mice. RESULTS: High expression and secretion of PLA2G7 were hallmarks of cachexia-inducing cancer cell lines. Circulating PLA2G7 activity was increased in different mouse models of CCx with various tumour entities and was associated with the severity of body wasting. Circulating PLA2G7 levels gradually rose during cachexia development. Genetic PLA2G7 knock-down in C26 tumours only partially reduced plasma PLA2G7 levels, suggesting that the host is also an important contributor. Chronic treatment with darapladib was not sufficient to counteract inflammation and tissue wasting despite a strong inhibition of the circulating PLA2G7 activity. Importantly, PLA2G7 levels were also increased in colorectal and pancreatic cancer patients with CCx. CONCLUSIONS: Overall, our data show that despite no immediate pathogenic role, at least when targeted as a single entity, PLA2G7 is a consistent marker of CCx in both mice and humans. The early increase in circulating PLA2G7 levels in pre-cachectic mice supports future prospective studies to assess its potential as biomarker for cancer patients.


Asunto(s)
Caquexia , Neoplasias Pancreáticas , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Animales , Benzaldehídos , Biomarcadores , Caquexia/tratamiento farmacológico , Caquexia/etiología , Humanos , Ratones , Oximas , Estudios Prospectivos
12.
Thromb Haemost ; 119(8): 1274-1282, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31254975

RESUMEN

Atherothrombosis is a frequent cause of cardiovascular mortality. It is mostly triggered by plaque rupture and exposure of the thrombogenic subendothelial matrix, which initiates platelet aggregation and clot formation. Current antithrombotic strategies, however, target both thrombosis and physiological hemostasis and thereby increase bleeding risk. Thus, there is an unmet clinical need for optimized therapies. Neutrophil activation and consecutive interactions of neutrophils and platelets contribute mechanistically to thromboinflammation and arterial thrombosis, and thus present a potential therapeutic target. Platelet-neutrophil interactions are mediated through adhesion molecules such as P-selectin and P-selectin glycoprotein ligand 1 as well as glycoprotein Ib and macrophage-1 antigen, which mediate physical cell interactions and intracellular signaling. Release of soluble mediators as well as direct signaling between platelets and neutrophils lead to their reciprocal activation and neutrophil release of extracellular traps, scaffolds of condensed chromatin that play a prothrombotic role in atherothrombosis. In this article, we review the role of neutrophils and neutrophil-derived prothrombotic molecules in platelet activation and atherothrombosis, and highlight potential therapeutic targets.


Asunto(s)
Aterosclerosis/metabolismo , Plaquetas/citología , Comunicación Celular , Neutrófilos/citología , Trombosis/metabolismo , Animales , Adhesión Celular , Trampas Extracelulares , Hemostasis , Humanos , Inflamación , Glicoproteínas de Membrana/metabolismo , Ratones , Selectina-P/metabolismo , Activación Plaquetaria , Agregación Plaquetaria , Transducción de Señal , Resultado del Tratamiento
13.
Thromb Res ; 122 Suppl 1: S19-22, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18691493

RESUMEN

Recent evidence suggests that protein disulfide isomerase (PDI) represents an injury response signal that can activate tissue factor (TF), a major initiator protein of blood coagulation. PDI was found to be specifically exposed at the site of vessel injury, originating both from disrupted vessel-wall cells and adhering platelets. The exposed PDI promotes TF-dependent fibrin deposition in different mouse models of thrombosis. In particular, PDI can mediate stimulation of circulating (intravascular) TF present on microparticles. It has been proposed that PDI activates TF by changing the disulfide status of the membrane-proximal Cys186-Cys209 pair of TF. Indeed, PDI was shown to cleave mixed disulfide bonds of TF with glutathione. This might enable the formation of an intrachain disulfide bond which is associated with an increased procoagulant efficiency of TF. The PDI-induced activation of TF could represent the primary step of the entire coagulation process.


Asunto(s)
Fibrina/biosíntesis , Proteína Disulfuro Isomerasas/metabolismo , Tromboplastina/metabolismo , Animales , Cisteína/química , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Glutatión/metabolismo , Humanos , Ratones , Tromboplastina/química
14.
FASEB J ; 19(8): 898-909, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15923400

RESUMEN

Lipid-rich atherosclerotic plaques are vulnerable, and their rupture can cause the formation of a platelet- and fibrin-rich thrombus leading to myocardial infarction and ischemic stroke. Although the role of plaque-based tissue factor as stimulator of blood coagulation has been recognized, it is not known whether plaques can cause thrombus formation through direct activation of platelets. We isolated lipid-rich atheromatous plaques from 60 patients with carotid stenosis and identified morphologically diverse collagen type I- and type III-positive structures in the plaques that directly stimulated adhesion, dense granule secretion, and aggregation of platelets in buffer, plasma, and blood. This material also elicited platelet-monocyte aggregation and platelet-dependent blood coagulation. Plaques exposed to flowing blood at arterial wall shear rate induced platelets to adhere to and spread on the collagenous structures, triggering subsequent thrombus formation. Plaque-induced platelet thrombus formation was observed in fully anticoagulated blood (i.e., in the absence of tissue factor-mediated coagulation). Mice platelets lacking glycoprotein VI (GPVI) were unable to adhere to atheromatous plaque or form thrombi. Human platelet thrombus formation onto plaques in flowing blood was completely blocked by GPVI inhibition with the antibody 10B12 but not affected by integrin alpha2beta1 inhibition with 6F1 mAb. Moreover, the initial platelet response, shape change, induced by plaque was blocked by GPVI inhibition but not with alpha2beta1 antagonists (6F1 mAb or GFOGER-GPP peptide). Pretreatment of plaques with collagenase or anti-collagen type I and anti-collagen type III antibodies abolished plaque-induced platelet activation. Our results indicate that morphologically diverse collagen type I- and collagen type III-containing structures in lipid-rich atherosclerotic plaques stimulate thrombus formation by activating platelet GPVI. This platelet collagen receptor, essential for plaque-induced thrombus formation, presents a promising new anti-thrombotic target for the prevention of ischemic cardiovascular diseases.


Asunto(s)
Aterosclerosis/complicaciones , Glicoproteínas de Membrana Plaquetaria/fisiología , Trombosis/etiología , Animales , Aterosclerosis/patología , Coagulación Sanguínea , Plaquetas/fisiología , Estenosis Carotídea/sangre , Colágeno Tipo I/análisis , Colágeno Tipo III/análisis , Humanos , Integrina alfa2beta1/fisiología , Lípidos/análisis , Ratones , Microscopía Fluorescente , Monocitos/fisiología , Activación Plaquetaria , Adhesividad Plaquetaria , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores
15.
Biochem J ; 385(Pt 3): 831-8, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15654766

RESUMEN

FSAP (Factor VII-activating protease) is a new plasma-derived serine protease with putative dual functions in haemostasis, including activation of coagulation Factor VII and generation of urinary-type plasminogen activator (urokinase). The (auto-)activation of FSAP is facilitated by polyanionic glycosaminoglycans, such as heparin or dextran sulphate, whereas calcium ions stabilize the active form of FSAP. In the present study, extracellular RNA was identified and characterized as a novel FSAP cofactor. The conditioned medium derived from various cell types such as smooth muscle cells, endothelial cells, osteosarcoma cells or CHO (Chinese-hamster ovary) cells contained an acidic factor that initiated (auto-)activation of FSAP. RNase A, but not other hydrolytic enzymes (proteases, glycanases and DNase), abolished the FSAP cofactor activity, which was subsequently isolated by anion-exchange chromatography and unequivocally identified as RNA. In purified systems, as well as in plasma, different forms of natural RNA (rRNA, tRNA, viral RNA and artificial RNA) were able to (auto-)activate FSAP into the two-chain enzyme form. The specific binding of FSAP to RNA (but not to DNA) was shown by mobility-shift assays and UV crosslinking, thereby identifying FSAP as a new extracellular RNA-binding protein, the K(D) estimated to be 170-350 nM. Activation of FSAP occurred through an RNA-dependent template mechanism involving a nucleic acid size of at least 100 nt. In a purified system, natural RNA augmented the FSAP-dependent Factor VII activation several-fold (as shown by subsequent Factor Xa generation), as well as the FSAP-mediated generation of urokinase. Our results provide evidence for the first time that extracellular RNA, present at sites of cell damage or vascular injury, can serve an important as yet unrecognized cofactor function in haemostasis by inducing (auto-)activation of FSAP through a novel surface-dependent mechanism.


Asunto(s)
ARN/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Células Cultivadas , Coenzimas/aislamiento & purificación , Coenzimas/metabolismo , Coenzimas/farmacología , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Unión Proteica , ARN/aislamiento & purificación , ARN/farmacología , Ribonucleasa Pancreática/metabolismo , Serina Endopeptidasas/sangre , Serina Endopeptidasas/aislamiento & purificación , Especificidad por Sustrato
16.
Sci Rep ; 6: 34440, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27694929

RESUMEN

The mechanisms protecting from immunopathology during acute bacterial infections are incompletely known. We found that in response to apoptotic immune cells and live or dead Listeria monocytogenes scavenger receptor BI (SR-BI), an anti-atherogenic lipid exchange mediator, activated internalization mechanisms with characteristics of macropinocytosis and, assisted by Golgi fragmentation, initiated autophagic responses. This was supported by scavenger receptor-induced local increases in membrane cholesterol concentrations which generated lipid domains particularly in cell extensions and the Golgi. SR-BI was a key driver of beclin-1-dependent autophagy during acute bacterial infection of the liver and spleen. Autophagy regulated tissue infiltration of neutrophils, suppressed accumulation of Ly6C+ (inflammatory) macrophages, and prevented hepatocyte necrosis in the core of infectious foci. Perifocal levels of Ly6C+ macrophages and Ly6C- macrophages were unaffected, indicating predominant regulation of the focus core. SR-BI-triggered autophagy promoted co-elimination of apoptotic immune cells and dead bacteria but barely influenced bacterial sequestration and survival or inflammasome activation, thus exclusively counteracting damage inflicted by immune responses. Hence, SR-BI- and autophagy promote a surveillance pathway that partially responds to products of antimicrobial defenses and selectively prevents immunity-induced damage during acute infection. Our findings suggest that control of infection-associated immunopathology can be based on a unified defense operation.


Asunto(s)
Autofagia/inmunología , Macrófagos/inmunología , Microdominios de Membrana/inmunología , Pinocitosis/inmunología , Receptores Depuradores de Clase B/inmunología , Animales , Autofagia/genética , Beclina-1/genética , Beclina-1/inmunología , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/inmunología , Listeriosis/patología , Hepatopatías/genética , Hepatopatías/inmunología , Hepatopatías/patología , Macrófagos/patología , Microdominios de Membrana/genética , Ratones , Ratones Noqueados , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Pinocitosis/genética , Receptores Depuradores de Clase B/genética , Enfermedades del Bazo/genética , Enfermedades del Bazo/inmunología , Enfermedades del Bazo/patología
17.
FASEB J ; 17(3): 476-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12514112

RESUMEN

Although tissue factor (TF), the principial initiator of physiological coagulation and pathological thrombosis, has recently been proposed to be present in human blood, the functional significance and location of the intravascular TF is unknown. In the plasma portion of blood, we found TF to be mainly associated with circulating microvesicles. By cell sorting with the specific marker CD42b, platelet-derived microvesicles were identified as a major location of the plasma TF. This was confirmed by the presence of full-length TF in microvesicles acutely shedded from the activated platelets. TF was observed to be stored in the alpha-granules and the open canalicular system of resting platelets and to be exposed on the cell surface after platelet activation. Functional competence of the blood-based TF was enabled when the microvesicles and platelets adhered to neutrophils, as mediated by P-selectin and neutrophil counterreceptor (PSGL-1, CD18 integrins) interactions. Moreover, neutrophil-secreted oxygen radical species supported the intravascular TF activity. The pools of platelet and microvesicle TF contributed additively and to a comparable extent to the overall blood TF activity, indicating a substantial participation of the microvesicle TF. Our results introduce a new concept of TF-mediated coagulation crucially dependent on TF associated with microvesicles and activated platelets, which principally enables the entire coagulation system to proceed on a restricted cell surface.


Asunto(s)
Coagulación Sanguínea , Plaquetas/química , Vesículas Citoplasmáticas/química , Tromboplastina/análisis , Tromboplastina/fisiología , Plaquetas/fisiología , Plaquetas/ultraestructura , Vasos Sanguíneos/química , Adhesión Celular , Vesículas Citoplasmáticas/ultraestructura , Fibrina/biosíntesis , Humanos , Modelos Biológicos , Neutrófilos/fisiología , Activación Plaquetaria , Especies Reactivas de Oxígeno/metabolismo
18.
Thromb Haemost ; 89(1): 3-8, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12540946

RESUMEN

The loss of blood through vessel wall ruptures is initially prevented by the rapid adhesion of platelets to the subendothelium, and the formation of a thrombus consisting of platelets and different types of leukocytes. Concomitantly, the coagulation process is thought to be activated by vascular wall tissue factor (TF). Here, a new model for the initiation of coagulation is presented, based on unexpected findings on the presence and functional activation of TF within the blood itself. TF was recently found to be stored in the alpha-granules of resting platelets under physiological conditions. Activation by collagen exposes TF on the platelet cell membrane and on platelet derived microvesicles. Adhesive interactions of the TF bearing platelets and microvesicles to neutrophils and monocytes support the functional activation of the blood based TF. The intravascular TF pathway is proposed to play a significant role during hemostasis by enabling the generation of fibrin at the site of the developing thrombus.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Tromboplastina/metabolismo , Plaquetas/química , Plaquetas/ultraestructura , Comunicación Celular , Humanos , Tromboplastina/fisiología
20.
Thromb Res ; 133 Suppl 1: S35-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24759139

RESUMEN

Microvascular thrombosis indicates a pathological occlusion of microvessels by fibrin- and/or platelet-rich thrombi. It is observed during systemic infections, cancer, myocardial infarction, stroke, neurodegenerative diseases and in thrombotic microangiopathies. Microvessel thrombosis can cause greatly differing symptoms that range from limited changes in plasma coagulation markers to severe multi-organ failure. Because microvessel thrombi are difficult to detect and often occur only transiently, their importance for disease development and host biology is likely markedly under-appreciated. Recently, clear indications for a biological basis of microvascular thrombosis have been obtained. During systemic infections microvessel thrombosis can mediate an intravascular innate immune response (immunothrombosis). This biological form of thrombosis is based on the generation of fibrin inside blood vessels and is critically triggered by neutrophils and their interactions with platelets which result in the release of neutrophil extracellular traps (extracellular nucleosomes). Immunothrombosis is critically supported by neutrophil elastase and the activator molecules of blood coagulation tissue factor and factor XII. Identification of the biological driving forces of microvascular thrombosis should help to elucidate the mechanisms promoting pathological vessel occlusions in both microvessels and large vessels.


Asunto(s)
Microvasos/inmunología , Microvasos/patología , Trombosis/inmunología , Trombosis/patología , Animales , Coagulación Sanguínea , Fibrina/metabolismo , Humanos , Inmunidad Innata , Microvasos/microbiología , Trombosis/sangre , Trombosis/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA