Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Heart Vessels ; 37(11): 1962-1970, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35545686

ABSTRACT

Transforming growth factor-ß-induced protein (TGFBIp) is released from activated platelets and promotes pro-thrombotic complications like pulmonary embolism. The role of TGFBIp in acute coronary syndrome, especially with a focus on platelets, has not been investigated so far. Using ELISA and immunoblotting, we demonstrate platelet TGFBIp release in patients with myocardial infarction (MI). We investigated TGFBIp-induced platelet adhesion and rolling by flow chamber and chemotactic effects of TGFBIp in transwell experiments. Immunochemistry staining of arterial vessels detected TGFBIp and the platelet-specific protein GPVI in the vessel wall.We demonstrate for the first time that platelet TGFBIp release is significantly increased in MI and correlates with the severity of acute coronary syndromes (STEMI, NSTEMI). After activation with TRAP, platelets release TGFBIp and TGFBIp itself activates platelets. Under flow, TGFBIp-mediated platelet rolling and adherence similarly to collagen. TGFBIp significantly increased platelet transmigration and we demonstrate TGFBIp deposits in the wall of human arteries. In this study, we add novel aspects to the role of TGFBIp in acute coronary syndrome by demonstrating that TGFBIp is partially released from platelets during MI and has activating, pro-adhesive and pro-migratory effects on platelets that could contribute to the disease development of coronary vascular inflammation and MI.


Subject(s)
Acute Coronary Syndrome , Extracellular Matrix Proteins/blood , Myocardial Infarction , Transforming Growth Factor beta/blood , Blood Platelets/metabolism , Collagen/metabolism , Humans , Myocardial Infarction/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factors/metabolism
2.
Int J Mol Sci ; 21(21)2020 Oct 31.
Article in English | MEDLINE | ID: mdl-33142786

ABSTRACT

Platelets are exposed to extracellular matrix (ECM) proteins like collagen and laminin and to fibrinogen during acute vascular events. However, beyond hemostasis, platelets have the important capacity to migrate on ECM surfaces, but the translational response of platelets to different extracellular matrix stimuli is still not fully characterized. Using 2D-gel electrophoresis, confocal microscopy, polysome analysis and protein sequencing by mass spectrometry, we demonstrate that platelets show a differential expression profile of newly synthesized proteins on laminin, collagen or fibrinogen. In this context, we observed a characteristic, ECM-dependent translocation phenotype of translation initiation factor eIF4E to the ribosomal site. eIF4E accumulated in polysomes with increased binding of mRNA and co-localization with vinculin, leading to de novo synthesis of important cytoskeletal regulator proteins. As the first study, we included a proteome analysis of laminin-adherent platelets and interestingly identified upregulation of essentially important proteins that mediate cytoskeletal regulation and mobility in platelets, such as filamin A, talin, vinculin, gelsolin, coronin or kindlin-3. In summary, we demonstrate that platelet activation with extracellular matrix proteins results in a distinct stimulus-specific translational response of platelets that will help to improve our understanding of the regulation of platelet mobility and migration.


Subject(s)
Blood Platelets/physiology , Cytoskeletal Proteins/metabolism , Extracellular Matrix/metabolism , Platelet Activation , Protein Biosynthesis , Protein Processing, Post-Translational , Blood Platelets/cytology , Collagen/metabolism , Cytoskeleton/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Fibrinogen/metabolism , Hemostasis , Humans
3.
Int J Mol Sci ; 20(24)2019 Dec 06.
Article in English | MEDLINE | ID: mdl-31817594

ABSTRACT

Kindlins are important proteins for integrin signaling and regulation of the cytoskeleton, but we know little about their precise function and regulation in platelets during acute ischemic events. In this work, we investigated kindlin-3 protein levels in platelets isolated from patients with ST-elevation myocardial infarction (STEMI) compared to patients with non-ischemic chest pain. Platelets from twelve patients with STEMI and twelve patients with non-ischemic chest pain were isolated and analyzed for kindlin-3 protein levels and intracellular localization by immunoblotting and two-dimensional gel electrophoresis. Platelet proteome analysis by two-dimensional gel electrophoresis and protein sequencing identified kindlin-3 as a protein that is cleaved in platelets from patients with myocardial infarction. Kindlin-3 full-length protein was significantly decreased in patients with STEMI compared to patients with non-ischemic chest pain (1.0 ± 0.2 versus 0.28 ± 0.2, p < 0.05) by immunoblotting. Kindlin-3 showed a differential distribution and was primarily cleaved in the cytosolic and membrane compartment of platelets in myocardial infarction. Platelet activation with thrombin alone did not affect kindlin-3 protein levels. The present study demonstrates that kindlin-3 protein levels become significantly reduced in platelets of patients with myocardial infarction compared to controls. The results suggest that kindlin-3 cleavage in platelets is associated with the ischemic event of myocardial infarction.


Subject(s)
Blood Platelets/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , ST Elevation Myocardial Infarction/metabolism , Aged , Blood Platelets/drug effects , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunoblotting , Male , Middle Aged , Thrombin/pharmacology
4.
Int J Mol Sci ; 20(23)2019 Nov 27.
Article in English | MEDLINE | ID: mdl-31783528

ABSTRACT

Heat-shock proteins are a family of proteins which are upregulated in response to stress stimuli including inflammation, oxidative stress, or ischemia. Protective functions of heat-shock proteins have been studied in vascular disease models, and malfunction of heat-shock proteins is associated with vascular disease development. Heat-shock proteins however have not been investigated in human platelets during acute myocardial infarction ex vivo. Using two-dimensional electrophoresis and immunoblotting, we observed that heat-shock protein 27 (HSPB1) levels and phosphorylation are significantly increased in platelets of twelve patients with myocardial infarction compared to patients with nonischemic chest pain (6.4 ± 1.0-fold versus 1.0 ± 0.9-fold and 5.9 ± 1.8-fold versus 1.0 ± 0.8-fold; p < 0.05). HSP27 (HSPB1) showed a distinct and characteristic intracellular translocation from the cytoskeletal fraction into the membrane fraction of platelets during acute myocardial infarction that did not occur in the control group. In this study, we could demonstrate for the first time that HSP27 (HSPB1) is upregulated and phosphorylated in human platelets during myocardial infarction on a cellular level ex vivo with a characteristic intracellular translocation pattern. This HSP27 (HSPB1) phenotype in platelets could thus represent a measurable stress response in myocardial infarction and potentially other acute ischemic events.


Subject(s)
Blood Platelets/metabolism , Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Phosphorylation/genetics , ST Elevation Myocardial Infarction/genetics , Up-Regulation/genetics , Cytoskeleton/genetics , Female , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , Humans , Male , Middle Aged , Oxidative Stress/genetics , Transcriptional Activation/genetics
6.
Cell Physiol Biochem ; 30(1): 259-68, 2012.
Article in English | MEDLINE | ID: mdl-22759972

ABSTRACT

Recent observations pointed to the ability of platelets to migrate and thus to invade the inflamed vascular wall. Platelet migration could be stimulated by stromal cell-derived factor-1 (SDF-1), an effect dependent on phosphatidylinositide-3-kinase (PI3K) and paralleled by activation and phosphorylation of Wiskott-Aldrich syndrome protein (WASP). Migration is inhibited by vinculin, which is similarly regulated by phosphorylation. PI3K-sensitive kinases include the serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored whether SGK1 modifies WASP and vinculin phosphorylation in murine platelets and participates in the regulation of platelet migration. Platelets were isolated from gene-targeted mice lacking SGK1 (sgk1(-/-)) and from their wild type littermates (sgk1(+/+)). Platelet migration stimulated with SDF-1 was significantly less pronounced in sgk1(-/-)platelets than in sgk1(+/+) platelets. Moreover, SDF-1 significantly induced WASP phosphorylation, an effect again reduced in platelets lacking SGK1. Phosphorylation of vinculin was significantly enhanced in sgk1(-/-)platelets and was significantly reduced following treatment of platelets with Ca(2+) chelator BAPTA. Immunohistochemical analysis of in vivo experiments in intestinal vessels after vascular inflammation revealed that transmigration of platelets into inflamed vessel walls was significantly less pronounced in sgk1(-/-)than in sgk1(+/+) mice. In conclusion, SGK1 is a powerful regulator of platelet migration.


Subject(s)
Blood Platelets/physiology , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transendothelial and Transepithelial Migration , Animals , Cells, Cultured , Chelating Agents/pharmacology , Chemokine CXCL12/physiology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , Immediate-Early Proteins/genetics , Intestines/blood supply , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Vinculin/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism
7.
Platelets ; 23(1): 1-6, 2012.
Article in English | MEDLINE | ID: mdl-21671729

ABSTRACT

Vascular lesion formation and collagen presentation are key events leading to the development of vulnerable plaques. Glycoprotein VI (GPVI) significantly contributes to plaque-associated collagen binding and thrombus formation. The aim of this study was to image endothelial injury using fluorescence-labelled GPVI-Fc (Fc, fragment crystallized), a soluble form of GPVI that was generated by cloning and fusing GPVI to an Fc-domain, in an ex-vivo rabbit model. This study serves as a proof-of-principle study to demonstrate that GPVI-Fc is a useful tool for detecting endothelial damage. The carotid and femoral arteries and the aorta abdominalis were isolated from rabbits and perfused with phosphate buffered saline (PBS) to remove all blood, and a catheter was placed into the vessels in situ. Endothelial damage was achieved by pulling an inflated balloon approximately 1 inch through the vessels, while control vessels were not balloon-treated. After balloon deflation, the catheter was removed. Fluorescence-labelled GPVI-Fc (50 µg/mL) was injected into the injured and control intact vessels, and the opened vessels were sealed by clamps. After incubation, the vessels were rinsed with PBS, and optical imaging was performed to measure GPVI-Fc binding to injured endothelium. The optical data corresponding to the mean detected optical signal of the regions of interest were corrected by subtracting the mean data of the background fluorescence (arbitrary units). After denudation, fluorescence was enhanced in injured femoral and carotid arteries when compared to intact femoral (41.1 ± 17.5 vs. 14.6 ± 6.5; P = 0.021) and carotid (30.2 ± 7.6 vs. 7.9 ± 3.9; P = 0.005) arteries. This preclinical GPVI-Fc-based vascular lesion imaging approach may be the first step towards a method that allows identification of vascular lesions in vivo.


Subject(s)
Carotid Artery Injuries/pathology , Endothelium, Vascular/pathology , Femoral Artery/pathology , Fluorescein Angiography/methods , Immunoglobulin Fc Fragments/pharmacology , Platelet Membrane Glycoproteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Carotid Arteries/pathology , Endothelium, Vascular/injuries , Female , Femoral Artery/injuries , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
8.
Toxins (Basel) ; 3(2): 120-33, 2011 02.
Article in English | MEDLINE | ID: mdl-22069700

ABSTRACT

The frequency and severity of bacteremic infections has increased over the last decade and bacterial endovascular infections (i.e., sepsis or endocarditis) are associated with high morbidity and mortality. Bacteria or secreted bacterial products modulate platelet function and, as a result, affect platelet accumulation at sites of vascular infection and inflammation. However, whether bacterial products regulate synthetic events in platelets is not known. In the present study, we determined if prolonged contact with staphylococcal α-toxin signals platelets to synthesize B-cell lymphoma (Bcl-3), a protein that regulates clot retraction in murine and human platelets. We show that α-toxin induced α(IIb)ß(3)-dependent aggregation (EC(50) 2.98 µg/mL ± 0.64 µg/mL) and, over time, significantly altered platelet morphology and stimulated de novo accumulation of Bcl-3 protein in platelets. Adherence to collagen or fibrinogen also increased the expression of Bcl-3 protein by platelets. α-toxin altered Bcl-3 protein expression patterns in platelets adherent to collagen, but not fibrinogen. Pretreatment of platelets with inhibitors of protein synthesis or the mammalian Target of Rapamycin (mTOR) decreased Bcl-3 protein expression in α-toxin stimulated platelets. In conclusion, Staphylococcusaureus-derived α-toxin, a pore forming exotoxin, exerts immediate (i.e., aggregation) and prolonged (i.e., protein synthesis) responses in platelets, which may contribute to increased thrombotic events associated with gram-positive sepsis or endocarditis.


Subject(s)
Bacterial Toxins/pharmacology , Blood Platelets/drug effects , Hemolysin Proteins/pharmacology , Platelet Activation/drug effects , Proto-Oncogene Proteins/biosynthesis , Transcription Factors/biosynthesis , B-Cell Lymphoma 3 Protein , Blood Platelets/physiology , Cells, Cultured , Collagen/chemistry , Fibrinogen/chemistry , Humans , Staphylococcus aureus
9.
PLoS Pathog ; 7(11): e1002355, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22102811

ABSTRACT

Human ß-defensins (hBD) are antimicrobial peptides that curb microbial activity. Although hBD's are primarily expressed by epithelial cells, we show that human platelets express hBD-1 that has both predicted and novel antibacterial activities. We observed that activated platelets surround Staphylococcus aureus (S. aureus), forcing the pathogens into clusters that have a reduced growth rate compared to S. aureus alone. Given the microbicidal activity of ß-defensins, we determined whether hBD family members were present in platelets and found mRNA and protein for hBD-1. We also established that hBD-1 protein resided in extragranular cytoplasmic compartments of platelets. Consistent with this localization pattern, agonists that elicit granular secretion by platelets did not readily induce hBD-1 release. Nevertheless, platelets released hBD-1 when they were stimulated by α-toxin, a S. aureus product that permeabilizes target cells. Platelet-derived hBD-1 significantly impaired the growth of clinical strains of S. aureus. hBD-1 also induced robust neutrophil extracellular trap (NET) formation by target polymorphonuclear leukocytes (PMNs), which is a novel antimicrobial function of ß-defensins that was not previously identified. Taken together, these data demonstrate that hBD-1 is a previously-unrecognized component of platelets that displays classic antimicrobial activity and, in addition, signals PMNs to extrude DNA lattices that capture and kill bacteria.


Subject(s)
Bacterial Toxins/immunology , Blood Platelets/metabolism , Hemolysin Proteins/immunology , Neutrophils/immunology , Staphylococcus aureus/immunology , beta-Defensins/metabolism , Bacterial Toxins/metabolism , Blood Platelets/enzymology , Blood Platelets/immunology , Cell Line, Tumor , HeLa Cells , Hemolysin Proteins/metabolism , Humans , Neutrophils/metabolism , Platelet Activation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction , Staphylococcus aureus/growth & development , beta-Defensins/genetics
10.
Biochem Biophys Res Commun ; 415(1): 54-60, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22005466

ABSTRACT

Platelets have been shown to migrate and thus to invade the vascular wall. Platelet migration is stimulated by SDF-1. In other cell types, migration is dependent on Ca(2+) entry via Ca(2+) channels. Ca(2+) influx is sensitive to cell membrane potential which is maintained by K(+) channel activity and/or Cl(-) channel activity. The present study explored the role of ion channels in the regulation of SDF-1 induced migration. Platelets were isolated from human volunteers as well as from gene targeted mice lacking the Ca(2+) activated K(+) channel SK4 (sk4(-/-)) and their wild type littermates (sk4(+/+)). According to confocal microscopy human platelets expressed the Ca(2+) channel Orai1 and the Ca(2+)-activated K(+) channel K(Ca)3.1 (SK4). SDF-1 (100 ng/ml) stimulated migration in human platelets, an effect blunted by Orai1 inhibitors 2-aminoethoxydiphenyl borate 2-APB (10 µM) and SKF-96365 (10 µM), by unspecific K(+) channel inhibitor TEA (30 mM), by SK4 specific K(+) channel blocker clotrimazole (10 µM), but not by Cl(-) channel inhibitor 5-nitro-2-(3-phenylpropylamino) benzoic acid NPPB (100 µM). Significant stimulation of migration by SDF-1 was further observed in sk4(+/+) platelets but was virtually absent in sk4(-/-) platelets. In conclusion, platelet migration requires activity of the Ca(2+) channel Orai1 and of the Ca(2+) activated K(+) channel SK4, but not of NPPB-sensitive Cl(-) channels.


Subject(s)
Blood Platelets/physiology , Cell Movement , Chemokine CXCL12/metabolism , Ion Channels/metabolism , Animals , Blood Platelets/drug effects , Calcium Channels/metabolism , Chloride Channels/metabolism , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Mice , Nitrobenzoates/pharmacology , ORAI1 Protein
11.
Platelets ; 22(6): 415-21, 2011.
Article in English | MEDLINE | ID: mdl-21413844

ABSTRACT

Shear forces are generated in all parts of the vascular system and contribute directly and indirectly to vascular disease progression. Endothelial cells are able to adapt to flow conditions, and are known to polarize and migrate in response to shear forces. Platelets exposed to shear stress are activated and release bioactive molecules from their alpha granules. So far, platelets have been considered to be static cells that do not leave the site of tight adhesion. However, we have recently been able to demonstrate the capacity of platelets to migrate in response to stromal derived factor-1 (SDF-1). In this project, we have demonstrated that platelets accumulate in areas with a high concentration of SDF-1 under flow conditions and respond to high shear stress by cellular polarization, cytoskeletal reorganisation, and flow-directed migration. In this context, we have shown increased Wiskott-Aldrich Syndrome protein (WASP) phosphorylation and intracellular redistribution of focal adhesion kinase (FAK) under high-shear stress conditions. The effect of flow-induced platelet migration has not previously been recognized and offers a new role for platelets as mobile cells. Their migratory potential may enable platelets to cover intimal lesions and contribute to vascular repair.


Subject(s)
Blood Platelets , Cell Movement , Chemokine CXCL12/pharmacology , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Cell Polarity , Chemokine CXCL12/metabolism , Endothelium, Vascular/metabolism , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Hemodynamics/drug effects , Humans , Microscopy, Confocal , Phosphorylation/drug effects , Stress, Mechanical , Wiskott-Aldrich Syndrome Protein/metabolism
12.
Int J Cardiol ; 149(2): 164-168, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-20071043

ABSTRACT

BACKGROUND: The purpose of the study was to test whether an elevated surface expression of platelet collagen receptor glycoprotein VI (GPVI) is an appropriate marker for the diagnosis of the acute coronary syndrome (ACS), especially when the electrocardiogram (ECG) is ambiguous. METHODS: Between 2007 to 2008, we consecutively evaluated 378 patients with ACS and ambiguous ECG on hospital admission. In all patients, GPVI surface expression was determined by flow cytometry. In addition, the myocardial necrosis markers troponin-I (Tn-I) and creatine kinase-MB (CKMB) were measured. RESULTS: We found that in patients with ACS and unclear ECG in whom GPVI levels (mean fluorescence intensity (MFI) ≥ 18.6) were elevated, the relative risk for ACS was 2.6-fold enhanced. Binary logistic regression analysis revealed that an elevated platelet GPVI level is indicating an ACS independent of biomarkers of myocardial necrosis including Tn-I, creatine kinase (CK), CKMB (GPVI: p=0.011; Tn-I: p=0.180; CKMB: p=0.250; CK: p=0.127). Patients with evident T-wave inversion and/or ST-depression showed a strong association between ACS and GPVI expression. CONCLUSIONS: Platelet GPVI surface expression is enhanced in patients with ACS with unclear ECG findings and is strongly associated with myocardial ischemia. Additional to the classical markers of myocardial necrosis Tn-I and CK, GPVI is an early biomarker for the diagnosis of ACS, especially when the ECG is ambiguous.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/physiopathology , Electrocardiography , Platelet Membrane Glycoproteins/physiology , Acute Coronary Syndrome/diagnosis , Aged , Biomarkers/blood , Electrocardiography/methods , Female , Humans , Male , Middle Aged , Myocardial Ischemia/blood , Myocardial Ischemia/diagnosis , Myocardial Ischemia/physiopathology , Survival Rate/trends
13.
Clin Appl Thromb Hemost ; 17(4): 340-5, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20460343

ABSTRACT

Thoracic mobile aortic mural thrombus (TAMT) of the aortic arch is a rare condition. We report 3 cases of symptomatic TAMT treated with systemic alteplase (tissue plasminogen activator [t-PA]) thrombolysis. The first patient was symptomatic with repetitive thromboembolism to the left brachial artery. She was treated with repetitive thrombolysis after surgical embolectomy of the brachial artery. The second patient was symptomatic with splenic infarction and mesenteric ischemia. She was treated with a single cycle of systemic thrombolysis followed by ileocoecal resection. The third patient presented with a TAMT obstructing the left common carotid artery, causing ischemic stroke. After systemic thrombolysis, a reduction in thrombus size was documented; however, the patient died later, of acute heart failure, during the clinical course. On follow-up 6 months after the incidences, the 2 surviving patients were in good condition and free of thromboembolic events. We show that systemic thrombolytic therapy can be performed successfully in patients with TAMT.


Subject(s)
Aorta, Thoracic/pathology , Thrombolytic Therapy/methods , Thrombosis/drug therapy , Aged, 80 and over , Aorta, Thoracic/diagnostic imaging , Echocardiography , Female , Humans , Middle Aged , Thrombosis/diagnostic imaging , Thrombosis/pathology , Tomography, X-Ray Computed , Treatment Outcome
14.
J Mol Med (Berl) ; 88(12): 1277-88, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20852838

ABSTRACT

Platelets have been regarded as static cells that do not move once they adhere to a matrix. The present study explored, whether platelets are able to migrate. In contrast to the current opinion, we found that platelets were mobile, able to migrate over a surface, and transmigrate through a transwell membrane and endothelium toward a source of stromal cell-derived factor 1 (SDF-1). Platelet migration was stimulated by SDF-1, which led to the downstream activation and phosphorylation of Wiskott-Aldrich syndrome protein. SDF-1 signaling and subsequent platelet migration could be inhibited by CXCR4-receptor blocker AMD3100, pertussis toxin, inhibition of phosphoinositol 3-kinase (PI3 kinase) with LY294002 or wortmannin, and disruption of actin polymerization with cytochalasin B. The potential of platelets to migrate in an SDF-1-mediated fashion may redefine the role of platelets in the pathophysiology of vascular inflammation, subsequent atherosclerotic degeneration, and vascular regeneration.


Subject(s)
Blood Platelets/cytology , Blood Platelets/enzymology , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Blood Platelets/drug effects , Blood Vessels/drug effects , Blood Vessels/pathology , Chemotaxis/drug effects , Collagen/pharmacology , Endothelium/drug effects , Endothelium/metabolism , Fibrinogen/pharmacology , GTP-Binding Proteins/metabolism , Humans , Ischemia/pathology , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Porosity/drug effects , Receptors, CXCR4/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism
15.
J Mol Med (Berl) ; 88(9): 909-20, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20454888

ABSTRACT

The appearance of lipid-rich foam cells is a major feature of vulnerable atherosclerotic plaque formation. The transformation of macrophages into foam cells results from excessive uptake of cholesterol-rich particles by scavenger receptors such as CD68. We cloned a CD68-Fc immunoadhesin, a fusion protein consisting of the extracellular domain of the human CD68 and a human Fc domain, and investigated the function in vitro. Specific binding of CD68-Fc to OxLDL with an affinity of 10 nmol/L was determined by surface plasmon resonance and increased binding to lipid-rich human and ApoE(-/-) mice plaque tissue. This was confirmed both by immunohistochemical staining of CD68-Fc-treated paraffin sections from human plaques and by ELISA-based quantification of CD68-Fc binding to human atherosclerotic plaque extracts. In an in vitro model of macrophage/foam cell formation, CD68-Fc reduced foam cell formation significantly. This was caused both by interference of CD68-Fc with OxLDL uptake into macrophages and platelets and by the inhibition of platelet/OxLDL phagocytosis. Finally, expression of metalloproteinases by macrophages/foam cells was inhibited by CD68-Fc. In conclusion, CD68-Fc seems to be a promising new tool for preventing macrophage/foam cell formation. Thus, CD68-Fc might offer a novel therapeutic strategy for patients with acute coronary syndrome by modulating the generation of vulnerable plaques.


Subject(s)
Antigens, CD/chemistry , Antigens, Differentiation, Myelomonocytic/chemistry , Foam Cells/cytology , Immunoglobulin Fc Fragments/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Apolipoproteins E/metabolism , CHO Cells , Cricetinae , Cricetulus , Foam Cells/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice , Plaque, Atherosclerotic/metabolism , Receptors, Scavenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Surface Plasmon Resonance , Transfection
16.
Arterioscler Thromb Vasc Biol ; 30(6): 1127-36, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20378847

ABSTRACT

OBJECTIVE: To investigate the role of junctional adhesion molecule A (JAM-A) on adhesion and differentiation of human CD34(+) cells into endothelial progenitor cells. METHODS AND RESULTS: Tissue healing and vascular regeneration is a multistep process requiring firm adhesion of circulating progenitor cells to the vascular wall and their further differentiation into endothelial cells. The role of JAM-A in platelet-mediated adhesion of progenitor cells was investigated by adhesion assays in vitro and with the help of intravital fluorescence microscopy in mice. Preincubation of human CD34(+) progenitor cells with soluble JAM-A-Fc (sJAM-A-Fc) resulted in significantly decreased adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Human CD34(+) cells express JAM-A, as defined by flow cytometry and Western blot analysis. JAM-A mediates differentiation of CD34(+) cells to endothelial progenitor cells and facilitates CD34(+) cell-induced reendothelialization in vitro. Pretreatment of human CD34(+) cells with sJAM-A-Fc resulted in increased neointima formation 3 weeks after endothelial denudation in the carotid arteries of nonobese diabetic/severe combined immunodeficient mice. CONCLUSIONS: These results indicate that the expression of JAM-A on CD34(+) cells mediates adhesion to the vascular wall after injury and differentiation into endothelial progenitor cells, a mechanism potentially involved in vascular regeneration. Human CD34(+) cells express JAM-A, mediating their interaction with platelets and endothelial cells. Specifically, JAM-A expressed on human CD34(+) progenitor cells regulates their adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Moreover, it mediates differentiation of CD34(+) cells to endothelial progenitor cells and facilitates reendothelialization.


Subject(s)
Antigens, CD34/analysis , Carotid Artery Injuries/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion , Cell Differentiation , Endothelial Cells/metabolism , Immunoglobulins/metabolism , Intestines/blood supply , Reperfusion Injury/metabolism , Stem Cells/metabolism , Animals , Blood Platelets/metabolism , Blotting, Western , CHO Cells , Carotid Artery Injuries/blood , Carotid Artery Injuries/immunology , Carotid Artery Injuries/pathology , Carotid Artery Injuries/physiopathology , Cell Adhesion Molecules/genetics , Cell Proliferation , Cricetinae , Cricetulus , Endothelial Cells/immunology , Endothelial Cells/transplantation , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/metabolism , Immunoglobulins/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence , Microscopy, Video , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Receptors, Cell Surface , Recombinant Fusion Proteins/metabolism , Reperfusion Injury/blood , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Stem Cell Transplantation , Stem Cells/immunology , Time Factors , Transfection , Wound Healing
17.
Semin Thromb Hemost ; 36(2): 146-56, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20414829

ABSTRACT

Platelets are involved in the initiation of atherosclerosis by adherence to inflamed endothelium. Monocytes bind to these platelets and transmigrate into the vessel wall, transforming into macrophages and foam cells. We have previously shown that lipid-laden platelets are phagocytosed by macrophages. In this study we investigated the functional consequences of oxidized low-density lipoprotein (oxLDL) uptake on platelet function and interaction with the endothelium. Human platelets were isolated from healthy donors and activated by adenosine diphosphate. Immunofluorescence microscopy and flow cytometry revealed that oxLDL is located intracellularly in vesicles. With mepacrine costaining and confocal microtomography, we were able to identify dense granules as the vesicles that contain oxLDL. OxLDL-laden platelets induced intercellular adhesion molecule 1 expression in endothelial cells more than exogenous native LDL, oxLDL, and oxLDL-negative platelets. Furthermore, oxLDL-laden platelets induced foam cell development from CD34(+) progenitor cells. On endothelial regeneration, oxLDL-laden platelets had the opposite effect: The number of CD34(+) progenitor cells (colony-forming units) able to transform into endothelial cells was significantly reduced in the presence of oxLDL-platelets, whereas native LDL had no effect. Our results demonstrate that activated platelets internalize oxLDL and that oxLDL-laden platelets activate endothelium, inhibit endothelial regeneration, and promote foam cell development. Platelet oxLDL contributes significantly to vascular inflammation and is able to promote atherosclerosis.


Subject(s)
Blood Platelets/physiology , Endothelium, Vascular/pathology , Inflammation/etiology , Lipoproteins, LDL/physiology , Blood Platelets/pathology , Endocytosis , Endothelial Cells/pathology , Foam Cells/cytology , Hematopoietic Stem Cells/cytology , Humans , Platelet Activation , Regeneration
18.
Semin Thromb Hemost ; 36(2): 203-11, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20414836

ABSTRACT

Platelets respond immediately to vascular injury by adhesion, aggregation, and thrombus formation. Disruption of the endothelial cell layer exposes extracellular matrix to the bloodstream. Collagen binding to platelet glycoprotein VI (GPVI) mediates the initial adhesion of the rolling platelet to the vascular wound. Signaling by GPVI leads to the onset of the platelet activation cascade that is finally crowned by a firm and shear-resistant integrin-based adhesive clot. Blockade of collagen binding to GPVI would prevent initial adhesion and further activation of the platelet and would have an enormous impact in antithrombotic therapy. Besides the therapeutical implication, radiolabeled GPVI gives us a valuable diagnostic means that may be used clinically for plaque imaging in the near future.


Subject(s)
Platelet Membrane Glycoproteins/analysis , Platelet Membrane Glycoproteins/metabolism , Atherosclerosis/diagnosis , Atherosclerosis/drug therapy , Collagen/metabolism , Fibrinolytic Agents/pharmacology , Humans , Platelet Activation/drug effects , Protein Binding/drug effects , Solubility
19.
Clin Res Cardiol ; 99(4): 227-33, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20049463

ABSTRACT

BACKGROUND: Platelets play a critical role in arterial thrombosis, acute coronary syndrome (ACS) and stroke. Platelet collagen receptor glycoprotein VI (GPVI) is associated with acute coronary events and a poor clinical outcome. METHODS: Between January 2006 and March 2009, we evaluated 2,213 consecutive patients in a prospective study design, who presented with chest pain. Among 1,819 patients (82.2%) who underwent coronary angiography, 1,652 patients (90.8%) showed a coronary and 167 patients (9.2%) a non-coronary origin of chest pain. 901 patients (54.5%) presented with an ACS, 751 (45.5%) with a stable angina pectoris. Clinical outcome was performed by a predefined structured telephone interview after a 3-month follow-up (92.8%:1,533 of 1,652 patients). RESULTS: Kaplan-Meier analysis for cumulative event-free survival revealed that patients with a elevated baseline GPVI expression had a poorer clinical outcome for cardiovascular death than patients with decreased GPVI levels (log rank; P = 0.040). These results were paralleled in composite cumulative survival that included myocardial infarction, stroke, and cardiovascular death (log rank; P = 0.002). Relative risk of cardiovascular death was found at 1.41 (95% CI 1.08-1.85) for platelet GPVI, 1.64 (95% CI 1.31-2.05) for brain natriuretic peptide, 1.16 (95% CI 0.81-1.64) for troponin-I and 1.33 (95% CI 0.97-1.82) for C-reactive protein. CONCLUSIONS: Patients with CAD presenting increased levels of platelet GPVI had a poorer outcome and may profit from a dual antiplatelet therapy. Thus, GPVI may be a useful prognostic tool for adverse cardiovascular events. Future studies should substantiate GPVI for its potential role of risk prediction and consider its prognostic value to improve risk stratification.


Subject(s)
Coronary Artery Disease/physiopathology , Platelet Membrane Glycoproteins/metabolism , Aged , Aged, 80 and over , Biomarkers/metabolism , Chest Pain/etiology , Coronary Angiography , Coronary Artery Disease/mortality , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prospective Studies
20.
Atherosclerosis ; 209(1): 51-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19758589

ABSTRACT

UNLABELLED: Upon coincubation with platelets, CD34(+) progenitor cells have the potential to differentiate into foam cells, and thereby may promote the progression of atherosclerosis. The exact mechanism of MMP-regulation during the cellular differentiation process to foam cells is still unclear. Thus, we investigated the role of EMMPRIN (CD147) and its ligand cyclophilin A (CyPA) during foam cell formation originating from both monocytes/macrophages and CD34(+) progenitor cells. METHODS AND RESULTS: Differentiation of CD34(+) progenitor to foam cells was analyzed in a coculture model of progenitor cells and platelets. While CD34(+) cells did not express EMMPRIN or MT1-MMP, mature foam cells strongly expressed EMMPRIN, which was associated with MT1-MMP expression as well as MMP-9. Gene silencing of EMMPRIN by siRNA during the cell differentiation process hindered not only the upregulation of MMPs (MT1-MMP, MMP-9), but also the secretion of the cytokine M-CSF. During the differentiation process CyPA was substantially released into the supernatant. The presence of the CyPA inhibitor NIM811 significantly reduced MMP-9 secretion during the differentiation process. Similar results were obtained using the classical pathway of foam cell formation by coincubating human macrophages with AcLDL. Additionally, the presence of soluble EMMPRIN ligands (CyPA, recombinant EMMPRIN) further enhanced MMP-9 secretion by mature foam cells. Consistently, CyPA and EMMPRIN were found in atherosclerotic plaques of ApoE-deficient mice by immunohistochemistry. CONCLUSION: EMMPRIN is upregulated during the differentiation process from CD34(+) progenitor cells to foam cells, whereas its ligand, CyPA, is released. The CyPA/EMMPRIN activation pathway may play a relevant role in promoting the vulnerability of atherosclerotic plaques.


Subject(s)
Atherosclerosis/pathology , Basigin/metabolism , Cyclophilin A/metabolism , Foam Cells/pathology , Macrophage Colony-Stimulating Factor/metabolism , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Antigens, CD34/metabolism , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Cell Differentiation , Cell Movement , Collagen , Cyclophilin A/antagonists & inhibitors , Cyclosporine/pharmacology , Drug Combinations , Foam Cells/metabolism , Humans , Laminin , Ligands , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Mutant Strains , Proteoglycans , Stem Cells/metabolism , Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...