Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 958
Filter
1.
J Thromb Haemost ; 20(11): 2617-2631, 2022 11.
Article in English | MEDLINE | ID: mdl-35894121

ABSTRACT

BACKGROUND: The collagen receptor glycoprotein VI (GPVI) is an attractive antiplatelet target due to its critical role in thrombosis but minor involvement in hemostasis. OBJECTIVE: To investigate GPVI receptor involvement in platelet activation by collagen-I and atherosclerotic plaque using novel blocking and non-blocking anti-GPVI nanobodies (Nbs). METHODS: Nb effects on GPVI-mediated signaling and function were assessed by western blot and whole blood thrombus formation under flow. GPVI clustering was visualized in thrombi using fluorescently labeled Nb28. RESULTS: Under arterial shear, inhibitory Nb2 blocks thrombus formation and platelet activation on collagen and plaque, but only reduces adhesion on plaque. In contrast, adhesion on collagen, but not plaque, is decreased by blocking integrin α2ß1. Adhesion on plaque is maintained despite inhibition of integrins αvß3, α5ß1, α6ß1, and αIIbß3. Only combined αIIbß3 and α2ß1 blockade inhibits adhesion and thrombus formation to the same extent as Nb2 alone. Nb2 prevents GPVI signaling, with loss of Syk, Lat, and PLCÉ£2 phosphorylation, especially to plaque stimulation. Non-blocking fluorescently labeled Nb28 reveals distinct GPVI distribution patterns on collagen and plaque, with GPVI clustering clearly apparent on collagen fibers and less frequent on plaque. Clustering on collagen fibers is lost in the presence of Nb2. CONCLUSIONS: This work emphasizes the critical difference in GPVI-mediated platelet activation by plaque and collagen; it highlights the importance of GPVI clustering for downstream signaling and thrombus formation. Labeled Nb28 is a novel tool for providing mechanistic insight into this process and the data suggest Nb2 warrants further investigation as a potential anti-thrombotic agent.


Subject(s)
Plaque, Atherosclerotic , Single-Domain Antibodies , Thrombosis , Humans , Platelet Membrane Glycoproteins/physiology , Phospholipase C gamma , Integrin alpha2beta1 , Single-Domain Antibodies/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex , Collagen/pharmacology , Cluster Analysis , Blood Platelets , Platelet Aggregation
2.
Cells ; 10(9)2021 09 07.
Article in English | MEDLINE | ID: mdl-34571986

ABSTRACT

The platelet-activating factor receptor (PAFR) and its ligand (PAF) are important inflammatory mediators that are overexpressed in ovarian cancer. The receptor is an important player in ovarian cancer development. In this study, we aimed to evaluate the prognostic value of PAFR in epithelial ovarian cancer (EOC) and the potential use of its antagonist, rupatadine, as an experimental treatment. Tissue microarrays of ovarian cancer patients, most markedly those with a non-mucinous subtype, immunohistochemically overexpressed PAFR. Elevated cytoplasmic PAFR expression was found to significantly and independently impair patients' overall and recurrence-free survival (OS: median 83.48 vs. 155.03 months; p = 0.022; RFS: median 164.46 vs. 78.03 months; p = 0.015). In vitro, the serous ovarian cancer subtypes especially displayed an elevated PAFR gene and protein expression. siRNA knockdown of PAFR decreased cell proliferation significantly, thus confirming the receptor's protumorigenic effect on ovarian cancer cells. The clinically approved PAFR antagonist rupatadine effectively inhibited in vitro cell proliferation and migration of ovarian cancer cells. PAFR is a prognostic marker in ovarian cancer patients and its inhibition through rupatadine may have important therapeutic implications in the therapy of ovarian cancer patients.


Subject(s)
Cyproheptadine/analogs & derivatives , Ovarian Neoplasms/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Aged , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyproheptadine/metabolism , Cyproheptadine/pharmacology , ErbB Receptors/metabolism , Female , Humans , Middle Aged , Ovarian Neoplasms/genetics , Ovary/metabolism , Ovary/pathology , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/physiology , Prognosis , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/physiology , Signal Transduction/drug effects , Treatment Outcome
3.
Hepatology ; 74(6): 3345-3361, 2021 12.
Article in English | MEDLINE | ID: mdl-34320243

ABSTRACT

BACKGROUND AND AIMS: Liver regeneration after extreme hepatocyte loss occurs through transdifferentiation of biliary epithelial cells (BECs), which includes dedifferentiation of BECs into bipotential progenitor cells (BPPCs) and subsequent redifferentiation into nascent hepatocytes and BECs. Although multiple molecules and signaling pathways have been implicated to play roles in the BEC-mediated liver regeneration, mechanisms underlying the dedifferentiation-redifferentiation transition and the early phase of BPPC redifferentiation that is pivotal for both hepatocyte and BEC directions remain largely unknown. APPROACH AND RESULTS: The zebrafish extreme liver damage model, genetic mutation, pharmacological inhibition, transgenic lines, whole-mount and fluorescent in situ hybridizations and antibody staining, single-cell RNA sequencing, quantitative real-time PCR, and heat shock-inducible overexpression were used to investigate roles and mechanisms of farnesoid X receptor (FXR; encoded by nuclear receptor subfamily 1, group H, member 4 [nr1h4]) in regulating BPPC redifferentiation. The nr1h4 expression was significantly up-regulated in response to extreme liver injury. Genetic mutation or pharmacological inhibition of FXR was ineffective to BEC-to-BPPC dedifferentiation but blocked the redifferentiation of BPPCs to both hepatocytes and BECs, leading to accumulation of undifferentiated or less-differentiated BPPCs. Mechanistically, induced overexpression of extracellular signal-related kinase (ERK) 1 (encoded by mitogen-activated protein kinase 3) rescued the defective BPPC-to-hepatocyte redifferentiation in the nr1h4 mutant, and ERK1 itself was necessary for the BPPC-to-hepatocyte redifferentiation. The Notch activities in the regenerating liver of nr1h4 mutant attenuated, and induced Notch activation rescued the defective BPPC-to-BEC redifferentiation in the nr1h4 mutant. CONCLUSIONS: FXR regulates BPPC-to-hepatocyte and BPPC-to-BEC redifferentiations through ERK1 and Notch, respectively. Given recent applications of FXR agonists in the clinical trials for liver diseases, this study proposes potential underpinning mechanisms by characterizing roles of FXR in the stimulation of dedifferentiation-redifferentiation transition and BPPC redifferentiation.


Subject(s)
Liver Regeneration , Platelet Membrane Glycoproteins/physiology , Stem Cells/physiology , Animals , Biliary Tract/cytology , Cell Differentiation , Liver Regeneration/physiology , Real-Time Polymerase Chain Reaction , Zebrafish
5.
Thromb Haemost ; 120(11): 1492-1504, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32772352

ABSTRACT

The pathogenesis of atherosclerotic vascular disease is driven by a multitude of risk factors intertwining metabolic and inflammatory pathways. Increasing knowledge about platelet biology sheds light on how platelets take part in these processes from early to later stages of plaque development. Recent insights from experimental studies and mouse models substantiate platelets as initiators and amplifiers in atherogenic leukocyte recruitment. These studies are complemented by results from genetics studies shedding light on novel molecular mechanisms which provide an interesting prospect as novel targets. For instance, experimental studies provide further details how platelet-decorated von Willebrand factor tethered to activated endothelial cells plays a role in atherogenic monocyte recruitment. Novel aspects of platelets as atherogenic inductors of neutrophil extracellular traps and particularities in signaling pathways such as cyclic guanosine monophosphate and the inhibitory adaptor molecule SHB23/LNK associating platelets with atherogenesis are shared. In summary, it was our intention to balance insights from recent experimental data that support a plausible role for platelets in atherogenesis against a paucity of clinical evidence needed to validate this concept in humans.


Subject(s)
Atherosclerosis/drug therapy , Blood Platelets/physiology , Animals , Blood Platelets/drug effects , Chemotaxis, Leukocyte , Coronary Disease/blood , Coronary Disease/genetics , Drug Evaluation, Preclinical , Endothelial Cells/pathology , Extracellular Traps/physiology , Genetic Predisposition to Disease , Lipids/blood , Lipids/physiology , Mice , Nitric Oxide/physiology , P-Selectin/physiology , Plaque, Atherosclerotic/metabolism , Platelet Adhesiveness , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/physiology , Risk , von Willebrand Factor/physiology
6.
Thromb Haemost ; 120(6): 977-993, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32492725

ABSTRACT

Glycoprotein VI (GPVI), a platelet collagen receptor, is crucial in mediating atherothrombosis. Besides collagen, injured plaques expose tissue factor (TF) that triggers fibrin formation. Previous studies reported that GPVI also is a platelet receptor for fibrinogen and fibrin. We studied the effect of anti-GPVI antibodies and inhibitors of GPVI signaling kinases (Syk and Btk) on platelet adhesion and aggregate formation onto immobilized fibrinogen and different types of fibrin under arterial flow conditions. Fibrin was prepared from isolated fibrinogen ("pure fibrin"), recombinant fibrinogen ("recombinant fibrin"), or generated more physiologically from endogenous fibrinogen in plasma ("plasma fibrin") or by exposing TF-coated surfaces to flowing blood ("blood fibrin"). Inhibition of GPVI and Syk did not inhibit platelet adhesion and aggregate formation onto fibrinogen. In contrast anti-GPVI antibodies, inhibitors of Syk and Btk and the anti-GPIb antibody 6B4 inhibited platelet aggregate formation onto pure and recombinant fibrin. However, inhibition of GPVI and GPVI signaling did not significantly reduce platelet coverage of plasma fibrin and blood fibrin. Plasma fibrin contained many proteins incorporated during clot formation. Advanced optical imaging revealed plasma fibrin as a spongiform cushion with thicker, knotty, and long fibers and little activation of adhering platelets. Albumin intercalated in plasma fibrin fibers left only little space for platelet attachment. Pure fibrin was different showing a dense mesh of thin fibers with strongly activated platelets. We conclude that fibrin formed in plasma and blood contains plasma proteins shielding GPVI-activating epitopes. Our findings do not support a role of GPVI for platelet activation by physiologic fibrin.


Subject(s)
Blood Platelets/metabolism , Fibrin/metabolism , Platelet Membrane Glycoproteins/physiology , Receptors, Peptide/metabolism , Agammaglobulinaemia Tyrosine Kinase/blood , Agammaglobulinaemia Tyrosine Kinase/physiology , Enzyme Activation , Fibrinogen/metabolism , Hemorheology , Humans , Microscopy, Confocal/methods , Plasma , Platelet Adhesiveness , Platelet Aggregation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/immunology , Protein Binding , Recombinant Proteins/metabolism , Syk Kinase/antagonists & inhibitors , Syk Kinase/blood , Syk Kinase/physiology , Thromboplastin/metabolism
7.
J Thromb Haemost ; 17(9): 1500-1510, 2019 09.
Article in English | MEDLINE | ID: mdl-31145836

ABSTRACT

OBJECTIVE: Clinical and laboratory studies have demonstrated that platelets become hyperactive and prothrombotic in conditions of inflammation. We have previously shown that the proinflammatory cytokine interleukin (IL)-6 forms a complex with soluble IL-6 receptor α (sIL-6Rα) to prime platelets for activation by subthreshold concentrations of collagen. Upon being stimulated with collagen, the transcription factor signal transducer and activator of transcription (STAT) 3 in platelets is phosphorylated and dimerized to act as a protein scaffold to facilitate the catalytic action between the kinase Syk and the substrate phospholipase Cγ2 (PLCγ2) in collagen-induced signaling. However, it remains unknown how collagen induces phosphorylation and dimerization of STAT3. METHODS AND RESULTS: We conducted complementary in vitro experiments to show that the IL-6 receptor subunit glycoprotein 130 (GP130) was in physical proximity to the collagen receptor glycoprotein VI (GPVI in membrane lipid rafts of platelets. This proximity allows collagen to induce STAT3 activation and dimerization, and the IL-6-sIL-6Rα complex to activate the kinase Syk and the substrate PLCγ2 in the GPVI signal pathway, resulting in an enhanced platelet response to collagen. Disrupting lipid rafts or blocking GP130-Janus tyrosine kinase (JAK)-STAT3 signaling abolished the cross-activation and reduced platelet reactivity to collagen. CONCLUSION: These results demonstrate cross-talk between collagen and IL-6 signal pathways. This cross-talk could potentially provide a novel mechanism for inflammation-induced platelet hyperactivity, so the IL-6-GP130-JAK-STAT3 pathway has been identified as a potential target to block this hyperactivity.


Subject(s)
Blood Platelets/metabolism , Cytokine Receptor gp130/blood , Membrane Microdomains/physiology , Platelet Membrane Glycoproteins/physiology , Blood Coagulation/drug effects , Collagen/pharmacology , Cytokine Receptor gp130/chemistry , Hemorheology , Humans , Immunoprecipitation , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/blood , Phospholipase C gamma/blood , Phosphorylation , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/chemistry , Protein Interaction Mapping , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , STAT3 Transcription Factor/blood
8.
BMC Cancer ; 18(1): 999, 2018 Oct 19.
Article in English | MEDLINE | ID: mdl-30340558

ABSTRACT

BACKGROUND: The tumour microenvironment conferred by mesenchymal stem cells (MSCs) plays a key role in tumour development and progression. We previously determined that platelet-activating factor receptor (PAFR) was overexpressed in ovarian cancer cells (OCCs) and that PAF can promote ovarian cancer progression via PAF/PAFR-mediated inflammatory signalling pathways. Evidence suggests that MSCs can secrete high concentrations of PAF. Here, we investigated the role of PAF/PAFR signalling in the microenvironment mediated by MSCs and OCCs and its effect on cancer progression. METHODS: The PAF concentrations in the culture media of MSCs, OCCs and co-cultured MSCs and OCCs were determined by ELISA. The effects of MSCs on OCCs in vitro were assessed on cells treated with conditioned medium (CM). The expression and phosphorylation of key proteins in the PAF/PAFR signalling pathway were evaluated. In vivo, MSCs/RFP and SKOV3 cells were co-administered at different proportions to nude mice by interscapular injection. Mice in the WEB2086 group were intraperitoneally injected with the PAFR antagonist WEB2086 at a dose of 1 mg/kg.d for the duration of the animal experiments. Tumour progression was observed, and the weight and survival time of mice were measured. The PAF concentration in peripheral and tumour site blood was determined by ELISA. RESULTS: High concentrations of PAF were detected in CM from MSCs and MSCs co-cultured with OCCs. Both types of medium promoted non-mucinous OCC proliferation and migration but had no effect on mucinous-type OCCs. These effects could be blocked by PAFR inhibitors. The expression and phosphorylation of key proteins in the PAF/PAFR pathway significantly increased upon treatment with PAF and MSC-CM. In vivo, the tumour volume was larger following co-injection of SKOV3 cells and MSCs/RFP than following injection of SKOV3 cells alone. The tumour-promoting effect of MSCs/RFP was blocked by the PAFR antagonist WEB2086. Serum PAF concentrations significantly increased in co-injected mice. CONCLUSION: Our results suggest that the tumour-promoting effect of MSCs on OCCs via their cross-talk in the tumour microenvironment was, at least in part, mediated by the PAF/PAFR pathway, suggesting a new target for the treatment of ovarian cancer.


Subject(s)
Disease Progression , Mesenchymal Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Platelet Activating Factor/physiology , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/physiology , Tumor Microenvironment/drug effects , Animals , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Coculture Techniques , Female , Humans , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , Platelet Activating Factor/antagonists & inhibitors , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Triazoles/pharmacology , Tumor Microenvironment/physiology
9.
Circ Res ; 122(3): 506-522, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29420211

ABSTRACT

Platelets play a vital role in normal hemostasis to stem blood loss at sites of vascular injury by tethering and adhering to sites of injury, recruiting other platelets and blood cells to the developing clot, releasing vasoactive small molecules and proteins, and assembling and activating plasma coagulation proteins in a tightly regulated temporal and spatial manner. In synchrony with specific end products of coagulation, primarily cross-linked fibrin, a stable thrombus quickly forms. Far beyond physiological hemostasis and pathological thrombosis, emerging evidence supports platelets playing a pivotal role in vascular homeostasis, inflammation, cellular repair, regeneration, and wide range of autocrine and paracrine functions. In essence, platelets play both structural and functional roles as reporters, messengers, and active transporters surveying the vasculature for cues of environmental or developmental stimuli and participating as first responders.1 In this review, we will provide a contemporary perspective of platelet physiology, including fundamental, translational, and clinical constructs that apply directly to human health and disease.


Subject(s)
Blood Platelets/physiology , Hemostasis/physiology , Animals , Atherosclerosis/blood , Atherosclerosis/physiopathology , Blood Proteins/physiology , Cell-Derived Microparticles/physiology , Drug Delivery Systems , Drug Development , Endothelium, Vascular/physiology , Extracellular Matrix Proteins/physiology , Extracellular Traps/physiology , Host-Pathogen Interactions , Humans , Infections/blood , Infections/physiopathology , Inflammation/blood , Inflammation/physiopathology , Lymphangiogenesis , MicroRNAs/blood , Neovascularization, Physiologic , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/physiology , Platelet-Rich Plasma , Regeneration/physiology , Thromboembolism/blood , Thromboembolism/drug therapy , Thromboembolism/physiopathology , Thrombopoiesis
10.
Blood ; 131(8): 864-876, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29187378

ABSTRACT

Platelet collagen receptor glycoprotein VI (GPVI) and podoplanin receptor C-type lectin-like receptor 2 (CLEC2) are receptors implicated in platelet activation that both signal via an immunoreceptor tyrosine-based activation motif. Platelets are necessary for host defense and prevention of hemorrhage during sepsis, but the role of platelet GPVI and CLEC2 herein is unknown. To investigate this, we infected mice depleted of platelet GPVI or CLEC2 by antibody treatment or GPVI-/- mice with the common human sepsis pathogen Klebsiella pneumoniae via the airways to induce pneumonia-derived sepsis. The GPVI ligand collagen and the CLEC2 ligand podoplanin were constitutively present in the lung, whereas the GPVI ligands fibrin and histone were induced during pneumonia. During late-stage infection, both mice depleted of GPVI and GPVI-/- mice showed increased bacterial growth in lungs, and GPVI-/- mice also showed increased bacterial growth in distant body sites. Despite higher bacterial loads, GPVI-depleted mice showed reduced platelet numbers, platelet activation, and platelet-leukocyte complex formation in the bronchoalveolar space. Consistently, in human whole blood, GPVI stimulation of platelets increased platelet-leukocyte complex formation and leukocyte activation, which was accompanied by enhanced phagocytosis of Klebsiella GPVI-depleted mice showed increased lung hemorrhage during infection, but not to the extent observed in platelet-depleted mice, and lung bleeding was not significantly different between GPVI-/- and wild-type mice. CLEC2 depletion did not affect any of the responses during pneumonia. These results suggest that platelet GPVI, but not CLEC2, contributes to local host defense during pneumonia-derived sepsis by enhancing leukocyte function.


Subject(s)
Blood Platelets/immunology , Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacterial Infections/microbiology , Platelet Membrane Glycoproteins/physiology , Pneumonia/complications , Sepsis/immunology , Animals , Blood Platelets/metabolism , Blood Platelets/microbiology , Female , Gram-Negative Bacterial Infections/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , Pneumonia/microbiology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sepsis/etiology , Sepsis/pathology
11.
Int J Cardiol ; 235: 1-5, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28302320

ABSTRACT

Atrial fibrillation (AF) is associated with increased risk of thromboembolic complications. One of the markers of the increased risk of hypercoagulable state is platelet hyperreactivity. The aim of the study was to assess impact of arrhythmia on platelet reactivity. METHODS: The study included 36 (mean age 48,3; range 21-60) male patients with lone atrial fibrillation, with exclusion of concomitant diseases known to trigger hypercoagulable state. The AF patients underwent cardioversion to restore sinus rhythm and were subsequently under observation for 1month. Echocardiography, ECG and blood collection was performed before cardioversion (T0) and 4weeks after successful cardioversion (T1). During the study period patients have been contacted and examined every week and 24h ECG monitoring was performed. Platelet reactivity was assessed based on changes of CD62 and CD42b expression on platelet surface after stimulation with thrombin. Also changes in MPV were assessed. RESULTS: In all patients sinus rhythm was maintained at the end of the study period, however in 14 patients recurrences of AF were observed, confirmed by 24h ECG monitoring (atrial fibrillation recurrence group - AFR) and 22 patients maintained sinus rhythm throughout the whole study period (SR group). Mean fluorescence intensity (MFI) of CD62 on thrombin stimulated platelets decreased significantly 4weeks after electrical cardioversion as compared to T0 (48.04±22.42 vs 41.47±16.03; p<0.01). Also MFI of CD42b on thrombin stimulated platelets decreased significantly 4weeks after electrical cardioversion as compared to T0 (22.16±10.82 vs 12.06±5.99; p<0.0001). Platelets reactivity estimated by CD 62 expression in SR group decreased significantly after 4weeks observation (58.01±15.26 vs 46.57±13.44; p<0.001) opposite to AFR group 35.66±21.87 vs 34.54±16.4; p-ns). Moreover there were significant differences between basal reactivity during AF between SR and AFR groups (58.01±15.26 vs 35.66±21.87; p-0.01). MFI of CD42b on thrombin stimulated platelets decreased significantly both in AFR and SR groups (22.05±11.36 vs 13.8±6.03; p<0.001 and 21.87±14.18 vs 10.04±5.09; p<0005). MPV decreased significantly 4weeks after electrical cardioversion as compared to T0 (8.81±0.19 vs 8.42±0.14; p<0.0001). CONCLUSION: The changes of platelet reactivity to thrombin observed after restoration of sinus rhythm in patients prove that arrhythmia intrinsically leads to increased reactivity of platelets.


Subject(s)
Atrial Fibrillation , Electric Countershock/methods , Mean Platelet Volume/methods , Platelet Activation/physiology , Platelet Membrane Glycoproteins , Thrombin/physiology , Thromboembolism , Thrombosis , Atrial Fibrillation/blood , Atrial Fibrillation/complications , Atrial Fibrillation/physiopathology , Atrial Fibrillation/therapy , Biomarkers/analysis , Blood Coagulation/physiology , Echocardiography/methods , Electrocardiography/methods , Female , Humans , Male , Middle Aged , Platelet Membrane Glycoproteins/analysis , Platelet Membrane Glycoproteins/physiology , Poland , Risk Factors , Statistics as Topic , Thromboembolism/blood , Thromboembolism/etiology , Thrombophilia/diagnosis , Thrombosis/blood , Thrombosis/etiology
12.
J Thromb Thrombolysis ; 43(1): 54-59, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27416950

ABSTRACT

Cirrhosis is a consequence of prolonged liver injury and is characterised by extensive tissue fibrosis: the deposition of collagen-rich extracellular matrix. The haemostatic balance is disordered in cirrhosis and coagulation activation appears to promote fibrosis. In spite of recent studies demonstrating a role for anticoagulant therapy in preventing cirrhosis progression, there has not been a change in clinical practice, suggesting that physicians are reluctant to anticoagulate patients with cirrhosis due to bleeding risks. Platelets play an important role in facilitating coagulation. Glycoprotein VI (GPVI) is a platelet-specific collagen receptor that is shed from the platelet surface in a metalloproteinase-dependent manner in response to GPVI ligation and coagulation activation. Our aim was to use soluble GPVI levels to determine whether there was evidence for collagen and coagulation-induced platelet activation in early, well-compensated cirrhosis. Plasma soluble GPVI levels were quantified in 46 patients with mixed aetiology cirrhosis and 55 healthy controls using an immunoassay. In the cirrhosis group, soluble GPVI levels were significantly increased (5.8 ± 4.4 ng/ml, n = 46) compared to healthy controls (3.3 ± 3.4 ng/ml, n = 55, p < 0.05). This increase in soluble GPVI levels was still evident when levels were adjusted for platelet count (Healthy controls; 0.015 ± 0.018 ng/106 platelets/ml vs. cirrhosis; 0.048 ± 0.04 ng/106 platelets/ml, p < 0.0001). This study provides evidence for early platelet activation in patients with well-compensated cirrhosis. This may have translational implications for prognosis, treatment, and risk stratification.


Subject(s)
Liver Cirrhosis/blood , Platelet Activation , Platelet Membrane Glycoproteins/analysis , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Platelet Count , Platelet Membrane Glycoproteins/physiology , Solubility
13.
Oncotarget ; 7(15): 20788-800, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26959112

ABSTRACT

Pro-oxidative stressors can suppress host immunity due to their ability to generate oxidized lipid agonists of the platelet-activating factor-receptor (PAF-R). As radiation therapy also induces reactive oxygen species, the present studies were designed to define whether ionizing radiation could generate PAF-R agonists and if these lipids could subvert host immunity. We demonstrate that radiation exposure of multiple tumor cell lines in-vitro, tumors in-vivo, and human subjects undergoing radiation therapy for skin tumors all generate PAF-R agonists. Structural characterization of radiation-induced PAF-R agonistic activity revealed PAF and multiple oxidized glycerophosphocholines that are produced non-enzymatically. In a murine melanoma tumor model, irradiation of one tumor augmented the growth of the other (non-treated) tumor in a PAF-R-dependent process blocked by a cyclooxygenase-2 inhibitor. These results indicate a novel pathway by which PAF-R agonists produced as a byproduct of radiation therapy could result in tumor treatment failure, and offer important insights into potential therapeutic strategies that could improve the overall antitumor effectiveness of radiation therapy regimens.


Subject(s)
Antioxidants/pharmacology , Melanoma/therapy , Platelet Activating Factor/agonists , Platelet Membrane Glycoproteins/agonists , Receptors, G-Protein-Coupled/agonists , Skin Neoplasms/therapy , Ultraviolet Rays , Animals , Female , Humans , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Oxidative Stress , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/secondary , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
J Clin Invest ; 125(10): 3878-90, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26426079

ABSTRACT

Regulation of neutrophil activity is critical for immune evasion among extracellular pathogens, yet the mechanisms by which many bacteria disrupt phagocyte function remain unclear. Here, we have shown that the respiratory pathogen Streptococcus pneumoniae disables neutrophils by exploiting molecular mimicry to degrade platelet-activating factor (PAF), a host-derived inflammatory phospholipid. Using mass spectrometry and murine upper airway infection models, we demonstrated that phosphorylcholine (ChoP) moieties that are shared by PAF and the bacterial cell wall allow S. pneumoniae to leverage a ChoP-remodeling enzyme (Pce) to remove PAF from the airway. S. pneumoniae-mediated PAF deprivation impaired viability, activation, and bactericidal capacity among responding neutrophils. In the absence of Pce, neutrophils rapidly cleared S. pneumoniae from the airway and impeded invasive disease and transmission between mice. Abrogation of PAF signaling rendered Pce dispensable for S. pneumoniae persistence, reinforcing that this enzyme deprives neutrophils of essential PAF-mediated stimulation. Accordingly, exogenous activation of neutrophils overwhelmed Pce-mediated phagocyte disruption. Haemophilus influenzae also uses an enzyme, GlpQ, to hydrolyze ChoP and subvert PAF function, suggesting that mimicry-driven immune evasion is a common paradigm among respiratory pathogens. These results identify a mechanism by which shared molecular structures enable microbial enzymes to subvert host lipid signaling, suppress inflammation, and ensure bacterial persistence at the mucosa.


Subject(s)
Cell Wall/chemistry , Immune Evasion/physiology , Molecular Mimicry , Nasal Cavity/microbiology , Neutrophils/immunology , Phosphorylcholine/metabolism , Platelet Activating Factor/metabolism , Pneumococcal Infections/microbiology , Receptors, Cell Surface/physiology , Streptococcus pneumoniae/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Carrier State/microbiology , Cell Wall/immunology , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Haemophilus influenzae/enzymology , Haemophilus influenzae/genetics , Haemophilus influenzae/immunology , Humans , Immunity, Innate , Immunoglobulin D/deficiency , Immunoglobulin D/genetics , Immunoglobulin D/physiology , Lipoproteins/deficiency , Lipoproteins/genetics , Lipoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Nasal Cavity/immunology , Neutropenia/chemically induced , Neutropenia/immunology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Phagocytosis , Phosphorylcholine/chemistry , Platelet Activating Factor/chemistry , Platelet Activating Factor/deficiency , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/physiology , Pneumococcal Infections/immunology , Proteolysis , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/physiology , Species Specificity , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology
15.
Cancer Res ; 75(19): 4198-210, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26359459

ABSTRACT

Platelet-activating factor receptor (PAFR), a G-protein-coupled receptor, has been implicated in tumorigenesis, but its contributions to metastatic progression have not been investigated. Here, we show that PAFR is overexpressed in non-small cell lung cancer (NSCLC) as well as in breast, colorectal, and gastric carcinomas. Expression of PAFR correlates closely with clinical stages, survival time, and distant metastasis. In human NSCLC cells, activation of the PAF/PAFR signaling axis accentuated malignant character, including by stimulating epithelial-mesenchymal transition (EMT). In contrast, silencing PAFR in aggressive NSCLC cells inhibited these effects. Mechanistic investigations showed that PAFR stimulated EMT by activating STAT3 via upregulation of G-protein-dependent SRC or JAK2 kinase activity. Notably, STAT3 transcriptionally elevated PAFR expression. Thus, activation of PAFR in NSCLC cells initiated a forward feedback loop responsible for mediating the aggressive malignant character of NSCLC cells in vitro and in vivo. Reinforcing this reciprocal activation loop, PAF/PAFR signaling also upregulated IL6 expression and thereby STAT3 activation. Overall, our results elucidated an important role for PAFR dysregulation in the pathogenicity of NSCLC and unraveled a forward feedback loop between PAFR and STAT3 that acts to drive the malignant progression of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Epithelial-Mesenchymal Transition/physiology , Lung Neoplasms/pathology , Neoplasm Proteins/physiology , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/physiology , STAT3 Transcription Factor/physiology , Adult , Aged , Animals , Breast Neoplasms/metabolism , Carcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Disease Progression , Enzyme Activation , Feedback, Physiological , Female , GTP-Binding Proteins/physiology , Heterografts , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Kinases/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction , Stomach Neoplasms/metabolism , Up-Regulation
16.
Thromb Haemost ; 114(6): 1175-88, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26245230

ABSTRACT

The ability of cellular fibronectin, found in the vessel wall in a fibrillar conformation, to regulate platelet functions and trigger thrombus formation remains largely unknown. In this study, we evaluated how parietal cellular fibronectin can modulate platelet responses under flow conditions. A fibrillar network was formed by mechanically stretching immobilised dimeric cellular fibronectin. Perfusion of anticoagulated whole blood over this surface resulted in efficient platelet adhesion and thrombus growth. The initial steps of platelet adhesion and activation, as evidenced by filopodia extension and an increase in intracellular calcium levels (419 ± 29 nmol/l), were dependent on integrins α5ß1 and αIIbß3. Subsequent thrombus growth was mediated by these integrins together with the GPIb-V-IX complex, GPVI and Toll-like receptor 4. The involvement of Toll-like receptor 4 could be conveyed via its binding to the EDA region of cellular fibronectin. Upon thrombus formation, the platelets became procoagulant and generated fibrin as revealed by video-microscopy. This work provides evidence that fibrillar cellular fibronectin is a strong thrombogenic surface which supports efficient platelet adhesion, activation, aggregation and procoagulant activity through the interplay of a series of receptors including integrins α5ß1 and αIIbß3, the GPIb-V-IX complex, GPVI and Toll-like receptor 4.


Subject(s)
Blood Coagulation/physiology , Fibronectins/physiology , Platelet Aggregation/physiology , Animals , Annexin A5/metabolism , Extracellular Matrix , Fibrin/biosynthesis , Fibroblasts , Fibronectins/chemistry , Immobilized Proteins , Integrin beta1/genetics , Integrins/physiology , Lab-On-A-Chip Devices , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfluidic Analytical Techniques , Microscopy, Electron, Scanning , Platelet Adhesiveness , Platelet Membrane Glycoprotein IIb/genetics , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/physiology , Rheology , Stress, Mechanical , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology
17.
Biochim Biophys Acta ; 1853(8): 1879-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25960397

ABSTRACT

Phosphatidylinositol 3-kinaseß (PI3Kß) plays a predominant role in integrin outside-in signaling and in platelet activation by GPVI engagement. We have shown that the tyrosine kinase Pyk2 mediates PI3Kß activation downstream of integrin αIIbß3, and promotes the phosphorylation of the PI3K-associated adaptor protein c-Cbl. In this study, we compared the functional correlation between Pyk2 and PI3Kß upon recruitment of the two main platelet collagen receptors, integrin α2ß1 and GPVI. PI3Kß-mediated phosphorylation of Akt was inhibited in Pyk2-deficient platelets adherent to monomeric collagen through integrin α2ß1, but occurred normally upon GPVI ligation. Integrin α2ß1 engagement led to Pyk2-independent association of c-Cbl with PI3K. However, c-Cbl was not phosphorylated in adherent platelets, and phosphorylation of Akt occurred normally in c-Cbl-deficient platelets, indicating that the c-Cbl is dispensable for Pyk2-mediated PI3Kß activation. Stimulation of platelets with CRP, a selective GPVI ligand, induced c-Cbl phosphorylation in the absence of Pyk2, but failed to promote its association with PI3K. Pyk2 activation was completely abrogated in PI3KßKD, but not in PI3KγKD platelets, and was strongly inhibited by Src kinases and phospholipase C inhibitors, and by BAPTA-AM. The absence of PI3Kß activity also hampered GPVI-induced tyrosine-phosphorylation and activation of PLCγ2, preventing intracellular Ca2+ increase and phosphorylation of pleckstrin. Moreover, GPVI-induced intracellular Ca2+ increase and pleckstrin phosphorylation were also strongly inhibited in human platelets treated with the PI3Kß inhibitor TGX-221. These results outline important differences in the regulation of PI3Kß by GPVI and integrin α2ß1 and suggest that inhibition of Pyk2 may target PI3Kß activation in a selective context of platelet stimulation.


Subject(s)
Focal Adhesion Kinase 2/physiology , Integrin alpha2beta1/physiology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Membrane Glycoproteins/physiology , Proto-Oncogene Proteins c-cbl/physiology , Animals , Cells, Cultured , Enzyme Activation , Humans , Mice , Mice, Knockout , Signal Transduction
18.
Thromb Haemost ; 114(2): 313-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25994029

ABSTRACT

Platelet cold agglutinins (PCA) cause pseudothrombocytopenia, spurious thrombocytopenia due to ex vivo platelet clumping, complicating clinical diagnosis, but mechanisms and consequences of PCA are not well defined. Here, we characterised an atypical immunoglobulin (Ig)M PCA in a 37-year-old woman with lifelong bleeding and chronic moderate thrombocytopenia, that induces activation and aggregation of autologous or allogeneic platelets via interaction with platelet glycoprotein (GP)VI. Patient temperature-dependent pseudothrombocytopenia was EDTA-independent, but was prevented by integrin αIIbß3 blockade. Unstimulated patient platelets revealed elevated levels of bound IgM, increased expression of activation markers (P-selectin and CD63), low GPVI levels and abnormally high thromboxane (TX)A2 production. Patient serum induced temperature- and αIIbß3-dependent decrease of platelet count in allogeneic donor citrated platelet-rich plasma (PRP), but not in PRP from Glanzmann's thrombasthenia or afibrinogenaemia patients. In allogeneic platelets, patient plasma induced shape change, P-selectin and CD63 expression, (14)C-serotonin release, and TXA2 production. Activation was not inhibited by aspirin, cangrelor or blocking anti-Fc receptor (FcγRIIA) antibody, but was abrogated by inhibitors of Src and Syk, and by a soluble GPVI-Fc fusion protein. GPVI-deficient platelets were not activated by patient plasma. These data provide the first evidence for an IgM PCA causing platelet activation/aggregation via GPVI. The PCA activity persisted over a five-year follow-up period, supporting a causative role in patient chronic thrombocytopenia and bleeding.


Subject(s)
Hemorrhagic Disorders/immunology , Immunoglobulin M/immunology , Platelet Aggregation/immunology , Platelet Membrane Glycoproteins/physiology , Thrombocytopenia/immunology , Adult , Afibrinogenemia/blood , Autoantibodies/blood , Biomarkers/blood , Cold Temperature/adverse effects , Cryoglobulins/pharmacology , Female , Humans , Platelet Activation/immunology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein-Tyrosine Kinases/blood , Thrombasthenia/blood , Thrombocytopenia/blood
19.
Cardiovasc Res ; 107(1): 9-19, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25902782

ABSTRACT

AIMS: Patients with conditions that are associated with insulin resistance such as obesity, type 2 diabetes mellitus, and polycystic ovary syndrome have an increased risk of thrombosis and a concurrent hyperactive platelet phenotype. Our aim was to determine whether insulin resistance of megakaryocytes/platelets promotes platelet hyperactivation. METHODS AND RESULTS: We generated a conditional mouse model where the insulin receptor (IR) was specifically knocked out in megakaryocytes/platelets and performed ex vivo platelet activation studies in wild-type (WT) and IR-deficient platelets by measuring aggregation, integrin αIIbß3 activation, and dense and α-granule secretion. Deletion of IR resulted in an increase in platelet count and volume, and blocked the action of insulin on platelet signalling and function. Platelet aggregation, granule secretion, and integrin αIIbß3 activation in response to the glycoprotein VI (GPVI) agonist collagen-related peptide (CRP) were significantly reduced in platelets lacking IR. This was accompanied by a reduction in the phosphorylation of effectors downstream of GPVI. Interestingly, loss of IR also resulted in a reduction in insulin-like growth factor-1 (IGF-1)- and insulin-like growth factor-2 (IGF-2)-mediated phosphorylation of IRS-1, Akt, and GSK3ß and priming of CRP-mediated platelet activation. Pharmacological inhibition of IR and the IGF-1 receptor in WT platelets recapitulated the platelet phenotype of IR-deficient platelets. CONCLUSIONS: Deletion of IR (i) increases platelet count and volume, (ii) does not cause platelet hyperactivity, and (iii) reduces GPVI-mediated platelet function and platelet priming by IGF-1 and IGF-2.


Subject(s)
Blood Platelets/physiology , Insulin-Like Growth Factor II/physiology , Insulin-Like Growth Factor I/physiology , Megakaryocytes/physiology , Receptor, Insulin/physiology , Signal Transduction/physiology , Thrombocytosis/etiology , Animals , Insulin/pharmacology , Mice , Mice, Inbred C57BL , Platelet Membrane Glycoproteins/physiology
20.
Adv Exp Med Biol ; 844: 263-75, 2014.
Article in English | MEDLINE | ID: mdl-25480646

ABSTRACT

At the clinical level, recent studies reveal the link between coagulation and other pathophysiological processes, including platelet activation, inflammation, cancer, the immune response, and/or infectious diseases. These links are likely to underpin the coagulopathy associated with risk factors for venous thromboembolic (VTE) and deep vein thrombosis (DVT). At the molecular level, the interactions between platelet-specific receptors and coagulation factors could help explain coagulopathy associated with aberrant platelet function, as well as revealing new approaches targeting platelet receptors in diagnosis or treatment of VTE or DVT. Glycoprotein (GP)Ibα, the major ligand-binding subunit of the platelet GPIb-IX-V complex, that binds the adhesive ligand, von Willebrand factor (VWF), is co-associated with the platelet-specific collagen receptor, GPVI. The GPIb-IX-V/GPVI adheso-signaling complex not only initiates platelet activation and aggregation (thrombus formation) in response to vascular injury or disease but GPIbα also regulates coagulation through a specific interaction with thrombin and other coagulation factors. Here, we discuss the structure and function of key platelet receptors involved in thrombus formation and coagulation in health and disease, with a particular focus on platelet GPIbα.


Subject(s)
Blood Coagulation , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/physiology , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/physiology , Animals , Blood Platelets/physiology , Humans , Protein Binding , Protein Conformation , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...