Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 12: 854126, 2022.
Article in English | MEDLINE | ID: mdl-36061862

ABSTRACT

The pathophysiology of sepsis and its accompanying hyper-inflammatory response are key events that lead to multi-organ failure and death. A growing body of literature now suggests that the vascular endothelium plays a critical role in driving early events of sepsis progression. In this study, we demonstrate how endothelial-derived exosomes contribute to a successive pro-inflammatory phenotype of monocytes. Exosomes isolated from S. aureus infected endothelial cells drive both CD11b and MHCII expression in monocytes and contribute dysregulated cytokine production. Conversely, healthy endothelial exosomes had no major effect. microRNA (miRNA) profiling of exosomes identified miR-99 upregulation which we hypothesised as driving this phenotypic change through mechanistic target of rapamycin (mTOR). Knockdown of mTOR with miR-99a and miR-99b mimetics in S. aureus infected monocytes increased IL-6 and decreased IL-10 production. Interestingly, inhibition of miRNAs with antagomirs has the opposing effect. Collectively, endothelial exosomes are driving a pro-inflammatory phenotype in monocytes through dysregulated expression of miR-99a and miR-99b.


Subject(s)
Exosomes , MicroRNAs/metabolism , Sepsis , TOR Serine-Threonine Kinases/genetics , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , MicroRNAs/genetics , Sepsis/genetics , Sepsis/metabolism , Sepsis/pathology , Staphylococcus aureus/genetics
3.
Res Social Adm Pharm ; 18(7): 3204-3209, 2022 07.
Article in English | MEDLINE | ID: mdl-34483082

ABSTRACT

The impact of the COVID-19 pandemic on pharmacy education worldwide has been immense, affecting students, educators and regulatory agencies. Pharmacy programmes have had to rapidly adapt in their delivery of education, maintaining standards while also ensuring the safety of all stakeholders. In this commentary, we describe the challenges, compromises and solutions adopted by our institution throughout the pandemic, the lessons learnt, adaptive measures taken, and strategies to develop and future-proof our curricula.


Subject(s)
COVID-19 , Education, Pharmacy , Pharmacy , Students, Pharmacy , COVID-19/epidemiology , Curriculum , Humans , Pandemics
4.
J Thromb Haemost ; 19(2): 582-587, 2021 02.
Article in English | MEDLINE | ID: mdl-34396675

ABSTRACT

In vitro flow-based assays are widely used to investigate the role of platelets and coagulation in hemostasis and thrombosis. Their main advantage over other assays relies on the fact that they integrate blood flow that regulates many aspects of platelet function, including adhesion, activation, and aggregation. Blood flow is also central in the regulation of coagulation through its ability to modulate the local concentrations of coagulation factors within and around thrombi. The most broadly used assay to study thrombus formation consists in perfusing whole blood over immobilized fibrillar collagen through a single channel, which helps to reproduce thrombus formation as it occurs in vivo after vascular injury, with platelets adhering, becoming activated, and forming a mural thrombus. This process can also be studied under conditions of thrombin generation, notably by recalcifying blood collected in sodium citrate. In this manuscript, we briefly discuss the advantages and limits of this broadly used "in vitro thrombus formation model." The main emphasis is on the description of the most recent developments regarding design of new flow models and new techniques, and how these may advance the landscape of in vitro studies into the formation of physiological or pathophysiological thrombi. Challenges linked to mimicking the formation of a hemostatic plug in a healthy vessel or a thrombus in diseased arteries and the complexity of reproducing the various aspects of venous thrombosis are discussed. Future directions are proposed to improve the physiological or pathophysiological relevance of current flow-based assays.


Subject(s)
Hemostasis , Thrombosis , Blood Coagulation , Blood Platelets , Humans , Platelet Function Tests
5.
PLoS One ; 16(6): e0253347, 2021.
Article in English | MEDLINE | ID: mdl-34161337

ABSTRACT

The unprecedented global COVID-19 pandemic has prompted a desperate international effort to accelerate the development of anti-viral candidates. For unknown reasons, COVID-19 infections are associated with adverse cardiovascular complications, implicating that vascular endothelial cells are essential in viral propagation. The etiological pathogen, SARS-CoV-2, has a higher reproductive number and infection rate than its predecessors, indicating it possesses novel characteristics that infers enhanced transmissibility. A unique K403R spike protein substitution encodes an Arg-Gly-Asp (RGD) motif, introducing a potential role for RGD-binding host integrins. Integrin αVß3 is widely expressed across the host, particularly in the endothelium, which acts as the final barrier before microbial entry into the bloodstream. This mutagenesis creates an additional binding site, which may be sufficient to increase SARS-CoV-2 pathogenicity. Here, we investigate how SARS-CoV-2 passes from the epithelium to endothelium, the effects of αVß3 antagonist, Cilengitide, on viral adhesion, vasculature permeability and leakage, and also report on a simulated interaction between the viral and host protein in-silico.


Subject(s)
Endothelium, Vascular/virology , Integrin alphaVbeta3/metabolism , SARS-CoV-2/pathogenicity , Snake Venoms/pharmacology , Antigens, CD/metabolism , Binding Sites , COVID-19/metabolism , COVID-19/physiopathology , Caco-2 Cells , Cadherins/metabolism , Computer Simulation , Endothelium, Vascular/cytology , Endothelium, Vascular/physiopathology , Host-Pathogen Interactions/drug effects , Humans , Integrin alphaVbeta3/chemistry , Models, Molecular , Mutation , Permeability , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization
6.
Antibiotics (Basel) ; 10(5)2021 May 14.
Article in English | MEDLINE | ID: mdl-34068975

ABSTRACT

The bacterial pathogen Staphylococcus aureus is a leading cause of bloodstream infections, where patients often suffer from relapse despite antibiotic therapy. Traditional anti-staphylococcal drugs display reduced effectivity against internalised bacteria, but nanoparticles conjugated with antibiotics can overcome these challenges. In the present study, we aimed to characterise the internalisation and re-emergence of S. aureus from human endothelial cells and construct a new formulation of nanoparticles that target intracellular bacteria. Using an in vitro infection model, we demonstrated that S. aureus invades and persists within endothelial cells, mediated through bacterial extracellular surface adhesion, Fibronectin-binding protein A/B. After internalising, S. aureus localises to vacuoles as determined by transmission electron microscopy. Viable S. aureus emerges from endothelial cells after 48 h, supporting the notion that intracellular persistence contributes to infection relapses during bloodstream infections. Poly lactic-co-glycolic acid nanoparticles were formulated using a water-in-oil double emulsion method, which loaded 10% vancomycin HCl with 82.85% ± 12 encapsulation efficiency. These non-toxic nanoparticles were successfully taken up by cells and demonstrated a biphasic controlled release of 91 ± 4% vancomycin. They significantly reduced S. aureus intracellular growth within infected endothelial cells, which suggests future potential applications for targeting internalised bacteria and reducing mortality associated with bacteraemia.

7.
Sci Rep ; 11(1): 5265, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664277

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) have demonstrated efficacy in pre-clinical models of inflammation and tissue injury, including in models of lung injury and infection. Rolling, adhesion and transmigration of MSCs appears to play a role during MSC kinetics in the systemic vasculature. However, a large proportion of MSCs become entrapped within the lungs after intravenous administration, while the initial kinetics and the site of arrest of MSCs in the pulmonary vasculature are unknown. We examined the kinetics of intravascularly administered MSCs in the pulmonary vasculature using a microfluidic system in vitro and intra-vital microscopy of intact mouse lung. In vitro, MSCs bound to endothelium under static conditions but not under laminar flow. VCAM-1 antibodies did not affect MSC binding. Intravital microscopy demonstrated MSC arrest at pulmonary micro-vessel bifurcations due to size obstruction. Retention of MSCs in the pulmonary microvasculature was increased in Escherichia coli-infected animals. Trapped MSCs deformed over time and appeared to release microvesicles. Labelled MSCs retained therapeutic efficacy against pneumonia. Our results suggest that MSCs are physically obstructed in pulmonary vasculature and do not display properties of rolling/adhesion, while retention of MSCs in the infected lung may require receptor interaction.


Subject(s)
Blood Vessels/transplantation , Lung/diagnostic imaging , Mesenchymal Stem Cell Transplantation , Pneumonia/therapy , Administration, Intravenous , Animals , Blood Vessels/diagnostic imaging , Blood Vessels/pathology , Cardiovascular System/metabolism , Disease Models, Animal , Humans , Kinetics , Lung/blood supply , Lung/metabolism , Lung/pathology , Mesenchymal Stem Cells/cytology , Mice , Pneumonia/diagnostic imaging , Pneumonia/metabolism , Pneumonia/pathology
8.
Drug Discov Today ; 25(12): 2317-2325, 2020 12.
Article in English | MEDLINE | ID: mdl-33035665

ABSTRACT

Sepsis is a life-threatening condition caused by the response of the body to an infection, and has recently been regarded as a global health priority because of the lack of effective treatments available. Vascular endothelial cells have a crucial role in sepsis and are believed to be a major target of pathogens during the early stages of infection. Accumulating evidence suggests that common sepsis pathogens, including bacteria, fungi, and viruses, all contain a critical integrin recognition motif, Arg-Gly-Asp (RGD), in their major cell wall-exposed proteins that might act as ligands to crosslink to vascular endothelial cells, triggering systemic dysregulation resulting in sepsis. In this review, we discuss the potential of anti-integrin therapy in the treatment of sepsis and septic shock.


Subject(s)
Bacterial Infections/drug therapy , Integrins/antagonists & inhibitors , Mycoses/drug therapy , Oligopeptides/antagonists & inhibitors , Sepsis/drug therapy , Virus Diseases/drug therapy , Humans
9.
Res Pract Thromb Haemost ; 4(5): 680-713, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32685876

ABSTRACT

The 2020 Congress of the International Society of Thrombosis and Haemostasis (ISTH) was held virtually July 12-15, 2019, due to the coronavirus disease 2019 pandemic. The congress convenes annually to discuss clinical and basic topics in hemostasis and thrombosis. Each year, the program includes State of Art (SOA) lectures given by prominent scientists. Presenters are asked to create Illustrated Capsules of their talks, which are concise illustrations with minimal explanatory text. Capsules cover major themes of the presentation, and these undergo formal peer review for inclusion in this article. Owing to the shift to a virtual congress this year, organizers reduced the program size. There were 39 SOA lectures virtually presented, and 29 capsules (9 from talks omitted from the virtual congress) were both submitted and successful in peer review, and are included in this article. Topics include the roles of the hemostatic system in inflammation, infection, immunity, and cancer, platelet function and signaling, platelet function disorders, megakaryocyte biology, hemophilia including gene therapy, phenotype tests in hemostasis, von Willebrand factor, anticoagulant factor V, computational driven discovery, endothelium, clinical and basic aspects of thrombotic microangiopathies, fibrinolysis and thrombolysis, antithrombotics in pediatrics, direct oral anticoagulant management, and thrombosis and hemostasis in pregnancy. Capsule authors invite virtual congress attendees to refer to these capsules during the live presentations and participate on Twitter in discussion. Research and Practice in Haemostasis and Thrombosis will release 2 tweets from @RPTHJournal during each presentation, using #IllustratedReview, #CoagCapsule and #ISTH2020. Readers are also welcome to utilize capsules for teaching and ongoing education.

10.
J Thromb Haemost ; 18(3): 748-752, 2020 03.
Article in English | MEDLINE | ID: mdl-32112535

ABSTRACT

Experimental videomicroscopic in vitro assays of thrombus formation based on blood perfusion are instrumental in a wide range of basic studies in thrombosis, screening for hereditary or acquired plateletrelated pathologies, and assessing the effectiveness of novel anti-platelet therapies. Here, we discuss application of the broadly used "in vitro thrombosis model": a frequently used assay to study the formation of 3D aggregates under flow, which involves perfusing anticoagulated whole blood over fibrillar collagen in a flow geometry of rectangular cross-section, such as glass microcapillaries or parallel-plate flow chambers. Major advantaged of this assay are simplicity and ability to reproduce the four main stages of platelet thrombus formation, i.e. platelet tethering, adhesion, activation and aggregation under a wide range of hemodynamic conditions. On the other hand, these devices represent, at best, simple reductive models of thrombosis. We also describe how blood flow assays can be used to study various aspects of platelet function on adhesive proteins and discuss the relevance of such flow models. Finally, we propose recommendations for standardization related to the use of this assay that cover collagen source, coating methods, micropatterning, sample composition, anticoagulation, choice of flow device, hemodynamic conditions, quantification challenges, variability, pre-analytical conditions and other issues.


Subject(s)
Thrombosis , Blood Platelets , Communication , Hemostasis , Humans , Platelet Function Tests , Reference Standards
11.
Front Immunol ; 10: 1748, 2019.
Article in English | MEDLINE | ID: mdl-31447831

ABSTRACT

Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. While many individual cells and systems in the body are involved in driving the excessive and sometimes sustained host response, pathogen engagement with endothelial cells and platelets early in sepsis progression, are believed to be key. Significant progress has been made in establishing key molecular interactions between platelets and pathogens and endothelial cells and pathogens. This review will explore the growing number of compensatory connections between bacteria and viruses with platelets and endothelial cells and how a better understanding of these interactions are informing the field of potential novel ways to treat the dysregulated host response during sepsis.


Subject(s)
Blood Platelets/physiology , Endothelial Cells/physiology , Host-Pathogen Interactions , Sepsis/immunology , Animals , Blood Platelets/immunology , Cytokines/physiology , Humans , Leukocytes/physiology , Signal Transduction/physiology
12.
Clin Microbiol Rev ; 31(2)2018 04.
Article in English | MEDLINE | ID: mdl-29444953

ABSTRACT

Osteomyelitis is an inflammatory bone disease that is caused by an infecting microorganism and leads to progressive bone destruction and loss. The most common causative species are the usually commensal staphylococci, with Staphylococcus aureus and Staphylococcus epidermidis responsible for the majority of cases. Staphylococcal infections are becoming an increasing global concern, partially due to the resistance mechanisms developed by staphylococci to evade the host immune system and antibiotic treatment. In addition to the ability of staphylococci to withstand treatment, surgical intervention in an effort to remove necrotic and infected bone further exacerbates patient impairment. Despite the advances in current health care, osteomyelitis is now a major clinical challenge, with recurrent and persistent infections occurring in approximately 40% of patients. This review aims to provide information about staphylococcus-induced bone infection, covering the clinical presentation and diagnosis of osteomyelitis, pathophysiology and complications of osteomyelitis, and future avenues that are being explored to treat osteomyelitis.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Osteomyelitis/drug therapy , Osteomyelitis/pathology , Staphylococcal Infections/pathology , Disease Progression , Host-Pathogen Interactions , Humans , Staphylococcal Infections/drug therapy , Staphylococcus/physiology
13.
Cell Microbiol ; 19(3)2017 03.
Article in English | MEDLINE | ID: mdl-27598716

ABSTRACT

Blood-brain barrier (BBB) disruption constitutes a hallmark event during pathogen-mediated neurological disorders such as bacterial meningitis. As a prevalent opportunistic pathogen, Staphylococcus aureus (SA) is of particular interest in this context, although our fundamental understanding of how SA disrupts the BBB is very limited. This paper employs in vitro infection models to address this. Human brain microvascular endothelial cells (HBMvECs) were infected with formaldehyde-fixed (multiplicity of infection [MOI] 0-250, 0-48 hr) and live (MOI 0-100, 0-3 hr) SA cultures. Both Fixed-SA and Live-SA could adhere to HBMvECs with equal efficacy and cause elevated paracellular permeability. In further studies employing Fixed-SA, infection of HBMvECs caused dose-dependent release of cytokines/chemokines (TNF-α, IL-6, MCP-1, IP-10, and thrombomodulin), reduced expression of interendothelial junction proteins (VE-Cadherin, claudin-5, and ZO-1), and activation of both canonical and non-canonical NF-κB pathways. Using N-acetylcysteine, we determined that these events were coupled to the SA-mediated induction of reactive oxygen species (ROS) within HBMvECs. Finally, treatment of HBMvECs with Fixed-ΔSpA (MOI 0-250, 48 hr), a gene deletion mutant of Staphylococcal protein A associated with bacterial infectivity, had relatively similar effects to Newman WT Fixed-SA. In conclusion, these findings provide insight into how SA infection may activate proinflammatory mechanisms within the brain microvascular endothelium to elicit BBB failure.


Subject(s)
Blood-Brain Barrier/injuries , Endothelial Cells/microbiology , Endothelial Cells/physiology , Staphylococcus aureus/pathogenicity , Bacterial Adhesion , Cells, Cultured , Cytokines/metabolism , Humans , Models, Biological , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Tight Junction Proteins/metabolism
14.
Drug Discov Today ; 21(9): 1437-1445, 2016 09.
Article in English | MEDLINE | ID: mdl-27117348

ABSTRACT

The field of tissue engineering is developing biomimetic biomaterial scaffolds that are showing increasing therapeutic potential for the repair of cardiovascular tissues. However, a major opportunity exists to use them as 3D in vitro models for the study of cardiovascular tissues and disease in addition to drug development and testing. These in vitro models can span the gap between 2D culture and in vivo testing, thus reducing the cost, time, and ethical burden of current approaches. Here, we outline the progress to date and the requirements for the development of ideal in vitro 3D models for blood vessels, heart valves, and myocardial tissue.


Subject(s)
Blood Vessels , Heart Valves , Models, Biological , Myocardium , Animals , Cardiovascular Diseases , Humans , Tissue Engineering
15.
Platelets ; 27(6): 535-40, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27025455

ABSTRACT

Gram-negative Escherichia coli cause diseases such as sepsis and hemolytic uremic syndrome in which thrombotic disorders can be found. Direct platelet-bacterium interactions might contribute to some of these conditions; however, mechanisms of human platelet activation by E. coli leading to thrombus formation are poorly understood. While the IgG receptor FcγRIIA has a key role in platelet response to various Gram-positive species, its role in activation to Gram-negative bacteria is poorly defined. This study aimed to investigate the molecular mechanisms of human platelet activation by E. coli, including the potential role of FcγRIIA. Using light-transmission aggregometry, measurements of ATP release and tyrosine-phosphorylation, we investigated the ability of two E. coli clinical isolates to activate platelets in plasma, in the presence or absence of specific receptors and signaling inhibitors. Aggregation assays with washed platelets supplemented with IgGs were performed to evaluate the requirement of this plasma component in activation. We found a critical role for the immune receptor FcγRIIA, αIIbß3, and Src and Syk tyrosine kinases in platelet activation in response to E. coli. IgG and αIIbß3 engagement was required for FcγRIIA activation. Moreover, feedback mediators adenosine 5'-diphosphate (ADP) and thromboxane A2 (TxA2) were essential for platelet aggregation. These findings suggest that human platelet responses to E. coli isolates are similar to those induced by Gram-positive organisms. Our observations support the existence of a central FcγRIIA-mediated pathway by which human platelets respond to both Gram-negative and Gram-positive bacteria.


Subject(s)
Blood Platelets/immunology , Blood Platelets/metabolism , Escherichia coli/immunology , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, IgG/metabolism , Adenosine Diphosphate/metabolism , Humans , Platelet Activation/immunology , Platelet Aggregation/immunology , Platelet Function Tests , Protein Binding , Syk Kinase/metabolism , Thromboxane A2/metabolism , src-Family Kinases/metabolism
16.
Pathogens ; 4(4): 869-82, 2015 Dec 05.
Article in English | MEDLINE | ID: mdl-26690226

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen often carried asymptomatically on the human body. Upon entry to the otherwise sterile environment of the cardiovascular system, S. aureus can lead to serious complications resulting in organ failure and death. The success of S. aureus as a pathogen in the bloodstream is due to its ability to express a wide array of cell wall proteins on its surface that recognise host receptors, extracellular matrix proteins and plasma proteins. Endothelial cells and platelets are important cells in the cardiovascular system and are a major target of bloodstream infection. Endothelial cells form the inner lining of a blood vessel and provide an antithrombotic barrier between the vessel wall and blood. Platelets on the other hand travel throughout the cardiovascular system and respond by aggregating around the site of injury and initiating clot formation. Activation of either of these cells leads to functional dysregulation in the cardiovascular system. In this review, we will illustrate how S. aureus establish intimate interactions with both endothelial cells and platelets leading to cardiovascular dysregulation.

17.
Biochem Soc Trans ; 43(4): 715-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26551718

ABSTRACT

The cardiovascular system is typically a sterile environment; however entry of a microorganism into the circulation can cause potentially life threatening cardiac and/or vascular disease. Staphylococcus aureus endothelial cell interactions are arguably the most important interactions in the pathogenesis of cardiovascular infection. These interactions can trigger cardiac valve destruction in the case of endocarditis, multi-organ dysfunction in the case of sepsis and coagulopathy. Here, we review the interactions between S. aureus and endothelial cells and discuss the implications of these interactions in the progression of cardiovascular infection.


Subject(s)
Endothelium, Vascular/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Bacterial Adhesion , Cardiovascular Infections/microbiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiopathology , Humans
18.
Platelets ; 26(4): 293-301, 2015.
Article in English | MEDLINE | ID: mdl-25734214

ABSTRACT

Given their small size, platelets are emerging as being one of the most important entities in the bloodstream. Not only do they play a key role in maintaining thrombosis and haemostasis, platelets also play a critical role in orchestrating the immune response. Being the first cell at the site of injury, they are perfectly placed to assess the extent of the damage and recruit immune cells as is necessary. As a first line of defence, platelets can act as primitive immune cells themselves by interacting with invading pathogens. A number of platelet receptors have been shown to interact with bacteria either directly or indirectly, involving a plasma protein bridge. This review will discuss the molecular mechanisms that exist between platelets and bacteria and the functional response to the interaction. We will also discuss the importance of considering animal models of disease and the use of physiological shear when studying platelet-bacterial interactions.


Subject(s)
Bacteria/immunology , Blood Platelets/immunology , Infections/immunology , Inflammation/immunology , Humans , Infections/blood , Inflammation/blood
19.
Thromb Haemost ; 113(2): 290-304, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25413489

ABSTRACT

The integrin αIIbß3 on resting platelets can bind to immobilised fibrinogen resulting in platelet spreading and activation but requires activation to bind to soluble fibrinogen. αIIbß3 is known to interact with the general integrin-recognition motif RGD (arginine-glycine-aspartate) as well as the fibrinogen-specific γ-chain dodecapeptide; however, it is not known how fibrinogen binding triggers platelet activation. NGR (asparagine-glycine-arginine) is another integrin-recognition sequence present in fibrinogen and this study aims to determine if it plays a role in the interaction between fibrinogen and αIIbß3. NGR-containing peptides inhibited resting platelet adhesion to fibrinogen with an IC50 of 175 µM but failed to inhibit the adhesion of activated platelets to fibrinogen (IC50> 500 µM). Resting platelet adhesion to mutant fibrinogens lacking the NGR sequences was reduced compared to normal fibrinogen under both static and shear conditions (200 s⁻¹). However, pre-activated platelets were able to fully spread on all types of fibrinogen. Thus, the NGR motif in fibrinogen is the site that is primarily responsible for the interaction with resting αIIbß3 and is responsible for triggering platelet activation.


Subject(s)
Blood Platelets/physiology , Fibrinogen/chemistry , Oligopeptides/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Amino Acid Motifs , Animals , Blood Coagulation , CD13 Antigens/chemistry , CHO Cells , COS Cells , Cell Adhesion , Chlorocebus aethiops , Cricetulus , Enzyme-Linked Immunosorbent Assay , Healthy Volunteers , Humans , Inhibitory Concentration 50 , Ligands , Microscopy, Confocal , Microscopy, Fluorescence , Peptides/chemistry , Platelet Activation , Platelet Adhesiveness , Platelet Aggregation/physiology , Platelet Function Tests , Platelet Membrane Glycoproteins/chemistry , Protein Binding , Recombinant Proteins/chemistry
20.
Blood ; 123(20): 3166-74, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24642751

ABSTRACT

Bacterial adhesion to platelets is mediated via a range of strain-specific bacterial surface proteins that bind to a variety of platelet receptors. It is unclear how these interactions lead to platelet activation. We demonstrate a critical role for the immune receptor FcγRIIA, αIIbß3, and Src and Syk tyrosine kinases in platelet activation by Staphylococcus aureus, Streptococcus sanguinis, Streptococcus gordonii, Streptococcus oralis, and Streptococcus pneumoniae. FcγRIIA activation is dependent on immunoglobulin G (IgG) and αIIbß3 engagement. Moreover, feedback agonists adenosine 5'-diphosphate and thromboxane A2 are mandatory for platelet aggregation. Additionally, platelet factor 4 (PF4) binds to bacteria and reduces the lag time for aggregation, and gray platelet syndrome α-granule-deficient platelets do not aggregate to 4 of 5 bacterial strains. We propose that FcγRIIA-mediated activation is a common response mechanism used against a wide range of bacteria, and that release of secondary mediators and PF4 serve as a positive feedback mechanism for activation through an IgG-dependent pathway.


Subject(s)
Blood Platelets/microbiology , Host-Pathogen Interactions , Platelet Factor 4/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Receptors, IgG/immunology , Staphylococcus aureus/physiology , Streptococcus/physiology , Adenosine Diphosphate/immunology , Animals , Blood Platelets/immunology , Humans , Mice , Mice, Transgenic , Platelet Activation , Staphylococcal Infections/immunology , Streptococcal Infections/immunology , Thromboxane A2/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...