181 results on '"Gibbins JM"'
Search Results
2. Exploring the constituent mechanisms of hepatitis: a dynamical systems approach
- Author
-
Dunster, JL, Gibbins, JM, and Nelson, MR
- Subjects
Pharmacology ,General Immunology and Microbiology ,Applied Mathematics ,Modeling and Simulation ,General Neuroscience ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,General Environmental Science - Abstract
Hepatitis is the term used to describe inflammation in the liver. It is associated with a high rate of mortality, but the underlying disease mechanisms are not completely understood and treatment options are limited. We present a mathematical model of hepatitis that captures the complex interactions between hepatocytes (liver cells), hepatic stellate cells (cells in the liver that produce hepatitis-associated fibrosis) and the immune components that mediate inflammation. The model is in the form of a system of ordinary differential equations. We use numerical techniques and bifurcation analysis to characterize and elucidate the physiological mechanisms that dominate liver injury and its outcome to a healthy or unhealthy, chronic state. This study reveals the complex interactions between the multiple cell types and mediators involved in this complex disease and highlights potential problems in targeting inflammation in the liver therapeutically.
- Published
- 2022
3. The voltage-gated K+ channel Kv1.3 modulates platelet motility and α2β1 integrin-dependent adhesion to collagen
- Author
-
Wright, JR, Jones, S, Parvathy, S, Kaczmarek, LK, Forsythe, I, Farndale, RW, Gibbins, JM, Mahaut-Smith, MP, Wright, JR, Jones, S, Parvathy, S, Kaczmarek, LK, Forsythe, I, Farndale, RW, Gibbins, JM, and Mahaut-Smith, MP
- Abstract
Kv1.3 is a voltage-gated K+-selective channel with roles in immunity, insulin-sensitivity, neuronal excitability and olfaction. Despite being one of the largest ionic conductances of the platelet surface membrane, its contribution to platelet function is poorly understood. Here we show that Kv1.3-deficient platelets display enhanced ADP-evoked platelet aggregation and secretion, and an increased surface expression of platelet integrin αIIb. In contrast, platelet adhesion and thrombus formation in vitro under arterial shear conditions on surfaces coated with collagen were reduced for samples from Kv1.3−/- compared to wild type mice. Use of collagen-mimetic peptides revealed a specific defect in the engagement with α2β1. Kv1.3−/- platelets developed significantly fewer, and shorter, filopodia than wild type platelets during adhesion to collagen fibrils. Kv1.3−/- mice displayed no significant difference in thrombus formation within cremaster muscle arterioles using a laser-induced injury model, thus other pro-thrombotic pathways compensate in vivo for the adhesion defect observed in vitro. This may include the increased platelet counts of Kv1.3−/- mice, due in part to a prolonged lifespan. The ability of Kv1.3 to modulate integrin-dependent platelet adhesion has important implications for understanding its contribution to normal physiological platelet function in addition to its reported roles in auto-immune diseases and thromboinflammatory models of stroke.
- Published
- 2021
4. Non-genomic effects of the Pregnane X Receptor negatively regulate platelet functions, thrombosis and haemostasis
- Author
-
Flora, GD, Sahli, KA, Sasikumar, P, Holbrook, LM, Stainer, AR, AlOuda, SK, Crescente, M, Sage, T, Unsworth, AJ, Gibbins, JM, Flora, GD, Sahli, KA, Sasikumar, P, Holbrook, LM, Stainer, AR, AlOuda, SK, Crescente, M, Sage, T, Unsworth, AJ, and Gibbins, JM
- Abstract
© 2019, The Author(s). The pregnane X receptor (PXR) is a nuclear receptor (NR), involved in the detoxification of xenobiotic compounds. Recently, its presence was reported in the human vasculature and its ligands were proposed to exhibit anti-atherosclerotic effects. Since platelets contribute towards the development of atherosclerosis and possess numerous NRs, we investigated the expression of PXR in platelets along with the ability of its ligands to modulate platelet activation. The expression of PXR in human platelets was confirmed using immunoprecipitation analysis. Treatment with PXR ligands was found to inhibit platelet functions stimulated by a range of agonists, with platelet aggregation, granule secretion, adhesion and spreading on fibrinogen all attenuated along with a reduction in thrombus formation (both in vitro and in vivo). The effects of PXR ligands were observed in a species-specific manner, and the human-specific ligand, SR12813, was observed to attenuate thrombus formation in vivo in humanised PXR transgenic mice. PXR ligand-mediated inhibition of platelet function was found to be associated with the inhibition of Src-family kinases (SFKs). This study identifies acute, non-genomic regulatory effects of PXR ligands on platelet function and thrombus formation. In combination with the emerging anti-atherosclerotic properties of PXR ligands, these anti-thrombotic effects may provide additional cardio-protective benefits.
- Published
- 2019
5. The chaperone protein HSP47: a platelet collagen binding protein that contributes to thrombosis and hemostasis.
- Author
-
Sasikumar, P, AlOuda, KS, Kaiser, WJ, Holbrook, LM, Kriek, N, Unsworth, AJ, Bye, AP, Sage, T, Ushioda, R, Nagata, K, Farndale, RW, Gibbins, JM, Sasikumar, P, AlOuda, KS, Kaiser, WJ, Holbrook, LM, Kriek, N, Unsworth, AJ, Bye, AP, Sage, T, Ushioda, R, Nagata, K, Farndale, RW, and Gibbins, JM
- Abstract
Essentials Heat shock protein 47 (HSP47), a collagen specific chaperone is present on the platelet surface. Collagen mediated platelet function was reduced following blockade or deletion of HSP47. GPVI receptor regulated signalling was reduced in HSP47 deficient platelets. Platelet HSP47 tethers to exposed collagen thus modulating thrombosis and hemostasis.Objective Heat shock protein 47 (HSP47) is an intracellular chaperone protein that is vital for collagen biosynthesis in collagen secreting cells. This protein has also been shown to be present on the surface of platelets. Given the importance of collagen and its interactions with platelets in triggering hemostasis and thrombosis, in this study we sought to characterize the role of HSP47 in these cells. Methods and Results The deletion of HSP47 in mouse platelets or its inhibition in human platelets reduced their function in response to collagen and the GPVI agonist (CRP-XL), but responses to thrombin were unaltered. In the absence of functional HSP47, the interaction of collagen with platelets was reduced, and this was associated with reduced GPVI-collagen binding, signalling and platelet activation. Thrombus formation on collagen, under arterial flow conditions, was also decreased following the inhibition or deletion of HSP47, in the presence or absence of eptifibatide, consistent with a role for HSP47 in enhancing platelet adhesion to collagen. Platelet adhesion under flow to von Willebrand factor was unaltered following HSP47 inhibition. Laser-induced thrombosis in cremaster muscle arterioles was reduced and bleeding time was prolonged in HSP47-deficient mice or following inhibition of HSP47. Conclusions Our study demonstrates the presence of HSP47 on the platelet surface, where it interacts with collagen, stabilizes platelet adhesion and increases collagen-mediated signalling and therefore thrombus formation and hemostasis.
- Published
- 2018
6. PPARγ agonists negatively regulate αIIbβ3 integrin outside-in signaling and platelet function through up-regulation of protein kinase A activity
- Author
-
Unsworth, AJ, Kriek, N, Bye, AP, Naran, K, Sage, T, Flora, GD, and Gibbins, JM
- Abstract
Essentials peroxisome proliferator-activated receptor γ (PPARγ) agonists inhibit platelet function. PPARγ agonists negatively regulate outside-in signaling via integrin αIIbβ3. PPARγ agonists disrupt the interaction of Gα13 with integrin β3. This is attributed to an upregulation of protein kinase A activity.Background Agonists for the peroxisome proliferator-activated receptor (PPARγ) have been shown to have inhibitory effects on platelet activity following stimulation by GPVI and GPCR agonists. Objectives Profound effects on thrombus formation led us to suspect a role for PPARγ agonists in the regulation of integrin αIIbβ3 mediated signaling. Both GPVI and GPCR signaling pathways lead to αIIbβ3 activation, and signaling through αIIbβ3 plays a critical role in platelet function and normal hemostasis. Methods The effects of PPARγ agonists on the regulation of αIIbβ3 outside-in signaling was determined by monitoring the ability of platelets to adhere and spread on fibrinogen and undergo clot retraction. Effects on signaling components downstream of αIIbβ3 activation were also determined following adhesion to fibrinogen by Western blotting. Results Treatment of platelets with PPARγ agonists inhibited platelet adhesion and spreading on fibrinogen and diminished clot retraction. A reduction in phosphorylation of several components of αIIbβ3 signaling, including the integrin β3 subunit, Syk, PLCγ2, focal adhesion kinase (FAK) and Akt, was also observed as a result of reduced interaction of the integrin β3 subunit with Gα13. Studies of VASP phosphorylation revealed that this was because of an increase in PKA activity following treatment with PPARγ receptor agonists. Conclusions This study provides further evidence for antiplatelet actions of PPARγ agonists, identifies a negative regulatory role for PPARγ agonists in the control of integrin αIIbβ3 outside-in signaling, and provides a molecular basis by which the PPARγ agonists negatively regulate platelet activation and thrombus formation.
- Published
- 2017
7. Platelet-Derived Inhibitors of Platelet Activation
- Author
-
Gresele, P, Kleiman, NS, Lopez, JA, Page, CP, Unsworth, AJ, Bye, AP, Gibbins, JM, Gresele, P, Kleiman, NS, Lopez, JA, Page, CP, Unsworth, AJ, Bye, AP, and Gibbins, JM
- Abstract
When blood vessels are damaged, circulating platelets come into contact with activating stimuli that trigger aggregation and enable them to form a haemostatic plug. This process is subject to both positive and negative feedback to ensure that platelets respond appropriately to damage and do not form thrombi that totally occlude the vessel. Dysregulation of negative feedback mechanisms is believed to contribute to the increased risk of thrombosis associated with some diseases. Despite the association with thrombosis, platelet derived negative regulators of platelet activation are relatively poorly understood in comparison to mediators of platelet activation. However, it is increasingly apparent that the mechanisms by which platelets restrain activation are diverse and of equal complexity to those that mediate positive signalling. Some regulators, such as RASA3 and JAM-A, act as gatekeepers that must be deactivated for platelet activation to occur. In contrast, regulators that contain ITIMs, such as PECAM-1, are activated following stimulation and mediate negative regulation via phosphatases that restrain activation. Wnt3a and ESAM are thought to directly limit plateletplatelet adhesion by blocking activation of the fibrinogen receptor, integrin αIIbβ3. The various isoforms of PKC expressed by platelets provide a diverse and complex array of inhibitory effects including receptor desensitisation. Many platelet derived inhibitors have been identified but not fully characterised and so questions remain regarding the mechanisms that underlie their effects on platelet activity following their activation, inhibition or genetic disruption. In this chapter the current understanding and recent developments in the field of platelet-derived inhibitors of platelet activation will be discussed.
- Published
- 2017
8. Mass spectrometry of platelet lipid raft fractions reveals a substantial pool of active integrin aIIb beta 3
- Author
-
Jones, CI, Tucker, KL, Barrett, EN, Wen, Y, Leake, DS, and Gibbins, JM
- Published
- 2016
9. A Role for Peripheral Tachykinins and the Neurokinin 1 Receptor in the Regulation of Platelet Thrombus Formation
- Author
-
Jones, S, Kaiser, WJ, Tucker, KL, Sage, T, Barrett, NE, Lowry, PJ, Zimmer, A, Hunt, SP, Emerson, M, and Gibbins, JM
- Published
- 2016
10. Syncytiotrophoblast Extracellular Vesicles from Pre-Eclampsia Placentas Differentially Affect Platelet Function
- Author
-
Tannetta, DS, Hunt, K, Jones, CI, Davidson, N, Coxon, CH, Ferguson, D, Redman, CW, Gibbins, JM, Sargent, IL, Tucker, KL, Oudejans, C, and Oudejans, C
- Subjects
Adult ,Blood Platelets ,medicine.medical_specialty ,Platelet Aggregation ,Placenta ,lcsh:Medicine ,030204 cardiovascular system & hematology ,Biology ,Systemic inflammation ,03 medical and health sciences ,Extracellular Vesicles ,0302 clinical medicine ,Syncytiotrophoblast ,Microscopy, Electron, Transmission ,Pre-Eclampsia ,Pregnancy ,Internal medicine ,medicine ,Humans ,Platelet ,Platelet activation ,lcsh:Science ,030304 developmental biology ,0303 health sciences ,Aspirin ,Multidisciplinary ,Eclampsia ,lcsh:R ,Thrombosis ,medicine.disease ,Platelet Activation ,Pathophysiology ,3. Good health ,Trophoblasts ,Endocrinology ,medicine.anatomical_structure ,Case-Control Studies ,embryonic structures ,lcsh:Q ,Female ,medicine.symptom ,medicine.drug ,Research Article - Abstract
Pre-eclampsia (PE) complicates around 3% of all pregnancies and is one of the most common causes of maternal mortality worldwide. The pathophysiology of PE remains unclear however its underlying cause originates from the placenta and manifests as raised blood pressure, proteinuria, vascular or systemic inflammation and hypercoagulation in the mother. Women who develop PE are also at significantly higher risk of subsequently developing cardiovascular (CV) disease. In PE, the failing endoplasmic reticulum, oxidative and inflammatory stressed syncytiotrophoblast layer of the placenta sheds increased numbers of syncytiotrophoblast extracellular vesicles (STBEV) into the maternal circulation. Platelet reactivity, size and concentration are also known to be altered in some women who develop PE, although the underlying reasons for this have not been determined. In this study we show that STBEV from disease free placenta isolated ex vivo by dual placental perfusion associate rapidly with platelets. We provide evidence that STBEV isolated from normal placentas cause platelet activation and that this is increased with STBEV from PE pregnancies. Furthermore, treatment of platelets with aspirin, currently prescribed for women at high risk of PE to reduce platelet aggregation, also inhibits STBEV-induced reversible aggregation of washed platelets. Increased platelet reactivity as a result of exposure to PE placenta derived STBEVs correlates with increased thrombotic risk associated with PE. These observations establish a possible direct link between the clotting disturbances of PE and dysfunction of the placenta, as well as the known increased risk of thromboembolism associated with this condition.
- Published
- 2015
11. Platelet signaling: a complex interplay between inhibitory and activatory networks
- Author
-
Bye, AP, Unsworth, AJ, Gibbins, JM, Bye, AP, Unsworth, AJ, and Gibbins, JM
- Published
- 2016
12. THE RELATIONSHIP BETWEEN THE METABOLITES OF THE DIETARY FLAVONOID QUERCETIN AND HAEMOSTASIS, THROMBOSIS AND PLATELET FUNCTION
- Author
-
Stainer, AR, primary, Lovegrove, JA, additional, and Gibbins, JM, additional
- Published
- 2014
- Full Text
- View/download PDF
13. PDI AND ERP57 CO-CLUSTER IN PLATELETS AND THEIR MOVEMENT IS REGULATED BY ACTIN POLYMERIZATION
- Author
-
Crescente, M, primary, Pluthero, FG, additional, Schenk, MP, additional, Ali, MS, additional, Kahr, WHA, additional, and Gibbins, JM, additional
- Published
- 2014
- Full Text
- View/download PDF
14. GAP JUNCTIONS AND CONNEXIN HEMICHANNELS UNDERPIN HAEMOSTASIS AND THROMBOSIS
- Author
-
Vaiyapuri, S, primary, Jones, CI, additional, Sasikumar, P, additional, Moraes, LA, additional, Ali, MS, additional, Sage, T, additional, Simon, AM, additional, Mahaut-Smith, M, additional, and Gibbins, JM, additional
- Published
- 2014
- Full Text
- View/download PDF
15. Ingestion of onion soup high in quercetin inhibits platelet aggregation and essential components of the collagen-stimulated platelet activation pathway in man: a pilot study.
- Author
-
Hubbard GP, Wolffram S, de Vos R, Bovy A, Gibbins JM, and Lovegrove JA
- Published
- 2006
- Full Text
- View/download PDF
16. The role of polyphenolic compounds in the diet as inhibitors of platelet function.
- Author
-
Hubbard GP, Wolffram S, Lovegrove JA, Gibbins JM, Hubbard, Gary P, Wolffram, Siegfried, Lovegrove, Julie A, and Gibbins, Jonathan M
- Abstract
Platelets play a substantial role in cardiovascular disease, and for many years there has been a search for dietary componentsthat are able to inhibit platelet function and therefore decrease the risk of cardiovasculardisease. Platelets can be inhibited by alcohol, dietary fats and some antioxidants, including agroup of compounds, the polyphenols, found in fruits and vegetables. A number of these compounds have been shown to inhibit platelet function both in vitro and in vivo. In the present study the effects of the hydroxycinnamates and the flavonoid quercetin on platelet activation and cell signalling in vitro were investigated. The hydroxycinnamates inhibited platelet function, although not at levels that can be achieved in human plasma by dietary intervention. However, quercetin inhibited platelet aggregation at levels lower than those previously reported. Quercetin was also found to inhibit intracellular Ca mobilisation and whole-cell tyrosine protein phosphorylation in platelets, which are both processes essential for platelet activation. The effect of polyphenols on platelet aggregation in vivo was also investigated. Twenty subjects followed a low-polyphenol diet for 3 d before and also during supplementation. All subjects were supplemented with a polyphenol-rich meal every lunchtime for 5d. Platelet aggregation and plasma flavonols were measured at baseline and after 5d of dietary supplementation. Total plasma flavonoids increased significantly after the dietary intervention period (P = 0.001). However, no significant changes in ex vivo platelet aggregation were observed. Further investigation of the effects of individual polyphenolic compoundson platelet function, both in vitro and in vivo, is required in order to elucidate their role in the relationship between diet and the risk of cardiovascular disease. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
17. Dietary n-3 polyunsaturated fatty acids alter the number, fatty acid profile and coagulatory activity of circulating and platelet-derived extracellular vesicles: a randomized, controlled crossover trial.
- Author
-
Bozbas E, Zhou R, Soyama S, Allen-Redpath K, Mitchell JL, Fisk HL, Calder PC, Jones C, Gibbins JM, Fischer R, Hester S, and Yaqoob P
- Subjects
- Humans, Male, Female, Middle Aged, Double-Blind Method, Dietary Supplements, Cardiovascular Diseases prevention & control, Adult, Fish Oils pharmacology, Fish Oils administration & dosage, Aged, Fatty Acids metabolism, Extracellular Vesicles metabolism, Fatty Acids, Omega-3 pharmacology, Cross-Over Studies, Blood Coagulation drug effects, Blood Platelets metabolism, Blood Platelets drug effects
- Abstract
Background: Extracellular vesicles (EVs) are proposed to play a role in the development of cardiovascular diseases (CVDs) and are considered emerging markers of CVDs. n-3 PUFAs are abundant in oily fish and fish oil and are reported to reduce CVD risk, but there has been little research to date examining the effects of n-3 PUFAs on the generation and function of EVs., Objectives: We aimed to investigate the effects of fish oil supplementation on the number, generation, and function of EVs in subjects with moderate risk of CVDs., Methods: A total of 40 participants with moderate risk of CVDs were supplemented with capsules containing either fish oil (1.9 g/d n-3 PUFAs) or control oil (high-oleic safflower oil) for 12 wk in a randomized, double-blind, placebo-controlled crossover intervention study. The effects of fish oil supplementation on conventional CVD and thrombogenic risk markers were measured, along with the number and fatty acid composition of circulating and platelet-derived EVs (PDEVs). PDEV proteome profiles were evaluated, and their impact on coagulation was assessed using assays including fibrin clot formation, thrombin generation, fibrinolysis, and ex vivo thrombus formation., Results: n-3 PUFAs decreased the numbers of circulating EVs by 27%, doubled their n-3 PUFA content, and reduced their capacity to support thrombin generation by >20% in subjects at moderate risk of CVDs. EVs derived from n-3 PUFA-enriched platelets in vitro also resulted in lower thrombin generation, but did not alter thrombus formation in a whole blood ex vivo assay., Conclusions: Dietary n-3 PUFAs alter the number, composition, and function of EVs, reducing their coagulatory activity. This study provides clear evidence that EVs support thrombin generation and that this EV-dependent thrombin generation is reduced by n-3 PUFAs, which has implications for prevention and treatment of thrombosis., Clinical Trial Registry: This trial was registered at clinicaltrials.gov as NCT03203512., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Endothelium-mediated regulation of platelet activation: Involvement of multiple protein kinases.
- Author
-
Provenzale I, Solari FA, Schönichen C, Brouns SLN, Fernández DI, Kuijpers MJE, van der Meijden PEJ, Gibbins JM, Sickmann A, Jones C, and Heemskerk JWM
- Subjects
- Humans, Protein Kinases metabolism, Nitric Oxide metabolism, Endothelial Cells metabolism, Platelet Activation physiology, Blood Platelets metabolism, Endothelium metabolism, Prostaglandins I, Platelet Membrane Glycoproteins metabolism, Thrombin metabolism
- Abstract
The endothelial regulation of platelet activity is incompletely understood. Here we describe novel approaches to find molecular pathways implicated on the platelet-endothelium interaction. Using high-shear whole-blood microfluidics, employing coagulant or non-coagulant conditions at physiological temperature, we observed that the presence of human umbilical vein endothelial cells (HUVEC) strongly suppressed platelet adhesion and activation, via the collagen receptor glycoprotein VI (GPVI) and the PAR receptors for thrombin. Real-time monitoring of the cytosolic Ca
2+ rises in the platelets indicated no major improvement of inhibition by prostacyclin or nitric oxide. Similarly under stasis, exposure of isolated platelets to HUVEC reduced the Ca2+ responses by collagen-related peptide (CRP-XL, GPVI agonist) and thrombin (PAR agonist). We then analyzed the label-free phosphoproteome of platelets (three donors), exposed to HUVEC, CRP-XL, and/or thrombin. High-resolution mass spectrometry gave 5463 phosphopeptides, corresponding to 1472 proteins, with good correlation between biological and technical replicates (R > .86). Stringent filtering steps revealed 26 regulatory pathways (Reactome) and 143 regulated kinase substrates (PhosphoSitePlus), giving a set of protein phosphorylation sites that was differentially (44) or similarly (110) regulated by HUVEC or agonist exposure. The differential regulation was confirmed by stable-isotope analysis of platelets from two additional donors. Substrate analysis indicated major roles of poorly studied protein kinase classes (MAPK, CDK, DYRK, STK, PKC members). Collectively, these results reveal a resetting of the protein phosphorylation profile in platelets exposed to endothelium or to conventional agonists and to endothelium-promoted activity of a multi-kinase network, beyond classical prostacyclin and nitric oxide actors, that may contribute to platelet inhibition., (© 2024 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)- Published
- 2024
- Full Text
- View/download PDF
19. Evidence that inositol 1,4,5-trisphosphate 3-kinase and inositol 1,3,4,5-tetrakisphosphate are negative regulators of platelet function.
- Author
-
Authi KS, Khan S, Gibbins JM, and Brain SD
- Abstract
Background: Inositol 1,3,4,5-tetrakisphosphate (IP
4 ) is formed from inositol 1,4,5-trisphosphate (IP3 ) by IP3 3-kinase (ITPK) in most cells. Its function is unknown but has been suggested to be involved in Ca2+ entry, IP3 regulation, and phosphoinositide 3-kinase antagonism., Objectives: To better elucidate a function for IP4 , we tested a specific inhibitor of ITPK (GNF362) on platelets, the effects of IP4 directly in permeabilized platelets and its effect on phosphatidylinositol 3,4,5-trisphosphate (PIP3 ) binding to pleckstrin-homology (PH) domain-containing proteins in platelets., Methods: Human platelets were utilized in whole blood for thrombus formation, in platelet-rich plasma and washed suspensions for aggregation, and for Ca2+ studies, or resuspended in high K+ and low Na+ buffers for permeabilization experiments. Phosphorylation of AKT-Ser473 and Rap1-GTP formation were measured by Western blotting and PIP3 binding using PIP3 beads., Results: GNF362-enhanced platelet aggregation stimulated by low concentrations of ADP, collagen, thrombin, U46619, and thrombus formation in collagen-coated capillaries. GNF362 induced a transient elevation of Ca2+ concentration, elevated basal levels of IP3 , and enhanced the peak height of Ca2+ elevated by agonists. In permeabilized platelets, IP4 inhibited GTPγS induced formation of AKT-Ser473 phosphorylation and platelet aggregation. IP4 reduced GTPγS-stimulated Rap1-GTP levels and potently reduced extraction of RASA3 and BTK by PIP3 beads., Conclusion: ITPK and IP4 are negative regulators of platelet function. IP4 regulation of PH domain-containing proteins may represent a pathway by which platelet activation may be controlled during thrombosis., (© 2024 The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
20. Multiple myeloma and its treatment contribute to increased platelet reactivity.
- Author
-
Mitchell JL, Khan D, Rana RH, Kriek N, Unsworth AJ, Sage T, Bye AP, Laffan M, Shapiro S, Thakurta A, Grech H, Ramasamy K, and Gibbins JM
- Subjects
- Humans, Lenalidomide pharmacology, Lenalidomide therapeutic use, Pilot Projects, Multiple Myeloma drug therapy, Multiple Myeloma complications, Thrombosis complications, Monoclonal Gammopathy of Undetermined Significance complications
- Abstract
Multiple myeloma (MM) and its precursor states, smoldering myeloma (SM) and monoclonal gammopathy of undetermined significance (MGUS) are associated with increased incidence of thrombosis, however the cause of this is unknown. Lenalidomide treatment of MM substantially improves patient survival, although significantly increases thrombotic risk by an unknown mechanism. This pilot study aimed to establish the impact of MM and its treatment with Lenalidomide on platelet function. We analyzed platelet function in MGUS, SM and MM compared to healthy controls. We report an increase in platelet reactivity in MGUS, SM, and MM where increases in fibrinogen binding, P-selectin exposure, altered receptor expression, elevated levels of aggregation and enhanced sensitivity to agonist stimulation were observed. We also demonstrate an increase in patient platelet reactivity post Lenalidomide treatment compared to pre-treatment. We show Lenalidomide treatment of platelets ex vivo increased reactivity that was associated with formation of larger thrombi at arterial shear rates but not venous shear rates. This study demonstrates a clear increase in platelet reactivity and prothrombotic potential in patients with MGUS, SM and MM which is elevated further upon treatment with Lenalidomide. Our observations suggest that more detailed studies are warranted to determine mechanisms of thrombotic complications to enable the development of new preventative strategies that specifically target platelets.
- Published
- 2023
- Full Text
- View/download PDF
21. Identification of new drugs to counteract anti-spike IgG-induced hyperinflammation in severe COVID-19.
- Author
-
Geyer CE, Chen HJ, Bye AP, Manz XD, Guerra D, Caniels TG, Bijl TP, Griffith GR, Hoepel W, de Taeye SW, Veth J, Vlaar AP, Vidarsson G, Bogaard HJ, Aman J, Gibbins JM, van Gils MJ, de Winther MP, and den Dunnen J
- Subjects
- Humans, SARS-CoV-2, Antibodies, Viral, Inflammation drug therapy, Immunoglobulin G pharmacology, COVID-19
- Abstract
Previously, we and others have shown that SARS-CoV-2 spike-specific IgG antibodies play a major role in disease severity in COVID-19 by triggering macrophage hyperactivation, disrupting endothelial barrier integrity, and inducing thrombus formation. This hyperinflammation is dependent on high levels of anti-spike IgG with aberrant Fc tail glycosylation, leading to Fcγ receptor hyperactivation. For development of immune-regulatory therapeutics, drug specificity is crucial to counteract excessive inflammation whereas simultaneously minimizing the inhibition of antiviral immunity. We here developed an in vitro activation assay to screen for small molecule drugs that specifically counteract antibody-induced pathology. We identified that anti-spike-induced inflammation is specifically blocked by small molecule inhibitors against SYK and PI3K. We identified SYK inhibitor entospletinib as the most promising candidate drug, which also counteracted anti-spike-induced endothelial dysfunction and thrombus formation. Moreover, entospletinib blocked inflammation by different SARS-CoV-2 variants of concern. Combined, these data identify entospletinib as a promising treatment for severe COVID-19., (© 2023 Geyer et al.)
- Published
- 2023
- Full Text
- View/download PDF
22. Regulation of Glycoprotein VI-Dependent Platelet Activation and Thrombus Formation by Heparan Sulfate Proteoglycan Perlecan.
- Author
-
Provenzale I, De Simone I, Gibbins JM, Heemskerk JWM, van der Meijden PEJ, and Jones CI
- Subjects
- Humans, Extracellular Matrix Proteins, Platelet Adhesiveness, Heparan Sulfate Proteoglycans, Thrombosis
- Abstract
Proteoglycans form a heterogeneous family of proteins with covalently bound sulfated glycosaminoglycans. The extracellular matrix proteoglycan perlecan has been proposed to bind to the platelet- and megakaryocyte-specific receptor G6bB, co-regulating platelet glycoprotein VI (GPVI) signaling. The derived non-sulfate proteoglycan endorepellin was previously shown to enhance platelet adhesion via the collagen receptor, integrin α2β1. Here, we compared the roles of perlecan and other matrix proteoglycans in platelet responses and thrombus formation. We used multi-color flow cytometry to measure the degranulation and integrin αIIbβ3 activation of washed platelets in response to various proteoglycans and collagen-related peptide (CRP), the GPVI agonist. Perlecan, but not endorepellin, enhanced the CRP-induced activation of platelets in a time- and concentration-dependent manner. Similar to collagen, immobilized perlecan, but not other proteoglycans, supported static platelet adhesion and spreading. In-flowed whole-blood perlecan diminished shear-dependent platelet adhesion, while it enforced GPVI-dependent thrombus formation-to a larger extent than endorepellin-to induce more contracted aggregates of activated platelets. We concluded that the sulfated proteoglycan perlecan enhances GPVI-dependent platelet responses extending to thrombus formation, but it does so at the expense of reduced adhesion of platelets under flow.
- Published
- 2023
- Full Text
- View/download PDF
23. Role of heat shock protein 47 in platelet glycoprotein VI dimerization and signaling.
- Author
-
AlOuda SK, Sasikumar P, AlThunayan T, Alaajam F, Khan S, Sahli KA, Abohassan MS, Pollitt A, Jung SM, and Gibbins JM
- Abstract
Background: Heat shock protein 47 (HSP47) is an intracellular chaperone protein with an indispensable role in collagen biosynthesis in collagen-secreting cells. This chaperone has also been shown to be released and present on the surface of platelets. The inhibition of HSP47 in human platelets or its ablation in mouse platelets reduces platelet function in response to collagen and the glycoprotein (GP) VI collagen receptor agonist CRP-XL., Objectives: In this study, we sought, through experiments, to explore cellular distribution, trafficking, and influence on GPVI interactions to understand how HSP47 modulates collagen receptor signaling., Methods: HSP47-deficient mouse platelets and SMIH- treated human platelets were used to study the role of HSP47 in collagen mediated responses and signaling., Results: Using subcellular fractionation analysis and immunofluorescence microscopy, HSP47 was found to be localized to the platelet-dense tubular system. Following platelet stimulation, HSP47 mobilization to the cell surface was shown to be dependent on actin polymerization, a feature common to other dense tubular system resident platelet proteins that are released to the cell surface during activation. In this location, HSP47 was found to contribute to platelet adhesion to collagen or CRP-XL but not to GFOGER peptide (an integrin α2β1-binding sequence within collagens), indicating selective effects of HSP47 on GPVI function. Dimerization of GPVI on the platelet surface increases its affinity for collagen. GPVI dimerization was reduced following HSP47 inhibition, as was collagen and CRP-XL-mediated signaling., Conclusion: The present study identifies a role for cell surface-localized HSP47 in modulating platelet responses to collagen through dimerization of GPVI, thereby enhancing platelet signaling and activation., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
24. The rate of platelet activation determines thrombus size and structure at arterial shear.
- Author
-
Mitchell JL, Dunster JL, Kriek N, Unsworth AJ, Sage T, Mohammed YMM, De Simone I, Taylor KA, Bye AP, Ólafsson G, Brunton M, Mark S, Dymott LD, Whyte A, Ruparelia N, Mckenna C, Gibbins JM, and Jones CI
- Subjects
- Humans, Blood Platelets metabolism, Platelet Function Tests, Arteries, Platelet Aggregation, Platelet Activation, Thrombosis metabolism
- Abstract
Background: The response of platelets to activating stimuli and pharmaceutical agents varies greatly within the normal population. Current platelet function tests are used to measure end-point levels of platelet activation without taking the speed at which platelets activate into account, potentially missing vital metrics to characterize platelet reactivity., Objectives: To identify variability, to agonists and among individuals, in platelet activation kinetics and assess the impact of this on thrombus formation., Methods: We have developed a bespoke real-time flow cytometry assay and analysis package to measure the rate of platelet activation over time using 2 parameters of platelet activation, fibrinogen binding and P-selectin exposure., Results: The rate of platelet activation varied considerably within the normal population but did not correlate with maximal platelet activation, demonstrating that platelet activation rate is a separate and novel metric to describe platelet reactivity. The relative rate of platelet response between agonists was strongly correlated, suggesting that a central control mechanism regulates the rate of platelet response to all agonists., Conclusion: For the first time, we have shown that platelet response rate corresponds to thrombus size and structure, wherein faster responders form larger, more densely packed thrombi at arterial, but crucially not venous, shear. We have demonstrated that the rate of platelet activation is an important metric in stratifying individual platelet responses and will provide a novel focus for the design and development of antiplatelet therapy, targeting high-shear thrombosis without exacerbating bleeding at low shear., Competing Interests: Funding information British Heart Foundation; Grants: PG/16/36/31967 (C.I.J. and J.M.G.) RG/20/7/34866 and RG/15/2/31224 (J.M.G and C.I.J.). European Union's Horizon 2020 research and innovation program under the Marie Sklodowska-Curie grant agreement No. 766118 (IDS). Declaration of competing interests There are no competing interests to disclose., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. Delineating Zinc Influx Mechanisms during Platelet Activation.
- Author
-
Kuravi SJ, Ahmed NS, Taylor KA, Capes EM, Bye A, Unsworth AJ, Gibbins JM, and Pugh N
- Subjects
- Zinc pharmacology, Zinc metabolism, Endoplasmic Reticulum metabolism, Platelet Activation, Blood Platelets metabolism, Cations metabolism, Calcium metabolism, Cation Transport Proteins metabolism
- Abstract
Zinc (Zn
2+ ) is released by platelets during a hemostatic response to injury. Extracellular zinc ([Zn2+ ]o ) initiates platelet activation following influx into the platelet cytosol. However, the mechanisms that permit Zn2+ influx are unknown. Fluctuations in intracellular zinc ([Zn2+ ]i ) were measured in fluozin-3-loaded platelets using fluorometry and flow cytometry. Platelet activation was assessed using light transmission aggregometry. The detection of phosphoproteins was performed by Western blotting. [Zn2+ ]o influx and subsequent platelet activation were abrogated by blocking the sodium/calcium exchanged, TRP channels, and ZIP7. Cation store depletion regulated Zn2+ influx. [Zn2+ ]o stimulation resulted in the phosphorylation of PKC substates, MLC, and β3 integrin. Platelet activation via GPVI or Zn2+ resulted in ZIP7 phosphorylation in a casein kinase 2-dependent manner and initiated elevations of [Zn2+ ]i that were sensitive to the inhibition of Orai1, ZIP7, or IP3 R-mediated pathways. These data indicate that platelets detect and respond to changes in [Zn2+ ]o via influx into the cytosol through TRP channels and the NCX exchanger. Platelet activation results in the externalization of ZIP7, which further regulates Zn2+ influx. Increases in [Zn2+ ]i contribute to the activation of cation-dependent enzymes. Sensitivity of Zn2+ influx to thapsigargin indicates a store-operated pathway that we term store-operated Zn2+ entry (SOZE). These mechanisms may affect platelet behavior during thrombosis and hemostasis.- Published
- 2023
- Full Text
- View/download PDF
26. Adding fuel to the flames in preeclampsia: the platelet connection.
- Author
-
Gibbins JM
- Subjects
- Pregnancy, Female, Humans, Blood Platelets, Pre-Eclampsia
- Published
- 2023
- Full Text
- View/download PDF
27. Developing Biomimetic Hydrogels of the Arterial Wall as a Prothrombotic Substrate for In Vitro Human Thrombosis Models.
- Author
-
Ranjbar J, Njoroge W, Gibbins JM, Roach P, Yang Y, and Harper AGS
- Abstract
Current in vitro thrombosis models utilise simplistic 2D surfaces coated with purified components of the subendothelial matrix. The lack of a realistic humanised model has led to greater study of thrombus formation in in vivo tests in animals. Here we aimed to develop 3D hydrogel-based replicas of the medial and adventitial layers of the human artery to produce a surface that can optimally support thrombus formation under physiological flow conditions. These tissue-engineered medial- (TEML) and adventitial-layer (TEAL) hydrogels were developed by culturing human coronary artery smooth muscle cells and human aortic adventitial fibroblasts within collagen hydrogels, both individually and in co-culture. Platelet aggregation upon these hydrogels was studied using a custom-made parallel flow chamber. When cultured in the presence of ascorbic acid, the medial-layer hydrogels were able to produce sufficient neo-collagen to support effective platelet aggregation under arterial flow conditions. Both TEML and TEAL hydrogels possessed measurable tissue factor activity and could trigger coagulation of platelet-poor plasma in a factor VII-dependent manner. Biomimetic hydrogel replicas of the subendothelial layers of the human artery are effective substrates for a humanised in vitro thrombosis model that could reduce animal experimentation by replacing current in vivo models.
- Published
- 2023
- Full Text
- View/download PDF
28. Platelet factor XIII-A regulates platelet function and promotes clot retraction and stability.
- Author
-
Mitchell JL, Little G, Bye AP, Gaspar RS, Unsworth AJ, Kriek N, Sage T, Stainer A, Sangowawa I, Morrow GB, Bastos RN, Shapiro S, Desborough MJR, Curry N, Gibbins JM, Whyte CS, Mutch NJ, and Jones CI
- Abstract
Background: Factor XIII (FXIII) is an important proenzyme in the hemostatic system. The plasma-derived enzyme activated FXIII cross-links fibrin fibers within thrombi to increase their mechanical strength and cross-links fibrin to fibrinolytic inhibitors, specifically α
2 -antiplasmin, to increase resistance to fibrinolysis. We have previously shown that cellular FXIII (factor XIII-A [FXIII-A]), which is abundant in the platelet cytoplasm, is externalized onto the activated membrane and cross-links extracellular substrates. The contribution of cellular FXIII-A to platelet activation and platelet function has not been extensively studied., Objectives: This study aims to identify the role of platelet FXIII-A in platelet function., Methods: We used normal healthy platelets with a cell permeable FXIII inhibitor and platelets from FXIII-deficient patients as a FXIII-free platelet model in a range of platelet function and clotting tests., Results: Our data demonstrate that platelet FXIII-A enhances fibrinogen binding to the platelet surface upon agonist stimulation and improves the binding of platelets to fibrinogen and aggregation under flow in a whole-blood thrombus formation assay. In the absence of FXIII-A, platelets show reduced sensitivity to agonist stimulation, including decreased P-selectin exposure and fibrinogen binding. We show that FXIII-A is involved in platelet spreading where a lack of FXIII-A reduces the ability of platelets to fully spread on fibrinogen and collagen. Our data demonstrate that platelet FXIII-A is important for clot retraction where clots formed in its absence retracted to a lesser extent., Conclusion: Overall, this study shows that platelet FXIII-A functions during thrombus formation by aiding platelet activation and thrombus retraction in addition to its antifibrinolytic roles., (© 2023 The Authors.)- Published
- 2023
- Full Text
- View/download PDF
29. Pirtobrutinib results in reversible platelet dysfunction compared to ibrutinib and acalabrutinib.
- Author
-
Bye AP, Kriek N, Sage T, Rawlings SJ, Prodger C, Kesavan M, Lees C, Booth S, Cowen LG, Shefferd K, Desborough MJ, Gibbins JM, and Eyre TA
- Subjects
- Humans, Benzamides, Protein Kinase Inhibitors, Pyrazines, Leukemia, Lymphocytic, Chronic, B-Cell
- Published
- 2023
- Full Text
- View/download PDF
30. Repeated platelet activation and the potential of previously activated platelets to contribute to thrombus formation.
- Author
-
De Simone I, Baaten CCFMJ, Gibbins JM, Ten Cate H, Heemskerk JWM, Jones CI, and van der Meijden PEJ
- Subjects
- Humans, Thrombin metabolism, Platelet Activation, Blood Platelets metabolism, Platelet Aggregation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Receptors, Thrombin metabolism, Adenosine Diphosphate pharmacology, Adenosine Diphosphate metabolism, Platelet Membrane Glycoproteins metabolism, Thrombosis metabolism
- Abstract
Background: Especially in disease conditions, platelets can encounter activating agents in circulation., Objectives: To investigate the extent to which previously activated platelets can be reactivated and whether in-and reactivation applies to different aspects of platelet activation and thrombus formation., Methods: Short-and long-term effects of glycoprotein VI (GPVI) and G protein-coupled receptor (GPCR) stimulation on platelet activation and aggregation potential were compared via flow cytometry and plate-based aggregation. Using fluorescence and electron microscopy, we assessed platelet morphology and content, as well as thrombus formation., Results: After 30 minutes of stimulation with thrombin receptor activator peptide 6 (TRAP6) or adenosine diphosphate (ADP), platelets secondarily decreased in PAC-1 binding and were less able to aggregate. The reversibility of platelets after thrombin stimulation was concentration dependent. Reactivation was possible via another receptor. In contrast, cross-linked collagen-related peptide (CRP-XL) or high thrombin stimulation evoked persistent effects in α
IIb β3 activation and platelet aggregation. However, after 60 minutes of CRP-XL or high thrombin stimulation, when αIIb β3 activation slightly decreased, restimulation with ADP or CRP-XL, respectively, increased integrin activation again. Compatible with decreased integrin activation, platelet morphology was reversed. Interestingly, reactivation of reversed platelets again resulted in shape change and if not fully degranulated, additional secretion. Moreover, platelets that were previously activated with TRAP6 or ADP regained their potential to contribute to thrombus formation under flow. On the contrary, prior platelet triggering with CRP-XL was accompanied by prolonged platelet activity, leading to a decreased secondary platelet adhesion under flow., Conclusion: This work emphasizes that prior platelet activation can be reversed, whereafter platelets can be reactivated through a different receptor. Reversed, previously activated platelets can contribute to thrombus formation., (Crown Copyright © 2023. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
31. Immobility-associated thromboprotection is conserved across mammalian species from bear to human.
- Author
-
Thienel M, Müller-Reif JB, Zhang Z, Ehreiser V, Huth J, Shchurovska K, Kilani B, Schweizer L, Geyer PE, Zwiebel M, Novotny J, Lüsebrink E, Little G, Orban M, Nicolai L, El Nemr S, Titova A, Spannagl M, Kindberg J, Evans AL, Mach O, Vogel M, Tiedt S, Ormanns S, Kessler B, Dueck A, Friebe A, Jørgensen PG, Majzoub-Altweck M, Blutke A, Polzin A, Stark K, Kääb S, Maier D, Gibbins JM, Limper U, Frobert O, Mann M, Massberg S, and Petzold T
- Subjects
- Animals, Humans, Mice, Fibrinolytic Agents therapeutic use, Pulmonary Embolism drug therapy, Pulmonary Embolism ethnology, Pulmonary Embolism metabolism, Risk Factors, Spinal Cord Injuries complications, Ursidae metabolism, Venous Thromboembolism etiology, Venous Thromboembolism metabolism, Hypokinesia complications, HSP47 Heat-Shock Proteins metabolism, Blood Platelets metabolism
- Abstract
Venous thromboembolism (VTE) comprising deep venous thrombosis and pulmonary embolism is a major cause of morbidity and mortality. Short-term immobility-related conditions are a major risk factor for the development of VTE. Paradoxically, long-term immobilized free-ranging hibernating brown bears and paralyzed spinal cord injury (SCI) patients are protected from VTE. We aimed to identify mechanisms of immobility-associated VTE protection in a cross-species approach. Mass spectrometry-based proteomics revealed an antithrombotic signature in platelets of hibernating brown bears with heat shock protein 47 (HSP47) as the most substantially reduced protein. HSP47 down-regulation or ablation attenuated immune cell activation and neutrophil extracellular trap formation, contributing to thromboprotection in bears, SCI patients, and mice. This cross-species conserved platelet signature may give rise to antithrombotic therapeutics and prognostic markers beyond immobility-associated VTE.
- Published
- 2023
- Full Text
- View/download PDF
32. Exploring the constituent mechanisms of hepatitis: a dynamical systems approach.
- Author
-
Dunster JL, Gibbins JM, and Nelson MR
- Subjects
- Humans, Hepatocytes metabolism, Inflammation, Systems Analysis, Liver, Hepatitis metabolism
- Abstract
Hepatitis is the term used to describe inflammation in the liver. It is associated with a high rate of mortality, but the underlying disease mechanisms are not completely understood and treatment options are limited. We present a mathematical model of hepatitis that captures the complex interactions between hepatocytes (liver cells), hepatic stellate cells (cells in the liver that produce hepatitis-associated fibrosis) and the immune components that mediate inflammation. The model is in the form of a system of ordinary differential equations. We use numerical techniques and bifurcation analysis to characterize and elucidate the physiological mechanisms that dominate liver injury and its outcome to a healthy or unhealthy, chronic state. This study reveals the complex interactions between the multiple cell types and mediators involved in this complex disease and highlights potential problems in targeting inflammation in the liver therapeutically., (© The Author(s) 2019. Published by Oxford University Press on behalf of the Institute of Mathematics and its Applications.)
- Published
- 2023
- Full Text
- View/download PDF
33. An agent-based approach for modelling and simulation of glycoprotein VI receptor diffusion, localisation and dimerisation in platelet lipid rafts.
- Author
-
Tantiwong C, Dunster JL, Cavill R, Tomlinson MG, Wierling C, Heemskerk JWM, and Gibbins JM
- Subjects
- Cell Membrane metabolism, Collagen metabolism, Membrane Microdomains metabolism, Platelet Membrane Glycoproteins metabolism, Blood Platelets metabolism
- Abstract
Receptor diffusion plays an essential role in cellular signalling via the plasma membrane microenvironment and receptor interactions, but the regulation is not well understood. To aid in understanding of the key determinants of receptor diffusion and signalling, we developed agent-based models (ABMs) to explore the extent of dimerisation of the platelet- and megakaryocyte-specific receptor for collagen glycoprotein VI (GPVI). This approach assessed the importance of glycolipid enriched raft-like domains within the plasma membrane that lower receptor diffusivity. Our model simulations demonstrated that GPVI dimers preferentially concentrate in confined domains and, if diffusivity within domains is decreased relative to outside of domains, dimerisation rates are increased. While an increased amount of confined domains resulted in further dimerisation, merging of domains, which may occur upon membrane rearrangements, was without effect. Modelling of the proportion of the cell membrane which constitutes lipid rafts indicated that dimerisation levels could not be explained by these alone. Crowding of receptors by other membrane proteins was also an important determinant of GPVI dimerisation. Together, these results demonstrate the value of ABM approaches in exploring the interactions on a cell surface, guiding the experimentation for new therapeutic avenues., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
34. Activation of Human Platelets by Staphylococcus aureus Secreted Protease Staphopain A.
- Author
-
Waller AK, Birch K, Gibbins JM, and Clarke SR
- Abstract
Infection by Staphylococcus aureus is the leading cause of infective endocarditis (IE). Activation of platelets by this pathogen results in their aggregation and thrombus formation which are considered to be important steps in the development and pathogenesis of IE. Here, we show that a secreted cysteine protease, staphopain A, activates human platelets and induces their aggregation. The culture supernatant of a scpA mutant deficient in staphopain A production was reduced in its ability to trigger platelet aggregation. The platelet agonist activity of purified staphopain A was inhibited by staphostatin A, a specific inhibitor, thus implicating its protease activity in the agonism. In whole blood, using concentrations of staphopain A that were otherwise insufficient to induce platelet aggregation, increased binding to collagen and thrombus formation was observed. Using antagonists specific to protease-activated receptors 1 and 4, we demonstrate their role in mediating staphopain A induced platelet activation.
- Published
- 2022
- Full Text
- View/download PDF
35. Coagulation Factor XIIIa and Activated Protein C Activate Platelets via GPVI and PAR1.
- Author
-
De Simone I, Baaten CCFMJ, Jandrot-Perrus M, Gibbins JM, Ten Cate H, Heemskerk JWM, Jones CI, and van der Meijden PEJ
- Subjects
- Blood Platelets metabolism, Factor XIIIa metabolism, Fibrin metabolism, Hirudins metabolism, Hirudins pharmacology, Phosphatidylserines metabolism, Platelet Activation, Platelet Aggregation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Protein C metabolism, Receptor, PAR-1 metabolism, Syk Kinase metabolism, Thrombin metabolism, Thrombin pharmacology, P-Selectin metabolism, Platelet Membrane Glycoproteins metabolism
- Abstract
Platelet and coagulation activation are highly reciprocal processes driven by multi-molecular interactions. Activated platelets secrete several coagulation factors and expose phosphatidylserine, which supports the activation of coagulation factor proteins. On the other hand, the coagulation cascade generates known ligands for platelet receptors, such as thrombin and fibrin. Coagulation factor (F)Xa, (F)XIIIa and activated protein C (APC) can also bind to platelets, but the functional consequences are unclear. Here, we investigated the effects of the activated (anti)coagulation factors on platelets, other than thrombin. Multicolor flow cytometry and aggregation experiments revealed that the 'supernatant of (hirudin-treated) coagulated plasma' (SCP) enhanced CRP-XL-induced platelet responses, i.e., integrin α
IIb β3 activation, P-selectin exposure and aggregate formation. We demonstrated that FXIIIa in combination with APC enhanced platelet activation in solution, and separately immobilized FXIIIa and APC resulted in platelet spreading. Platelet activation by FXIIIa was inhibited by molecular blockade of glycoprotein VI (GPVI) or Syk kinase. In contrast, platelet spreading on immobilized APC was inhibited by PAR1 blockade. Immobilized, but not soluble, FXIIIa and APC also enhanced in vitro adhesion and aggregation under flow. In conclusion, in coagulation, factors other than thrombin or fibrin can induce platelet activation via GPVI and PAR receptors.- Published
- 2022
- Full Text
- View/download PDF
36. Cucurbitacins Elicit Anti-Platelet Activity via Perturbation of the Cytoskeleton and Integrin Function.
- Author
-
Kriek N, Nock SH, Sage T, Khalifa B, Bye AP, Mitchell JL, Thomson S, McLaughlin MG, Jones S, Gibbins JM, and Unsworth AJ
- Subjects
- Blood Platelets metabolism, Cucurbitacins metabolism, Cucurbitacins pharmacology, Cytoskeleton metabolism, Humans, Microtubules metabolism, Platelet Aggregation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Thrombosis metabolism
- Abstract
Cucurbitacins are dietary compounds that have been shown to elicit a range of anti-tumour, anti-inflammatory and anti-atherosclerotic activities. Originally identified as signal transducer and activator of transcription, STAT, inhibitors, a variety of mechanisms of action have since been described, including dysregulation of the actin cytoskeleton and disruption of integrin function. Integrin outside-in signalling and cytoskeletal rearrangements are critical for the propagation of stable thrombus formation and clot retraction following platelet adhesion at the site of vessel damage. The effects of cucurbitacins on platelet function and thrombus formation are unknown. We report for the first time anti-platelet and anti-thrombotic effects of cucurbitacins B, E and I in human platelets. Treatment of platelets with cucurbitacins resulted in attenuation of platelet aggregation, secretion and fibrinogen binding following stimulation by platelet agonists. Cucurbitacins were also found to potently inhibit other integrin- and cytoskeleton-mediated events, including adhesion, spreading and clot retraction. Further investigation of cytoskeletal dynamics found treatment with cucurbitacins altered cofilin phosphorylation, enhanced activation and increased F actin polymerisation and microtubule assembly. Disruption to cytoskeletal dynamics has been previously shown to impair integrin activation, platelet spreading and clot retraction. Anti-platelet properties of cucurbitacins were found to extend to a disruption of stable thrombus formation, with an increase in thrombi instability and de-aggregation under flow. Our research identifies novel, anti-platelet and anti-thrombotic actions of cucurbitacins that appear to be linked to dysregulation of cytoskeletal dynamics and integrin function., Competing Interests: None declared., (The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).)
- Published
- 2022
- Full Text
- View/download PDF
37. G protein-coupled receptor kinase 5 regulates thrombin signaling in platelets via PAR-1.
- Author
-
Downes K, Zhao X, Gleadall NS, McKinney H, Kempster C, Batista J, Thomas PL, Cooper M, Michael JV, Kreuzhuber R, Wedderburn K, Waller K, Varney B, Verdier H, Kriek N, Ashford SE, Stirrups KE, Dunster JL, McKenzie SE, Ouwehand WH, Gibbins JM, Yang J, Astle WJ, and Ma P
- Subjects
- Animals, Genome-Wide Association Study, Mice, Platelet Activation, Receptor, PAR-1 genetics, Receptor, PAR-1 metabolism, Blood Platelets metabolism, Thrombin metabolism, Thrombin pharmacology
- Abstract
The interindividual variation in the functional response of platelets to activation by agonists is heritable. Genome-wide association studies (GWASs) of quantitative measures of platelet function have identified fewer than 20 distinctly associated variants, some with unknown mechanisms. Here, we report GWASs of pathway-specific functional responses to agonism by adenosine 5'-diphosphate, a glycoprotein VI-specific collagen mimetic, and thrombin receptor-agonist peptides, each specific to 1 of the G protein-coupled receptors PAR-1 and PAR-4, in subsets of 1562 individuals. We identified an association (P = 2.75 × 10-40) between a common intronic variant, rs10886430, in the G protein-coupled receptor kinase 5 gene (GRK5) and the sensitivity of platelets to activate through PAR-1. The variant resides in a megakaryocyte-specific enhancer that is bound by the transcription factors GATA1 and MEIS1. The minor allele (G) is associated with fewer GRK5 transcripts in platelets and the greater sensitivity of platelets to activate through PAR-1. We show that thrombin-mediated activation of human platelets causes binding of GRK5 to PAR-1 and that deletion of the mouse homolog Grk5 enhances thrombin-induced platelet activation sensitivity and increases platelet accumulation at the site of vascular injury. This corroborates evidence that the human G allele of rs10886430 is associated with a greater risk for cardiovascular disease. In summary, by combining the results of pathway-specific GWASs and expression quantitative trait locus studies in humans with the results from platelet function studies in Grk5-/- mice, we obtain evidence that GRK5 regulates the human platelet response to thrombin via the PAR-1 pathway., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
38. The voltage-gated K + channel Kv1.3 modulates platelet motility and α 2 β 1 integrin-dependent adhesion to collagen.
- Author
-
Wright JR, Jones S, Parvathy S, Kaczmarek LK, Forsythe I, Farndale RW, Gibbins JM, and Mahaut-Smith MP
- Subjects
- Humans, Blood Platelets metabolism, Collagen metabolism, Integrin alpha2beta1 metabolism, Platelet Adhesiveness physiology, Platelet Aggregation physiology, Potassium Channels, Voltage-Gated metabolism
- Abstract
Kv1.3 is a voltage-gated K
+ -selective channel with roles in immunity, insulin-sensitivity, neuronal excitability and olfaction. Despite being one of the largest ionic conductances of the platelet surface membrane, its contribution to platelet function is poorly understood. Here we show that Kv1.3-deficient platelets display enhanced ADP-evoked platelet aggregation and secretion, and an increased surface expression of platelet integrin αIIb . In contrast, platelet adhesion and thrombus formation in vitro under arterial shear conditions on surfaces coated with collagen were reduced for samples from Kv1.3-/- compared to wild type mice. Use of collagen-mimetic peptides revealed a specific defect in the engagement with α2 β1 . Kv1.3-/- platelets developed significantly fewer, and shorter, filopodia than wild type platelets during adhesion to collagen fibrils. Kv1.3-/- mice displayed no significant difference in thrombus formation within cremaster muscle arterioles using a laser-induced injury model, thus other pro-thrombotic pathways compensate in vivo for the adhesion defect observed in vitro . This may include the increased platelet counts of Kv1.3-/- mice, due in part to a prolonged lifespan. The ability of Kv1.3 to modulate integrin-dependent platelet adhesion has important implications for understanding its contribution to normal physiological platelet function in addition to its reported roles in auto-immune diseases and thromboinflammatory models of stroke.- Published
- 2022
- Full Text
- View/download PDF
39. Fully automated platelet differential interference contrast image analysis via deep learning.
- Author
-
Kempster C, Butler G, Kuznecova E, Taylor KA, Kriek N, Little G, Sowa MA, Sage T, Johnson LJ, Gibbins JM, and Pollitt AY
- Subjects
- Image Processing, Computer-Assisted, Platelet Activation, Blood Platelets, Deep Learning
- Abstract
Platelets mediate arterial thrombosis, a leading cause of myocardial infarction and stroke. During injury, platelets adhere and spread over exposed subendothelial matrix substrates of the damaged blood vessel wall. The mechanisms which govern platelet activation and their interaction with a range of substrates are therefore regularly investigated using platelet spreading assays. These assays often use differential interference contrast (DIC) microscopy to assess platelet morphology and analysis performed using manual annotation. Here, a convolutional neural network (CNN) allowed fully automated analysis of platelet spreading assays captured by DIC microscopy. The CNN was trained using 120 generalised training images. Increasing the number of training images increases the mean average precision of the CNN. The CNN performance was compared to six manual annotators. Significant variation was observed between annotators, highlighting bias when manual analysis is performed. The CNN effectively analysed platelet morphology when platelets spread over a range of substrates (CRP-XL, vWF and fibrinogen), in the presence and absence of inhibitors (dasatinib, ibrutinib and PRT-060318) and agonist (thrombin), with results consistent in quantifying spread platelet area which is comparable to published literature. The application of a CNN enables, for the first time, automated analysis of platelet spreading assays captured by DIC microscopy., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
40. Thiol Isomerases Orchestrate Thrombosis and Hemostasis.
- Author
-
Gaspar RS and Gibbins JM
- Subjects
- Animals, Humans, Hemostasis, Membrane Proteins metabolism, Thioredoxins metabolism, Thrombosis metabolism
- Abstract
Significance: Since protein disulfide isomerase (PDI) was first described in 1963, researchers have shown conclusively that PDI and sibling proteins are quintessential for thrombus formation. PDI, endoplasmic reticulum protein (ERp)5, ERp57, and ERp72 are released from platelets and vascular cells and interact with integrin αIIbβ3 on the outer surface of platelets. Recent Advances: At the cell surface they influence protein folding and function, propagating thrombosis and maintaining hemostasis. TMX1, which is a transmembrane thiol isomerase, is the first family member shown to negatively regulate platelets. Targets of thiol isomerases have been identified, including integrin α2β1, Von Willebrand Factor, GpIbα, nicotinamide adenine dinucleotide phosphate oxidase (Nox)-1, Nox-2, and tissue factor, all of which are pro-thrombotic, and several of which are on the cell surface. In spite of this, PDI can paradoxically catalyze the delivery of nitric oxide to platelets, which decrease thrombus formation. Critical Issues: Although the overall effect of PDI is to positively regulate platelet activation, it is still unclear how thiol isomerases function in pro-thrombotic states, such as obesity, diabetes, and cancer. In parallel, there has been a surge in the development of novel thiol isomerase inhibitors, which display selectivity, potency and modulate thrombosis and hemostasis. The availability of selective thiol isomerase inhibitors has culminated in clinical trials, with promising outcomes for the prevention of cancer-associated thrombosis. Future Directions: Altogether, thiol isomerases are perceived as an orchestrating force that regulates thrombus development. In the current review, we will explore the history of PDI in cardiovascular biology, detail known mechanisms of action, and summarize known thiol isomerase inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
41. Multiparameter phenotyping of platelet reactivity for stratification of human cohorts.
- Author
-
Dunster JL, Bye AP, Kriek N, Sage T, Mitchell JL, Kempster C, Batista J, McKinney H, Thomas P, Jones CI, Downes K, Unsworth AJ, and Gibbins JM
- Subjects
- Humans, Platelet Aggregation Inhibitors, Platelet Function Tests, Blood Platelets, Thrombosis
- Abstract
Accurate and comprehensive assessment of platelet function across cohorts of donors may be key to understanding the risk of thrombotic events associated with cardiovascular disease, and, hence, to help personalize the application of antiplatelet drugs. However, platelet function tests can be difficult to perform and analyze; they also can be unreliable or uninformative and poorly standardized across studies. The Platelet Phenomic Analysis (PPAnalysis) assay and associated open-source software platform were developed in response to these challenges. PPAnalysis utilizes preprepared freeze-dried microtiter plates to provide a detailed characterization of platelet function. The automated analysis of the high-dimensional data enables the identification of subpopulations of donors with distinct platelet function phenotypes. Using this approach, we identified that the sensitivity of a donor's platelets to an agonist and their capacity to generate a functional response are distinct independent metrics of platelet reactivity. Hierarchical clustering of these metrics identified 6 subgroups with distinct platelet phenotypes within healthy cohorts, indicating that platelet reactivity does not fit into the traditional simple categories of "high" and "low" responders. These platelet phenotypes were found to exist in 2 independent cohorts of healthy donors and were stable on recall. PPAnalysis is a powerful tool for stratification of cohorts on the basis of platelet reactivity that will enable investigation of the causes and consequences of differences in platelet function and drive progress toward precision medicine., (© 2021 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
42. Aberrant glycosylation of anti-SARS-CoV-2 spike IgG is a prothrombotic stimulus for platelets.
- Author
-
Bye AP, Hoepel W, Mitchell JL, Jégouic S, Loureiro S, Sage T, Vidarsson G, Nouta J, Wuhrer M, de Taeye S, van Gils M, Kriek N, Cooper N, Jones I, den Dunnen J, and Gibbins JM
- Subjects
- Antibodies, Viral blood, Antibodies, Viral immunology, Antigen-Antibody Complex immunology, Blood Platelets immunology, Blood Platelets metabolism, COVID-19 immunology, COVID-19 virology, Glycosylation, Humans, Platelet Activation immunology, Thrombosis immunology, Thrombosis virology, von Willebrand Factor genetics, Blood Platelets pathology, COVID-19 complications, Immunoglobulin G immunology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus metabolism, Thrombosis pathology, von Willebrand Factor metabolism
- Abstract
A subset of patients with coronavirus disease 2019 (COVID-19) become critically ill, suffering from severe respiratory problems and also increased rates of thrombosis. The causes of thrombosis in severely ill patients with COVID-19 are still emerging, but the coincidence of critical illness with the timing of the onset of adaptive immunity could implicate an excessive immune response. We hypothesized that platelets might be susceptible to activation by anti-severe acute respiratory syndrome coronavirus 2 (anti-SARS-CoV-2) antibodies and might contribute to thrombosis. We found that immune complexes containing recombinant SARS-CoV-2 spike protein and anti-spike immunoglobulin G enhanced platelet-mediated thrombosis on von Willebrand factor in vitro, but only when the glycosylation state of the Fc domain was modified to correspond with the aberrant glycosylation previously identified in patients with severe COVID-19. Furthermore, we found that activation was dependent on FcγRIIA, and we provide in vitro evidence that this pathogenic platelet activation can be counteracted by the therapeutic small molecules R406 (fostamatinib) and ibrutinib, which inhibit tyrosine kinases Syk and Btk, respectively, or by the P2Y12 antagonist cangrelor., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
43. Kinetx: A Combined Flow Cytometry Assay and Analysis Software Framework to Quantitatively Measure and Categorize Platelet Activation in Real-time.
- Author
-
Dunster JL, Mitchell JL, Mohammed YMM, Taylor KA, Gibbins JM, and Jones CI
- Subjects
- Flow Cytometry, Hemostasis, Software, Blood Platelets metabolism, Platelet Activation
- Abstract
Platelets react rapidly to vascular injury and undergo activation in response to a range of stimuli to limit blood loss. Many platelet function tests measure endpoint responses after a defined time period and not the rate of platelet activation. However, the rate at which platelets convert extracellular stimuli into a functional response is an essential factor in determining how efficiently they can respond to injury, bind to a forming thrombus, and signal to recruit other platelets. This paper describes a flow cytometry-based platelet function assay that enables simultaneous data acquisition and sample stimulation and utilizes newly developed bespoke open-source software (Kinetx) to enable quantitative kinetic measurements of platelet granule release, fibrinogen binding, and intracellular calcium flux. Kinetix was developed in R so that users can alter parameters such as degree of smoothing, identification of outlying data points, or time scales. To aid users unfamiliar with the R environment, Kinetix analysis of data can be performed by a single command. Together, this allows real-time platelet activation metrics, such as rate, acceleration, time to peak-rate, time to peak-calcium, and qualitative shape changes, to be accurately and reproducibly measured and categorized. Kinetic measurements of platelet activation give a unique insight into platelets' behavior during the first stages of activation and may provide a method of predicting the recruitment of platelets into a forming thrombus.
- Published
- 2021
- Full Text
- View/download PDF
44. Mind the gap: connexins and pannexins in platelet function.
- Author
-
Taylor KA, Little G, and Gibbins JM
- Subjects
- Animals, Humans, Mice, Blood Platelets metabolism, Connexins metabolism, Platelet Function Tests methods
- Abstract
Connexins are a family of gap junction forming proteins widely expressed by mammalian cells. They assemble into hexameric hemichannels, which can either function independently or dock with opposing hemichannels on apposite cells, forming a gap junction. Pannexins are structurally related to the connexins but extensive glycosylation of these channels prevents docking to form gap junctions and they function as membrane channels. Platelets express pannexin-1 and several connexin family members (Cx37, Cx40 and Cx62). These channels are permeable to molecules up to 1,000 Daltons in molecular mass and functional studies demonstrate their role in non-vesicular ATP release. Channel activation is regulated by (patho)physiological stimuli, such as mechanical stimulation, making them attractive potential drug targets for the management of arterial thrombosis. This review explores the structure and function of platelet pannexin-1 and connexins, the mechanisms by which they are gated and their therapeutic potential.
- Published
- 2021
- Full Text
- View/download PDF
45. The protein disulphide isomerase inhibitor CxxCpep modulates oxidative burst and mitochondrial function in platelets.
- Author
-
Gaspar RS, Mansilla S, Vieira VA, da Silva LB, Gibbins JM, Castro L, Trostchansky A, and Paes AMA
- Subjects
- Chromatography, Liquid, Mitochondria metabolism, Platelet Activation, Platelet Aggregation, Platelet Membrane Glycoproteins metabolism, Respiratory Burst, Tandem Mass Spectrometry, Blood Platelets metabolism, Protein Disulfide-Isomerases metabolism
- Abstract
Background: We have previously described CxxCpep, a peptide with anti-platelet properties that inhibits peri/epicellular protein disulphide isomerase (pecPDI) by forming a mixed disulfide bond with Cys400 within the pecPDI active site., Objectives: Here we sought to determine if pecPDI targeted by CxxCpep is relevant to redox mechanisms downstream of the collagen receptor GPVI in platelets., Methods and Results: Restriction of effects of CxxCpep to the platelet surface was confirmed by LC-MS/MS following cell fractionation. Platelet aggregation was measured in platelet-rich plasma (PRP) incubated with 30 μM CxxCpep or vehicle. CxxCpep inhibited collagen-induced platelet aggregation but exerted no effect in TRAP-6-stimulated platelets. PRP was incubated with DCFDA to measure oxidative burst upon platelet adhesion to collagen. Results showed that CxxCpep decreased oxidative burst in platelets adhered to immobilized collagen while the number of adherent cells was unaffected. Furthermore, flow cytometry studies using a FITC-maleimide showed that the GPVI agonist CRP stimulated an increase in free thiols on the platelet outer membrane, which was inhibited by CxxCpep. Finally, CxxCpep inhibited platelet mitochondrial respiration upon activation with collagen, but not with thrombin., Conclusions: Our data suggest that pecPDI is a potential modulator of GPVI-mediated redox regulation mechanisms and that CxxCpep can be further exploited as a template for new antiplatelet compounds., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
46. Targeting platelet inhibition receptors for novel therapies: PECAM-1 and G6b-B.
- Author
-
Soriano Jerez EM, Gibbins JM, and Hughes CE
- Subjects
- Animals, Humans, Ligands, Mice, Mice, Knockout, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Blood Platelets metabolism, Platelet Activation genetics, Receptors, Immunologic metabolism
- Abstract
While current oral antiplatelet therapies benefit many patients, they deregulate the hemostatic balance leaving patients at risk of systemic side-effects such as hemorrhage. Dual antiplatelet treatment is the standard approach, combining aspirin with P2Y
12 blockers. These therapies mainly target autocrine activation mechanisms (TxA2 , ADP) and, more recently, the use of thrombin or thrombin receptor antagonists have been added to the available approaches. Recent efforts to develop new classes of anti-platelet drugs have begun to focus on primary platelet activation pathways such as through the immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor GPVI/FcRγ-chain complex. There are already encouraging results from targeting GPVI, with reduced aggregation and smaller arterial thrombi, without major bleeding complications, likely due to overlapping activation signaling pathways with other receptors such as the GPIb-V-IX complex. An alternative approach to reduce platelet activation could be to inhibit this signaling pathway by targeting the inhibitory pathways intrinsic to platelets. Stimulation of endogenous negative modulators could provide more specific inhibition of platelet function, but is this feasible? In this review, we explore the potential of the two major platelet immunoreceptor tyrosine-based inhibitory motif (ITIM)-containing inhibitory receptors, G6b-B and PECAM-1, as antithrombotic targets.- Published
- 2021
- Full Text
- View/download PDF
47. Identification of HSP47 Binding Site on Native Collagen and Its Implications for the Development of HSP47 Inhibitors.
- Author
-
Cai H, Sasikumar P, Little G, Bihan D, Hamaia SW, Zhou A, Gibbins JM, and Farndale RW
- Subjects
- Binding Sites, Collagen metabolism, HSP47 Heat-Shock Proteins metabolism, Humans, Collagen chemistry, HSP47 Heat-Shock Proteins antagonists & inhibitors, HSP47 Heat-Shock Proteins chemistry, Molecular Docking Simulation
- Abstract
HSP47 (heat shock protein 47) is a collagen-specific molecular chaperone that is essential for procollagen folding and function. Previous studies have shown that HSP47 binding requires a critical Arg residue at the Y position of the (Gly-Xaa-Yaa) repeats of collagen; however, the exact binding sites of HSP47 on native collagens are not fully defined. To address this, we mapped the HSP47 binding sites on collagens through an ELISA binding assay using collagen toolkits, synthetic collagen peptides covering the entire amino acid sequences of collagen types II and III assembled in triple-helical conformation. Our results showed that HSP47 binds to only a few of the GXR motifs in collagen, with most of the HSP47 binding sites identified located near the N-terminal part of the triple-helical region. Molecular modelling and binding energy calculation indicated that residues flanking the key Arg in the collagen sequence also play an important role in defining the high-affinity HSP47 binding site of collagen. Based on this binding mode of HSP47 to collagen, virtual screening targeting both the Arg binding site and its neighboring area on the HSP47 surface, and a subsequent bioassay, we identified two novel compounds with blocking activity towards HSP47 binding of collagen. Overall, our study revealed the native HSP47 binding sites on collagen and provided novel information for the design of small-molecule inhibitors of HSP47.
- Published
- 2021
- Full Text
- View/download PDF
48. Antiplatelet properties of Pim kinase inhibition are mediated through disruption of thromboxane A2 receptor signaling.
- Author
-
Unsworth AJ, Bye AP, Sage T, Gaspar RS, Eaton N, Drew C, Stainer A, Kriek N, Volberding PJ, Hutchinson JL, Riley R, Jones S, Mundell SJ, Cui W, Falet H, and Gibbins JM
- Subjects
- Blood Platelets, Humans, Platelet Aggregation, Proto-Oncogene Proteins c-pim-1 genetics, Receptors, Thromboxane A2, Prostaglandin H2 genetics, Thrombosis drug therapy
- Abstract
Pim kinases are upregulated in several forms of cancer, contributing to cell survival and tumour development, but their role in platelet function and thrombotic disease has not been explored. We report for the first time that Pim-1 is expressed in human and mouse platelets. Genetic deletion or pharmacological inhibition of Pim kinase results in reduced thrombus formation but is not associated with impaired haemostasis. Attenuation of thrombus formation was found to be due to inhibition of the thromboxane A2 receptor as effects on platelet function was non-additive to inhibition caused by the cyclooxygenase inhibitor indomethacin or thromboxane A2 receptor antagonist GR32191. Treatment with Pim kinase inhibitors caused reduced surface expression of the thromboxane A2 receptor and resulted in reduced responses to thromboxane A2 receptor agonists, indicating a role for Pim kinase in the regulation of thromboxane A2 receptor function. Our research identifies a novel, Pim kinase dependent regulatory mechanism for the thromboxane A2 receptor and represents a new targeting strategy that is independent of COX-1 inhibition or direct antagonism of the thromboxane A2 receptor that whilst attenuating thrombosis does not increase bleeding.
- Published
- 2021
- Full Text
- View/download PDF
49. Zinc regulates reactive oxygen species generation in platelets.
- Author
-
Lopes-Pires ME, Ahmed NS, Vara D, Gibbins JM, Pula G, and Pugh N
- Subjects
- Animals, Humans, Reactive Oxygen Species, Blood Platelets metabolism, Zinc therapeutic use
- Abstract
Vascular complications resulting from atherosclerosis development are a major cause of death. Reactive oxygen species (ROS) are produced by platelets during activation, and have been demonstrated to positively regulate platelet activatory responses. Zn
2+ is also an important hemostatic cofactor in platelets, acting both as a platelet agonist and second messenger. Whilst the effect of Zn2+ -dependent signaling mechanisms on ROS production in nucleated cells has been demonstrated, comparable roles in platelets have yet to be investigated. In this study we investigated the relationship between fluctuations in cytosolic Zn2 [Zn2+ ]i and platelet ROS production. Agonist-evoked ROS production, GSH levels and GPx activity are abrogated in platelets treated with the Zn2+ -chelator, TPEN. Conversely, increasing platelet [Zn2+ ]i using Zn2+ ionophores potentiated ROS generation and decreased GSH levels and GPx activity. Zn2+ -dependent ROS production was sensitive to pretreatment with DPI or mitoTEMPO, NADPH oxidase and mitochondria inhibitors respectively. Increasing [Zn2+ ]i resulted in increases of Erk1/2 and JNK phosphorylation. Our data are consistent with a functional association between [Zn2+ ]i and ROS production in platelets that could influence thrombus formation in a clinical context.- Published
- 2021
- Full Text
- View/download PDF
50. Protein Disulphide Isomerase and NADPH Oxidase 1 Cooperate to Control Platelet Function and Are Associated with Cardiometabolic Disease Risk Factors.
- Author
-
Gaspar RS, Sage T, Little G, Kriek N, Pula G, and Gibbins JM
- Abstract
Background: Protein disulphide isomerase (PDI) and NADPH oxidase 1 (Nox-1) regulate platelet function and reactive oxygen species (ROS) generation, suggesting potentially interdependent roles. Increased platelet reactivity and ROS production have been correlated with cardiometabolic disease risk factors., Objectives: To establish whether PDI and Nox-1 cooperate to control platelet function., Methods: Immunofluorescence microscopy was utilised to determine expression and localisation of PDI and Nox-1. Platelet aggregation, fibrinogen binding, P-selectin exposure, spreading and calcium mobilization were measured as markers of platelet function. A cross-sectional population study ( n = 136) was conducted to assess the relationship between platelet PDI and Nox-1 levels and cardiometabolic risk factors., Results: PDI and Nox-1 co-localized upon activation induced by the collagen receptor GPVI. Co-inhibition of PDI and Nox-1 led to additive inhibition of GPVI-mediated platelet aggregation, activation and calcium flux. This was confirmed in murine Nox-1
-/- platelets treated with PDI inhibitor bepristat, without affecting bleeding. PDI and Nox-1 together contributed to GPVI signalling that involved the phosphorylation of p38 MAPK, p47phox, PKC and Akt. Platelet PDI and Nox-1 levels were upregulated in obesity, with platelet Nox-1 also elevated in hypertensive individuals., Conclusions: We show that PDI and Nox-1 cooperate to control platelet function and are associated with cardiometabolic risk factors.- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.