Next Article in Journal
Is There Enough Evidence to Support the Role of Glycosaminoglycans and Proteoglycans in Thoracic Aortic Aneurysm and Dissection?—A Systematic Review
Next Article in Special Issue
Association of Circulating Platelet Extracellular Vesicles and Pulse Wave Velocity with Cardiovascular Risk Estimation
Previous Article in Journal
mTOR: A Potential New Target in Nonalcoholic Fatty Liver Disease
Previous Article in Special Issue
The Role of Platelets in Diabetic Kidney Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oxidised Low-Density Lipoprotein-Induced Platelet Hyperactivity—Receptors and Signalling Mechanisms

1
Department of Internal Medicine 1, University Hospital Aachen, Pauwelstraße 30, 52074 Aachen, Germany
2
Leeds Institute of Cardiovascular and Metabolic Medicine, The LIGHT Laboratories, Clarendon Way, University of Leeds, Leeds LS2 9JT, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(16), 9199; https://doi.org/10.3390/ijms23169199
Submission received: 2 July 2022 / Revised: 26 July 2022 / Accepted: 9 August 2022 / Published: 16 August 2022
(This article belongs to the Special Issue Advances in Platelet Biology and Functions)

Abstract

:
Dyslipidaemia leads to proatherogenic oxidative lipid stress that promotes vascular inflammation and thrombosis, the pathologies that underpin myocardial infarction, stroke, and deep vein thrombosis. These prothrombotic states are driven, at least in part, by platelet hyperactivity, and they are concurrent with the appearancxe of oxidatively modified low-density lipoproteins (LDL) in the circulation. Modified LDL are heterogenous in nature but, in a general sense, constitute a prototype circulating transporter for a plethora of oxidised lipid epitopes that act as danger-associated molecular patterns. It is well-established that oxidatively modified LDL promote platelet activation and arterial thrombosis through a number of constitutively expressed scavenger receptors, which transduce atherogenic lipid stress to a complex array of proactivatory signalling pathways in the platelets. Stimulation of these signalling events underlie the ability of modified LDL to induce platelet activation and blunt platelet inhibitory pathways, as well as promote platelet-mediated coagulation. Accumulating evidence from patients at risk of arterial thrombosis and experimental animal models of disease suggest that oxidised LDL represents a tangible link between the dyslipidaemic environment and increased platelet activation. The aim of this review is to summarise recent advances in our understanding of the pro-thrombotic signalling events induced in platelets by modified LDL ligation, describe the contribution of individual platelet scavenger receptors, and highlight potential future challenges of targeting these pathways.

1. Introduction

Thrombotic events associated with atherosclerotic plaque rupture and unwanted platelet activation account for almost 25% of deaths worldwide and represent a major burden on the health of most countries [1]. Dyslipidaemia, secondary to conditions such as type 2 diabetes, obesity, chronic kidney disease, and metabolic syndrome, is associated with both atherogenesis and thrombosis [2]. Studies in mice and humans strongly suggest that dyslipidaemia induces a prothrombotic phenotype, in which platelet hyperactivity plays a major role [3,4,5,6,7,8]. Indeed, the increased thrombotic risk associated with dyslipidaemia may contribute equally to its atherogenic effects the predisposition of disease [9]. A biochemical characteristic of dyslipidaemia is increased oxidative stress and the accumulation of oxidised lipids in the vessel wall, in the form of oxidised low-density lipoproteins (oxLDL), as well as in the plasma [10]. These particles play an important role in promoting atherogenesis through endothelial cell activation, the proliferation of vascular smooth muscle cells, and the formation of lipid-laden foam cells [11]. However, there is compelling evidence to indicate that these modified forms of the lipoproteins also exist in the circulation, particularly in subjects at risk of atherothromboic disease [12,13,14,15,16]. In humans, the presence of circulating oxidatively modified LDL was first reported by Avogaro and colleagues, who described a modified sub-fraction of LDL with increased electrophoretic mobility on ion exchange chromatography and elevated levels of lipid peroxidation-induced conjugated dienes [17]. The presence of circulating oxLDL was given further credence by studies that identified the autoantibodies of oxLDL in subjects with coronary artery disease (CAD) and animal disease models [18,19,20,21,22]. Autoantibodies of oxLDL isolated from ApoE-deficient hyperlipidaemic mice bind exclusively to oxidised phospholipids [19] and lead to the development of several monoclonal antibodies against oxidatively-modified epitopes on LDL, thus allowing for the determination of concentrations of oxLDL plasma of human subjects [12,23,24,25,26]. The presence of elevated levels of modified LDL has been observed in end-stage renal disease, obesity, hypercholesterolemia, diabetes, and coronary artery disease (CAD) [27,28,29,30,31]. More recently, a similar electronegative LDL representing the fifth quintile of the electrophoretically separated LDL (L5), with similar biochemical properties to LDL that had been oxidised in vitro, was identified in subjects with both CAD and stroke [32,33]; it was critically shown to be biologically active ex vivo. Moreover, in subjects with acute coronary syndrome (ACS), the platelets circulate with oxLDL bound to their surface, suggesting a functional relationship in the disease environment [34,35].
The link between dyslipidaemia and elevated platelet activation was first described by Carvalho in subjects with familial hypercholesterolaemia [8]. These critical findings were confirmed in animal models of hyperlipidaemia and shown to impact directly on thrombosis [7], with even mild hyperlipidaemia leading to accelerated platelet activation [4]. The levels of oxLDL in the circulation predicted future atherothrombotic events and the presence and severity of atherosclerotic plaque [12,31,36]. Given this context, a significant number of investigators have shown that oxLDL can both induce and promote the activity of platelets when tested in vitro (Table 1). Thus, there is strong evidence to suggest that oxidised LDL may be a primary causative factor in inducing platelet hyperactivity and the associated prothrombotic phenotype observed in a number of disease cohorts [37,38,39], both in the acute setting of plaque rupture and chronically while the platelets are in circulation. In the current article, we review the biochemical mechanisms that translate atherogenic lipid stress, in the form of oxLDL from the circulation, into platelet hyperactivity.

2. Oxidised LDL Are Heterogeneous Particles

LDL are circulating lipid-rich particles whose primary function is the delivery of cholesterol, in the form of cholesteryl esters and fat-soluble vitamins, to non-hepatic cells. They are composed of a hydrophobic core of cholesteryl esters and triglycerides, surrounded by a monolayer shell of phospholipids. Native LDL also contain a single polypeptide chain, apolipoprotein B100, which is required by the ApoB/E receptors on the cell surface for recognition and endocytosis. It is important to recognise that LDL represent a heterogenous group of particles whose biochemical composition is determined by diet, genetics, and physical activity [98]. This biochemical heterogeneity occurs within individuals and between individuals with any given population. The composition of the particles also has a direct influence on both their resistance to oxidation and the types of biochemical lipid species that are produced in response to oxidative attack. The oxidation of LDL in vivo is thought to occur in the subendothelial space, when the particles can become anchored by glycosaminoglycans and are subsequently exposed to a number of factors that induce oxidative modifications, including reactive oxygen species, reactive nitrogen species, and cellular enzymes [98]. In vitro this can be mimicked by exposing isolated LDL to a range oxidizing systems, including cells, free radical generating systems, non-radical oxidants, and enzymes [98,99]. While, in this review, we focus on LDL that has been altered by oxidation, it is important to note that other forms of modification have been reported including glycation (diabetes mellitus), carbamylation (chronic kidney disease), and acetylation, all of which may contribute to platelet activation in different circumstances [27,30,74,100].
The term oxidised LDL is often used as a “catch all” term used to describe LDL preparations, either isolated directly from the circulation or, more commonly, preparations that have been oxidised under very defined experimental conditions. Given the complex and varied biochemical composition of LDL particles, a vast array of bioactive compounds have been reported to be present, including modified phospholipids, sphingolipids, free fatty acids, cholesterol esters, oxysterols, and aldehydes [99]. The generation of various combinations of these species creates a complex population of particles, with potentially distinct biological properties. Oxidation is a progressive event, beginning with the loss of endogenous antioxidants, including β-carotene and α-tocopherol, progressing to lipid oxidation, and finally, the modification of the apoB100, as measured by ELISA or mass-spectrometry [101,102]. From an experimental perspective, oxLDL preparations are generally divided into two types, minimally modified LDL (mmLDL or mildly oxidised LDL) and oxLDL (fully oxidised). MmLDL represents particles that have undergone lipid peroxidation, but not protein modification. The process begins with the peroxidation of the fatty acid chains of polyunsaturated fatty acids of the particle shell before progressing to cholesteryl esters in the core. These mmLDL particles can still be recognised by the apoB/E receptor and could, therefore, exist in circulation for extensive periods [103]. In contrast, oxLDL have undergone extensive lipid oxidation coupled to protein modification and most likely represents the type of particles present in atherosclerotic plaques that may be released upon rupture. Here, the continued oxidation of the lipids leads to the generation of aldehydes, such as malondialdehyde and 4-hydroxynonenal, which form adducts with the lysine residues of the ApoB100, thus resulting a net loss of the overall positive charge [99,101]. These progressive biochemical changes diminish the recognition of LDL by the apoB/E receptor and lead to the increased recognition by a series of scavenger receptors. Given the continual generation of LDL in the blood stream, as part of the endogenous lipid metabolism pathway, atherosclerotic plaques probably contain a spectrum of modified LDL species, although it is unclear if the same is true in the circulation. Therefore, it is critically important to appreciate that, when assessing the effects of modified LDL on platelets, regardless of their origin or method of oxidation, the cells are being exposed to a diverse population of particles with non-uniform biochemical composition and properties. The difference in bioactive compounds produced by these varied approaches probably account for the plethora of effects these particles have been shown to induce in platelets.

3. The Role of oxLDL in the Pathogenesis of Metabolic Disease

The presence of oxLDL is linked to several disease states, and elevated levels of oxLDL in the plasma have been detected in a varity of disorders, including coronary artery disease, dyslipidaemia, stroke, acute coronary syndrome, metabolic syndrome, chronic kidney disease, and several autoimmune diseases (Figure 1) [14,15,28,29,104]. Interestingly, the many lines of evidence suggest that oxLDL has a causal relationship with the pathogenesis of atherosclerosis, diabetes, and certain autoimmune disease (reviewed in [98]). This causal role is particularly well-established in the context of atherosclerosis, and oxLDL has been found to affect the early stages of the disease, including the endothelial dysfunction by endothelial cell network formation, dysregulation of endothelial nitric oxide production, and alignment of endothelial cells under flow conditions [105,106,107]. Later, the accumulation of the modified cholesterol species, derived from oxLDL in macrophages and leading to foam cell formation, in the intima is critical to driving proinflammatory processes in the vessel wall [108,109]. The causality is underpinned by three observations—(i) oxLDL is only found in diseased arteries [110], (ii) downregulation of scavenger receptors that serve as ligands for oxLDL greatly reduce atherosclerosis disease burden in mice [111], and (iii) in Apo-E deficient mice presence of oxLDL precedes the development of an atherosclerotic plaque [112]. In patients with diabetes, the glycaemic environment increases the susceptibility of LDL to oxidation and, therefore, increases its atherogenic potential [113]. In addition, the presence of oxLDL leads to an impairment of glucose transporter 4 (GLUT4) and desensitises adipocytes against insulin, therefore increasing the hyperglycaemic state [113,114]. The notion that inflammation promotes LDL oxidation has led many researchers to investigate the role of oxLDL in various autoimmune disease. Even though the causal link in pathogenesis is less clear in these patients, the presence of oxidised LDL or anti-oxLDL immunocomplexes is linked to disease severity and progression in Rheumatoid arthritis, Systemic Lupus erythematodes, Sjogrens’s syndrome, and several vasculitides [115,116,117]. In summary, oxLDL has been linked to many disease states and, therefore, serves as an interesting biomarker; however, and while uncertainties still exist, evidence suggests a causal link, particularly in atherosclerosis, which conceptually makes oxLDL an interesting therapeutic target.

4. Functional Responses of Platelets to Modified Low-Density Lipoproteins

The ability of oxidatively modified forms of LDL to activate and sensitise platelets for activation has been appreciated for over forty years; since that time, our knowledge of these effects and the associated mechanisms have increased dramatically. The study by Avogaro, identifying oxidised LDL in the plasma of patient with atherosclerosis [17], suggested that these modified lipoproteins may be the link between dysregulated lipid metabolism and platelet hyperactivity. A large number of studies have been performed examining platelet function in response to various forms of laboratory oxidised LDL. These atherogenic lipoproteins activate a plethora of signalling events in the platelets that underpin their ability to augment platelet functions. Examination of the literature demonstrate that oxLDL can influence many critical platelet functions, including secretion [34,56,57,58,59], adhesion [34,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54], microvesicle release and pro-coagulant phosphatidylserine expression [96], Ca2+ mobilisation [63,68,83], reactive oxygen species generation [42,89,90], and act synergistically with physiological agonists to promote secretion and aggregation [33,63,68,79,80,81,118] (Table 1).
Furthermore, immobilised oxLDL supports platelet adhesion and spreading, both as independent ligands and through the potentiation of the adhesive capacity of extracellular matrix proteins [44,89]. It is noteworthy that there is robust evidence suggesting that the exposure of platelets to mmLDL and oxLDL primes the cells for activation and potentiates the effects of haemostatic agonists; the evidence supporting oxLDLs as agonists capable of induce aggregation is inconsistent. The reasons for this are unclear, but likely reflects the use of distinct LDL preparations that have a specific combination of lipid oxidation products.
The two major considerations when evaluating the potential for oxLDL augmented platelet functions are the platelet model used and type of LDL preparation. The vast majority of studies highlighted in Table 1 use washed platelets and occasionally platelet-rich plasma (PRP), but not whole blood. Experiments with washed platelets allow for the investigation of lipoproteins in a controlled environment and represent important proof-of-principle; however, it is clear that platelet phenotypes differ according to the presence of buffer, plasma, or whole blood (Table 1). The effects of oxLDL are clearly most potent when incubated with washed platelets and less so with PRP. It possible that the antioxidant environment of the plasma protects agonist potent oxidised lipids, thus diminishing their activatory capacity. Nevertheless, the oxLDL has been shown to be effective in whole blood, where it can act as a mild agonist that influences platelet adhesion, secretion, and thrombosis [79,88]. Therefore, LDL are likely to have a major impact at sites of atherosclerotic plaque rupture, where platelets would be exposed to both various forms of modified LDL and a plethora of physiological platelet activators. While this has not been demonstrated directly, a recent study demonstrated that the infusion of oxLDL into mice significantly enhanced thrombosis using a ferric chloride model [88]. Given that oxLDL may induce procoagulant effects on the endothelium [119,120], these experiments need to be interpreted with caution; nevertheless, they are an important proof of principle that oxLDL promotes thrombosis in vivo.
The second important consideration is the method and extent of oxidation. It has been proposed that mildly oxidised LDL are more proficient in driving platelet activation, and they may have more functional relevance, given that they could potentially exist for extended times in circulation. Korporaal and colleagues compared several degrees of LDL oxidation, ranging from 0–60%, based on the number of conjugated dienes, and found that 60% lipid peroxidation (likely classified as mmLDL) is required to achieve a significant aggregatory response [94]. Similarly, both Weideman et al. and Naseem et al. showed that the same LDL preparation, oxidised to different extents, produced distinct functional responses with the mildly oxidised inducing aggregation, which would fully oxidise LDL, with little or no effect [73,75]. This picture is further complicated by the type of oxidation, with early studies suggesting that hypochlorous acid modified LDL was a more potent platelet activator than Cu2SO4-induced oxLDL, with LDL inducing full platelet aggregation, while copper-oxidised LDL only showed minor effects [63,74]. These studies present an extremely complex picture of the activatory effects, influenced by an array of biologically active compounds found in different quantities that are produced under these conditions. Whilst these various modified LDL preparations can present a confusing picture, with regard to their effects on platelets, it is important recognise that the heterogeneity of modified LDL closely resembles the in vivo situation, where there will be LDL at various stages of oxidation that have been exposed to multiple sources of oxidative insult. In this regard, several critically important studies have examined the clinical relevance of these studies using LDL oxidised in vitro by isolating oxLDL from patients and testing their effect on platelet function. Definitive links to in vivo modified LDL were shown much later, with Colas demonstrating that LDL taken from obese subjects with metabolic syndrome or subjects with type 2 diabetes were peroxidised and had the ability to potentiate platelet aggregation and TxA2 formation [28]. Two studies from Chan and colleagues isolated an electronegative LDL from patients with ACS and ischaemic stroke, which had a similar biochemical composition to in vitro copper oxidised LDL [32,33]. In vivo studies demonstrated that this modified electronegative faction of LDL could potentiate platelet activation, but not induce aggregation. These important observations justify the enormous efforts to identify the mechanisms of oxLDL-induced platelet activation and suggest that targeting this type of platelet activation may provide context-dependent platelet inhibition, without compromising key haemostatic pathways.

5. Scavenger Receptors—The Transducers of oxLDL into Platelet Hyperactivity

The cellular recognition of modified LDL was initially described in the landmark study by Goldstein and colleagues, who demonstrated the ability of macrophages to recognise and engulf acetylated LDL, independently of the apoB/E receptor [109]. The binding of these modified lipid bearing particles is facilitated by a number of different scavenger receptors, of which, there are five different classes with the capacity to bind to a plethora of danger- and pathogen-associated molecular patterns (DAMPs, PAMPs) [121]. The binding of modified LDL to platelets has been demonstrated by a variety of techniques, including radioactive labelling, flow cytometry, and fluorescence microscopy [74,92,122]. Ligation has been proposed to occur through several platelet scavenger receptors, including cluster of differentiation 36 (CD36), lectin-like oxidised low-density lipoprotein receptor-1 (LOX-1), scavenger receptor B1 (SR-B1), G-coupled receptor lysophosphatidic acid receptor 1 (LPAR1), and integrins [123]. The recognition of oxLDL by the scavenger receptors is dependent on specific lipid oxidation products, their relative amounts, and the modification of the apoB100 by lipid derivatives. These receptors show distinct preferences for particular forms of oxidatively modified LDL; although, given the complex and progressive nature of the biochemical modification, there is a likely overlap, where modified LDL may interact with more than one type of scavenger receptor (Figure 1). The presence of multiple scavenger receptors as binding sites for oxLDL, combined with the heterogenous composition of the particles and differences in oxidation methods, underpins the varied responses to these lipoproteins observed in platelets. However, despite this complexity, the ligation of all of scavenger receptors by modified LDL is generally linked to an activatory signalling in platelets. Understanding the contribution of these individual receptors to the prothrombotic phenotype in disease states will allow for a better understanding of how and why oxLDL-induced signalling promotes unwanted platelet activation.

5.1. CD36—Class B Scavenger Receptor

CD36 is a heavily glycosylated transmembrane receptor consisting of two short intracellular segments at the amino and carboxy termini, two transmembrane domains, and a large extracellular loop [124,125]. CD36 is highly expressed on the platelet surface, with up to 20,000 copies per platelet [126,127]. It was first identified as GPIV as a receptor for collagen and then later found to have a high affinity for plasma protein thrombospondin-1 [128,129]. The identification of CD36 as a receptor for oxLDL was reported by Endemann et al., who demonstrated that CD36-transfected HEK cells bind oxLDL [130]. CD36 does not recognise delipidated LDL, thus indicating that the ligand recognition moieties within oxLDL are primarily oxidised phospholipids [131]. This was confirmed by Terpstra and colleagues, who showed that the recognition by CD36 was mediated by oxidised lipids present in oxLDL, which appeared early in the oxidative process, and were likely ligands for CD36, since these were effective antagonists of oxLDL binding to macrophage CD36 [132]. A critical breakthrough in our understanding of CD36 ligation was the systematic investigation of phospholipids present in oxLDL, as reported by Podrez and colleagues. This study demonstrated that oxidation of the two most abundant molecular species, i.e., choline glycerophospholipids 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphocholine (PAPC) and 1-palmitoyl-2-linoleoyl-sn-glycero-3-phosphocholine (PLPC), lead to generation of several highly CD36-specific phospholipids [133]. These phospholipids have been shown to be dramatically increased in the plasma of genetic models of hyperlipidaemia, including both apoE and LDLr deficient mice. Importantly, it was recognised that only one or two of the oxidised molecules needed to be present in the LDL to enable recognition and binding, thus reiterating that only minor changes in the oxidative modification process are required to alter platelet function. Elegant translational studies from Podrez and colleagues build on these observations to demonstrate the key link between oxidative lipid stress, CD36, and platelet hyperactivity. Using genetically modified mice hyperlipidemic mice, the authors demonstrated the presence of increased plasma levels in these oxidised phospholipids, which was associated with a platelet-driven prothrombotic phenotype and corrected by the deletion of CD36 [61].
OxLDL binds to CD36 in a region located between amino acids 155–183, with a particular requirement of the positively charged, highly conserved amino acids lysine164 and lysine166 [134,135]. The ligation of CD36 by oxLDL or oxidised phospholipids leads to the activation of a complex and interlinked signalling pathway that is linked to multiple aspects of platelet function. Critical to the ability to transduce oxLDL binding to the intracellular signalling machinery of the platelets are the Src family kinases (SFK), two of which, Fyn and Lyn, are constitutively associated with platelet CD36 [136]. We, and others, have shown that ligation of oxLDL to CD36 leads to activation of a tyrosine kinase signalling pathway that is initiated by the activation of SKF [44,46,59], which links CD36 to multiple downstream signalling pathways (Figure 1). The use of SFK kinase inhibitors indicates that Fyn and/or Lyn are functionally linked to ERK, p38, Mkk4/JNK, Vav1/3, and Syk [46,52,59,90,137]. Mice that are deficient in Vav1 and Vav3 are protected against the prothrombotic phenotype displayed in ApoE−/− mice, suggesting a critical role for this protein in promoting atherothrombosis [137]. The establishment of a role of serine/threonine kinases of mitogen-activated protein kinases (MAPK) in oxLDL-mediated macrophage foam cell-formation led Chen et al. to describe the sequential activation of Src, MKK-4, and JNK in response to oxLDL, thus leading to activation of aIIbb3 and P-selectin expression [59]. Korporaal and colleagues linked oxLDL activatory signalling to the MAPK p38, but the necessity of SFKs was not investigated [52]. Consistent with these studies, Karimi et al. showed that inhibition of p38 and JNK lead to a reduced functional response to oxLDL treatment [138]. Most recently, SFK signalling has been associated with increased ROS generation and subsequent Caspase-3 cleavage, which lead to increased phosphatidylserine expression and might link oxLDL ligation and SFK signalling to pro-coagulant activity of platelets [139].
Consistent with canonical signalling from GPVI, SFK couple CD36 to the activation of Syk, which leads to the activation of a RhoA-dependent inhibition of myosin light chain phosphatase that underpins spreading and shape change [46]. Downstream signalling from Syk leads to phosphorylation of phospholipase Cγ2 (PLCγ2), which appears to be critical for oxLDL-induced platelet activation, since either pharmacological inhibition or genetic deletion blunted platelet hyperactivity [46,89,140]. The role of Syk in activating the other key signalling elements, such as ERK, p38, Mkk4/JNK, and Vav1/3, is unclear; although, it is likely, given that Syk activation lies downstream of other tyrosine kinase-linked receptors in platelets, for example, GPIb-IX-V, GPVI, Clec-2, and αIIbβ3 [141,142,143,144]. The sequential activation of Syk and PLCg2 leads to the activation of protein kinase C (PKC) isoforms. While it remains unclear which PKC isoforms lie downstream of CD36, their activation is critical to its ability to modulate cyclic nucleotide signalling in platelets. PKC-mediated activation of NADPH oxidase 2 (NOX2)-dependent ROS production inhibits PKG signalling [140], while activation of the phosphorylation and activation of PDE3A modulates of PKA signalling (Figure 1) [88]. This process has been termed disinhibition and leads to a reduced threshold for platelet activation and provides the molecular mechanisms for earlier observations [87]. Thus, a substantial body of work suggests that oxLDL ligation to CD36 promotes platelet activation, blunts inhibitory signalling, and contributes to the generation of procoagulant platelets; the combination of these interrelated pathways promotes the potential for platelet hyperactivity and contribute to dyslipidaemia-induced procoagulant state.
The role of CD36 as a critical receptor for platelet hyperactivity, in the context of lipid imbalances, has gained further importance with two very recent studies. Proprotein convertase subtilisin/kexin type 9 (PCSK9), which can regulate cholesterol uptake in the liver, is elevated in subjects at risk of cardiovascular events and associated with increased platelet reactivity [145].

5.2. Lectin-like Oxidised Low-Density Receptor 1—Class E Scavenger Receptor

Lectin-like oxidised low-density receptor 1 belongs to the C-type lectin family and consists of four domains, i.e., a short N-terminal cytoplasmic domain, single transmembrane domain, connecting neck domain, and lectin-like domain at the C-terminus [146]. LOX-1 was initially identified as a receptor for oxLDL in endothelial cells [147]. LOX-1 differs in its recognition of oxLDL from CD36, since it can bind to both mildly oxidised and delipidated oxLDL, suggesting it has the capacity to recognise oxidised phospholipids and modified apoB100. In platelets, LOX-1 is found in the alpha-granules and relocated to the cell surface upon platelet activation [147]. This suggests that LOX-1 may facilitate the effects of oxLDL secondary to initial platelet activation and makes understanding the signalling pathways that follow LOX-1 ligation difficult to investigate, since any preactivation by alternative agonists could contribute directly to the signalling outcomes. Nevertheless, LOX-1 has been shown to play a role in platelets, in response to chromatographically separated LDL from ST-elevation myocardial infarction (STEMI) and stroke patients [32,33]. It is worth noting that electronegative LDL resembles the bioactive properties of mildly oxidised LDL and was characterised by increased levels of thiobarbituric acid-reactive substances (TBARS), a measure of lipid oxidation. This highly electronegative fraction potentiated the activatory effects of ADP or amyloid β (Aβ), in order to increase platelet aggregation, P-selectin expression, and integrin expression. Mechanistically, L5-LDL-mediated activation of platelet was linked to a signalling pathway involving PI3-kinase and AKT [32,33]. In a separate study, L5-LDL isolated from stroke patients synergised with Aβ to promote platelet activation through an IKK2, IκBα, p65, and c-Jun N-terminal kinase 1 [32]. Given that Aβ is a ligand for CD36 [148], it is possible that this pathway is activatory in response to cooperative signalling between these receptors. In contrast to other scavenger receptors, the role of oxLDL-LOX-1 in platelets has been exclusively studied, in the context of severe atherothrombotic events, including myocardial infarction and stroke. These patients have elevated levels of platelet LOX-1 expression, which may suggest the presence of preactivated platelets in these conditions. This is consistent with many in vitro studies, which demonstrated that oxLDL potentiates the activation of preactivated platelets and could suggest that the minor platelet activation associated with disease may facilitate increased responsive to circulating oxLDL. However, given the absent expression of LOX-1 under resting conditions, the role of LOX-1 under stable conditions remains unclear and deserves further investigation.

5.3. Scavenger Receptor A1 (SR-A)—Class A Scavenger Receptor

SR-A was identified in macrophages as a receptor capable of recognising and endocytosing oxLDL [149]. SR-A is a trimeric membrane glycoprotein with broad ligand specificity that includes oxidised lipoproteins, heat shock proteins, and bacteria. This receptor has a high affinity for fully oxidised LDL and is thought to bind to the modified apoB100 through recognition that involves the loss of positively charged lysine residues. Currently, definitive evidence for the expression of SR-A on platelets is lacking, since it has not been identified in proteomic studies [127], although it is present in the human platelet transcriptome [150]. A functional role for SR-A in oxLDL-mediated platelet activation has been suggested, based on a combination of pharmacological and genetic studies. The inhibition of SR-A in human platelets and double deletion of CD36 and SR-A completely inhibited p38MAPK signalling in response to oxLDL [52]. Furthermore, the inhibition of both receptors abolished platelet spreading on fibrinogen and mimicked the phenotype of pharmacological inhibition of p38MAPK. Unfortunately, knock out mice of the individual receptors were not used in these experiments. However, the fact that pharmacological inhibition of both receptors was required to prevent spreading suggests that, under some circumstances, SR-A may serve as a coreceptor of CD36.

5.4. LPA Receptor

Lysophosphatic acid (LPA) associated with mmLDL has been shown to activate washed platelets [53]. The presence of the three Edg family LPA receptor 1–3 has been suggested by RT-PCR [151]. However, formal identification of the receptor on the platelets by other methodologies is lacking to date. Nevertheless, Maschberger et al. implied that functional copies of the LPA receptor might be present on the platelets by showing that LPA receptor desensitization and the LPA receptor antagonist NPTyrPA blunted the activatory consequences of mildly oxidised LDL exposure [48]. However, given the lack of studies showing receptor expression and the lack of any LPA receptor knock-out animal model, these data need further confirmation.

5.5. Platelet Activating Factor (PAF) Receptor

Mass-spectroscopy data indicates that the PAF receptor, i.e. a G-protein-coupled receptor that binds the derivative of phosphorylcholine PAF, is likely to be expressed at low levels on platelets [127]. Chen et al. found that oxidatively modified phospholipids present in oxLDL can activate platelets in a PAF receptor-dependent manner, based on pharmacological studies with PAF receptor antagonists [68]. OxLDL and a range of oxidised phospholipids potentiated thrombin-induced Ca2+ mobilisation, P-selectin expression, and αIIbβ3 activation [68]. Corroboration of the potential role of the PAF receptor in platelet hyperactivity is provide by Chan and colleagues, who found that the proaggregatory effects of patient-derived electronegative LDL was blocked by the PAF receptor inhibitor ABT-491 [33]. Given the uncertainly around the expression of PAF receptors, further work in this area is required. It is possible that very low levels of expression only become functionally relevant when presented with the appropriate oxidised phospholipid species.

5.6. Toll-like Receptors (TLR)

TLR are highly conserved pattern recognition receptors that recognise either danger-associated (e.g., apoptotic cells) or pathogen-associated (e.g., bacterial peptides) molecular patterns (DAMPs and PAMPs). The presence of several TLR has been shown on platelets and megakaryocytes; however, until recently, their functions remained unclear [152]. Studies in macrophages demonstrates that several constituents of oxLDL, including cholesteryl esters (CE) and oxidised phospholipids (oxPL), can bind to TLRs-mediated signalling. Biswas et al. demonstrated that both TLR-2 and TLR-6 can contribute to both human and murine platelet activation induced by a oxidised phospholipids species found in oxLDL [62]. A complex picture emerges from this work, with TLR-2 contributing independently to platelet activity, while TLR-6 may utilise CD36 as a coreceptor [62]. Critically, this study did not use oxLDL; nevertheless, it proposes some interesting questions around the potentially cooperative signalling between platelet scavenger receptors. It is possible that minimally modified LDL could act as a ligand for TLRs, as evidenced in macrophages [153].

6. Concluding Remarks

It is now established that oxidatively modified versions of LDL circulate in subjects who are at high risk of major adverse cardiovascular events, including obesity, diabetes, stable coronary artery disease, acute coronary syndromes, and ischemic stroke. Increased circulating levels of these particles are associated with elevated risk of atherothrombotic events and linked to heightened platelet activity. Therefore, understanding the mechanisms how these pathogenic lipid particles induce maladaptive platelets phenotypes is critical to appreciating the prothrombotic phenotype in high-risk groups. Currently, mechanisms targeting scavenger receptors to control unwanted platelet activation seems unlikely, given their widespread expression; other key functional roles will need more well-designed studies to evaluate its effect. However, a key to furthering this agenda is developing a greater understanding of how these receptors function under pathological conditions. A number of key areas require further exploration
i. Functional reprogramming of platelets in disease cohorts. In mice, dyslipidaemia is associated with altered haematopoiesis and increased numbers of reticulated platelets, which are thought to be more thrombogenic [154]. However, it is unclear whether this changes the expression profile of platelet scavenger receptors. Platelets from subjects with ACS express elevated levels of LOX-1 on their surface, suggesting that scavenger receptor function may be upregulated in disease [47]. Similarly, the surface expression levels of CD36 can vary by up to 50 times, which, in turn, can affect the functional response to oxLDL [126]. Understanding the expression profiles and functionality of the receptors in disease cohorts may be critical to linking oxLDL-induced platelet hyperactivity and thrombosis in disease subjects.
ii. Receptor crosstalk. There are several examples in the literature that suggest cooperative partnerships between scavenger receptors, particularly CD36, which can induce many of its cellular effects through the formation of a diverse array of heteromeric signalling complexes. A range of transmembrane proteins, such as CD9, TLR2, TLR4, TLR6, CD47, and β1 integrin, can be associated with CD36 in a context-dependent manner [121], although only CD9 interacts in platelets [155]. The formation of hetromultimeric signalling complexes between different scavenger receptors or individual scavenger receptors with other adaptor proteins could be critical to the orchestration of distinct platelet responses in response to oxLDL.
iii. Scavenger receptor-linked signalling proteins as biomarkers. A recent report indicated that the phosphorylation of acethyl-CoA carboxylase (ACC) in platelets, potentially downstream of CD36, was causally linked to poorer outcomes in ACS patients [156]. This is the first example of a scavenger receptor-linked biomarker for thrombotic risk and could open up the possibility of further specific early appearance biomarkers for preventive of thrombosis.
It is noteworthy that current anti-platelet medication is geared towards treating the consequences of platelet activation. A better understanding of the role of scavenger receptor may open up the possibility of targeting the cause of platelet hyperactivity in high-risk groups.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jackson, S.P. Arterial thrombosis—Insidious, unpredictable and deadly. Nat. Med. 2011, 17, 1423–1436. [Google Scholar] [CrossRef] [PubMed]
  2. Musunuru, K. Atherogenic dyslipidemia: Cardiovascular risk and dietary intervention. Lipids 2010, 45, 907–914. [Google Scholar] [CrossRef] [PubMed]
  3. Eitzman, D.T.; Westrick, R.J.; Xu, Z.; Tyson, J.; Ginsburg, D. Hyperlipidemia promotes thrombosis after injury to atherosclerotic vessels in apolipoprotein E-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 1831–1834. [Google Scholar] [CrossRef] [PubMed]
  4. Van Geffen, J.P.; Swieringa, F.; van Kuijk, K.; Tullemans, B.M.E.; Solari, F.A.; Peng, B.; Clemetson, K.J.; Farndale, R.W.; Dubois, L.J.; Sickmann, A.; et al. Mild hyperlipidemia in mice aggravates platelet responsiveness in thrombus formation and exploration of platelet proteome and lipidome. Sci. Rep. 2020, 10, 21407. [Google Scholar] [CrossRef] [PubMed]
  5. Pawelczyk, M.; Kaczorowska, B.; Baj, Z. The impact of hyperglycemia and hyperlipidemia on plasma P-selectin and platelet markers after ischemic stroke. Arch. Med. Sci. 2017, 13, 1049–1056. [Google Scholar] [CrossRef]
  6. Broijersen, A.; Hamsten, A.; Eriksson, M.; Angelin, B.; Hjemdahl, P. Platelet activity in vivo in hyperlipoproteinemia—Importance of combined hyperlipidemia. Thromb. Haemost. 1998, 79, 268–275. [Google Scholar] [CrossRef]
  7. Suehiro, A.; Kakishita, E.; Nagai, K. The role of platelet hyperfunction in thrombus formation in hyperlipidemia. Thromb. Res. 1982, 25, 331–339. [Google Scholar] [CrossRef]
  8. Carvalho, A.C.; Colman, R.W.; Lees, R.S. Platelet function in hyperlipoproteinemia. N. Engl. J. Med. 1974, 290, 434–438. [Google Scholar] [CrossRef]
  9. Lacoste, L.; Lam, J.Y.; Hung, J.; Letchacovski, G.; Solymoss, C.B.; Waters, D. Hyperlipidemia and coronary disease. Correction of the increased thrombogenic potential with cholesterol reduction. Circulation 1995, 92, 3172–3177. [Google Scholar] [CrossRef]
  10. Lu, J.; Chen, B.; Chen, T.; Guo, S.; Xue, X.; Chen, Q.; Zhao, M.; Xia, L.; Zhu, Z.; Zheng, L.; et al. Comprehensive metabolomics identified lipid peroxidation as a prominent feature in human plasma of patients with coronary heart diseases. Redox Biol. 2017, 12, 899–907. [Google Scholar] [CrossRef]
  11. Lusis, A.J. Atherosclerosis. Nature 2000, 407, 233–241. [Google Scholar] [CrossRef] [PubMed]
  12. Toshima, S.; Hasegawa, A.; Kurabayashi, M.; Itabe, H.; Takano, T.; Sugano, J.; Shimamura, K.; Kimura, J.; Michishita, I.; Suzuki, T.; et al. Circulating oxidized low density lipoprotein levels. A biochemical risk marker for coronary heart disease. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 2243–2247. [Google Scholar] [CrossRef] [PubMed]
  13. Ehara, S.; Ueda, M.; Naruko, T.; Haze, K.; Itoh, A.; Otsuka, M.; Komatsu, R.; Matsuo, T.; Itabe, H.; Takano, T.; et al. Elevated levels of oxidized low density lipoprotein show a positive relationship with the severity of acute coronary syndromes. Circulation 2001, 103, 1955–1960. [Google Scholar] [CrossRef] [PubMed]
  14. Holvoet, P.; Mertens, A.; Verhamme, P.; Bogaerts, K.; Beyens, G.; Verhaeghe, R.; Collen, D.; Muls, E.; Van de Werf, F. Circulating oxidized LDL is a useful marker for identifying patients with coronary artery disease. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 844–848. [Google Scholar] [CrossRef] [PubMed]
  15. Holvoet, P.; Collen, D.; Van de Werf, F. Malondialdehyde-modified LDL as a marker of acute coronary syndromes. JAMA 1999, 281, 1718–1721. [Google Scholar] [CrossRef]
  16. Tsimikas, S.; Bergmark, C.; Beyer, R.W.; Patel, R.; Pattison, J.; Miller, E.; Juliano, J.; Witztum, J.L. Temporal increases in plasma markers of oxidized low-density lipoprotein strongly reflect the presence of acute coronary syndromes. J. Am. Coll. Cardiol. 2003, 41, 360–370. [Google Scholar] [CrossRef]
  17. Avogaro, P.; Bon, G.B.; Cazzolato, G. Presence of a modified low density lipoprotein in humans. Arteriosclerosis 1988, 8, 79–87. [Google Scholar] [CrossRef]
  18. Wu, R.; Lefvert, A.K. Autoantibodies against oxidized low density lipoproteins (oxLDL): Characterization of antibody isotype, subclass, affinity and effect on the macrophage uptake of oxLDL. Clin. Exp. Immunol. 1995, 102, 174–180. [Google Scholar] [CrossRef]
  19. Palinski, W.; Horkko, S.; Miller, E.; Steinbrecher, U.P.; Powell, H.C.; Curtiss, L.K.; Witztum, J.L. Cloning of monoclonal autoantibodies to epitopes of oxidized lipoproteins from apolipoprotein E-deficient mice. Demonstration of epitopes of oxidized low density lipoprotein in human plasma. J. Clin. Investig. 1996, 98, 800–814. [Google Scholar] [CrossRef]
  20. Tsimikas, S.; Palinski, W.; Witztum, J.L. Circulating autoantibodies to oxidized LDL correlate with arterial accumulation and depletion of oxidized LDL in LDL receptor-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 95–100. [Google Scholar] [CrossRef]
  21. Erkkila, A.T.; Narvanen, O.; Lehto, S.; Uusitupa, M.I.; Yla-Herttuala, S. Autoantibodies against oxidized low-density lipoprotein and cardiolipin in patients with coronary heart disease. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 204–209. [Google Scholar] [CrossRef] [PubMed]
  22. Van den Berg, V.J.; Vroegindewey, M.M.; Kardys, I.; Boersma, E.; Haskard, D.; Hartley, A.; Khamis, R. Anti-Oxidized LDL Antibodies and Coronary Artery Disease: A Systematic Review. Antioxidants 2019, 8, 484. [Google Scholar] [CrossRef] [PubMed]
  23. Itabe, H.; Takeshima, E.; Iwasaki, H.; Kimura, J.; Yoshida, Y.; Imanaka, T.; Takano, T. A monoclonal antibody against oxidized lipoprotein recognizes foam cells in atherosclerotic lesions. Complex formation of oxidized phosphatidylcholines and polypeptides. J. Biol. Chem. 1994, 269, 15274–15279. [Google Scholar] [CrossRef]
  24. Itabe, H.; Yamamoto, H.; Imanaka, T.; Shimamura, K.; Uchiyama, H.; Kimura, J.; Sanaka, T.; Hata, Y.; Takano, T. Sensitive detection of oxidatively modified low density lipoprotein using a monoclonal antibody. J. Lipid Res. 1996, 37, 45–53. [Google Scholar] [CrossRef]
  25. Itabe, H.; Yamamoto, H.; Suzuki, M.; Kawai, Y.; Nakagawa, Y.; Suzuki, A.; Imanaka, T.; Takano, T. Oxidized phosphatidylcholines that modify proteins. Analysis by monoclonal antibody against oxidized low density lipoprotein. J. Biol. Chem. 1996, 271, 33208–33217. [Google Scholar] [CrossRef]
  26. Imazu, M.; Ono, K.; Tadehara, F.; Kajiwara, K.; Yamamoto, H.; Sumii, K.; Tasaki, N.; Oiwa, J.; Shimohara, Y.; Gomyo, Y.; et al. Plasma levels of oxidized low density lipoprotein are associated with stable angina pectoris and modalities of acute coronary syndrome. Int. Heart J. 2008, 49, 515–524. [Google Scholar] [CrossRef]
  27. Holy, E.W.; Akhmedov, A.; Speer, T.; Camici, G.G.; Zewinger, S.; Bonetti, N.; Beer, J.H.; Luscher, T.F.; Tanner, F.C. Carbamylated Low-Density Lipoproteins Induce a Prothrombotic State Via LOX-1: Impact on Arterial Thrombus Formation In Vivo. J. Am. Coll. Cardiol. 2016, 68, 1664–1676. [Google Scholar] [CrossRef]
  28. Colas, R.; Sassolas, A.; Guichardant, M.; Cugnet-Anceau, C.; Moret, M.; Moulin, P.; Lagarde, M.; Calzada, C. LDL from obese patients with the metabolic syndrome show increased lipid peroxidation and activate platelets. Diabetologia 2011, 54, 2931–2940. [Google Scholar] [CrossRef]
  29. Van Tits, L.; De Graaf, J.; Hak-Lemmers, H.; Bredie, S.; Demacker, P.; Holvoet, P.; Stalenhoef, A. Increased levels of low-density lipoprotein oxidation in patients with familial hypercholesterolemia and in end-stage renal disease patients on hemodialysis. Lab. Investig. 2003, 83, 13–21. [Google Scholar] [CrossRef]
  30. Calzada, C.; Coulon, L.; Halimi, D.; Le Coquil, E.; Pruneta-Deloche, V.; Moulin, P.; Ponsin, G.; Véricel, E.; Lagarde, M. In vitro glycoxidized low-density lipoproteins and low-density lipoproteins isolated from type 2 diabetic patients activate platelets via p38 mitogen-activated protein kinase. J. Clin. Endocrinol. Metab. 2007, 92, 1961–1964. [Google Scholar] [CrossRef]
  31. Meisinger, C.; Baumert, J.; Khuseyinova, N.; Loewel, H.; Koenig, W. Plasma oxidized low-density lipoprotein, a strong predictor for acute coronary heart disease events in apparently healthy, middle-aged men from the general population. Circulation 2005, 112, 651–657. [Google Scholar] [CrossRef] [PubMed]
  32. Shen, M.Y.; Chen, F.Y.; Hsu, J.F.; Fu, R.H.; Chang, C.M.; Chang, C.T.; Liu, C.H.; Wu, J.R.; Lee, A.S.; Chan, H.C.; et al. Plasma L5 levels are elevated in ischemic stroke patients and enhance platelet aggregation. Blood 2016, 127, 1336–1345. [Google Scholar] [CrossRef] [PubMed]
  33. Chan, H.C.; Ke, L.Y.; Chu, C.S.; Lee, A.S.; Shen, M.Y.; Cruz, M.A.; Hsu, J.F.; Cheng, K.H.; Chan, H.C.; Lu, J.; et al. Highly electronegative LDL from patients with ST-elevation myocardial infarction triggers platelet activation and aggregation. Blood 2013, 122, 3632–3641. [Google Scholar] [CrossRef]
  34. Stellos, K.; Sauter, R.; Fahrleitner, M.; Grimm, J.; Stakos, D.; Emschermann, F.; Panagiota, V.; Gnerlich, S.; Perk, A.; Schonberger, T.; et al. Binding of oxidized low-density lipoprotein on circulating platelets is increased in patients with acute coronary syndromes and induces platelet adhesion to vascular wall in vivo—Brief report. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 2017–2020. [Google Scholar] [CrossRef] [PubMed]
  35. Nording, H.; Giesser, A.; Patzelt, J.; Sauter, R.; Emschermann, F.; Stellos, K.; Gawaz, M.; Langer, H.F. Platelet bound oxLDL shows an inverse correlation with plasma anaphylatoxin C5a in patients with coronary artery disease. Platelets 2016, 27, 593–597. [Google Scholar] [CrossRef] [PubMed]
  36. Moriel, P.; Okawabata, F.S.; Abdalla, D.S. Oxidized lipoproteins in blood plasma: Possible marker of atherosclerosis progression. IUBMB Life 1999, 48, 413–417. [Google Scholar] [CrossRef]
  37. Trip, M.D.; Cats, V.M.; van Capelle, F.J.; Vreeken, J. Platelet hyperreactivity and prognosis in survivors of myocardial infarction. N. Engl. J. Med. 1990, 322, 1549–1554. [Google Scholar] [CrossRef] [PubMed]
  38. Johnston, N.; Jernberg, T.; Lagerqvist, B.; Siegbahn, A.; Wallentin, L. Oxidized low-density lipoprotein as a predictor of outcome in patients with unstable coronary artery disease. Int. J. Cardiol. 2006, 113, 167–173. [Google Scholar] [CrossRef]
  39. Fuchs, I.; Frossard, M.; Spiel, A.; Riedmuller, E.; Laggner, A.N.; Jilma, B. Platelet function in patients with acute coronary syndrome (ACS) predicts recurrent ACS. J. Thromb. Haemost. 2006, 4, 2547–2552. [Google Scholar] [CrossRef]
  40. Dardik, R.; Varon, D.; Tamarin, I.; Zivelin, A.; Salomon, O.; Shenkman, B.; Savion, N. Homocysteine and oxidized low density lipoprotein enhanced platelet adhesion to endothelial cells under flow conditions: Distinct mechanisms of thrombogenic modulation. Thromb. Haemost. 2000, 83, 338–344. [Google Scholar]
  41. Liu, B.; Sidiropoulos, A.; Zhao, B.; Dierichs, R. Oxidized LDL damages endothelial cell monolayer and promotes thrombocyte adhesion. Am. J. Hematol. 1998, 57, 341–343. [Google Scholar] [CrossRef]
  42. Chatterjee, M.; Rath, D.; Schlotterbeck, J.; Rheinlaender, J.; Walker-Allgaier, B.; Alnaggar, N.; Zdanyte, M.; Muller, I.; Borst, O.; Geisler, T.; et al. Regulation of oxidized platelet lipidome: Implications for coronary artery disease. Eur. Heart J. 2017, 38, 1993–2005. [Google Scholar] [CrossRef] [PubMed]
  43. Vink, H.; Constantinescu, A.A.; Spaan, J.A.E. Oxidized Lipoproteins Degrade the Endothelial Surface Layer: Implications for Platelet-Endothelial Cell Adhesion. Circulation 2000, 101, 1500–1502. [Google Scholar] [CrossRef] [PubMed]
  44. Nergiz-Unal, R.; Lamers, M.M.E.; Kruchten, V.R.; Luiken, J.J.; Cosemans, J.; Glatz, J.F.C.; Kuijpers, M.J.E.; Heekerk, J.W.M. Signaling role of CD36 in platelet activation and thrombus formation on immobilized thrombospondin or oxidized low-density lipoprotein. J. Thromb. Haemost. 2011, 9, 1835–1846. [Google Scholar] [CrossRef]
  45. Zhao, B.; Rickert, C.H.; Filler, T.J.; Liu, B.; Verhallen, P.F.J.; Dierichs, R. Adhesion of washed blood platelets in vitro is advanced, accelerated, and enlarged by oxidized low-density lipoprotein. Am. J. Hematol. 1995, 49, 177–182. [Google Scholar] [CrossRef] [PubMed]
  46. Wraith, K.S.; Magwenzi, S.; Aburima, A.; Wen, Y.; Leake, D.; Naseem, K.M. Oxidized low-density lipoproteins induce rapid platelet activation and shape change through tyrosine kinase and Rho kinase–signaling pathways. Blood 2013, 122, 580–589. [Google Scholar] [CrossRef]
  47. Hartwich, J.; Dembinska-Kieć, A.; Gruca, A.; Motyka, M.; Partyka, L.; Skrzeczyńska, J.; Bzowska, M.; Pryjma, J.; Huber, J.; Leitinger, N.; et al. Regulation of platelet adhesion by oxidized lipoproteins and oxidized phospholipids. Platelets 2002, 13, 141–151. [Google Scholar] [CrossRef]
  48. Maschberger, P.; Bauer, M.; Baumann-Siemons, J.; Zangl, K.J.; Negrescu, E.V.; Reininger, A.J.; Siess, W. Mildly oxidized low density lipoprotein rapidly stimulates via activation of the lysophosphatidic acid receptor Src family and Syk tyrosine kinases and Ca2+ influx in human platelets. J. Biol. Chem. 2000, 275, 19159–19166. [Google Scholar] [CrossRef]
  49. Zhao, B.; Filler, T.J.; Rickert, C.H.; Dierichs, R. Decreased adhesion of oxidized LDL-stimulated platelets caused by cytochalasin D. Cell Tissue Res. 1995, 280, 183–188. [Google Scholar] [CrossRef]
  50. Sutipornpalangkul, W.; Unchen, S.; Sanvarinda, Y.; Chantharaksri, U. Modification of platelet shape change parameter by modified phospholipids in oxidized LDL. J. Med. Assoc. Thail. = Chotmaihet Thangphaet 2009, 92, 229–235. [Google Scholar]
  51. Zhao, B.; Dierichs, R.; Liu, B.; Holling-Rauβ, M. Functional morphological alterations of human blood platelets induced by oxidized low density lipoprotein. Thromb. Res. 1994, 74, 293–301. [Google Scholar] [CrossRef]
  52. Korporaal, S.J.; Van Eck, M.; Adelmeijer, J.; Ijsseldijk, M.; Out, R.; Lisman, T.; Lenting, P.J.; Van Berkel, T.J.; Akkerman, J.-W.N. Platelet activation by oxidized low density lipoprotein is mediated by CD36 and scavenger receptor-A. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2476–2483. [Google Scholar] [CrossRef] [PubMed]
  53. Siess, W.; Zangl, K.J.; Essler, M.; Bauer, M.; Brandl, R.; Corrinth, C.; Bittman, R.; Tigyi, G.; Aepfelbacher, M. Lysophosphatidic acid mediates the rapid activation of platelets and endothelial cells by mildly oxidized low density lipoprotein and accumulates in human atherosclerotic lesions. Proc. Natl. Acad. Sci. USA 1999, 96, 6931–6936. [Google Scholar] [CrossRef]
  54. Haserück, N.; Erl, W.; Pandey, D.; Tigyi, G.; Ohlmann, P.; Ravanat, C.; Gachet, C.; Siess, W. The plaque lipid lysophosphatidic acid stimulates platelet activation and platelet-monocyte aggregate formation in whole blood: Involvement of P2Y1 and P2Y12 receptors. Blood 2004, 103, 2585–2592. [Google Scholar] [CrossRef] [PubMed]
  55. Suttnar, J.; Otáhalová, E.; Cermák, J.; Dyr, J.E. Effects of malondialdehyde content in low density lipoproteins on platelet adhesion. Platelets 2006, 17, 92–99. [Google Scholar] [CrossRef] [PubMed]
  56. Daub, K.; Seizer, P.; Stellos, K.; Krämer, B.F.F.; Bigalke, B.; Schaller, M.; Fateh-Moghadam, S.; Gawaz, M.; Lindemann, S. Oxidized LDL-activated platelets induce vascular inflammation. Semin. Thromb. Hemost. 2010, 36, 146–156. [Google Scholar] [CrossRef]
  57. Murohara, T.; Scalia, R.; Lefer, A.M. Lysophosphatidylcholine promotes P-selectin expression in platelets and endothelial cells. Possible involvement of protein kinase C activation and its inhibition by nitric oxide donors. Circ. Res. 1996, 78, 780–789. [Google Scholar] [CrossRef]
  58. Takahashi, Y.; Fuda, H.; Yanai, H.; Akita, H.; Shuping, H.; Chiba, H.; Matsuno, K. Significance of Membrane Glycoproteins in Platelet Interaction with Oxidized Low-Density Lipoprotein. Semin. Thromb. Hemost. 1998, 24, 251–253. [Google Scholar] [CrossRef]
  59. Chen, K.; Febbraio, M.; Li, W.; Silverstein, R.L. A specific CD36-dependent signaling pathway is required for platelet activation by oxidized low-density lipoprotein. Circ. Res. 2008, 102, 1512–1519. [Google Scholar] [CrossRef]
  60. Nomura, S.; Shouzu, A.; Omoto, S.; Nishikawa, M.; Iwasaka, T.; Fukuhara, S. Activated platelet and oxidized LDL induce endothelial membrane vesiculation: Clinical significance of endothelial cell-derived microparticles in patients with type 2 diabetes. Clin. Appl. Thromb. Hemost. Off. J. Int. Acad. Clin. Appl. Thromb. Hemost. 2004, 10, 205–215. [Google Scholar] [CrossRef]
  61. Podrez, E.A.; Byzova, T.V.; Febbraio, M.; Salomon, R.G.; Ma, Y.; Valiyaveettil, M.; Poliakov, E.; Sun, M.; Finton, P.J.; Curtis, B.R.; et al. Platelet CD36 links hyperlipidemia, oxidant stress and a prothrombotic phenotype. Nat. Med. 2007, 13, 1086–1095. [Google Scholar] [CrossRef] [PubMed]
  62. Biswas, S.; Zimman, A.; Gao, D.; Byzova, T.V.; Podrez, E.A. TLR2 Plays a Key Role in Platelet Hyperreactivity and Accelerated Thrombosis Associated With Hyperlipidemia. Circ. Res. 2017, 121, 951–962. [Google Scholar] [CrossRef] [PubMed]
  63. Coleman, L.G.; Polanowska-Grabowska, R.K.; Marcinkiewicz, M.; Gear, A.R. LDL oxidized by hypochlorous acid causes irreversible platelet aggregation when combined with low levels of ADP, thrombin, epinephrine, or macrophage-derived chemokine (CCL22). Blood 2004, 104, 380–389. [Google Scholar] [CrossRef] [PubMed]
  64. Zhao, B.; Dierichs, R.; Harrach-Ruprecht, B.; Winterhorff, H. Oxidized LDL induces serotonin release from blood platelets. Am. J. Hematol. 1995, 48, 285–287. [Google Scholar] [CrossRef] [PubMed]
  65. Yanai, H.; Yoshida, H.; Hirowatari, Y.; Tomono, Y.; Tada, N. Oxidized low density lipoprotein elevates platelet serotonin release. Am. J. Hematol. 2007, 82, 686–687. [Google Scholar] [CrossRef] [PubMed]
  66. Puccetti, L.; Sawamura, T.; Pasqui, A.L.; Pastorelli, M.; Auteri, A.; Bruni, F. Atorvastatin reduces platelet-oxidized-LDL receptor expression in hypercholesterolaemic patients. Eur. J. Clin. Investig. 2005, 35, 47–51. [Google Scholar] [CrossRef]
  67. Bruni, F.; Pasqui, A.L.; Pastorelli, M.; Bova, G.; Cercignani, M.; Palazzuoli, A.; Sawamura, T.; Gioffre, W.R.; Auteri, A.; Puccetti, L. Different effect of statins on platelet oxidized-LDL receptor (CD36 and LOX-1) expression in hypercholesterolemic subjects. Clin. Appl. Thromb. Hemost. Off. J. Int. Acad. Clin. Appl. Thromb. Hemost. 2005, 11, 417–428. [Google Scholar] [CrossRef]
  68. Chen, R.; Chen, X.; Salomon, R.G.; McIntyre, T.M. Platelet Activation by Low Concentrations of Intact Oxidized LDL Particles Involves the PAF Receptor. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 363–371. [Google Scholar] [CrossRef]
  69. Yang, S.-H.H.; Li, Y.-T.T.; Du, D.-Y.Y. Oxidized low-density lipoprotein-induced CD147 expression and its inhibition by high-density lipoprotein on platelets in vitro. Thromb. Res. 2013, 132, 702–711. [Google Scholar] [CrossRef]
  70. Mahfouz, M.M.; Kummerow, F.A. Oxysterols and TBARS are among the LDL oxidation products which enhance thromboxane A2 synthesis by platelets. Prostaglandins Other Lipid Mediat. 1998, 56, 197–217. [Google Scholar] [CrossRef]
  71. Mahfouz, M.M.; Kummerow, F.A. Oxidized low-density lipoprotein (LDL) enhances thromboxane A(2) synthesis by platelets, but lysolecithin as a product of LDL oxidation has an inhibitory effect. Prostaglandins Other Lipid Mediat. 2000, 62, 183–200. [Google Scholar] [CrossRef]
  72. Strüßmann, T.; Tillmann, S.; Wirtz, T.; Bucala, R.; von Hundelshausen, P.; Bernhagen, J. Platelets are a previously unrecognised source of MIF. Thromb. Haemost. 2013, 110, 1004–1013. [Google Scholar] [PubMed]
  73. Naseem, K.M.; Goodall, A.H.; Bruckdorfer, K.R. Differential effects of native and oxidatively modified low-density lipoproteins on platelet function. Platelets 1997, 8, 163–173. [Google Scholar] [CrossRef] [PubMed]
  74. Volf, I.; Roth, A.; Cooper, J.; Moeslinger, T.; Koller, E. Hypochlorite modified LDL are a stronger agonist for platelets than copper oxidized LDL. FEBS Lett. 2000, 483, 155–159. [Google Scholar] [CrossRef]
  75. Weidtmann, A.; Scheithe, R.; Hrboticky, N.; Pietsch, A.; Lorenz, R.; Siess, W. Mildly oxidized LDL induces platelet aggregation through activation of phospholipase A2. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 1131–1138. [Google Scholar] [CrossRef]
  76. Vlasova, I.I.; Azizova, O.A.; Lopukhin Yu, M. Inhibitor analysis of LDL-induced platelet aggregation. Biochem. Biokhimiia 1997, 62, 307–311. [Google Scholar]
  77. Szuwart, T.; Dierichs, R.B.; Zhao, B. Vitamin E (α-tocopherol) does not inhibit platelet stimulation by oxidized low density lipoprotein in vitro. Am. J. Hematol. 1999, 60, 242–244. [Google Scholar] [CrossRef]
  78. Mehta, J.L.; Chen, L.Y. Reversal by high-density lipoprotein of the effect of oxidized low-density lipoprotein on nitric oxide synthase protein expression in human platelets. J. Lab. Clin. Med. 1996, 127, 287–295. [Google Scholar] [CrossRef]
  79. Tornvall, P.; Chirkova, L.; Toverud, K.D.; Horowitz, J.D.; Chirkov, Y. Native and oxidized low density lipoproteins enhance platelet aggregation in whole blood. Thromb. Res. 1999, 95, 177–183. [Google Scholar] [CrossRef]
  80. Ardlie, N.G.; Selley, M.L.; Simons, L.A. Platelet activation by oxidatively modified low density lipoproteins. Atherosclerosis 1989, 76, 117–124. [Google Scholar] [CrossRef]
  81. Rother, E.; Brandl, R.; Baker, D.L.; Goyal, P.; Gebhard, H.; Tigyi, G.; Siess, W. Subtype-selective antagonists of lysophosphatidic Acid receptors inhibit platelet activation triggered by the lipid core of atherosclerotic plaques. Circulation 2003, 108, 741–747. [Google Scholar] [CrossRef] [PubMed]
  82. Schmid, W.; Assinger, A.; Lee, A.; Bielek, E.; Koller, E.; Volf, I. Platelet-stimulating effects of oxidized LDL are not attributable to toxic properties of the lipoproteins. Thromb. Res. 2008, 122, 630–639. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, B.; Dierichs, R.; Miller, F.N.; Dean, W.L. Oxidized low density lipoprotein inhibits platelet plasma membrane Ca(2+)-ATPase. Cell Calcium 1996, 19, 453–458. [Google Scholar] [CrossRef]
  84. Schmidt, K.; Graier, W.F.; Kostner, G.M.; Mayer, B.; Kukovetz, W.R. Activation of soluble guanylate cyclase by nitrovasodilators is inhibited by oxidized low-density lipoprotein. Biochem. Biophys. Res. Commun. 1990, 172, 614–619. [Google Scholar] [CrossRef]
  85. Zuliani, V.; Tommasol, R.; Gaino, S.; Degan, M.; Cominacini, L.; Davoli, A.; Lechi, C.; Lechi, A.; Minuz, P. Oxidized low density lipoprotein (LDL) and platelet intracellular calcium: Interaction with nitric oxide. Platelets 1998, 9, 269–272. [Google Scholar] [CrossRef] [PubMed]
  86. Bruckdorfer, K.R.; Naseem, K.M.; Mohammadi, B.; Chirico, S.; Ettelaie, C.; Gleeson, A.; Young, T.; Adam, J.M.; Wilbourn, B.; Skinner, V.O.; et al. The influence of oxidized lipoproteins, oxidation products and antioxidants on the release of nitric oxide from the endothelium and the response of platelets to nitric oxide. BioFactors 1997, 6, 191–199. [Google Scholar] [CrossRef] [PubMed]
  87. Minuz, P.; Lechi, C.; Gaino, S.; Bonapace, S.; Fontana, L.; Garbin, U.; Paluani, F.; Cominacini, L.; Zatti, M.; Lechi, A. Oxidized LDL and reduction of the antiaggregating activity of nitric oxide derived from endothelial cells. Thromb. Haemost. 1995, 74, 1175–1179. [Google Scholar] [CrossRef]
  88. Berger, M.; Raslan, Z.; Aburima, A.; Magwenzi, S.; Wraith, K.S.; Spurgeon, B.E.J.; Hindle, M.S.; Law, R.; Febbraio, M.; Naseem, K.M. Atherogenic lipid stress induces platelet hyperactivity through CD36-mediated hyposensitivity to prostacyclin: The role of phosphodiesterase 3A. Haematologica 2020, 105, 808–819. [Google Scholar] [CrossRef]
  89. Magwenzi, S.; Woodward, C.; Wraith, K.S.; Aburima, A.; Raslan, Z.; Jones, H.; McNeil, C.; Wheatcroft, S.; Yuldasheva, N.; Febbriao, M.; et al. Oxidized LDL activates blood platelets through CD36/NOX2-mediated inhibition of the cGMP/protein kinase G signaling cascade. Blood 2015, 125, 2693–2703. [Google Scholar] [CrossRef]
  90. Yang, M.; Cooley, B.C.; Li, W.; Chen, Y.; Vasquez-Vivar, J.; Scoggins, N.O.; Cameron, S.J.; Morrell, C.N.; Silverstein, R.L. Platelet CD36 promotes thrombosis by activating redox sensor ERK5 in hyperlipidemic conditions. Blood 2017, 129, 2917–2927. [Google Scholar] [CrossRef]
  91. Badrnya, S.; Butler, L.M.; Söderberg-Naucler, C.; Volf, I.; Assinger, A. Platelets directly enhance neutrophil transmigration in response to oxidised low-density lipoprotein. Thromb. Haemost. 2012, 108, 719–729. [Google Scholar] [CrossRef] [PubMed]
  92. Badrnya, S.; Schrottmaier, W.C.; Kral, J.B.; Yaiw, K.C.; Volf, I.; Schabbauer, G.; Soderberg-Naucler, C.; Assinger, A. Platelets mediate oxidized low-density lipoprotein-induced monocyte extravasation and foam cell formation. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 571–580. [Google Scholar] [CrossRef] [PubMed]
  93. Lehr, H.A.; Olofsson, A.M.; Carew, T.E.; Vajkoczy, P.; von Andrian, U.H.; Hübner, C.; Berndt, M.C.; Steinberg, D.; Messmer, K.; Arfors, K.E. P-selectin mediates the interaction of circulating leukocytes with platelets and microvascular endothelium in response to oxidized lipoprotein in vivo. Lab. Investig. J. Tech. Methods Pathol. 1994, 71, 380–386. [Google Scholar]
  94. Korporaal, S.J.; Gorter, G.; van Rijn, H.J.; Akkerman, J.W. Effect of oxidation on the platelet-activating properties of low-density lipoprotein. Arteroscler. Thromb. Vasc. Biol. 2005, 25, 867–872. [Google Scholar] [CrossRef]
  95. Holm, T.; Damås, J.K.; Holven, K.; Nordøy, I.; Brosstad, F.R.; Ueland, T.; Währe, T.; Kjekshus, J.; Frøland, S.S.; Eiken, H.G.; et al. CXC-chemokines in coronary artery disease: Possible pathogenic role of interactions between oxidized low-density lipoprotein, platelets and peripheral blood mononuclear cells. J. Thromb. Haemost. 2003, 1, 257–262. [Google Scholar] [CrossRef]
  96. Wang, H.; Wang, Z.-H.H.; Kong, J.; Yang, M.-Y.Y.; Jiang, G.-H.H.; Wang, X.-P.P.; Zhong, M.; Zhang, Y.; Deng, J.-T.T.; Zhang, W. Oxidized low-density lipoprotein-dependent platelet-derived microvesicles trigger procoagulant effects and amplify oxidative stress. Mol. Med. 2012, 18, 159–166. [Google Scholar] [CrossRef]
  97. Zieseniss, S.; Zahler, S.; Müller, I.; Hermetter, A.; Engelmann, B. Modified Phosphatidylethanolamine as the Active Component of Oxidized Low Density Lipoprotein Promoting Platelet Prothrombinase Activity. J. Biol. Chem. 2001, 276, 19828–19835. [Google Scholar] [CrossRef]
  98. Levitan, I.; Volkov, S.; Subbaiah, P.V. Oxidized LDL: Diversity, patterns of recognition, and pathophysiology. Antioxid. Redox Signal. 2010, 13, 39–75. [Google Scholar] [CrossRef]
  99. Esterbauer, H.; Dieber-Rotheneder, M.; Waeg, G.; Striegl, G.; Jurgens, G. Biochemical, structural, and functional properties of oxidized low-density lipoprotein. Chem. Res. Toxicol. 1990, 3, 77–92. [Google Scholar] [CrossRef]
  100. Tang, W.H.; Hazen, S.L. Carbamylated Low-Density Lipoprotein and Thrombotic Risk in Chronic Kidney Disease. J. Am. Coll. Cardiol. 2016, 68, 1677–1679. [Google Scholar] [CrossRef]
  101. Obama, T.; Kato, R.; Masuda, Y.; Takahashi, K.; Aiuchi, T.; Itabe, H. Analysis of modified apolipoprotein B-100 structures formed in oxidized low-density lipoprotein using LC-MS/MS. Proteomics 2007, 7, 2132–2141. [Google Scholar] [CrossRef] [PubMed]
  102. Itabe, H.; Ueda, M. Measurement of plasma oxidized low-density lipoprotein and its clinical implications. J. Atheroscler. Thromb. 2007, 14, 1–11. [Google Scholar] [CrossRef] [PubMed]
  103. Berliner, J.A.; Subbanagounder, G.; Leitinger, N.; Watson, A.D.; Vora, D. Evidence for a role of phospholipid oxidation products in atherogenesis. Trends Cardiovasc. Med. 2001, 11, 142–147. [Google Scholar] [CrossRef]
  104. Samouilidou, E.C.; Karpouza, A.P.; Kostopoulos, V.; Bakirtzi, T.; Pantelias, K.; Petras, D.; Tzanatou-Exarchou, H.; Grapsa, E.J. Lipid abnormalities and oxidized LDL in chronic kidney disease patients on hemodialysis and peritoneal dialysis. Ren. Fail. 2012, 34, 160–164. [Google Scholar] [CrossRef] [PubMed]
  105. Byfield, F.J.; Tikku, S.; Rothblat, G.H.; Gooch, K.J.; Levitan, I. OxLDL increases endothelial stiffness, force generation, and network formation. J. Lipid Res. 2006, 47, 715–723. [Google Scholar] [CrossRef]
  106. Kowalsky, G.B.; Byfield, F.J.; Levitan, I. oxLDL facilitates flow-induced realignment of aortic endothelial cells. Am. J. Physiol.-Cell Physiol. 2008, 295, C332–C340. [Google Scholar] [CrossRef]
  107. Uittenbogaard, A.; Shaul, P.W.; Yuhanna, I.S.; Blair, A.; Smart, E.J. High density lipoprotein prevents oxidized low density lipoprotein-induced inhibition of endothelial nitric-oxide synthase localization and activation in caveolae. J. Biol. Chem. 2000, 275, 11278–11283. [Google Scholar] [CrossRef]
  108. Witztum, J.L.; Steinberg, D. Role of oxidized low density lipoprotein in atherogenesis. J. Clin. Investig. 1991, 88, 1785–1792. [Google Scholar] [CrossRef]
  109. Goldstein, J.L.; Ho, Y.; Basu, S.K.; Brown, M.S. Binding site on macrophages that mediates uptake and degradation of acetylated low density lipoprotein, producing massive cholesterol deposition. Proc. Natl. Acad. Sci. USA 1979, 76, 333–337. [Google Scholar] [CrossRef]
  110. Rosenfeld, M.E.; Palinski, W.; Ylä-Herttuala, S.; Butler, S.; Witztum, J.L. Distribution of oxidation specific lipid-protein adducts and apolipoprotein B in atherosclerotic lesions of varying severity from WHHL rabbits. Arterioscler. Off. J. Am. Heart Assoc. Inc. 1990, 10, 336–349. [Google Scholar] [CrossRef]
  111. Manning-Tobin, J.J.; Moore, K.J.; Seimon, T.A.; Bell, S.A.; Sharuk, M.; Alvarez-Leite, J.I.; de Winther, M.P.; Tabas, I.; Freeman, M.W. Loss of SR-A and CD36 activity reduces atherosclerotic lesion complexity without abrogating foam cell formation in hyperlipidemic mice. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 19–26. [Google Scholar] [CrossRef] [PubMed]
  112. Kato, R.; Mori, C.; Kitazato, K.; Arata, S.; Obama, T.; Mori, M.; Takahashi, K.; Aiuchi, T.; Takano, T.; Itabe, H. Transient increase in plasma oxidized LDL during the progression of atherosclerosis in apolipoprotein E knockout mice. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 33–39. [Google Scholar] [CrossRef] [PubMed]
  113. Virella, G.; Thorpe, S.R.; Alderson, N.L.; Stephan, E.M.; Atchley, D.; Wagner, F.; Lopes-Virella, M.F. Autoimmune response to advanced glycosylation end-products of human LDL. J. Lipid Res. 2003, 44, 487–493. [Google Scholar] [CrossRef] [PubMed]
  114. Scazzocchio, B.; Varì, R.; D’Archivio, M.; Santangelo, C.; Filesi, C.; Giovannini, C.; Masella, R. Oxidized LDL impair adipocyte response to insulin by activating serine/threonine kinases. J. Lipid Res. 2009, 50, 832–845. [Google Scholar] [CrossRef]
  115. Lopez, D.; Kobayashi, K.; Merrill, J.T.; Matsuura, E.; Lopez, L.R. IgG Autoantibodies against β2-Glycoprotein I Complexed with a Lipid Ligand Derived from Oxidized Low-Density Lipoprotein are Associated with Arterial Thrombosis in Antiphospholipid Syndrome. J. Immunol. Res. 2003, 10, 149426. [Google Scholar]
  116. Cimen, F.; Yildirmak, S.T.; Ergen, A.; Cakmak, M.; Dogan, S.; Yenice, N.; Sezgin, F. Serum Lipid, Lipoprotein and Oxidatively Modified Low Density Lipoprotein Levels in Active or Inactive Patients with Behcet’s Disease. Indian J. Dermatol. 2012, 57, 97–101. [Google Scholar]
  117. Nowak, B.; Madej, M.; Luczak, A.; Malecki, R.; Wiland, P. Disease Activity, Oxidized-LDL Fraction and Anti-Oxidized LDL Antibodies Influence Cardiovascular Risk in Rheumatoid Arthritis. Adv. Clin. Exp. Med. 2016, 25, 43–50. [Google Scholar] [CrossRef]
  118. Volf, I.; Roth, A.; Moeslinger, T.; Cooper, J.; Schmid, W.; Zehetgruber, M.; Koller, E. Stimulating effect of biologically modified low density lipoproteins on ADP-induced aggregation of washed platelets persists in absence of specific binding. Thromb. Res. 2000, 97, 441–449. [Google Scholar] [CrossRef]
  119. Drake, T.A.; Hannani, K.; Fei, H.H.; Lavi, S.; Berliner, J.A. Minimally oxidized low-density lipoprotein induces tissue factor expression in cultured human endothelial cells. Am. J. Pathol. 1991, 138, 601–607. [Google Scholar]
  120. Bochkov, V.N.; Mechtcheriakova, D.; Lucerna, M.; Huber, J.; Malli, R.; Graier, W.F.; Hofer, E.; Binder, B.R.; Leitinger, N. Oxidized phospholipids stimulate tissue factor expression in human endothelial cells via activation of ERK/EGR-1 and Ca(++)/NFAT. Blood 2002, 99, 199–206. [Google Scholar] [CrossRef]
  121. Canton, J.; Neculai, D.; Grinstein, S. Scavenger receptors in homeostasis and immunity. Nat. Rev. Immunol. 2013, 13, 621–634. [Google Scholar] [CrossRef] [PubMed]
  122. Mazurov, A.V.; Preobrazhensky, S.N.; Leytin, V.L.; Repin, V.S.; Smirnov, V.N. Study of low density lipoprotein interaction with platelets by flow cytofluorimetry. FEBS Lett. 1982, 137, 319–322. [Google Scholar] [CrossRef]
  123. Zimman, A.; Podrez, E.A. Regulation of platelet function by class B scavenger receptors in hyperlipidemia. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 2350–2356. [Google Scholar] [CrossRef] [PubMed]
  124. Neculai, D.; Schwake, M.; Ravichandran, M.; Zunke, F.; Collins, R.F.; Peters, J.; Neculai, M.; Plumb, J.; Loppnau, P.; Pizarro, J.C.; et al. Structure of LIMP-2 provides functional insights with implications for SR-BI and CD36. Nature 2013, 504, 172–176. [Google Scholar] [CrossRef] [PubMed]
  125. Crombie, R.; Silverstein, R. Lysosomal integral membrane protein II binds thrombospondin-1. Structure-function homology with the cell adhesion molecule CD36 defines a conserved recognition motif. J. Biol. Chem. 1998, 273, 4855–4863. [Google Scholar] [CrossRef] [PubMed]
  126. Ghosh, A.; Murugesan, G.; Chen, K.; Zhang, L.; Wang, Q.; Febbraio, M.; Anselmo, R.M.; Marchant, K.; Barnard, J.; Silverstein, R.L. Platelet CD36 surface expression levels affect functional responses to oxidized LDL and are associated with inheritance of specific genetic polymorphisms. Blood 2011, 117, 6355–6366. [Google Scholar] [CrossRef]
  127. Burkhart, J.M.; Vaudel, M.; Gambaryan, S.; Radau, S.; Walter, U.; Martens, L.; Geiger, J.; Sickmann, A.; Zahedi, R.P. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood 2012, 120, e73–e82. [Google Scholar] [CrossRef]
  128. Tandon, N.N.; Lipsky, R.H.; Burgess, W.H.; Jamieson, G.A. Isolation and characterization of platelet glycoprotein IV (CD36). J. Biol. Chem. 1989, 264, 7570–7575. [Google Scholar] [CrossRef]
  129. Asch, A.S.; Barnwell, J.; Silverstein, R.L.; Nachman, R.L. Isolation of the thrombospondin membrane receptor. J. Clin. Investig. 1987, 79, 1054–1061. [Google Scholar] [CrossRef]
  130. Endemann, G.; Stanton, L.W.; Madden, K.S.; Bryant, C.M.; White, R.T.; Protter, A.A. CD36 is a receptor for oxidized low density lipoprotein. J. Biol. Chem. 1993, 268, 11811–11816. [Google Scholar] [CrossRef]
  131. Handberg, A.; Levin, K.; Hojlund, K.; Beck-Nielsen, H. Identification of the oxidized low-density lipoprotein scavenger receptor CD36 in plasma: A novel marker of insulin resistance. Circulation 2006, 114, 1169–1176. [Google Scholar] [CrossRef] [PubMed]
  132. Terpstra, V.; van Berkel, T.J. Scavenger receptors on liver Kupffer cells mediate the in vivo uptake of oxidatively damaged red blood cells in mice. Blood 2000, 95, 2157–2163. [Google Scholar] [CrossRef] [PubMed]
  133. Podrez, E.A.; Poliakov, E.; Shen, Z.; Zhang, R.; Deng, Y.; Sun, M.; Finton, P.J.; Shan, L.; Gugiu, B.; Fox, P.L.; et al. Identification of a novel family of oxidized phospholipids that serve as ligands for the macrophage scavenger receptor CD36. J. Biol. Chem. 2002, 277, 38503–38516. [Google Scholar] [CrossRef] [PubMed]
  134. Kar, N.S.; Ashraf, M.Z.; Valiyaveettil, M.; Podrez, E.A. Mapping and characterization of the binding site for specific oxidized phospholipids and oxidized low density lipoprotein of scavenger receptor CD36. J. Biol. Chem. 2008, 283, 8765–8771. [Google Scholar] [CrossRef]
  135. Puente Navazo, M.D.; Daviet, L.; Ninio, E.; McGregor, J.L. Identification on human CD36 of a domain (155–183) implicated in binding oxidized low-density lipoproteins (Ox-LDL). Arterioscler. Thromb. Vasc. Biol. 1996, 16, 1033–1039. [Google Scholar] [CrossRef]
  136. Huang, M.M.; Bolen, J.B.; Barnwell, J.W.; Shattil, S.J.; Brugge, J.S. Membrane glycoprotein IV (CD36) is physically associated with the Fyn, Lyn, and Yes protein-tyrosine kinases in human platelets. Proc. Natl. Acad. Sci. USA 1991, 88, 7844–7848. [Google Scholar] [CrossRef]
  137. Chen, K.; Li, W.; Major, J.; Rahaman, S.O.; Febbraio, M.; Silverstein, R.L. Vav guanine nucleotide exchange factors link hyperlipidemia and a prothrombotic state. Blood 2011, 117, 5744–5750. [Google Scholar] [CrossRef]
  138. Karimi, P.; Rashtchizadeh, N. Oxidative Versus Thrombotic Stimulation of Platelets Differentially activates Signalling Pathways. J. Cardiovasc. Thorac. Res. 2013, 5, 61–65. [Google Scholar]
  139. Yang, M.; Kholmukhamedov, A.; Schulte, M.L.; Cooley, B.C.; Scoggins, N.O.; Wood, J.P.; Cameron, S.J.; Morrell, C.N.; Jobe, S.M.; Silverstein, R.L. Platelet CD36 signaling through ERK5 promotes caspase-dependent procoagulant activity and fibrin deposition in vivo. Blood Adv. 2018, 2, 2848–2861. [Google Scholar] [CrossRef]
  140. Berger, M.; Wraith, K.; Woodward, C.; Aburima, A.; Raslan, Z.; Hindle, M.S.; Moellmann, J.; Febbraio, M.; Naseem, K.M. Dyslipidemia-associated atherogenic oxidized lipids induce platelet hyperactivity through phospholipase Cgamma2-dependent reactive oxygen species generation. Platelets 2019, 30, 467–472. [Google Scholar] [CrossRef]
  141. Nieswandt, B.; Watson, S.P. Platelet-collagen interaction: Is GPVI the central receptor? Blood 2003, 102, 449–461. [Google Scholar] [CrossRef] [PubMed]
  142. Suzuki-Inoue, K.; Wilde, J.I.; Andrews, R.K.; Auger, J.M.; Siraganian, R.P.; Sekiya, F.; Rhee, S.G.; Watson, S.P. Glycoproteins VI and Ib-IX-V stimulate tyrosine phosphorylation of tyrosine kinase Syk and phospholipase Cgamma2 at distinct sites. Biochem. J. 2004, 378 Pt 3, 1023–1029. [Google Scholar] [CrossRef] [PubMed]
  143. Watson, S.P.; Auger, J.M.; McCarty, O.J.; Pearce, A.C. GPVI and integrin alphaIIb beta3 signaling in platelets. J. Thromb. Haemost. 2005, 3, 1752–1762. [Google Scholar] [CrossRef]
  144. Navarro-Nunez, L.; Langan, S.A.; Nash, G.B.; Watson, S.P. The physiological and pathophysiological roles of platelet CLEC-2. Thromb. Haemost. 2013, 109, 991–998. [Google Scholar] [PubMed]
  145. Navarese, E.P.; Kolodziejczak, M.; Winter, M.P.; Alimohammadi, A.; Lang, I.M.; Buffon, A.; Lip, G.Y.; Siller-Matula, J.M. Association of PCSK9 with platelet reactivity in patients with acute coronary syndrome treated with prasugrel or ticagrelor: The PCSK9-REACT study. Int. J. Cardiol. 2017, 227, 644–649. [Google Scholar] [CrossRef] [PubMed]
  146. Aoyama, T.; Sawamura, T.; Furutani, Y.; Matsuoka, R.; Yoshida, M.C.; Fujiwara, H.; Masaki, T. Structure and chromosomal assignment of the human lectin-like oxidized low-density-lipoprotein receptor-1 (LOX-1) gene. Biochem. J. 1999, 339 Pt 1, 177–184. [Google Scholar] [CrossRef]
  147. Chen, M.; Kakutani, M.; Naruko, T.; Ueda, M.; Narumiya, S.; Masaki, T.; Sawamura, T. Activation-dependent surface expression of LOX-1 in human platelets. Biochem. Biophys. Res. Commun. 2001, 282, 153–158. [Google Scholar] [CrossRef]
  148. Moore, K.J.; El Khoury, J.; Medeiros, L.A.; Terada, K.; Geula, C.; Luster, A.D.; Freeman, M.W. A CD36-initiated signaling cascade mediates inflammatory effects of beta-amyloid. J. Biol. Chem. 2002, 277, 47373–47379. [Google Scholar] [CrossRef]
  149. Kodama, T.; Freeman, M.; Rohrer, L.; Zabrecky, J.; Matsudaira, P.; Krieger, M. Type I macrophage scavenger receptor contains alpha-helical and collagen-like coiled coils. Nature 1990, 343, 531–535. [Google Scholar] [CrossRef]
  150. Rowley, J.W.; Oler, A.J.; Tolley, N.D.; Hunter, B.N.; Low, E.N.; Nix, D.A.; Yost, C.C.; Zimmerman, G.A.; Weyrich, A.S. Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes. Blood 2011, 118, e101–e111. [Google Scholar] [CrossRef]
  151. Motohashi, K.; Shibata, S.; Ozaki, Y.; Yatomi, Y.; Igarashi, Y. Identification of lysophospholipid receptors in human platelets: The relation of two agonists, lysophosphatidic acid and sphingosine 1-phosphate. FEBS Lett. 2000, 468, 189–193. [Google Scholar] [CrossRef]
  152. Cognasse, F.; Hamzeh, H.; Chavarin, P.; Acquart, S.; Genin, C.; Garraud, O. Evidence of Toll-like receptor molecules on human platelets. Immunol. Cell Biol. 2005, 83, 196–198. [Google Scholar] [CrossRef] [PubMed]
  153. Miller, Y.I.; Choi, S.H.; Wiesner, P.; Bae, Y.S. The SYK side of TLR4: Signalling mechanisms in response to LPS and minimally oxidized LDL. Br. J. Pharmacol. 2012, 167, 990–999. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, W.; Oh, S.; Koester, M.; Abramowicz, S.; Wang, N.; Tall, A.R.; Welch, C.L. Enhanced Megakaryopoiesis and Platelet Activity in Hypercholesterolemic, B6-Ldlr-/-, Cdkn2a-Deficient Mice. Circ. Cardiovasc. Genet. 2016, 9, 213–222. [Google Scholar] [CrossRef] [PubMed]
  155. Miao, W.M.; Vasile, E.; Lane, W.S.; Lawler, J. CD36 associates with CD9 and integrins on human blood platelets. Blood 2001, 97, 1689–1696. [Google Scholar] [CrossRef]
  156. Kautbally, S.; Lepropre, S.; Onselaer, M.B.; Le Rigoleur, A.; Ginion, A.; De Meester de Ravenstein, C.; Ambroise, J.; Boudjeltia, K.Z.; Octave, M.; Wera, O.; et al. Platelet Acetyl-CoA Carboxylase Phosphorylation: A Risk Stratification Marker That Reveals Platelet-Lipid Interplay in Coronary Artery Disease Patients. JACC Basic Transl. Sci. 2019, 4, 596–610. [Google Scholar] [CrossRef]
Figure 1. Major signalling events downstream platelet scavenger receptors induced by oxidatively modified LDL. Platelets are exposed to different types of oxidatively modified LDL, which interact with distinct pattern recognition receptors on their surface. Given that these particles have a complex and varied chemical composition, there is potential for particular LDL species to interact with different receptors. OxLDL has been shown to interact with CD36, SRA1, and potentially LOX-1. In contrast, electronegative LDL (fraction L5) has currently only been shown to interact with LOX-1. TLR2 has not been shown to bind with oxLDL but does interact with specific oxidised phospholipids that are known to be present in oxLDL (oxPCCD36). LOX-1 is located in the alpha granules of resting platelets and recruited to the platelet surface upon platelet activation. Once at the platelet surface, LOX-1 can bind electronegative LDL to stimulate signalling events (yellow) through PI3K and JNK to increase P-selectin expression and integrin aIIbb3 activation. SR-A1 binds oxLDL to initiate signalling through JNK and p38 (red), which can drive P-selectin expression, aIIbb3 activation, shape change, and TxA2. CD36 is constitutively associated with the Src family kinases Fyn and Lyn and, once ligated by oxLDL, incites a multitude of pathways (Blue). These drive shape change through RhoA and MLCP, P-selectin expression, and aIIbb3 activation through Vav1/3 and PLCg2. This pathway also leads to the activation of a PKC isoform, which is critical to the control of membrane phospholipids and inhibitory signalling (green). PKC induces the ROS generation through NOX2, which activates both ERK and phosphatidyl serine exposure, as well as the inhibition of PKG. PKC also activates PDE3A, which reduces cAMP concentrations and the activation of PKA. TLR2 does not bind oxLDL, but interaction with oxidised phospholipids leads to the MyD88-mediated activation of tyrosine kinase signalling, P-selectin expression, and aIIbb3 activation. Abbreviations: Phosphoinositide 3-kinase (PI3K), c-Jun N-terminal kinase (JNK), Myosin light chain phosphatase (MLCP), extracellular-signal regulated kinases (ERK), Protein kinase G (PKG), Protein kinase A (PKA), Protein kinase C (PKC), Phospholipase C gamma 2 (PLCg2), Vav1/3, NADPH Oxidase 2 (NOX2), p38.
Figure 1. Major signalling events downstream platelet scavenger receptors induced by oxidatively modified LDL. Platelets are exposed to different types of oxidatively modified LDL, which interact with distinct pattern recognition receptors on their surface. Given that these particles have a complex and varied chemical composition, there is potential for particular LDL species to interact with different receptors. OxLDL has been shown to interact with CD36, SRA1, and potentially LOX-1. In contrast, electronegative LDL (fraction L5) has currently only been shown to interact with LOX-1. TLR2 has not been shown to bind with oxLDL but does interact with specific oxidised phospholipids that are known to be present in oxLDL (oxPCCD36). LOX-1 is located in the alpha granules of resting platelets and recruited to the platelet surface upon platelet activation. Once at the platelet surface, LOX-1 can bind electronegative LDL to stimulate signalling events (yellow) through PI3K and JNK to increase P-selectin expression and integrin aIIbb3 activation. SR-A1 binds oxLDL to initiate signalling through JNK and p38 (red), which can drive P-selectin expression, aIIbb3 activation, shape change, and TxA2. CD36 is constitutively associated with the Src family kinases Fyn and Lyn and, once ligated by oxLDL, incites a multitude of pathways (Blue). These drive shape change through RhoA and MLCP, P-selectin expression, and aIIbb3 activation through Vav1/3 and PLCg2. This pathway also leads to the activation of a PKC isoform, which is critical to the control of membrane phospholipids and inhibitory signalling (green). PKC induces the ROS generation through NOX2, which activates both ERK and phosphatidyl serine exposure, as well as the inhibition of PKG. PKC also activates PDE3A, which reduces cAMP concentrations and the activation of PKA. TLR2 does not bind oxLDL, but interaction with oxidised phospholipids leads to the MyD88-mediated activation of tyrosine kinase signalling, P-selectin expression, and aIIbb3 activation. Abbreviations: Phosphoinositide 3-kinase (PI3K), c-Jun N-terminal kinase (JNK), Myosin light chain phosphatase (MLCP), extracellular-signal regulated kinases (ERK), Protein kinase G (PKG), Protein kinase A (PKA), Protein kinase C (PKC), Phospholipase C gamma 2 (PLCg2), Vav1/3, NADPH Oxidase 2 (NOX2), p38.
Ijms 23 09199 g001
Table 1. Functional effects of oxLDL and derivates on platelets. OxLDL—oxidised LDL; CuSO4—copper (II) sulphate; HUVEC—human umbilical vein endothelial cells; WT—wildtype; Acyl-LPA—acylated lysophosphatidic acid; FeSO4—iron (II) sulphate; MDA—malondialdehyde; HOCL—hypochlorus acid; Lyso-PC—lysophosphatidylcholine; MPO—myeloperoxidase; oxPCCD36—oxidised phosphocholine specific for CD36; ADP—adenosine diphosphate; METS-LDL—metabolic syndrome-associated LDL; DMII-LDL—diabetes mellitus II-associated LDL; SIN-1-LDL—3-(N-morpholino)sydnonimine-LDL; Ac-LDL—acetylated LDL; PRP—platelet-rich plasma; CuCL2—copper(II) chloride; Ca2+—calcium; NO—nitrix oxide; Cu2+—copper; AAPH—2,2′-Azobis-(2-amidinopropane hydrochloride).
Table 1. Functional effects of oxLDL and derivates on platelets. OxLDL—oxidised LDL; CuSO4—copper (II) sulphate; HUVEC—human umbilical vein endothelial cells; WT—wildtype; Acyl-LPA—acylated lysophosphatidic acid; FeSO4—iron (II) sulphate; MDA—malondialdehyde; HOCL—hypochlorus acid; Lyso-PC—lysophosphatidylcholine; MPO—myeloperoxidase; oxPCCD36—oxidised phosphocholine specific for CD36; ADP—adenosine diphosphate; METS-LDL—metabolic syndrome-associated LDL; DMII-LDL—diabetes mellitus II-associated LDL; SIN-1-LDL—3-(N-morpholino)sydnonimine-LDL; Ac-LDL—acetylated LDL; PRP—platelet-rich plasma; CuCL2—copper(II) chloride; Ca2+—calcium; NO—nitrix oxide; Cu2+—copper; AAPH—2,2′-Azobis-(2-amidinopropane hydrochloride).
EnvironmentFunctional ResponseMethod of
Oxidation
SpeciesPlatelet
Preparation
Reference
AdhesionIn vitroOxLDL induces increased platelet adhesion to collagen and HUVECs under flow/static conditionsCuSO4HumanWhole blood/washed platelets[34,40,41,42]
In vitroOxLDL induces increased platelet adhesion to fibrinogen under flow conditionsElectronegative LDL—fraction L5, isolated from patientsHumanWashed platelets[33]
In vivoOxLDL injection increases platelet endothelial adhesion after carotid ligation in WT animalsCuSO4MouseWhole blood[34]
In vivoOxLDL injection increases platelet endothelial adhesions in cremaster muscle blood vesselsCuSO4HamsterWhole blood[43]
In vitroOxLDL increases platelet spreading and pseudopodia formation under static conditionsCuSO4/FeSO4HumanWashed platelets[44,45,46,47,48,49,50,51,52]
In vitroOxLDL increases adhesion of platelets to oxLDL co-coated collagen matrix under arterial flow conditionsCuSO4HumanWhole blood[44]
In vitroMildly/extensively oxidised LDL causes shape changes in platelet suspensionsO2 oxidation/CuSO4/Acyl-LPAHumanWashed platelets[46,53,54]
In vitroIncreased platelet adhesion to MDA/HOCL modified-oxLDL under static conditionsMDA modified oxLDL/HOCL-LDLHumanWashed platelets[55]
Degranulation & SecretionIn vitroOxLDL increases platelet P-selectin expressionCuSO4/LysoPCHuman/catWhole blood/washed platelets[34,56,57,58,59]
In vitroPositive correlation of platelet P-selectin, sCD40L and oxLDL levelsMPO-mediated LDL oxidationHumanWashed platelets[60]
In vitroOxPCCD36 induces increased platelet P-selectin expressionOxPCCD36HumanWashed platelets[61]
In vitroOxPCCD36 induces P-selectin expressionKODA-PCMouseWashed platelets[62]
In vitroOxLDL potentiates ADP-induced P-selectin expression in plateletsHOCL-LDLHumanWashed platelets[63]
In vitroOxLDL induces platelet serotonin secretionCuSO4HumanWashed platelets[64,65]
In vitroOxLDL potentiates ADP-induced serotonin secretion in plateletsHOCL–LDLHumanWashed platelets[63]
In vitroBinding of oxLDL on dyslipidaemic platelets linked to increased P-selectin expressionoxLDL-associated dyslipidaemiaHumanWashed platelets/whole blood[34,66,67]
In vitroOxLDL potentiates the effect of thrombin on platelet P-selectin expressionCuSO4HumanWashed platelets[68]
In vitroOxLDL induces platelet CD147 releaseCuSO4HumanWashed platelets[69]
In vitroOxLDL induces platelet thromboxane A2 generationCuSO4HumanWashed platelets[70,71]
In vitroOxLDL induces platelet thromboxane B2 generationMETS-LDL/DMII LDL/glycooxidised LDLHumanWashed platelets[28,30]
In vitroOxLDL induces platelet CXCL12 releaseCuSO4HumanWashed platelets[72]
AggregationIn vitroOxLDL induces platelet aggregationCuSO4/Ac-LDL/HOCL-LDL/SIN-1-LDL/Acyl-LPA/O2 oxidation/Electronegative LDL—Fraction L5/METS-LDL/DMII-LDL/HumanWashed platelets/
PRP
[28,46,54,73,74,75,76,77,78]
In vitroOxLDL potentiates the effects of ADP on platelet aggregationCuSO4/HOCL-LDL/Electronegative LDL/oxLDL-associated APOE−/− dyslipidaemiaHuman/mouseWashed plt./PRP/PRP and washed platelet in ratio 1:1[33,63,74,79,80,81]
In vitroOxLDL potentiates the effects of thrombin aggregationCuSO4/HOCL-LDLHumanWashed platelets[74,82]
In vitroOxPCCD36 induces increased fibrinogen binding in plateletsOxPCCD36HumanWashed platelets[61]
In vitroOxLDL potentiates the effects of epinephrine and thrombin in aggregationHOCL-LDLHumanWashed platelets[63]
In vitrooxLDL increases active conformation of aIIbß3CuSO4Human/mouseWashed platelets[42,62]
Ca2+
mobilisation
In vitroOxLDL induces intracellular Ca2+ mobilisationCuSO4/HOCL-LDLHumanWashed platelets[63,68,83]
DisinhibitionIn vitroOxLDL inhibits activation of guanylate cyclaseCuCl2BovineWashed platelets
In vitroOxLDL decreases platelets sensitivity to NO in thrombin aggregationCuSO4/O2 OxidationHumanWashed platelets[84,85,86]
In vitroOxLDL reduces the effect of endothelial cell-derived NO on plateletsNot definedHumanWashed platelets[87]
In vivo/In vitrooxLDL, oxPCCD36 and oxPCCD36-associated highfat diet induces PDE3A and leads to reduced cAMP signallingCuSO4/oxPCCD36/High fat dietHuman/mouseWashed platelet, PRP, whole blood[88]
In vivo/In vitrooxLDL-induced ROS production inhibits the effects of 8pCPT-cGMPCuSO4Human/mouseWhole blood[89]
ROS-generationIn vitrooxLDL induces ROS generationCuSO4HumanWashed platelets[42,89,90]
Cell-Cell-interactionsIn vitro/In vivoOxLDL increases platelet-leucocyte interaction and platelet-induced leucocyte transmigrationHOCL/CuSO4Human/mousePRP/leucocyte enriched PRP[91,92]
In vivoOxLDL increases platelet-leucocyte interactions and P-selectin-dependent leucocyte endothelial interactionsCu2+HamsterWhole blood[93]
In vitroOxLDL Increases platelet-monocyte interactionsAcyl-LPAHumanWhole blood[54]
In vitroOxLDL treated platelets induce ICAM-1 expression in endothelial cellsHOCLHumanWashed platelets[56]
MiscellaneousIn vivoInjection of OxLDL shortens tailbleeding timeElectronegative LDL, fraction L5, isolated from patientsMouseWhole blood[33]
In vitroOxidised LDL attenuates Fibrinogen bindingFeSO4 (>30% assessed by REM)HumanWashed platelets[94]
In vitroOxLDL increases CXC5 release in platelets from CAD patientsCuSO4HumanPRP[95]
In vitroOxLDL induces microvesicle release and increases platelet phosphatidylserine exposureCuSO4HumanWashed platelets[96]
In vitroOxLDL increases platelet prothrombinase activityCuSO4 + AAPH/MPO oxidised LDLHumanWashed platelets[97]
In vivooxLDL injection increases time to occlusion in a ferric chloride thrombosis model carotid arteryCuSO4MouseWhole blood[42]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Berger, M.; Naseem, K.M. Oxidised Low-Density Lipoprotein-Induced Platelet Hyperactivity—Receptors and Signalling Mechanisms. Int. J. Mol. Sci. 2022, 23, 9199. https://doi.org/10.3390/ijms23169199

AMA Style

Berger M, Naseem KM. Oxidised Low-Density Lipoprotein-Induced Platelet Hyperactivity—Receptors and Signalling Mechanisms. International Journal of Molecular Sciences. 2022; 23(16):9199. https://doi.org/10.3390/ijms23169199

Chicago/Turabian Style

Berger, Martin, and Khalid M. Naseem. 2022. "Oxidised Low-Density Lipoprotein-Induced Platelet Hyperactivity—Receptors and Signalling Mechanisms" International Journal of Molecular Sciences 23, no. 16: 9199. https://doi.org/10.3390/ijms23169199

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop