Next Article in Journal
EDS1-Dependent Cell Death and the Antioxidant System in Arabidopsis Leaves is Deregulated by the Mammalian Bax
Previous Article in Journal
Photochemotherapy Induces Interferon Type III Expression via STING Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Scavenger Receptors as Biomarkers and Therapeutic Targets in Cardiovascular Disease

by
Gary A. Cuthbert
1,*,
Faheem Shaik
2,
Michael A. Harrison
3,
Sreenivasan Ponnambalam
2 and
Shervanthi Homer-Vanniasinkam
1
1
Faculty of Medicine and Health, University of Leeds, Leeds LS2 9JT, UK
2
School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
3
School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
*
Author to whom correspondence should be addressed.
Cells 2020, 9(11), 2453; https://doi.org/10.3390/cells9112453
Submission received: 26 October 2020 / Revised: 6 November 2020 / Accepted: 9 November 2020 / Published: 10 November 2020
(This article belongs to the Section Cellular Immunology)

Abstract

:
The process of atherosclerosis leads to the formation of plaques in the arterial wall, resulting in a decreased blood supply to tissues and organs and its sequelae: morbidity and mortality. A class of membrane-bound proteins termed scavenger receptors (SRs) are closely linked to the initiation and progression of atherosclerosis. Increasing interest in understanding SR structure and function has led to the idea that these proteins could provide new routes for cardiovascular disease diagnosis, management, and treatment. In this review, we consider the main classes of SRs that are implicated in arterial disease. We consider how our understanding of SR-mediated recognition of diverse ligands, including modified lipid particles, lipids, and carbohydrates, has enabled us to better target SR-linked functionality in disease. We also link clinical studies on vascular disease to our current understanding of SR biology and highlight potential areas that are relevant to cardiovascular disease management and therapy.

Graphical Abstract

1. Introduction

1.1. Oxidized Low-Density Lipoprotein and Atherogenesis

Atherosclerosis is the main biological process underlying cardiovascular disease (CVD) development and pathology [1]. Atherosclerosis is a multifactorial, progressive, and chronic inflammatory process characterized by the accumulation of lipid and fibrous elements within the arterial wall, manifesting macroscopically as an atherosclerotic plaque (Figure 1). The key events in atherosclerotic plaque initiation and progression are depicted in Figure 1. Atherosclerosis is initiated by abnormally high circulating blood low-density lipoprotein (LDL) levels (LDL-cholesterol >40 mg/dL) [2], leading to LDL accumulation within the sub-endothelial matrix of arterial blood vessel walls, usually operating at a higher blood pressure of 80–120 mm Hg [3]. The LDL deposits within arterial vessel walls become chemically modified by free radicals produced by vascular cells, such as the endothelium, vascular smooth muscle, and infiltrating immune cells. These new lipid particle species are termed minimally modified LDL (mmLDL), modified LDL (mLDL), and oxidized LDL (oxLDL). Notably, oxLDL promotes a powerful proinflammatory response by different cell types, including the production of cell surface adhesion molecules and chemotactic factors by the endothelium. OxLDL indirectly promotes the recruitment of circulating blood monocytes by binding to the activated endothelium followed by trans-endothelial migration (TEM) into the sub-endothelial intima, where the monocytes differentiate into macrophages. Here, the accumulation of oxLDL within the vascular wall further causes macrophage scavenger receptors (SRs) to bind and promote oxLDL internalization, leading to the development of lipid-laden foam cells, i.e., the early stages of atherosclerosis (atherogenesis) [4,5]. The vascular smooth muscle cell (VSMC) layer underlying the intima can also respond to endothelial-derived factors secreted in response to oxLDL accumulation [6]. VSMCs also express SRs, which can enable oxLDL internalization and contribution to more lipid-laden foam cell development, albeit to a lesser extent than macrophages [6].

1.2. The Role of Atherosclerotic Plaque in CVD

The clinical consequences of atherosclerosis relate principally to ischaemia and acute thrombo-occlusive events at the sites of plaque deposition. Thrombo-occlusive events in the majority of cases result from plaque rupture [7]. The progression of atherosclerotic plaque from early asymptomatic lesions to those which are high risk of rupture (i.e., vulnerable plaque) is well described [8,9]. Vulnerable plaque is characterized by a large necrotic core with an overlying thin fibrous cap heavily infiltrated by foamy macrophages [10]. Observational studies suggest that expansion of the necrotic core precedes plaque rupture [10,11]. Inter-plaque haemorrhage has been identified as a critical step leading to expansion of the necrotic core, where the accumulation of erythrocytes within the core leads to an increased free cholesterol content and excessive macrophage infiltration [8]. Subsequent plaque rupture exposes leukocytes, platelets, and circulating blood factors to underlying thrombogenic proteins, e.g., Von Willebrand factor (VWF), leading to thrombus formation. Recent studies have highlighted the phenomena of coronary artery plaque erosion (without rupture) and the formation of “white thrombus” in patients with acute coronary syndrome (ACS) [12,13]. Plaque erosion has been previously understood to account for one fifth of ACS; however, the most recent data suggests this has increased to one third [14]. The working hypothesis to explain the rise of plaque erosion in ACS is that advances in lipid control have led to a shift in plaque morphology from the classical lipid-rich rupture-prone plaque to a plaque that is low in lipid content and rich in proteoglycans, glycosaminoglycans, and VSMCs. The mechanism of thrombus formation in eroded plaque differs from ruptured plaque. Briefly, the activation and subsequent desquamation and apoptosis of endothelial cells due to disturbed flow leads to the recruitment of neutrophils to the exposed basement membrane. Recruited neutrophils congregate and in turn degranulate, leading to the formation of neutrophil extracellular traps (NETs). NETs comprise mainly of myeloperoxidase and tissue factor amongst other extruded proteins, all of which contribute to the formation of leucocyte-rich white thrombus by promoting leucocyte and platelet recruitment [12,13,14]. The formation of NETs may also be promoted by circulating oxLDL as shown in a recently published in vitro study [15]. In addition, the authors found that oxLDL and NETs exerted synergistic proinflammatory effects on endothelial cells.

1.3. A Brief Introduction to SRs

SRs are defined as integral membrane proteins present at the cell surface, which typically bind multiple ligands to promote clearance or harmful non-self or altered-self substance. SR activity usually requires sensing and/or internalization of such substances, which ultimately leads to the elimination of these substances [16]. In 1979, Goldstein, Brown and co-workers first described scavenger receptors (SRs) whilst investigating how low-density lipoprotein (LDL) particles promote foam cell development in atherosclerosis [17,18]. This led to the discovery that macrophages exhibit high-affinity binding sites, which enable recognition of modified and negatively charged LDL particles, thus promoting lipid particle internalization and subsequent formation of lipid-enriched foam cells [18]. Different SRs have since been identified and categorized into 10 distinct classes based on primary structure (Table 1) [19]. SRs are defined by common properties, such as the recognition and binding to ligands, such as lipoprotein particles, phospholipids, apoptotic cells, carbohydrates, and cholesterol esters; however, the primary consensus protein sequence within each class bears little or no homology to other classes [19]. SRs are expressed largely in vascular and immune cells under physiological or pathological conditions. Different immune cells express SRs, such as macrophages, which are linked to the initiation and progression of atherosclerosis. The wide spectrum of SR expression has been linked to pathological conditions, such as degenerative brain disease [20,21], malignancy [22], and systemic inflammatory states, such as sepsis [23], rheumatoid arthritis [24], and renal vasculitis [25]. SR function in regulating atherosclerosis is a major focus of research in CVD diagnosis and therapy. Current work on SR regulation of atherosclerosis focuses largely on five SR classes: SR-A; SR-B (CD36 (cluster of differentiation 36)); SR-E (lectin-like oxidized LDL receptor-1 (LOX-1)); SR-G (CXCL16); SR-J (receptor for advanced glycation end-products (RAGEs)) (Figure 2). This review article aims to highlight SR properties and functionality in the diagnosis and treatment of atherosclerosis linked to CVD as summarized in Table 1.

2. SRs as Biomarkers in CVD

The association of SRs with CVD status is well established, and multiple genes and isoforms have been highlighted as potential markers for use in genetic screening for disease risk, or as diagnostic or prognostic biomarkers. Screening patients with a genetic predisposition to CVD has potentially far-reaching benefits in disease prevention and early management. Polymorphisms in genes encoding both CD36 and LOX-1 proteins have been associated with increased overall CVD risk [58,59,60]. A single-nucleotide polymorphism (SNP) of the OLR1 (LOX-1, SR-E1) allele has been shown to increase steady-state LOX-1 levels in vitro, suggesting that this polymorphism may contribute to disease initiation and/or progression; OLR1 genotyping could be used for CVD risk stratification [60]. Moreover, a study of a group of 150 patients with acute myocardial infarction (MI) vs. a control group of 103 healthy individuals identified SNPs in OLR1 associated with increased risk of MI [61]. Cohort studies comparing healthy individuals and coronary artery disease (CAD) patient groups have shown association between CD36 polymorphisms with increased lipid oxidation, CVD risk, and accelerated development of CAD [58,59,62].
Much has been done to investigate the diagnostic value of quantifying SR levels in ACS. Circulating concentrations of soluble SR-derived proteins in serum, and SR levels in specific immune cell types have been studied. The use of an “SR-A index” quantifies monocyte SR-A expression in peripheral blood smears from patients with CAD: this demonstrates that an increase in this SR-A index is associated with altered atherosclerotic plaque status, leading to destabilization, i.e., disruption, fissure, erosion [63]. Furthermore, such an SR-A index could be used to stratify plaque morphology, helping to inform therapeutic strategies for CAD [64,65]. Similar studies show an association between circulating monocyte CD36 levels and atheroma burden in CAD patients [66]. Overexpressed SR-A in macrophages in atherosclerotic plaque has been targeted using SR-A-specific nanoparticles in an apolipoprotein-E (APO-E)-null mouse model in order to identify vulnerable plaques using magnetic resonance imaging with some success [67]. A similar study using CD36-targeted nanovesicles to quantify plaque burden in low-density lipoprotein receptor (LDLR)-null mice showed similarly promising results [68]. The use of SR-targeted probes in plaque identification, risk stratification, and quantification is an interesting avenue of ongoing research.
Measuring biomarkers in serum is widely used in clinical settings to ACS diagnosis and stratification for treatment [69]. Proteolytically derived soluble fragments or splice isoforms of CD36, LOX-1, CXCL16, and RAGE are potential biomarkers for a range of CVD conditions, including ACS, cardiac failure, cardiomyopathy, and carotid artery disease. A summary of studies investigating the this potential application is seen in Table 2. Baseline serum levels of soluble CD36 (sCD36) have been shown in three recent cohort studies to have no association with the development of CAD in healthy subjects or plaque burden in known CAD patients [70,71]. Baseline levels of sCD36 were also found to not be associated with carotid atherosclerosis in a recently performed cross-sectional study [72]. In contrast, a recent case control study of patients with CAD found an association between increased sLOX-1 levels and burden of CAD, compared with healthy controls [73]. A similar study of metabolic syndrome patients found that baseline sLOX-1 levels were higher in patients with CAD, implying a potential benefit in the measurement of baseline sLOX-1 levels in at-risk cohorts to risk stratify for CAD development [74].
A small clinical study on 67 patients found that serum-soluble LOX-1 (sLOX-1) levels increased significantly in ACS patients compared to a group of patients with stable angina pectoris [75]. A combined index of serum sLOX-1 and cardiac troponin (cTn) levels improved diagnostic accuracy in ACS compared with diagnostic tests carried out in isolation. The same study also found that measuring sLOX-1 levels in ACS was a reliable test compared to a standard diagnosis of ACS [76]. A study on 107 patients undergoing coronary stenting found that measuring sLOX-1 levels was more informative and accurate compared with cTn measurements in ACS diagnosis; a combined sLOX-1/cTn index was a significantly better biomarker tool than measuring sLOX-1 levels alone [77]. The potential ability of sLOX-1 in identifying the most severe acute coronary events was demonstrated in a recently published study comparing levels of sLOX-1 in aspirated coronary thrombus from non-ST elevation and ST-elevation myocardial infarction patients [78]. The studies that compare cTn and sLOX-1 as biomarkers for ACS have consistently shown that sLOX-1 levels are raised earlier compared with cTn [76,77]. The hypothesis to explain this phenomenon is that sLOX-1 is released at the point of plaque rupture and platelet activation during thrombus formation. Based on this hypothesis and the findings from Lee et al. [78], there may be a role for sLOX-1 as a marker for coronary micro-thrombosis peri-procedurally at the point of percutaneous coronary intervention; however, there are no data currently to support this. Baseline sLOX-1 levels may also be of benefit as a prognostic biomarker in stable CAD. A recently published study of 833 stable CAD patients found a positive correlation between baseline sLOX-1 levels and major adverse cardiovascular events [79]. The relationship between LOX-1 and myocardial reperfusion injury has been investigated in several mouse models of myocardial ischaemia; however, this has not been further investigated in human studies. Mouse models have demonstrated that LOX-1-null mice exhibit significantly less reperfusion injury and significantly preserve cardiac function compared with wild-type mice [80,81]. In vitro and in vivo heart failure models suggest sLOX-1 release by cardiac myocytes promotes apoptosis [82,83]. To evaluate sLOX-1 as a biomarker for systolic heart failure, serum sLOX-1 levels were measured in 55 patients with systolic heart failure and compared to a control group of 25 individuals. Serum sLOX-1 levels were found to negatively correlate with left ventricular ejection fraction, particularly in patients with ischemia-related aetiology [84]. The relationship between LOX-1 and C-reactive protein (CRP) in atherogenesis and their role as biomarkers for cardiovascular events is an area of ongoing interest. CRP is a widely used biomarker for inflammation, which is also highly predictive of recurrent cardiovascular events [85]. In vitro studies have demonstrated that CRP induces endothelial cell LOX-1 expression, leading to increased monocyte adhesion and oxLDL uptake [86]. The release of sLOX-1 from monocytes is stimulated by incubation CRP, via the activation of tumour necrosis factor-α in vitro [87]. These studies demonstrate the synergistic effects of CRP and LOX-1 and their potential for use as biomarkers in tandem. The evidence supporting the potential use of sLOX-1 as a biomarker for stroke is mounting but is not as significant compared with CAD and ACS. Multiple small cohort studies comparing serum sLOX-1 levels between patients presenting with acute stroke and controls found that serum sLOX-1 levels were significantly raised in ischaemic stroke and haemorrhagic stroke, and that higher sLOX-1 levels independently predicted poorer neurological outcomes [88,89,90]. A recently published prospective population study of 4703 participants found that higher baseline sLOX-1 levels were associated with an increased incidence of ischaemic stroke over a 16.5-year mean follow-up [91]. In participants requiring carotid endarterectomy (n = 202), higher baseline sLOX-1 levels were associated with an increased incidence of recurrent ischaemic events post-operatively. The source of increased sLOX-1 levels and the timing of sampling in acute stroke are areas of controversy. Most studies have hypothesized that sLOX-1 is released from the offending atherosclerotic plaque; however, the contribution of cerebral ischaemia to sLOX-1 levels is unknown. Skarpengland et al. recently examined carotid plaque OLR1 gene expression and plasma sLOX-1 levels in patients referred for carotid endarterectomy [92]. Predictably, circulating sLOX-1 levels and arterial OLR1 expression were increased in patients with carotid plaque compared with healthy controls. Interestingly, sLOX-1 levels were comparably increased in patients with thrombotic ischaemic stroke and embolic stroke secondary to atrial fibrillation, raising the possibility that the ischaemic event may contribute to sLOX-1 release. The embolic stroke patients must have harboured a degree of carotid plaque, however, to warrant a referral for carotid endarterectomy.
The SR member, CXCL16, is a membrane-bound chemokine, which binds oxLDL and promotes proinflammatory responses linked to atherosclerosis development [93]. Serum-soluble CXCL16 (sCXCL16) has been evaluated as a potential biomarker in CAD, inflammatory cardiomyopathy, and carotid artery disease. In 118 patients presenting with an acute ischaemic stroke, serum sCXCL16 levels were significantly higher in patients with vulnerable carotid plaques, a higher degree of luminal stenosis, and increased intimal layer thickness [94]. Other clinical studies have also shown that measuring serum sCXCL16 concentration can improve diagnostic accuracy when combined with cTn and sLOX-1 in ACS, and that may be of benefit in with type 2 diabetes mellitus (T2DM) patients [49,95]. As a prognostic biomarker, a study of 1351 ACS patients found that a single measurement of serum sCXCL16 within 24 h of hospital admission indicated that higher sCXL16 levels increased risk of longer term mortality, suggesting that CXCL16 may be part of sustained proatherogenic inflammatory response in this patient cohort [96]. A recently published cohort study of 5142 ACS patients found that increasing serial CXCL16 levels were associated with composite adverse clinical outcomes and that the CXCL16 level on admission was independently associated with cardiovascular death [97]. A large cohort study of healthy individuals performed in Norway compared baseline CXCL16 levels between those who developed myocardial infarction and those who did not over the trial period of 11 years [98]. Increased baseline CXCL16 levels were associated with increased risk of myocardial infarction, further strengthening the case for CXCL16 as a prognostic biomarker for CAD. CXCL16 has also been proposed as a prognostic biomarker in inflammatory cardiomyopathy and heart failure. In 174 patients with heart failure, myocardial biopsy samples stained for CXCL16 showed significantly enhanced expression in inflammatory cardiomyopathy compared with non-inflammatory cardiomyopathy [99]. The same study found that increased CXCL16 expression was an independent predictor of death in both inflammatory and non-inflammatory cardiomyopathy patients.
The class J members include the receptor for advanced glycation end-products (RAGEs), which can undergo proteolytic cleavage to release a soluble fragment (sRAGE) present in extracellular fluids, including blood. sRAGE has been proposed as a novel diagnostic and prognostic biomarker in CVD; however, conflicting data has led to controversy. Multiple studies have found that increased serum sRAGE levels are predictive of serious clinical events in CAD and cardiovascular conditions [100,101,102,103]. The relationship is not entirely straightforward: a computed tomography angiography study of sRAGE levels in 127 consecutive patients with non-acute CAD found that higher sRAGE levels were inversely proportional to overall plaque burden [104]. Another study of 328 non-diabetic male patients found the same inverse relationship [105]. As a biomarker for the diagnosis of ACS, similar controversy exists. While numerous studies have found significantly higher levels of sRAGE in ACS compared with controls, and a positive correlation between sRAGE and troponin levels in ACS patients, conflicting studies report no significant differences [106]. The most recent study of sRAGE in ACS by Larsen et al. assessed the utility of sRAGE as a prognostic biomarker post-ACS and found that while baseline sRAGE was associated with greater risk of major adverse cardiac events post-ACS, sRAGE levels recorded at 6 weeks post-ACS were inversely related to the rate of recurrent ischaemic events [107]. The impact of diabetes mellitus on the diagnostic utility of sRAGE in ACS has been demonstrated by multiple studies to be non-significant [106].

3. SRs as Therapeutic Targets in CVD

3.1. Current Therapy

Owing to a central role for SR functionality in atherosclerotic plaque development and progression, targeting SRs is of great interest in CVD therapy. Multiple therapeutic avenues have been explored, including repurposing clinically approved drugs, herbal medicines, and emerging clinical modalities, such as gene therapy and nanoparticle administration. One well-established therapeutic regime worldwide is the long-term use of small-molecule inhibitors of hydroxymethylglutaryl-coenzyme A (HMG Co-A) reductase, commonly known as statins. These drugs are cholesterol-lowering agents that are well accepted in reducing the atherosclerosis burden and risk of CVD [108]. Statins not only reduce total serum cholesterol but also exhibit pleiotropic effects on the vascular wall, including improved endothelial function, a reduction in vascular inflammation, and enhanced plaque stability [109]. The effect of statin therapy on SR function is not yet understood. An in vitro study on a THP-1 human monocyte cell line found that atorvastatin caused dose-dependent attenuation of an oxLDL-stimulated increase in SR-A and monocyte chemoattractant protein-1 (MCP-1) levels [110]. Atorvastatin also reduced lipid-laden foam cell development, indicating that cholesterol levels and plaque development are functionally linked. Another compound, pitavastatin, caused a significant increase in SR-B1 in murine and human macrophages: this SR has been implicated as an anti-atherogenic factor [111,112]. Statins also promote endothelial-derived nitric oxide (NO) levels by activation of SR-B1 and downstream signalling events [112]. Various statins have been found to downregulate CD36 expression in VSMCs in vitro and in circulating monocytes in human subjects [113]. A recent APO-E-null mouse model of atherosclerosis found that simvastatin administration reduced CD36 expression as well as atherosclerosis and inflammatory signalling compared with controls [114]. Statins have been shown to downregulate LOX-1 levels and reduce serum sLOX-1 levels, thus reducing the cellular capacity to sense oxLDL, and activate proatherogenic signalling to downstream pathways [115,116]. Reduced LOX-1 levels caused by statin therapy also has additional anti-atherogenic properties, such as increased NO levels, and downregulation of adhesion molecules [117,118]. The importance of the link between LOX-1 and statins in arterial disease is further highlighted in a cohort study of 751 patients with hypercholesterolaemia receiving statin therapy: OLR1 (LOX-1) polymorphisms are linked to circulating LDL levels and risk of CVD [119].
Statin administration may also affect proatherogenic RAGE levels. Simvastatin treatment of diabetic patients causes the downregulation of RAGE levels in atherosclerotic plaques [120]. Atorvastatin treatment increases sRAGE levels in vitro. Here, sRAGE could act as a decoy to inhibit proatherogenic effects of advanced glycation end-product (AGE) species [121]. One issue is the structurally different statins have similar effects on SR activity and function; this has not been resolved at present. There are notable differences between the therapeutic effects of atorvastatin, which is less noticeable when pravastatin is used.
Monoclonal antibody proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are relatively novel LDL-lowering agents, which have recently been approved for clinical use. PCSK9 is a protease required for the degradation of the LDLR [122]. PCSK9 inhibitors work by inhibiting PCSK9–LDLR interaction, leading to decreased LDLR degradation and ultimately a reduction in serum LDL cholesterol. Studies by Ding et al. investigating the possibility of interactions between PCSK9 and LOX-1 suggest that cross-talk between PCSK9 and LOX-1 exists in the form of positive feedback [123]. The authors demonstrate LOX-1 ablation in cellular and transgenic mouse experiments results in reduced PCSK9 expression and vice versa with PCSK9 ablation. The activation of the PCSK9–LOX-1 axis has also been shown to be mediated by altered ROS generation at sites of shear stress [124]. The effects of monoclonal PCSK9 inhibitors on LOX-1 expression in humans is not known; however, it is conceivable that LOX-1 downregulation resulting from PCSK9 inhibition could be providing additional anti-atherogenic benefit.
Peroxisome proliferator-activated receptor-γ (PPAR-γ) exhibits anti-inflammatory and antidiabetic properties. Pioglitazone is a PPAR-γ agonist, which is commonly prescribed as a glucose-lowering agent for T2DM patients. Analysis of human and animal models treated with pioglitazone has effects on SR levels and function in the context of diabetes and metabolic syndrome. In a study of 30 patients with polycystic ovarian syndrome (PCOS) combined with either increased fasting glucose or obesity, serum sCD36 levels were significantly raised compared to healthy controls (n = 14) [125]. Furthermore, pioglitazone treatment significantly reduced circulating sCD36 and CRP levels whilst improving insulin sensitivity. This strengthens an association between sCD36 and insulin resistance in PCOS. In this cohort, pioglitazone treatment may have an added benefit of reducing risk of CVD. A different study on diabetic APOE-null mice subjected to pioglitazone treatment observed attenuation of atherosclerosis via RAGE-linked signalling events [126]. These studies showed that pioglitazone administration reduced atherosclerotic plaque burden in coronary arteries and inhibited RAGE expression in arterial and vascular smooth muscle cells.
Other clinically approved drugs that modulate LOX-1 expression and/or function include ursolic acid, histamine, spironolactone, and angiotensin-converting enzyme (ACE) inhibitors. Ursolic acid inhibits LOX-1 expression in vitro and significantly reduces both LOX-1 expression and atherosclerotic plaque burden in arteries of APOE-null mice [127]. Similar outcomes were observed using the same models treated with histamine [128]. Antihypertensive medications, such as spironolactone (potassium-sparing diuretic) and losartan (angiotensin II receptor inhibitor), have been shown to downregulate LOX-1 levels and functionality, thus showing wider benefits for targeting atherosclerosis [129,130]. Nifedipine, a calcium channel blocker used commonly in hypertension, has been found to reduce CD36 expression and lipid internalization in macrophages in vitro [113].

3.2. Novel Therapeutics

Deletion of a functional SR-A locus in transgenic mouse models causes a significant reduction in atherosclerosis plaque burden in the aorta, stimulating many groups to search for novel therapeutics or inhibitors that target SR-A function or activity [131]. Recent work investigating the role of SR-A activation in neutrophils and NET formation has found that NET release may be triggered by SR-A activation by reactive oxygen species (ROS) [132]. The role of NETs in plaque erosion and thrombo-occlusive complications of CVD is becoming more prominent and the role of SR-A targeting in this area is a potential area for future studies. Certain amphiphilic macromolecules (AMs) that exhibit selective binding to SR-A have shown promise in attenuating atherosclerosis [133,134]. SR-A-specific AMs not only reduce oxLDL binding and uptake in human embryonic kidney cells but also inhibit proinflammatory signalling in activated THP-1 monocytes [134]. Using rat models, this study also showed a reduction in intimal cholesterol accumulation and macrophage retention under proatherogenic conditions [134]. Higher serum levels of the anti-inflammatory cytokine interleukin-10 (IL-10) were associated with superior outcomes in ACS patients, but this was not fully understood at that time [135,136]. Since these studies, it has been found that IL-10 addition reduces oxLDL uptake and downregulates SR-A levels in activated THP-1 monocytes [137]. Such work provides further support for IL-10 as a therapeutic strategy aimed at SR-A for CVD, particularly in ACS.
Macrophage SR-A expression in macrophages is downregulated by treatment with intermedin, a novel member of the calcitonin gene-related peptide family: intermedin attenuates atherosclerosis plaque burden in APOE-null mice [138]. Intermedin is implicated in vascular homeostasis by reducing calcium deposition in atherosclerotic plaques [139]. Intermedin treatment caused increased expression of the phosphatase and tensin homolog (PTEN) whilst SR-A levels were reduced, further supporting intermedin use in CVD therapy [139]. SR-A is also a potential target in diabetic nephropathy, although no therapeutics are forthcoming for such therapy. SR-A-null mice subjected to streptozotocin-induced diabetes mellitus exhibit significant changes in markers of nephropathy, such as albuminuria, glomerular hypertrophy, mesangial matrix expansion, and parenchymal macrophage infiltration with increased levels of transforming growth factor-β [140].
Using RNA interference (RNAi) to downregulate SR-A and CD36 levels is effective in reducing oxLDL uptake in vitro and atherosclerotic load in vivo when either receptor is targeted in isolation; however, when both receptors are simultaneously targeted, there is no change compared to controls [141]. One likelihood is that downregulation of either SR-A or CD36 alone causes a reciprocal upregulation of the other; SR crosstalk could explain why inhibition of both receptors is less effective than targeting a single receptor. Further studies also indicate conflicting conclusions in targeting both SR-A and CD36 together. Monoclonal antibody-mediated targeting of either SR-A or CD36 separately reduces foam cell formation in vitro [142], but simultaneously targeting both receptors in this context was not studied. Double knockout of SR-A and CD36 in APO-E-null mice did not reduce aortic root atherosclerosis compared with APOE-null mice; however, the authors did not study SR-A- or CD36-null mice in isolation [143]. Single and double knockout mouse lines have been studied elsewhere, with findings that CD36 ablation significantly reduced aortic atherosclerotic plaque burden; however, SR-A-null mice were less effective in isolation, and SR-A/CD36 double-null animals showed comparable efficacy to CD36-null mice [144]. A recent study of insulin sensitivity in a mouse model of obesity found that SR-A knockout exacerbated insulin resistance compared with wildtype mice [145]. Interestingly, in a similar study, CD36 knockout provided protective effects against insulin resistance [146]. These studies highlight the importance of studying the off-target effects of SR targeting as each class of receptor is likely to exhibit unique and potentially opposing tissue-specific functions.
CD36 is a multi-ligand multifunctional scavenger receptor, which plays a complex role in CVD and cerebrovascular disease states [147]. CD36 exhibits both proatherogenic and anti-angiogenic properties in vitro and in vivo [148,149] and there is an emerging understanding that there may be an “optimal protective window” of CD36 expression [150]. CD36-null mice display a significant increase in angiogenesis and re-vascularization after acute stroke [151]. Treatment with a cell-permeable antioxidant peptide (SS31) reduces cerebral oxidative stress and infarct size in a mice subjected to transient middle cerebral artery occlusion, but this effect was absent in the CD36-null group [152], indicating that the therapeutic effects of SS31 is occurring via a CD36-regulated pathway. Nanoparticles, such as AMs and nanovesicles, have been identified, which bind to CD36 and inhibit oxLDL uptake in vitro [68,153]. CD36-targeted liposome-like nanovesicles show impressive binding affinities to CD36 in vitro and in vivo in atherosclerotic LDLR-null mice. CD36-targeted nanovesicles thus have potential for reducing atherosclerosis. Synthetic protein ligands that bind CD36, such as EP80317, have been shown to reduce oxLDL uptake by macrophages and atherosclerotic plaque burden in APOE-null mice [154]. Furthermore, EP80317 administration promotes reverse cholesterol transport in macrophages; gut receptors, such as LXRα and NPC1L1, further facilitate cholesterol removal through faecal excretion [155]. The administration of ectopic adipokines to ameliorate atherosclerosis by CD36 modulation has been recently investigated by Wang et al. [156]. The authors found that administration of C1q/tumour necrosis factor-related protein 13 (CTRP13) reduced CD36 expression in vitro and significantly reduced aortic atherosclerosis in vivo, thus defining a potential novel therapeutic in atherosclerosis by SR modulation.
LOX-1 targeting in atherosclerosis attracts much interest, due to its proatherogenic properties [157]. In vitro studies show that blocking LOX-1 reduces the proapoptotic effects of oxLDL; transgenic animal studies indicate that OLR1/LDLR double-null mice exhibit reduced atherosclerotic plaque burden in coronary arteries [158,159]. Administration of anti-LOX-1 antibody in animal models causes a significant reduction in arterial pathologies characteristic of neointimal hyperplasia and MI [160,161]. Furthermore, anti-LOX-1 antibody administration in a hypertensive rat model causes reduction of lipid deposition in mesenteric arteries; such treatment also attenuates renal vascular fibrosis in an obese diabetic hypertensive rat model [162,163].
Studies on microRNAs (miRNAs) linked to atherosclerosis and CVD is a relatively new but increasing area of interest. These miRNAs are usually tissue-specific relatively small (∼22 bases), non-protein coding RNAs, which bind one or more protein-coding mRNAs to regulate protein expression: miRNAs are implicated in many pathological processes, including atherosclerosis [164]. Dai and colleagues used bioinformatics to identify miRNA-98 as a LOX-1-specific agent and showed that miRNA-98 administration reduced lipid accumulation in vitro; this treatment also reduced LOX-1 expression and aortic plaque burden in APOE-null mice [165]. A similar study showed that miRNA let-7g-mediated targeting of LOX-1 attenuated the development of atherosclerosis [166]. As many miRNAs bind multiple mRNA species, off-target effects of miRNA therapy is a serious concern; however, LOX-1-specific miRNA therapy did not raise toxicity issues [165]. Small interfering RNAs (siRNAs), which target the LOX-1 mRNA and knockdown protein expression, can also inhibit LOX-1-regulated pathways in vitro [167]. Although initial studies are promising [167], further evaluation in animal models is needed to better evaluate such treatments to attenuate atherosclerosis and plaque burden. Similar to the use of miRNAs, antibodies, and synthetic proteins to target LOX-1, careful evaluation of off-target effects and absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters is needed before effective therapies can be implemented [168].
The three-dimensional structure of the LOX-1 extracellular domain has been determined using a combination of crystallography and NMR [169]. This 3-D model for LOX-1 was used to screen chemical libraries to identify new molecules that block LOX-1 function; two lead compounds could reduce oxLDL uptake and monocyte adhesion to endothelial cells [170]. Chemical compounds with cell- and tissue-penetrating properties have substantial advantages over antibody or synthetic protein-based therapies; however, such compounds require careful evaluation of ADMET parameters in whole animal models. Single-chain variable fragment (scFv) antibodies targeting LOX-1 have been recently reported [171]. These scFvs exhibit specific binding affinity, enhanced bio-distribution, and low immunogenicity compared with humanized monoclonal antibodies, whilst being substantially cheaper to produce using bacterial expression systems. LOX-1-specific scFvs are in the early stages of development and ongoing studies are aiming to improve the binding affinity, thermostability, and serum half-life, and using multimerization techniques [172,173].
The relationship between CXCL16 and atherogenesis is less defined compared with SR-A, CD36, and LOX-1. A sCXCL16 isoform is a potential biomarker for CAD, and membrane-bound CXCL16 is highly expressed in carotid and coronary artery atheromas [174]. Hofnagel and co-workers found that CXCL16 is expressed primarily in the endothelium at sites predisposed to atherosclerosis in the rabbit aorta; furthermore, anti-CXCL16 antibody treatment of primary human endothelial cells caused a significant reduction in endothelial-monocyte adhesion [175]. However, there is still a need to demonstrate that anti-CXCL16 monoclonal antibody therapy has efficacy in an animal model of atherosclerosis. Interestingly, LDLR/CXCL16 double-null transgenic mice displayed an increased atherosclerotic plaque burden compared to LDLR-null mice [93]; this work suggests that the CXCL16 locus is anti-atherogenic, contradicting previous studies.
AGEs have been suggested to be proatherogenic, especially in the context of atherosclerosis in diabetes mellitus patients. Inhibition in biochemical pathways that cause the production of AGEs using agents, such as aminoguanidine-HCl, which inhibits chemical crosslinking, can produce beneficial cardiovascular outcomes in vivo [176]. The binding and cellular uptake of oxLDL by RAGE is promoted by AGEs; this proatherogenic effect can be blocked by anti-RAGE treatment [177]. RNAi to target RAGE expression reduced endothelial-monocyte adhesion, and improved CVD-related outcomes [178]. Diabetic transgenic mice carrying the APOE/RAGE double-null genotype displayed reduced leucocyte adhesion, expression of proinflammatory mediators, and a statistically non-significant decrease in reduction in aortic atherosclerotic plaque burden [179]. Interestingly, administration of recombinant sRAGE in diabetic APOE-null mice suppresses atherosclerosis whilst having little or no effect on plasma lipids or serum glucose levels [180]. One hypothesis is that sRAGE acts as a circulating soluble “AGE sponge”, thus downregulating membrane-bound RAGE expression in vascular tissues, further reducing proatherogenic signalling and cellular responses [180].
Novel drug delivery systems, such as nanocarriers and nanovesicles, have been employed with some success in SR targeting in atherosclerosis [68,153]. The ideal delivery system is one that exhibits immunological inertness and non-cytotoxicity, high tissue permeability and specificity, excellent biodistribution, and adequate half-life in the circulation. A recently published study evaluating the use of M2 macrophage-derived exosomes as a novel drug delivery system in atherosclerosis describes their ability to target areas of atherosclerosis by inflammation-tropism via the expression of surface-bonded chemokine receptors [181]. Atherosclerosis-targeting drug delivery systems may have an important future role in the development of SR-targeted therapies.

4. Conclusions

SRs are a multi-ligand receptor super-group defined by the ability to recognize modified lipid particles; each class has a defined consensus sequence, which is usually unrelated to other classes. One common feature of this diverse group of proteins is the ability to bind potentially harmful substances to mediate clearance from circulation. Many SRs are linked to atherosclerosis, which is the main pathway involved in the development of CVD. The current view is that the main SRs involved in CVD are SR-A, CD36, LOX-1, CXCL16, and RAGE. Monitoring serum levels of soluble SRs, alongside cardiac troponins, in better diagnosis of MI and ACS is a potential prognostic tool. Current cholesterol-lowering therapies, e.g., statin treatment, have been also found to modulate SR expression and downstream signalling, suggesting wider implications of SR function linked to cholesterol metabolism. So far, numerous approaches for targeting SRs, including gene therapy, monoclonal antibodies, and nanoparticles, have shown differing levels of success in reducing CVD. A major issue is that SR studies have so far not progressed beyond animal models; only SR biomarker assessments in human patients have been carried out. We concur that the future application of SR-linked tools and therapies will play a central role in the diagnosis, management, and treatment of arterial pathologies, such as CVD.

5. Future Perspectives

SRs are attractive molecular targets in the development of targeted therapy for atherosclerosis. While the results from in vitro and in vivo models are promising, SR receptor modulation in humans has not been investigated. Given that SRs expression is widespread in multiple organ systems, the concept of systemic SR modulation should be considered with caution as there is a significant potential for off-target effects and reciprocal upregulation of SRs from the other classes. Local delivery of SR modulators utilizing novel delivery systems, such as exosomes and other nanocarrier technologies, may play an important role in translating SR-targeting agents to a clinical setting.

Author Contributions

Conceptualization, G.A.C. and S.P.; writing—original draft preparation, G.A.C. and F.S.; writing—review and editing, G.A.C., F.S., M.A.H., S.P., S.H.-V.; supervision, S.P. and S.H.-V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Falk, E. Pathogenesis of Atherosclerosis. J. Am. Coll. Cardiol. 2006, 47, C7–C12. [Google Scholar] [CrossRef] [Green Version]
  2. Ference, B.A.; Ginsberg, H.N.; Graham, I.; Ray, K.K.; Packard, C.J.; Bruckert, E.; Hegele, R.A.; Krauss, R.M.; Raal, F.J.; Schunkert, H.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2017, 38, 2459–2472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lusis, A.J. Atherosclerosis. Nature 2000, 407, 233–241. [Google Scholar] [CrossRef] [PubMed]
  4. Gistera, A.; Hansson, G.K. The immunology of atherosclerosis. Nat. Rev. Nephrol. 2017, 13, 368–380. [Google Scholar] [CrossRef] [PubMed]
  5. Di Pietro, N.; Formoso, G.; Pandolfi, A. Physiology and pathophysiology of oxLDL uptake by vascular wall cells in atherosclerosis. Vasc. Pharm. 2016, 84, 1–7. [Google Scholar] [CrossRef] [PubMed]
  6. Katakami, N. Mechanism of Development of Atherosclerosis and Cardiovascular Disease in Diabetes Mellitus. J. Atheroscler. Thromb. 2018, 25, 27–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Davies, M.J.; Thomas, A. Thrombosis and acute coronary-artery lesions in sudden cardiac ischemic death. N. Engl. J. Med. 1984, 310, 1137–1140. [Google Scholar] [CrossRef]
  8. Virmani, R.; Kolodgie Frank, D.; Burke Allen, P.; Finn Aloke, V.; Gold Herman, K.; Tulenko Thomas, N.; Wrenn Steven, P.; Narula, J. Atherosclerotic Plaque Progression and Vulnerability to Rupture. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 2054–2061. [Google Scholar] [CrossRef] [Green Version]
  9. Stary Herbert, C.; Chandler, A.B.; Dinsmore Robert, E.; Fuster, V.; Glagov, S.; Insull, W.; Rosenfeld Michael, E.; Schwartz Colin, J.; Wagner William, D.; Wissler Robert, W. A Definition of Advanced Types of Atherosclerotic Lesions and a Histological Classification of Atherosclerosis. Circulation 1995, 92, 1355–1374. [Google Scholar] [CrossRef]
  10. Virmani, R.; Burke, A.P.; Kolodgie, F.D.; Farb, A. Vulnerable plaque: The pathology of unstable coronary lesions. J. Interv. Cardiol. 2002, 15, 439–446. [Google Scholar] [CrossRef]
  11. Burke, A.P.; Virmani, R.; Galis, Z.; Haudenschild, C.C.; Muller, J.E. Task force #2—What is the pathologic basis for new atherosclerosis imaging techniques? J. Am. Coll. Cardiol. 2003, 41, 1874–1886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Libby, P.; Pasterkamp, G.; Crea, F.; Jang, I.K. Reassessing the Mechanisms of Acute Coronary Syndromes. Circ. Res. 2019, 124, 150–160. [Google Scholar] [CrossRef] [PubMed]
  13. Crea, F.; Libby, P. Acute Coronary Syndromes: The Way Forward From Mechanisms to Precision Treatment. Circulation 2017, 136, 1155–1166. [Google Scholar] [CrossRef] [PubMed]
  14. Partida, R.A.; Libby, P.; Crea, F.; Jang, I.K. Plaque erosion: A new in vivo diagnosis and a potential major shift in the management of patients with acute coronary syndromes. Eur. Heart J. 2018, 39, 2070–2076. [Google Scholar] [CrossRef] [PubMed]
  15. Obama, T.; Ohinata, H.; Takaki, T.; Iwamoto, S.; Sawada, N.; Aiuchi, T.; Kato, R.; Itabe, H. Cooperative Action of Oxidized Low-Density Lipoproteins and Neutrophils on Endothelial Inflammatory Responses Through Neutrophil Extracellular Trap Formation. Front. Immunol. 2019, 10, 1899. [Google Scholar] [CrossRef] [Green Version]
  16. Prabhudas, M.; Bowdish, D.; Drickamer, K.; Febbraio, M.; Herz, J.; Kobzik, L.; Krieger, M.; Loike, J.; Means, T.K.; Moestrup, S.K.; et al. Standardizing scavenger receptor nomenclature. J. Immunol. 2014, 192, 1997–2006. [Google Scholar] [CrossRef] [Green Version]
  17. Goldstein, J.L.; Ho, Y.K.; 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] [Green Version]
  18. Brown, M.S.; Basu, S.K.; Falck, J.R.; Ho, Y.K.; Goldstein, J.L. The scavenger cell pathway for lipoprotein degradation: Specificity of the binding site that mediates the uptake of negatively-charged LDL by macrophages. J. Supramol. Struct. 1980, 13, 67–81. [Google Scholar] [CrossRef]
  19. Zani, I.A.; Stephen, S.L.; Mughal, N.A.; Russell, D.; Homer-Vanniasinkam, S.; Wheatcroft, S.B.; Ponnambalam, S. Scavenger receptor structure and function in health and disease. Cells 2015, 4, 178–201. [Google Scholar] [CrossRef] [Green Version]
  20. Wilkinson, K.; El Khoury, J. Microglial scavenger receptors and their roles in the pathogenesis of Alzheimer’s disease. Int. J. Alzheimers Dis. 2012, 2012, 489456. [Google Scholar] [CrossRef] [Green Version]
  21. Singh, T.D.; Park, S.Y.; Bae, J.S.; Yun, Y.; Bae, Y.C.; Park, R.W.; Kim, I.S. MEGF10 functions as a receptor for the uptake of amyloid-beta. FEBS Lett. 2010, 584, 3936–3942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Bachli, E.B.; Schaer, D.J.; Walter, R.B.; Fehr, J.; Schoedon, G. Functional expression of the CD163 scavenger receptor on acute myeloid leukemia cells of monocytic lineage. J. Leukoc. Biol. 2006, 79, 312–318. [Google Scholar] [CrossRef] [PubMed]
  23. Leelahavanichkul, A.; Bocharov, A.V.; Kurlander, R.; Baranova, I.N.; Vishnyakova, T.G.; Souza, A.C.; Hu, X.; Doi, K.; Vaisman, B.; Amar, M.; et al. Class B scavenger receptor types I and II and CD36 targeting improves sepsis survival and acute outcomes in mice. J. Immunol. 2012, 188, 2749–2758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Matsushita, N.; Kashiwagi, M.; Wait, R.; Nagayoshi, R.; Nakamura, M.; Matsuda, T.; Hogger, P.; Guyre, P.M.; Nagase, H.; Matsuyama, T. Elevated levels of soluble CD163 in sera and fluids from rheumatoid arthritis patients and inhibition of the shedding of CD163 by TIMP-3. Clin. Exp. Immunol. 2002, 130, 156–161. [Google Scholar] [CrossRef]
  25. Nagai, M.; Hirayama, K.; Ebihara, I.; Higuchi, T.; Shimohata, H.; Kobayashi, M. Serum levels of the soluble haemoglobin scavenger receptor CD163 in MPO-ANCA-associated renal vasculitis. Scand. J. Rheumatol. 2016, 45, 397–403. [Google Scholar] [CrossRef]
  26. Hughes, D.A.; Fraser, I.P.; Gordon, S. Murine macrophage scavenger receptor: In vivo expression and function as receptor for macrophage adhesion in lymphoid and non-lymphoid organs. Eur. J. Immunol. 1995, 25, 466–473. [Google Scholar] [CrossRef]
  27. Plüddemann, A.; Neyen, C.; Gordon, S. Macrophage scavenger receptors and host-derived ligands. Methods 2007, 43, 207–217. [Google Scholar] [CrossRef]
  28. Gough, P.J.; Greaves, D.R.; Suzuki, H.; Hakkinen, T.; Hiltunen, M.O.; Turunen, M.; Herttuala, S.Y.; Kodama, T.; Gordon, S. Analysis of macrophage scavenger receptor (SR-A) expression in human aortic atherosclerotic lesions. Arterioscler. Thromb. Vasc. Biol. 1999, 19, 461–471. [Google Scholar] [CrossRef] [Green Version]
  29. Kelley, J.L.; Ozment, T.R.; Li, C.; Schweitzer, J.B.; Williams, D.L. Scavenger receptor-A (CD204): A two-edged sword in health and disease. Crit. Rev. Immunol. 2014, 34, 241–261. [Google Scholar] [CrossRef]
  30. Pluddemann, A.; Mukhopadhyay, S.; Gordon, S. Innate immunity to intracellular pathogens: Macrophage receptors and responses to microbial entry. Immunol. Rev. 2011, 240, 11–24. [Google Scholar] [CrossRef]
  31. Christie, R.H.; Freeman, M.; Hyman, B.T. Expression of the macrophage scavenger receptor, a multifunctional lipoprotein receptor, in microglia associated with senile plaques in Alzheimer’s disease. Am. J. Pathol. 1996, 148, 399–403. [Google Scholar] [PubMed]
  32. El Khoury, J.; Hickman, S.E.; Thomas, C.A.; Cao, L.; Silverstein, S.C.; Loike, J.D. Scavenger receptor-mediated adhesion of microglia to beta-amyloid fibrils. Nature 1996, 382, 716–719. [Google Scholar] [CrossRef] [PubMed]
  33. Talle, M.A.; Rao, P.E.; Westberg, E.; Allegar, N.; Makowski, M.; Mittler, R.S.; Goldstein, G. Patterns of antigenic expression on human monocytes as defined by monoclonal antibodies. Cell Immunol. 1983, 78, 83–99. [Google Scholar] [CrossRef]
  34. Yokoi, H.; Yanagita, M. Targeting the fatty acid transport protein CD36, a class B scavenger receptor, in the treatment of renal disease. Kidney Int. 2016, 89, 740–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Nicholson, A.C.; Han, J.; Febbraio, M.; Silversterin, R.L.; Hajjar, D.P. Role of CD36, the macrophage class B scavenger receptor, in atherosclerosis. Ann. N. Y. Acad. Sci. 2001, 947, 224–228. [Google Scholar] [CrossRef]
  36. Stewart, C.R.; Stuart, L.M.; Wilkinson, K.; van Gils, J.M.; Deng, J.; Halle, A.; Rayner, K.J.; Boyer, L.; Zhong, R.; Frazier, W.A.; et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. Immunol. 2010, 11, 155–161. [Google Scholar] [CrossRef] [Green Version]
  37. Greaves, D.R.; Gordon, S. Macrophage-Specific Gene Expression: Current Paradigms and Future Challenges. Int. J. Hematol. 2002, 76, 6–15. [Google Scholar] [CrossRef]
  38. Yoshida, H.; Quehenberger, O.; Kondratenko, N.; Green, S.; Steinberg, D. Minimally oxidized low-density lipoprotein increases expression of scavenger receptor A, CD36, and macrosialin in resident mouse peritoneal macrophages. Arterioscler. Thromb. Vasc. Biol. 1998, 18, 794–802. [Google Scholar] [CrossRef] [Green Version]
  39. De Beer, M.C.; Zhao, Z.; Webb, N.R.; van der Westhuyzen, D.R.; de Villiers, W.J. Lack of a direct role for macrosialin in oxidized LDL metabolism. J. Lipid Res. 2003, 44, 674–685. [Google Scholar] [CrossRef] [Green Version]
  40. Song, L.; Lee, C.; Schindler, C. Deletion of the murine scavenger receptor CD68. J. Lipid Res. 2011, 52, 1542–1550. [Google Scholar] [CrossRef] [Green Version]
  41. Herre, J.; Willment, J.A.; Gordon, S.; Brown, G.D. The role of Dectin-1 in antifungal immunity. Crit. Rev. Immunol. 2004, 24, 193–203. [Google Scholar] [CrossRef] [PubMed]
  42. Mango, R.; Predazzi, I.M.; Romeo, F.; Novelli, G. LOX-1/LOXIN: The yin/yang of atheroscleorosis. Cardiovasc. Drugs 2011, 25, 489–494. [Google Scholar] [CrossRef] [PubMed]
  43. Ishii, J.; Adachi, H.; Aoki, J.; Koizumi, H.; Tomita, S.; Suzuki, T.; Tsujimoto, M.; Inoue, K.; Arai, H. SREC-II, a new member of the scavenger receptor type F family, trans-interacts with SREC-I through its extracellular domain. J. Biol. Chem. 2002, 277, 39696–39702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Adachi, H.; Tsujimoto, M. Structure and function of a novel scavenger receptor expressed in human endothelial cells. Tanpakushitsu Kakusan Koso 1999, 44, 1282–1286. [Google Scholar]
  45. Hofnagel, O.; Luechtenborg, B.; Plenz, G.; Robenek, H. Expression of the novel scavenger receptor SR-PSOX in cultured aortic smooth muscle cells and umbilical endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 710–711. [Google Scholar] [CrossRef] [Green Version]
  46. Wågsäter, D.; Olofsson, P.S.; Norgren, L.; Stenberg, B.; Sirsjö, A. The chemokine and scavenger receptor CXCL16/SR-PSOX is expressed in human vascular smooth muscle cells and is induced by interferon gamma. Biochem. Biophys. Res. Commun. 2004, 325, 1187–1193. [Google Scholar] [CrossRef]
  47. Ma, Z.; Jin, X.; He, L.; Wang, Y. CXCL16 regulates renal injury and fibrosis in experimental renal artery stenosis. Am. J. Physiol. Heart Circ. Physiol. 2016, 311, H815–H821. [Google Scholar] [CrossRef] [Green Version]
  48. Hu, Z.B.; Chen, Y.; Gong, Y.X.; Gao, M.; Zhang, Y.; Wang, G.H.; Tang, R.N.; Liu, H.; Liu, B.C.; Ma, K.L. Activation of the CXCL16/CXCR6 Pathway by Inflammation Contributes to Atherosclerosis in Patients with End-stage Renal Disease. Int. J. Med. Sci. 2016, 13, 858–867. [Google Scholar] [CrossRef] [Green Version]
  49. Zhou, F.; Wang, J.; Wang, K.; Zhu, X.; Pang, R.; Li, X.; Zhu, G.; Pan, X. Serum CXCL16 as a Novel Biomarker of Coronary Artery Disease in Type 2 Diabetes Mellitus: A Pilot Study. Ann. Clin. Lab. Sci. 2016, 46, 184–189. [Google Scholar]
  50. Adachi, H.; Tsujimoto, M. FEEL-1, a novel scavenger receptor with in vitro bacteria-binding and angiogenesis-modulating activities. J. Biol. Chem. 2002, 277, 34264–34270. [Google Scholar] [CrossRef] [Green Version]
  51. Zhou, B.; Weigel, J.A.; Fauss, L.; Weigel, P.H. Identification of the hyaluronan receptor for endocytosis (HARE). J. Biol. Chem. 2000, 275, 37733–37741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Irjala, H.; Elima, K.; Johansson, E.L.; Merinen, M.; Kontula, K.; Alanen, K.; Grenman, R.; Salmi, M.; Jalkanen, S. The same endothelial receptor controls lymphocyte traffic both in vascular and lymphatic vessels. Eur. J. Immunol. 2003, 33, 815–824. [Google Scholar] [CrossRef] [PubMed]
  53. Irjala, H.; Alanen, K.; Grénman, R.; Heikkilä, P.; Joensuu, H.; Jalkanen, S. Mannose receptor (MR) and common lymphatic endothelial and vascular endothelial receptor (CLEVER)-1 direct the binding of cancer cells to the lymph vessel endothelium. Cancer Res. 2003, 63, 4671–4676. [Google Scholar] [PubMed]
  54. Etzerodt, A.; Maniecki, M.B.; Graversen, J.H.; Møller, H.J.; Torchilin, V.P.; Moestrup, S.K. Efficient intracellular drug-targeting of macrophages using stealth liposomes directed to the hemoglobin scavenger receptor CD163. J. Control Release 2012, 160, 72–80. [Google Scholar] [CrossRef]
  55. Kristiansen, M.; Graversen, J.H.; Jacobsen, C.; Sonne, O.; Hoffman, H.J.; Law, S.K.; Moestrup, S.K. Identification of the haemoglobin scavenger receptor. Nature 2001, 409, 198–201. [Google Scholar] [CrossRef]
  56. Ramasamy, R.; Yan, S.F.; Schmidt, A.M. RAGE: Therapeutic target and biomarker of the inflammatory response--the evidence mounts. J. Leukoc. Biol. 2009, 86, 505–512. [Google Scholar] [CrossRef]
  57. Ramasamy, R.; Yan, S.F.; Schmidt, A.M. The RAGE axis and endothelial dysfunction: Maladaptive roles in the diabetic vasculature and beyond. Trends Cardiovasc. Med. 2005, 15, 237–243. [Google Scholar] [CrossRef]
  58. Che, J.J.; Shao, Y.X.; Li, G.P. Association between rs1049673 polymorphism in CD36 and premature coronary heart disease. Genet. Mol. Res. 2014, 13, 7708–7717. [Google Scholar] [CrossRef]
  59. Jayewardene, A.F.; Gwinn, T.; Hancock, D.P.; Mavros, Y.; Rooney, K.B. The associations between polymorphisms in the CD36 gene, fat oxidation and cardiovascular disease risk factors in a young adult Australian population: A pilot study. Obes. Res. Clin. Pract. 2014, 8, e618–e621. [Google Scholar] [CrossRef]
  60. Morini, E.; Rizzacasa, B.; Pucci, S.; Polidoro, C.; Ferrè, F.; Caporossi, D.; Helmer Citterich, M.; Novelli, G.; Amati, F. The human rs1050286 polymorphism alters LOX-1 expression through modifying miR-24 binding. J. Cell. Mol. Med. 2016, 20, 181–187. [Google Scholar] [CrossRef] [Green Version]
  61. Mango, R.; Clementi, F.; Borgiani, P.; Forleo, G.B.; Federici, M.; Contino, G.; Giardina, E.; Garza, L.; Fahdi, I.E.; Lauro, R.; et al. Association of single nucleotide polymorphisms in the oxidised LDL receptor 1 (OLR1) gene in patients with acute myocardial infarction. J. Med. Genet. 2003, 40, 933–936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Du, Y.; Chen, K.; Liu, E.; Wang, X.; Li, F.; Liu, T.; Zheng, X.; Li, G.; Che, J. Gender-specific associations of CD36 polymorphisms with the lipid profile and susceptibility to premature multi-vessel coronary artery heart disease in the Northern Han Chinese. Gene 2020, 753, 144806. [Google Scholar] [CrossRef] [PubMed]
  63. Nakayama, M.; Kudoh, T.; Kaikita, K.; Yoshimura, M.; Oshima, S.; Miyamoto, Y.; Takeya, M.; Ogawa, H. Class A macrophage scavenger receptor gene expression levels in peripheral blood mononuclear cells specifically increase in patients with acute coronary syndrome. Atherosclerosis 2008, 198, 426–433. [Google Scholar] [CrossRef] [PubMed]
  64. Emura, I.; Usuda, H.; Fujita, T.; Ebe, K.; Nagai, T. Scavenger receptor A index and coronary thrombus in patients with acute ST elevation myocardial infarction. Pathol. Int. 2011, 61, 351–355. [Google Scholar] [CrossRef] [PubMed]
  65. Emura, I.; Usuda, H.; Fujita, T.; Ebe, K.; Nagai, T. Increase of scavenger receptor A-positive monocytes in patients with acute coronary syndromes. Pathol. Int. 2007, 57, 502–508. [Google Scholar] [CrossRef]
  66. Teupser, D.; Mueller, M.A.; Koglin, J.; Wilfert, W.; Ernst, J.; von Scheidt, W.; Steinbeck, G.; Seidel, D.; Thiery, J. CD36 mRNA expression is increased in CD14+ monocytes of patients with coronary heart disease. Clin. Exp. Pharmacol. Physiol. 2008, 35, 552–556. [Google Scholar] [CrossRef]
  67. Ye, M.; Zhou, J.; Zhong, Y.; Xu, J.; Hou, J.; Wang, X.; Wang, Z.; Guo, D. SR-A-Targeted Phase-Transition Nanoparticles for the Detection and Treatment of Atherosclerotic Vulnerable Plaques. ACS Appl. Mater. Interfaces 2019, 11, 9702–9715. [Google Scholar] [CrossRef]
  68. Nie, S.; Zhang, J.; Martinez-Zaguilan, R.; Sennoune, S.; Hossen, M.N.; Lichtenstein, A.H.; Cao, J.; Meyerrose, G.E.; Paone, R.; Soontrapa, S.; et al. Detection of atherosclerotic lesions and intimal macrophages using CD36-targeted nanovesicles. J. Control Release 2015, 220, 61–70. [Google Scholar] [CrossRef] [Green Version]
  69. Shah, A.S.V.; Anand, A.; Strachan, F.E.; Ferry, A.V.; Lee, K.K.; Chapman, A.R.; Sandeman, D.; Stables, C.L.; Adamson, P.D.; Andrews, J.P.M.; et al. High-sensitivity troponin in the evaluation of patients with suspected acute coronary syndrome: A stepped-wedge, cluster-randomised controlled trial. Lancet 2018, 392, 919–928. [Google Scholar] [CrossRef] [Green Version]
  70. Wang, Y.; Zhu, J.; Handberg, A.; Overvad, K.; Tjønneland, A.; Rimm, E.B.; Jensen, M.K. Association between plasma CD36 levels and incident risk of coronary heart disease among Danish men and women. Atherosclerosis 2018, 277, 163–168. [Google Scholar] [CrossRef] [Green Version]
  71. Rac, M.; Krzystolik, A.; Safranow, K.; Dziedziejko, V.; Goschorska, M.; Poncyljusz, W.; Chlubek, D. Is plasma-soluble CD36 associated with density of atheromatous plaque and ankle-brachial index in early-onset coronary artery disease patients? Kardiol. Pol. 2016, 74, 570–575. [Google Scholar] [CrossRef] [PubMed]
  72. Castelblanco, E.; Sanjurjo, L.; Barranco-Altirriba, M.; Falguera, M.; Hernández, M.; Soldevila, B.; Sarrias, M.-R.; Franch-Nadal, J.; Arroyo, J.A.; Fernandez-Real, J.-M.; et al. The Circulating Fatty Acid Transporter Soluble CD36 Is Not Associated with Carotid Atherosclerosis in Subjects with Type 1 and Type 2 Diabetes Mellitus. J. Clin. Med. 2020, 9, 1700. [Google Scholar] [CrossRef] [PubMed]
  73. Sheikh, S.A. Role of Plasma Soluble Lectin Like Oxidized Low-Density Lipoprotein Receptor-1 in Severity of CAD Patients and Relationship with Microrna-98. Balk. Med. J. 2020. [Google Scholar] [CrossRef] [PubMed]
  74. Stankova, T.; Delcheva, G.; Maneva, A.; Vladeva, S. Serum Levels of Carbamylated LDL and Soluble Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1 Are Associated with Coronary Artery Disease in Patients with Metabolic Syndrome. Medicina 2019, 55, 493. [Google Scholar] [CrossRef] [Green Version]
  75. Misaka, T.; Suzuki, S.; Sakamoto, N.; Yamaki, T.; Sugimoto, K.; Kunii, H.; Nakazato, K.; Saitoh, S.-i.; Sawamura, T.; Ishibashi, T.; et al. Significance of soluble lectin-like oxidized LDL receptor-1 levels in systemic and coronary circulation in acute coronary syndrome. BioMed Res. Int. 2014, 2014, 649185. [Google Scholar] [CrossRef]
  76. Kobayashi, N.; Hata, N.; Kume, N.; Shinada, T.; Tomita, K.; Shirakabe, A.; Kitamura, M.; Nozaki, A.; Inami, T.; Seino, Y.; et al. Soluble lectin-like oxidized LDL receptor-1 and high-sensitivity troponin T as diagnostic biomarkers for acute coronary syndrome. Improved values with combination usage in emergency rooms. Circ. J. 2011, 75, 2862–2871. [Google Scholar] [CrossRef] [Green Version]
  77. Kume, N.; Mitsuoka, H.; Hayashida, K.; Tanaka, M.; Kominami, G.; Kita, T. Soluble lectin-like oxidized LDL receptor-1 (sLOX-1) as a sensitive and specific biomarker for acute coronary syndrome—Comparison with other biomarkers. J. Cardiol. 2010, 56, 159–165. [Google Scholar] [CrossRef] [Green Version]
  78. Lee, A.S.; Wang, Y.C.; Chang, S.S.; Lo, P.H.; Chang, C.M.; Lu, J.; Burns, A.R.; Chen, C.H.; Kakino, A.; Sawamura, T.; et al. Detection of a High Ratio of Soluble to Membrane-Bound LOX-1 in Aspirated Coronary Thrombi From Patients With ST-Segment-Elevation Myocardial Infarction. J. Am. Heart Assoc. 2020, 9, e014008. [Google Scholar] [CrossRef]
  79. Zhao, Z.W.; Xu, Y.W.; Li, S.M.; Guo, J.J.; Yi, T.; Chen, L.L. Higher serum lectin-like oxidized low-density lipoprotein receptor-1 in patients with stable coronary artery disease is associated with major adverse cardiovascular events: A multicentre pilot study. Biochem. Med. (Zagreb) 2019, 29, 010705. [Google Scholar] [CrossRef]
  80. Hu, C.; Chen, J.; Dandapat, A.; Fujita, Y.; Inoue, N.; Kawase, Y.; Jishage, K.; Suzuki, H.; Li, D.; Hermonat, P.L.; et al. LOX-1 abrogation reduces myocardial ischemia-reperfusion injury in mice. J. Mol. Cell. Cardiol. 2008, 44, 76–83. [Google Scholar] [CrossRef]
  81. Hu, C.; Dandapat, A.; Chen, J.; Fujita, Y.; Inoue, N.; Kawase, Y.; Jishage, K.; Suzuki, H.; Sawamura, T.; Mehta, J.L. LOX-1 deletion alters signals of myocardial remodeling immediately after ischemia-reperfusion. Cardiovasc. Res. 2007, 76, 292–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Iwai-Kanai, E.; Hasegawa, K.; Sawamura, T.; Fujita, M.; Yanazume, T.; Toyokuni, S.; Adachi, S.; Kihara, Y.; Sasayama, S. Activation of Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1 Induces Apoptosis in Cultured Neonatal Rat Cardiac Myocytes. Circulation 2001, 104, 2948–2954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kobayashi, N.; Yoshida, K.; Nakano, S.; Ohno, T.; Honda, T.; Tsubokou, Y.; Matsuoka, H. Cardioprotective Mechanisms of Eplerenone on Cardiac Performance and Remodeling in Failing Rat Hearts. Hypertension 2006, 47, 671–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Besli, F.; Gullulu, S.; Sag, S.; Kecebas, M.; Acikgoz, E.; Sarandol, E.; Aydinlar, A. The relationship between serum lectin-like oxidized LDL receptor-1 levels and systolic heart failure. Acta Cardiol. 2016, 71, 185–190. [Google Scholar] [CrossRef]
  85. Koenig, W. High-sensitivity C-reactive protein and atherosclerotic disease: From improved risk prediction to risk-guided therapy. Int. J. Cardiol. 2013, 168, 5126–5134. [Google Scholar] [CrossRef]
  86. Li, L.; Roumeliotis, N.; Sawamura, T.; Renier, G. C-reactive protein enhances LOX-1 expression in human aortic endothelial cells: Relevance of LOX-1 to C-reactive protein-induced endothelial dysfunction. Circ. Res. 2004, 95, 877–883. [Google Scholar] [CrossRef] [Green Version]
  87. Zhao, X.Q.; Zhang, M.W.; Wang, F.; Zhao, Y.X.; Li, J.J.; Wang, X.P.; Bu, P.L.; Yang, J.M.; Liu, X.L.; Zhang, M.X.; et al. CRP enhances soluble LOX-1 release from macrophages by activating TNF-α converting enzyme. J. Lipid Res. 2011, 52, 923–933. [Google Scholar] [CrossRef] [Green Version]
  88. Hofmann, A.; Brunssen, C.; Wolk, S.; Reeps, C.; Morawietz, H. Soluble LOX-1: A Novel Biomarker in Patients with Coronary Artery Disease, Stroke, and Acute Aortic Dissection? J. Am. Heart Assoc. 2020, 9, e013803. [Google Scholar] [CrossRef]
  89. Jin, P.; Cong, S. LOX-1 and atherosclerotic-related diseases. Clin. Chim. Acta 2019, 491, 24–29. [Google Scholar] [CrossRef]
  90. Huang, W.; Li, Q.; Chen, X.; Lin, Y.; Xue, J.; Cai, Z.; Zhang, W.; Wang, H.; Jin, K.; Shao, B. Soluble lectin-like oxidized low-density lipoprotein receptor-1 as a novel biomarker for large-artery atherosclerotic stroke. Int. J. Neurosci. 2017, 127, 881–886. [Google Scholar] [CrossRef]
  91. Markstad, H.; Edsfeldt, A.; Yao Mattison, I.; Bengtsson, E.; Singh, P.; Cavalera, M.; Asciutto, G.; Björkbacka, H.; Fredrikson, G.N.; Dias, N.; et al. High Levels of Soluble Lectinlike Oxidized Low-Density Lipoprotein Receptor-1 Are Associated With Carotid Plaque Inflammation and Increased Risk of Ischemic Stroke. J. Am. Heart Assoc. 2019, 8, e009874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Skarpengland, T.; Skjelland, M.; Kong Xiang, Y.; Skagen, K.; Holm, S.; Otterdal, K.; Dahl Christen, P.; Krohg-Sørensen, K.; Sagen Ellen, L.; Bjerkeli, V.; et al. Increased Levels of Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1 in Ischemic Stroke and Transient Ischemic Attack. J. Am. Heart Assoc. 2018, 7, e006479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Aslanian Ara, M.; Charo Israel, F. Targeted Disruption of the Scavenger Receptor and Chemokine CXCL16 Accelerates Atherosclerosis. Circulation 2006, 114, 583–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Jin, G. The relationship between serum CXCL16 level and carotid vulnerable plaque in patients with ischemic stroke. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 3911–3915. [Google Scholar]
  95. Mitsuoka, H.; Toyohara, M.; Kume, N.; Hayashida, K.; Jinnai, T.; Tanaka, M.; Kita, T. Circulating Soluble SR-PSOX/CXCL16 as a Biomarker for Acute Coronary Syndrome -Comparison with High-Sensitivity C-Reactive Protein. J. Atheroscler. Thromb. 2009, 16, 586–593. [Google Scholar] [CrossRef] [Green Version]
  96. Jansson, A.M.; Aukrust, P.; Ueland, T.; Smith, C.; Omland, T.; Hartford, M.; Caidahl, K. Soluble CXCL16 Predicts Long-Term Mortality in Acute Coronary Syndromes. Circulation 2009, 119, 3181–3188. [Google Scholar] [CrossRef] [Green Version]
  97. Andersen, T.; Ueland, T.; Ghukasyan Lakic, T.; Åkerblom, A.; Bertilsson, M.; Aukrust, P.; Michelsen, A.E.; James, S.K.; Becker, R.C.; Storey, R.F.; et al. C-X-C Ligand 16 Is an Independent Predictor of Cardiovascular Death and Morbidity in Acute Coronary Syndromes. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 2402–2410. [Google Scholar] [CrossRef] [Green Version]
  98. Laugsand, L.E.; Åsvold, B.O.; Vatten, L.J.; Janszky, I.; Platou, C.; Michelsen, A.E.; Arain, F.; Damås, J.K.; Aukrust, P.; Ueland, T. Soluble CXCL16 and risk of myocardial infarction: The HUNT study in Norway. Atherosclerosis 2016, 244, 188–194. [Google Scholar] [CrossRef]
  99. Borst, O.; Schaub, M.; Walker, B.; Sauter, M.; Muenzer, P.; Gramlich, M.; Mueller, K.; Geisler, T.; Lang, F.; Klingel, K.; et al. CXCL16 is a novel diagnostic marker and predictor of mortality in inflammatory cardiomyopathy and heart failure. Int. J. Cardiol. 2014, 176, 896–903. [Google Scholar] [CrossRef]
  100. Nakamura, K.; Yamagishi, S.-i.; Adachi, H.; Kurita-Nakamura, Y.; Matsui, T.; Yoshida, T.; Sato, A.; Imaizumi, T. Elevation of soluble form of receptor for advanced glycation end products (sRAGE) in diabetic subjects with coronary artery disease. Diabetes Metab. Res. Rev. 2007, 23, 368–371. [Google Scholar] [CrossRef]
  101. Nin, J.W.M.; Jorsal, A.; Ferreira, I.; Schalkwijk, C.G.; Prins, M.H.; Parving, H.-H.; Tarnow, L.; Rossing, P.; Stehouwer, C.D.A. Higher Plasma Soluble Receptor for Advanced Glycation End Products (sRAGE) Levels Are Associated With Incident Cardiovascular Disease and All-Cause Mortality in Type 1 Diabetes. Diabetes 2010, 59, 2027–2032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Colhoun, H.M.; Betteridge, D.J.; Durrington, P.; Hitman, G.; Neil, A.; Livingstone, S.; Charlton-Menys, V.; Bao, W.; DeMicco, D.A.; Preston, G.M.; et al. Total Soluble and Endogenous Secretory Receptor for Advanced Glycation End Products as Predictive Biomarkers of Coronary Heart Disease Risk in Patients With Type 2 Diabetes. Diabetes 2011, 60, 2379–2385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Fujisawa, K.; Katakami, N.; Kaneto, H.; Naka, T.; Takahara, M.; Sakamoto, F.; Irie, Y.; Miyashita, K.; Kubo, F.; Yasuda, T.; et al. Circulating soluble RAGE as a predictive biomarker of cardiovascular event risk in patients with type 2 diabetes. Atherosclerosis 2013, 227, 425–428. [Google Scholar] [CrossRef] [PubMed]
  104. Basta, G.; Del Turco, S.; Navarra, T.; Mazzarisi, A.; Cocci, F.; Coceani, M.; Bianchi, M.; Schlueter, M.; Marraccini, P. Inverse Association between Circulating Levels of Soluble Receptor for Advanced Glycation End-Products and Coronary Plaque Burden. J. Atheroscler. Thromb. 2012, 19, 941–948. [Google Scholar] [CrossRef] [Green Version]
  105. Falcone, C.; Emanuele, E.; D’Angelo, A.; Buzzi Maria, P.; Belvito, C.; Cuccia, M.; Geroldi, D. Plasma Levels of Soluble Receptor for Advanced Glycation End Products and Coronary Artery Disease in Nondiabetic Men. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1032–1037. [Google Scholar] [CrossRef] [Green Version]
  106. Jensen, L.J.N.; Flyvbjerg, A.; Bjerre, M. Soluble Receptor for Advanced Glycation End Product: A Biomarker for Acute Coronary Syndrome. BioMed Res. Int. 2015, 2015, 815942. [Google Scholar] [CrossRef] [Green Version]
  107. Grauen Larsen, H.; Yndigegn, T.; Marinkovic, G.; Grufman, H.; Mares, R.; Nilsson, J.; Goncalves, I.; Schiopu, A. The soluble receptor for advanced glycation end-products (sRAGE) has a dual phase-dependent association with residual cardiovascular risk after an acute coronary event. Atherosclerosis 2019, 287, 16–23. [Google Scholar] [CrossRef]
  108. Hawkes, N. NICE guidelines could put 12 million UK adults on statins. BMJ 2017, 358, j3674. [Google Scholar] [CrossRef]
  109. Liao, J.K. Beyond lipid lowering: The role of statins in vascular protection. Int. J. Cardiol. 2002, 86, 5–18. [Google Scholar] [CrossRef]
  110. Zhu, G.Y.; Zhu, X.L.; Li, R.T.; Liu, T.B.; Shang, D.Y.; Zhang, Y. Atorvastatin inhibits scavenger receptor A and monocyte chemoattractant protein-1 expressions in foam cell. Zhonghua Xin Xue Guan Bing Za Zhi 2007, 35, 666–669. [Google Scholar]
  111. Han, J.; Parsons, M.; Zhou, X.; Nicholson Andrew, C.; Gotto Antonio, M.; Hajjar David, P. Functional Interplay Between the Macrophage Scavenger Receptor Class B Type I and Pitavastatin (NK-104). Circulation 2004, 110, 3472–3479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Arai, T.; Wang, N.; Bezouevski, M.; Welch, C.; Tall, A.R. Decreased Atherosclerosis in Heterozygous Low Density Lipoprotein Receptor-deficient Mice Expressing the Scavenger Receptor BI Transgene. J. Biol. Chem. 1999, 274, 2366–2371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tian, K.; Xu, Y.; Sahebkar, A.; Xu, S. CD36 in Atherosclerosis: Pathophysiological Mechanisms and Therapeutic Implications. Curr. Atheroscler. Rep. 2020, 22, 59. [Google Scholar] [CrossRef] [PubMed]
  114. Yin, M.; Liu, Q.; Yu, L.; Yang, Y.; Lu, M.; Wang, H.; Luo, D.; Rong, X.; Tang, F.; Guo, J. Downregulations of CD36 and Calpain-1, Inflammation, and Atherosclerosis by Simvastatin in Apolipoprotein E Knockout Mice. J. Vasc. Res. 2017, 54, 123–130. [Google Scholar] [CrossRef] [PubMed]
  115. Biocca, S.; Iacovelli, F.; Matarazzo, S.; Vindigni, G.; Oteri, F.; Desideri, A.; Falconi, M. Molecular mechanism of statin-mediated LOX-1 inhibition. Cell Cycle 2015, 14, 1583–1595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Uzui, H.; Hayashi, H.; Nakae, I.; Matsumoto, T.; Uenishi, H.; Hayasaki, H.; Asaji, T.; Matsui, S.; Miwa, K.; Lee, J.D.; et al. Pitavastatin decreases serum LOX-1 ligand levels and MT1-MMP expression in CD14-positive mononuclear cells in hypercholesterolemic patients. Int. J. Cardiol. 2014, 176, 1230–1232. [Google Scholar] [CrossRef]
  117. Mehta, J.L.; Li, D.Y.; Chen, H.J.; Joseph, J.; Romeo, F. Inhibition of LOX-1 by Statins May Relate to Upregulation of eNOS. Biochem. Biophys. Res. Commun. 2001, 289, 857–861. [Google Scholar] [CrossRef]
  118. Li, D.; Chen, H.; Romeo, F.; Sawamura, T.; Saldeen, T.; Mehta, J.L. Statins Modulate Oxidized Low-Density Lipoprotein-Mediated Adhesion Molecule Expression in Human Coronary Artery Endothelial Cells: Role of LOX-1. J. Pharmacol. Exp. Ther. 2002, 302, 601. [Google Scholar] [CrossRef] [Green Version]
  119. Puccetti, L.; Pasqui, A.L.; Bruni, F.; Pastorelli, M.; Ciani, F.; Palazzuoli, A.; Pontani, A.; Ghezzi, A.; Auteri, A. Lectin-like oxidized-LDL receptor-1 (LOX-1) polymorphisms influence cardiovascular events rate during statin treatment. Int. J. Cardiol. 2007, 119, 41–47. [Google Scholar] [CrossRef]
  120. Cuccurullo, C.; Iezzi, A.; Fazia Maria, L.; De Cesare, D.; Di Francesco, A.; Muraro, R.; Bei, R.; Ucchino, S.; Spigonardo, F.; Chiarelli, F.; et al. Suppression of Rage as a Basis of Simvastatin-Dependent Plaque Stabilization in Type 2 Diabetes. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 2716–2723. [Google Scholar] [CrossRef] [Green Version]
  121. Santilli, F.; Bucciarelli, L.; Noto, D.; Cefalù, A.B.; Davì, V.; Ferrante, E.; Pettinella, C.; Averna, M.R.; Ciabattoni, G.; Davì, G. Decreased plasma soluble RAGE in patients with hypercholesterolemia: Effects of statins. Free Radic. Biol. Med. 2007, 43, 1255–1262. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, Y.; Liu, Z.P. PCSK9 Inhibitors: Novel Therapeutic Strategies for Lowering LDLCholesterol. Mini Rev. Med. Chem. 2019, 19, 165–176. [Google Scholar] [CrossRef] [PubMed]
  123. Ding, Z.; Liu, S.; Wang, X.; Deng, X.; Fan, Y.; Shahanawaz, J.; Shmookler Reis, R.J.; Varughese, K.I.; Sawamura, T.; Mehta, J.L. Cross-talk between LOX-1 and PCSK9 in vascular tissues. Cardiovasc. Res. 2015, 107, 556–567. [Google Scholar] [CrossRef] [PubMed]
  124. Ding, Z.; Pothineni, N.V.K.; Goel, A.; Lüscher, T.F.; Mehta, J.L. PCSK9 and inflammation: Role of shear stress, pro-inflammatory cytokines, and LOX-1. Cardiovasc. Res. 2020, 116, 908–915. [Google Scholar] [CrossRef]
  125. Glintborg, D.; Højlund, K.; Andersen, M.; Henriksen, J.E.; Beck-Nielsen, H.; Handberg, A. Soluble CD36 and Risk Markers of Insulin Resistance and Atherosclerosis Are Elevated in Polycystic Ovary Syndrome and Significantly Reduced During Pioglitazone Treatment. Diabetes Care 2008, 31, 328–334. [Google Scholar] [CrossRef] [Green Version]
  126. Gao, H.; Li, H.; Li, W.; Shen, X.; Di, B. Pioglitazone Attenuates Atherosclerosis in Diabetic Mice by Inhibition of Receptor for Advanced Glycation End-Product (RAGE) Signaling. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2017, 23, 6121–6131. [Google Scholar] [CrossRef] [Green Version]
  127. Li, Q.; Zhao, W.; Zeng, X.; Hao, Z. Ursolic Acid Attenuates Atherosclerosis in ApoE(-/-) Mice: Role of LOX-1 Mediated by ROS/NF-κB Pathway. Molecules 2018, 23, 1101. [Google Scholar] [CrossRef] [Green Version]
  128. Song, G.; Tian, H.; Liu, J.; Zhang, H.; Sun, X.; Qin, S. H2 inhibits TNF-α-induced lectin-like oxidized LDL receptor-1 expression by inhibiting nuclear factor κB activation in endothelial cells. Biotechnol. Lett. 2011, 33, 1715–1722. [Google Scholar] [CrossRef]
  129. Taye, A.; Sawamura, T.; Morawietz, H. Aldosterone augments LOX-1-mediated low-density lipoprotein uptake in human umbilical artery endothelial cells. Pharmacol. Rep. 2010, 62, 311–318. [Google Scholar] [CrossRef]
  130. Morawietz, H.; Rueckschloss, U.; Niemann, B.; Duerrschmidt, N.; Galle, J.; Hakim, K.; Zerkowski, H.R.; Sawamura, T.; Holtz, J. Angiotensin II induces LOX-1, the human endothelial receptor for oxidized low-density lipoprotein. Circulation 1999, 100, 899–902. [Google Scholar] [CrossRef] [Green Version]
  131. Babaev, V.R.; Gleaves, L.A.; Carter, K.J.; Suzuki, H.; Kodama, T.; Fazio, S.; Linton, M.F. Reduced Atherosclerotic Lesions in Mice Deficient for Total or Macrophage-Specific Expression of Scavenger Receptor-A. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 2593–2599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Zhu, Y.; Yang, Y.; Li, F.; Fan, S.; Chen, X.; Lu, Y.; Wei, Y.; Chen, Q.; Xia, L.; Tang, J.; et al. Stimulation of the class-A scavenger receptor induces neutrophil extracellular traps (NETs) by ERK dependent NOX2 and ROMO1 activation. Biochem. Biophys. Res. Commun. 2019, 511, 847–854. [Google Scholar] [CrossRef] [PubMed]
  133. Hehir, S.; Plourde, N.M.; Gu, L.; Poree, D.E.; Welsh, W.J.; Moghe, P.V.; Uhrich, K.E. Carbohydrate composition of amphiphilic macromolecules influences physicochemical properties and binding to atherogenic scavenger receptor A. Acta Biomater. 2012, 8, 3956–3962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Iverson, N.M.; Plourde, N.M.; Sparks, S.M.; Wang, J.; Patel, E.N.; Shah, P.S.; Lewis, D.R.; Zablocki, K.R.; Nackman, G.B.; Uhrich, K.E.; et al. Dual use of amphiphilic macromolecules as cholesterol efflux triggers and inhibitors of macrophage athero-inflammation. Biomaterials 2011, 32, 8319–8327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Fichtlscherer, S.; Breuer, S.; Heeschen, C.; Dimmeler, S.; Zeiher, A.M. Interleukin-10 serum levels and systemic endothelial vasoreactivity in patients with coronary artery disease. J. Am. Coll. Cardiol. 2004, 44, 44–49. [Google Scholar] [CrossRef] [Green Version]
  136. Anguera, I.; Miranda-Guardiola, F.; Bosch, X.; Filella, X.; Sitges, M.; Marin, J.L.; Betriu, A.; Sanz, G. Elevation of serum levels of the anti-inflammatory cytokine interleukin-10 and decreased risk of coronary events in patients with unstable angina. Am. Heart J. 2002, 144, 811–817. [Google Scholar] [CrossRef]
  137. Yang, H.; Chen, S.; Tang, Y.; Dai, Y. Interleukin-10 down-regulates oxLDL induced expression of scavenger receptor A and Bak-1 in macrophages derived from THP-1 cells. Arch. Biochem. Biophys. 2011, 512, 30–37. [Google Scholar] [CrossRef]
  138. Dai, X.-Y.; Cai, Y.; Mao, D.-D.; Qi, Y.-F.; Tang, C.; Xu, Q.; Zhu, Y.; Xu, M.-J.; Wang, X. Increased stability of phosphatase and tensin homolog by intermedin leading to scavenger receptor A inhibition of macrophages reduces atherosclerosis in apolipoprotein E-deficient mice. J. Mol. Cell. Cardiol. 2012, 53, 509–520. [Google Scholar] [CrossRef]
  139. Cai, Y.; Xu, M.-J.; Teng, X.; Zhou, Y.B.; Chen, L.; Zhu, Y.; Wang, X.; Tang, C.S.; Qi, Y.F. Intermedin inhibits vascular calcification by increasing the level of matrix γ-carboxyglutamic acid protein. Cardiovasc. Res. 2009, 85, 864–873. [Google Scholar] [CrossRef] [Green Version]
  140. Usui, H.K.; Shikata, K.; Sasaki, M.; Okada, S.; Matsuda, M.; Shikata, Y.; Ogawa, D.; Kido, Y.; Nagase, R.; Yozai, K.; et al. Macrophage Scavenger Receptor-A–Deficient Mice Are Resistant against Diabetic Nephropathy Through Amelioration of Microinflammation. Diabetes 2007, 56, 363–372. [Google Scholar] [CrossRef] [Green Version]
  141. Mäkinen, P.I.; Lappalainen, J.P.; Heinonen, S.E.; Leppänen, P.; Lähteenvuo, M.T.; Aarnio, J.V.; Heikkilä, J.; Turunen, M.P.; Ylä-Herttuala, S. Silencing of either SR-A or CD36 reduces atherosclerosis in hyperlipidaemic mice and reveals reciprocal upregulation of these receptors. Cardiovasc. Res. 2010, 88, 530–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Seizer, P.; Schiemann, S.; Merz, T.; Daub, K.; Bigalke, B.; Stellos, K.; Muller, I.; Stockle, C.; Muller, K.; Gawaz, M.; et al. CD36 and macrophage scavenger receptor a modulate foam cell formation via inhibition of lipid-laden platelet phagocytosis. Semin. Thromb. Hemost. 2010, 36, 157–162. [Google Scholar] [CrossRef] [PubMed]
  143. 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] [Green Version]
  144. Kuchibhotla, S.; Vanegas, D.; Kennedy, D.J.; Guy, E.; Nimako, G.; Morton, R.E.; Febbraio, M. Absence of CD36 protects against atherosclerosis in ApoE knock-out mice with no additional protection provided by absence of scavenger receptor A I/II. Cardiovasc. Res. 2008, 78, 185–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Cavallari, J.F.; Anhê, F.F.; Foley, K.P.; Denou, E.; Chan, R.W.; Bowdish, D.M.E.; Schertzer, J.D. Targeting macrophage scavenger receptor 1 promotes insulin resistance in obese male mice. Physiol. Rep. 2018, 6, e13930. [Google Scholar] [CrossRef] [PubMed]
  146. Wilson, C.G.; Tran, J.L.; Erion, D.M.; Vera, N.B.; Febbraio, M.; Weiss, E.J. Hepatocyte-Specific Disruption of CD36 Attenuates Fatty Liver and Improves Insulin Sensitivity in HFD-Fed Mice. Endocrinology 2016, 157, 570–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Cho, S.; Kim, E. CD36: A multi-modal target for acute stroke therapy. J. Neurochem. 2009, 109 (Suppl. 1), 126–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Cho, S. CD36 as a therapeutic target for endothelial dysfunction in stroke. Curr. Pharm. Des. 2012, 18, 3721–3730. [Google Scholar] [CrossRef] [Green Version]
  149. Mwaikambo, B.R.; Yang, C.; Ong, H.; Chemtob, S.; Hardy, P. Emerging roles for the CD36 scavenger receptor as a potential therapeutic target for corneal neovascularization. Endocr. Metab. Immune Disord Drug Targets 2008, 8, 255–272. [Google Scholar] [CrossRef]
  150. Zhao, L.; Varghese, Z.; Moorhead, J.F.; Chen, Y.; Ruan, X.Z. CD36 and lipid metabolism in the evolution of atherosclerosis. Br. Med. Bull. 2018, 126, 101–112. [Google Scholar] [CrossRef] [Green Version]
  151. Qin, L.; Kim, E.; Ratan, R.; Lee, F.S.; Cho, S. Genetic Variant of BDNF (Val66Met) Polymorphism Attenuates Stroke-Induced Angiogenic Responses by Enhancing Anti-Angiogenic Mediator CD36 Expression. J. Neurosci. 2011, 31, 775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Cho, S.; Szeto, H.H.; Kim, E.; Kim, H.; Tolhurst, A.T.; Pinto, J.T. A Novel Cell-permeable Antioxidant Peptide, SS31, Attenuates Ischemic Brain Injury by Down-regulating CD36. J. Biol. Chem. 2007, 282, 4634–4642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Chnari, E.; Nikitczuk, J.S.; Wang, J.; Uhrich, K.E.; Moghe, P.V. Engineered Polymeric Nanoparticles for Receptor-Targeted Blockage of Oxidized Low Density Lipoprotein Uptake and Atherogenesis in Macrophages. Biomacromolecules 2006, 7, 1796–1805. [Google Scholar] [CrossRef] [PubMed]
  154. Marleau, S.; Harb, D.; Bujold, K.; Avallone, R.; Iken, K.; Wang, Y.; Demers, A.; Sirois, M.G.; Febbraio, M.; Silverstein, R.L.; et al. EP 80317, a ligand of the CD36 scavenger receptor, protects apolipoprotein E-deficient mice from developing atherosclerotic lesions. FASEB J. 2005, 19, 1869–1871. [Google Scholar] [CrossRef] [PubMed]
  155. Bujold, K.; Mellal, K.; Zoccal, K.F.; Rhainds, D.; Brissette, L.; Febbraio, M.; Marleau, S.; Ong, H. EP 80317, a CD36 selective ligand, promotes reverse cholesterol transport in apolipoprotein E-deficient mice. Atherosclerosis 2013, 229, 408–414. [Google Scholar] [CrossRef]
  156. Wang, C.; Xu, W.; Liang, M.; Huang, D.; Huang, K. CTRP13 inhibits atherosclerosis via autophagy-lysosome-dependent degradation of CD36. FASEB J. 2019, 33, 2290–2300. [Google Scholar] [CrossRef] [Green Version]
  157. De Siqueira, J.; Abdul Zani, I.; Russell, D.A.; Wheatcroft, S.B.; Ponnambalam, S.; Homer-Vanniasinkam, S. Clinical and Preclinical Use of LOX-1-Specific Antibodies in Diagnostics and Therapeutics. J. Cardiovasc. Transl. Res. 2015, 8, 458–465. [Google Scholar] [CrossRef]
  158. Mehta Jawahar, L.; Sanada, N.; Hu Chang, P.; Chen, J.; Dandapat, A.; Sugawara, F.; Satoh, H.; Inoue, K.; Kawase, Y.; Jishage, K.-I.; et al. Deletion of LOX-1 Reduces Atherogenesis in LDLR Knockout Mice Fed High Cholesterol Diet. Circ. Res. 2007, 100, 1634–1642. [Google Scholar] [CrossRef] [Green Version]
  159. Li, D.; Williams, V.; Liu, L.; Chen, H.; Sawamura, T.; Romeo, F.; Mehta, J.L. Expression of lectin-like oxidized low-density lipoprotein receptors during ischemia-reperfusion and its role in determination of apoptosis and left ventricular dysfunction. J. Am. Coll. Cardiol. 2003, 41, 1048–1055. [Google Scholar] [CrossRef] [Green Version]
  160. Hinagata, J.; Kakutani, M.; Fujii, T.; Naruko, T.; Inoue, N.; Fujita, Y.; Mehta, J.L.; Ueda, M.; Sawamura, T. Oxidized LDL receptor LOX-1 is involved in neointimal hyperplasia after balloon arterial injury in a rat model. Cardiovasc. Res. 2006, 69, 263–271. [Google Scholar] [CrossRef]
  161. Li, D.; Williams, V.; Liu, L.; Chen, H.; Sawamura, T.; Antakli, T.; Mehta, J.L. LOX-1 inhibition in myocardial ischemia-reperfusion injury: Modulation of MMP-1 and inflammation. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H1795–H1801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Nakano, A.; Inoue, N.; Sato, Y.; Nishimichi, N.; Takikawa, K.; Fujita, Y.; Kakino, A.; Otsui, K.; Yamaguchi, S.; Matsuda, H.; et al. LOX-1 mediates vascular lipid retention under hypertensive state. J. Hypertens. 2010, 28, 1273–1280. [Google Scholar] [CrossRef] [PubMed]
  163. Dominguez, J.H.; Mehta, J.L.; Li, D.; Wu, P.; Kelly, K.J.; Packer, C.S.; Temm, C.; Goss, E.; Cheng, L.; Zhang, S.; et al. Anti-LOX-1 therapy in rats with diabetes and dyslipidemia: Ablation of renal vascular and epithelial manifestations. Am. J. Physiol. Ren. Physiol. 2008, 294, F110–F119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. McDonald, R.A.; Hata, A.; MacLean, M.R.; Morrell, N.W.; Baker, A.H. MicroRNA and vascular remodelling in acute vascular injury and pulmonary vascular remodelling. Cardiovasc. Res. 2012, 93, 594–604. [Google Scholar] [CrossRef] [Green Version]
  165. Dai, Y.; Wu, X.; Dai, D.; Li, J.; Mehta, J.L. MicroRNA-98 regulates foam cell formation and lipid accumulation through repression of LOX-1. Redox Biol. 2018, 16, 255–262. [Google Scholar] [CrossRef]
  166. Liu, M.; Tao, G.; Liu, Q.; Liu, K.; Yang, X. MicroRNA let-7g alleviates atherosclerosis via the targeting of LOX-1 in vitro and in vivo. Int. J. Mol. Med. 2017, 40, 57–64. [Google Scholar] [CrossRef] [Green Version]
  167. Arjuman, A.; Chandra, N.C. LOX-1: A potential target for therapy in atherosclerosis; an in vitro study. Int. J. Biochem. Cell Biol. 2017, 91, 65–80. [Google Scholar] [CrossRef]
  168. Gavrilov, K.; Saltzman, W.M. Therapeutic siRNA: Principles, challenges, and strategies. Yale J. Biol. Med. 2012, 85, 187–200. [Google Scholar]
  169. Ishigaki, T.; Ohki, I.; Utsunomiya-Tate, N.; Tate, S.I. Chimeric structural stabilities in the coiled-coil structure of the NECK domain in human lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1). J. Biochem. 2007, 141, 855–866. [Google Scholar] [CrossRef]
  170. Thakkar, S.; Wang, X.; Khaidakov, M.; Dai, Y.; Gokulan, K.; Mehta, J.L.; Varughese, K.I. Structure-based Design Targeted at LOX-1, a Receptor for Oxidized Low-Density Lipoprotein. Sci. Rep. 2015, 5, 16740. [Google Scholar] [CrossRef] [Green Version]
  171. Fan, Q.; Cai, H.; Yang, H.; Li, L.; Yuan, C.; Lu, X.; Wan, L. Biological evaluation of 131I- and CF750-labeled Dmab(scFv)-Fc antibodies for xenograft imaging of CD25-positive tumors. BioMed Res. Int. 2014, 2014, 459676. [Google Scholar] [CrossRef] [PubMed]
  172. Hu, W.; Xie, Q.; Xiang, H. Improved scFv Anti-LOX-1 Binding Activity by Fusion with LOX-1-Binding Peptides. BioMed Res. Int. 2017, 2017, 8946935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Hu, W.; Xie, Q.; Liu, L.; Xiang, H. Enhanced Bioactivity of the Anti-LOX-1 scFv Engineered by Multimerization Strategy. Appl. Biochem. Biotechnol. 2018, 185, 233–247. [Google Scholar] [CrossRef] [PubMed]
  174. Minami, M.; Kume, N.; Shimaoka, T.; Kataoka, H.; Hayashida, K.; Akiyama, Y.; Nagata, I.; Ando, K.; Nobuyoshi, M.; Hanyuu, M.; et al. Expression of SR-PSOX, a Novel Cell-Surface Scavenger Receptor for Phosphatidylserine and Oxidized LDL in Human Atherosclerotic Lesions. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 1796–1800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Hofnagel, O.; Engel, T.; Severs, N.J.; Robenek, H.; Buers, I. SR-PSOX at sites predisposed to atherosclerotic lesion formation mediates monocyte-endothelial cell adhesion. Atherosclerosis 2011, 217, 371–378. [Google Scholar] [CrossRef]
  176. Bierhaus, A.; Hofmann, M.A.; Ziegler, R.; Nawroth, P.P. AGEs and their interaction with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept. Cardiovasc. Res. 1998, 37, 586–600. [Google Scholar] [CrossRef] [Green Version]
  177. Xu, L.; Wang, Y.-R.; Li, P.-C.; Feng, B. Advanced glycation end products increase lipids accumulation in macrophages through upregulation of receptor of advanced glycation end products: Increasing uptake, esterification and decreasing efflux of cholesterol. Lipids Health Dis. 2016, 15, 161. [Google Scholar] [CrossRef] [Green Version]
  178. Zhou, J.; Bai, W.; Liu, Q.; Cui, J.; Zhang, W. Intermittent Hypoxia Enhances THP-1 Monocyte Adhesion and Chemotaxis and Promotes M1 Macrophage Polarization via RAGE. BioMed Res. Int. 2018, 2018, 1650456. [Google Scholar] [CrossRef]
  179. Soro-Paavonen, A.; Watson, A.M.D.; Li, J.; Paavonen, K.; Koitka, A.; Calkin, A.C.; Barit, D.; Coughlan, M.T.; Drew, B.G.; Lancaster, G.I.; et al. Receptor for advanced glycation end products (RAGE) deficiency attenuates the development of atherosclerosis in diabetes. Diabetes 2008, 57, 2461–2469. [Google Scholar] [CrossRef] [Green Version]
  180. Park, L.; Raman, K.G.; Lee, K.J.; Lu, Y.; Ferran, L.J.; Chow, W.S.; Stern, D.; Schmidt, A.M. Suppression of accelerated diabetic atherosclerosis by the soluble receptor for advanced glycation endproducts. Nat. Med. 1998, 4, 1025–1031. [Google Scholar] [CrossRef]
  181. Wu, G.; Zhang, J.; Zhao, Q.; Zhuang, W.; Ding, J.; Zhang, C.; Gao, H.; Pang, D.W.; Pu, K.; Xie, H.Y. Molecularly Engineered Macrophage-Derived Exosomes with Inflammation Tropism and Intrinsic Heme Biosynthesis for Atherosclerosis Treatment. Angew. Chem. Int. Ed. Engl. 2020, 59, 4068–4074. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Key events in atherosclerotic plaque initiation. Blood LDL can accumulate within the arterial wall, in the sub-endothelial intima. This accumulated LDL can be chemically modified or oxidized: this new lipid particle species (e.g., oxLDL) promotes chronic inflammation, which promotes the trans-endothelial migration of immune cell types and foam cell development. Abbreviations: endothelial cell, EC; intercellular adhesion molecule-1, ICAM-1; interferon-γ, IFNγ; low-density lipoprotein, LDL; Neutrophil extracellular traps, NETS; oxidized low-density lipoprotein, oxLDL; scavenger receptor, SR; T- helper 1 cell, Th1; vascular cell adhesion molecule-1, VCAM-1; vascular smooth muscle cell, VSMC.
Figure 1. Key events in atherosclerotic plaque initiation. Blood LDL can accumulate within the arterial wall, in the sub-endothelial intima. This accumulated LDL can be chemically modified or oxidized: this new lipid particle species (e.g., oxLDL) promotes chronic inflammation, which promotes the trans-endothelial migration of immune cell types and foam cell development. Abbreviations: endothelial cell, EC; intercellular adhesion molecule-1, ICAM-1; interferon-γ, IFNγ; low-density lipoprotein, LDL; Neutrophil extracellular traps, NETS; oxidized low-density lipoprotein, oxLDL; scavenger receptor, SR; T- helper 1 cell, Th1; vascular cell adhesion molecule-1, VCAM-1; vascular smooth muscle cell, VSMC.
Cells 09 02453 g001
Figure 2. Five different SR classes (A, B, E, G, J) are most closely linked to the initiation and progression of atherosclerosis. These molecules are potential biomarkers and therapeutic targets. Their structures are represented in the sketches above. Abbreviations: N-terminus, N; C-terminus, C.
Figure 2. Five different SR classes (A, B, E, G, J) are most closely linked to the initiation and progression of atherosclerosis. These molecules are potential biomarkers and therapeutic targets. Their structures are represented in the sketches above. Abbreviations: N-terminus, N; C-terminus, C.
Cells 09 02453 g002
Table 1. SR expression and function.
Table 1. SR expression and function.
SR ClassNomenclature/IsoformsExpressionFunction
SR-AMacrophage SR
  • Macrophages and subtypes e.g., Kupffer cells, medullary thymic macrophages
  • Vascular smooth muscle cells
  • Endothelial cells
  • Microglia
[26,27,28]
  • Internalization of oxLDL in atherosclerosis.
  • Bacterial cell recognition in innate immunity.
  • Involved in beta-amyloid clearance in the brain.
[20,29,30,31,32]
SR-BSR-B1

SR-B2 (CD36)

SR-B3 (LIMP2)
  • Macrophages
  • Endothelial cells
  • Adipocytes
  • Renal tubular cells
  • Podocytes
[33,34]
  • Internalization of oxLDL in atherosclerosis.
  • Bacterial cell adhesion, internalization and lysosomal sequestration.
  • May participate in systemic inflammation associated with sepsis.
[23,35,36]
SR-CNot discussed (plant receptor)
SR-DCD68
  • Macrophages
  • Monocytes
  • Microglia
  • Osteoclasts
  • Myeloid dendritic cells
[37]
  • May play a role in internalization of oxLDL.
[38,39,40]
SR-ESR-E1 (LOX-1)

SR-E1.1 (LOXIN)

SR-E2
(Dectin-1)
  • Macrophages
  • Neutrophils
  • Endothelial cells
  • Smooth muscle cells
  • Platelets
[41]
  • Internalization of oxLDL in atherosclerosis.
  • Bacterial and fungal cell recognition in innate immunity.
  • LOXIN (alternatively spliced form of SR-E1) demonstrates no known scavenger receptor activity but has been shown to exert a dominant negative effect on LOX-1 function.
[41,42]
SR-FSR-F1 (SREC-1,SCARF-1)

SR-F2 (SREC-2, SCARF-2)

MEGF10
  • Neuronal cells
  • Sinusoidal Endothelial cells
[19,43]
  • SR-F1 binds and internalizes oxLDL, whilst formation of heterodimer with SR-F2 suppresses oxLDL binding activity.
  • MEGF10 is involved in beta-amyloid clearance in the brain.
[21,43,44]
SR-GSR-G1 (CXCL16, SR-PSOX)
  • Macrophages
  • Smooth muscle cells
[45,46]
  • Adhesion of cells expressing the CXCR6 receptor such as natural killer T cells and polarized T helper cells.
  • Internalization of oxLDL in atherosclerosis.
  • Bacterial cell recognition in innate immunity.
[47,48,49]
SR-HSR-H1 (FEEL-1, Stabilin-1, Clever-1)

SR-H2 (FEEL-2, Stabilin-2)
  • Macrophages
  • Splenic, hepatic and lymphatic endothelial cells
  • Monocytes
[50,51]
  • Internalization of oxLDL in atherosclerosis.
  • Bacterial cell recognition in innate immunity.
  • Binding of advanced glycation end-products (AGEs).
  • Potential role in the adhesion of metastatic tumour cells to lymphatic endothelial cells.
[50,52,53]
SR-ISR-I1 (CD163, haemoglobin SR)

SR-I2 (CD163B)
  • Circulating and tissue specific macrophages and monocytes
  • Leukemic blasts
[54,55]
  • Mediates haptoglobin-haemoglobin complex endocytosis during intravascular haemolysis.
[55]
SR-JRAGE
  • Endothelial cells
  • Hepatocytes
  • Smooth muscle cells
  • Monocytes
[56]
  • Binds AGEs.
  • Amplification of immune and inflammatory responses, cell mobility, arterial injury, and atherogenesis via sustained post-receptor signalling.
[57]
Table 2. SRs as biomarkers in CVD.
Table 2. SRs as biomarkers in CVD.
Disease StateSR BiomarkerHuman Studies
CAD/ACSSR-A
  • SR-A index (monocyte SR-A expression in peripheral blood film) associated with advanced plaque morphology [63,64,65].
  • SR-A-targeted probes used for plaque imaging [67].
SR-B
  • Monocyte CD36 expression associated with increased atheroma burden in CAD patients [66].
  • CD36-targeted probes used for plaque imaging [68].
  • Baseline sCD36 not associated with CAD development or plaque burden [70,71].
SR-E
  • Raised serum levels of sLOX-1 used as a diagnostic biomarker in ACS [75,76,77].
  • sLOX-1 levels combined with troponin levels provide more diagnostic accuracy compared with either in isolation [76,77].
  • Raised sLOX-1 levels correlate with incidence of major adverse cardiovascular events in stable CAD [79].
SR-G
  • sCXCL16 levels improve diagnostic accuracy in ACS when combined with troponin/LOX-1 [49,95].
  • sCXCL16 levels are associated with increased adverse events and mortality in ACS [96,97].
  • Baseline sCXCL16 levels associated with increased risk of ACS in non-CAD patients [98].
SR-J
  • Due to conflicting reports, the potential use of sRAGE as a biomarker for ACS remains controversial.
CardiomyopathySR-E
  • sLOX-1 levels negatively correlate with left ventricular ejection fraction [84].
SR-G
  • Increased sCXCL16 levels independently predict mortality in inflammatory and non-inflammatory cardiomyopathy [99].
  • Enhanced CXCL16 expression in cardiac myocytes in inflammatory cardiomyopathy [99].
StrokeSR-E
  • sLOX-1 levels increased in ischaemic and haemorrhagic stroke [88,89].
  • Increased baseline sLOX-1 levels associated with increased incidence of ischaemic stroke and secondary ischaemic events post-carotid intervention [91].
SR-G
  • Increased sCXCL16 associated with more advanced carotid lesions in patients presenting with ischaemic stroke [94].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cuthbert, G.A.; Shaik, F.; Harrison, M.A.; Ponnambalam, S.; Homer-Vanniasinkam, S. Scavenger Receptors as Biomarkers and Therapeutic Targets in Cardiovascular Disease. Cells 2020, 9, 2453. https://doi.org/10.3390/cells9112453

AMA Style

Cuthbert GA, Shaik F, Harrison MA, Ponnambalam S, Homer-Vanniasinkam S. Scavenger Receptors as Biomarkers and Therapeutic Targets in Cardiovascular Disease. Cells. 2020; 9(11):2453. https://doi.org/10.3390/cells9112453

Chicago/Turabian Style

Cuthbert, Gary A., Faheem Shaik, Michael A. Harrison, Sreenivasan Ponnambalam, and Shervanthi Homer-Vanniasinkam. 2020. "Scavenger Receptors as Biomarkers and Therapeutic Targets in Cardiovascular Disease" Cells 9, no. 11: 2453. https://doi.org/10.3390/cells9112453

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