Next Article in Journal
A Paternal Methylation Error in the Congenital Hydrocephalic Texas (H-Tx) Rat Is Partially Rescued with Natural Folate Supplements
Next Article in Special Issue
Pathophysiological Involvement of Mast Cells and the Lipid Mediators in Pulmonary Vascular Remodeling
Previous Article in Journal
Gut Microbial Perturbation and Host Response Induce Redox Pathway Upregulation along the Gut–Liver Axis during Giardiasis in C57BL/6J Mouse Model
Previous Article in Special Issue
APOE Genotypes Modulate Inflammation Independently of Their Effect on Lipid Metabolism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis

1
Fondazione CNR-Regione Toscana G Monasterio, 56124 Pisa, Italy
2
Institute of Clinical Physiology, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1637; https://doi.org/10.3390/ijms24021637
Submission received: 2 December 2022 / Revised: 6 January 2023 / Accepted: 8 January 2023 / Published: 13 January 2023
(This article belongs to the Special Issue Lipid and Fatty Acid Metabolism in Cardiovascular Diseases)

Abstract

:
It is commonly believed that the inactivation of inflammation is mainly due to the decay or cessation of inducers. In reality, in connection with the development of atherosclerosis, spontaneous decay of inducers is not observed. It is now known that lipid mediators originating from polyunsaturated fatty acids (PUFAs), which are important constituents of all cell membranes, can act in the inflamed tissue and bring it to resolution. In fact, PUFAs, such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), are precursors to both pro-inflammatory and anti-inflammatory compounds. In this review, we describe the lipid mediators of vascular inflammation and resolution, and their biochemical activity. In addition, we highlight data from the literature that often show a worsening of atherosclerotic disease in subjects deficient in lipid mediators of inflammation resolution, and we also report on the anti-proteasic and anti-thrombotic properties of these same lipid mediators. It should be noted that despite promising data observed in both animal and in vitro studies, contradictory clinical results have been observed for omega-3 PUFAs. Many further studies will be required in order to clarify the observed conflicts, although lifestyle habits such as smoking or other biochemical factors may often influence the normal synthesis of lipid mediators of inflammation resolution.

1. Introduction

Atherosclerosis is a lipoprotein-driven, non-resolving chronic inflammatory disease that could cause heart attacks [1,2] and cardiovascular disease [3]. Atherosclerosis begins with the retention of apolipoprotein B–containing lipoproteins within the subendothelial intima of arteries, which leads to the activation of endothelial cells, recruitment of leukocytes, and accumulation of cells, extracellular matrix, and lipids [4].
Macrophages, the major leukocyte type ingesting retained lipoproteins in the subendothelial region, in some cases modified by oxidation [5], become foam cells that help to repair and to clear the tissue from the necrotic cells. However, the prolonged pathogenesis of atherosclerosis is marked by endothelial cell dysfunction, followed by the deposition of lipid-laden macrophages and VSMC proliferation, leading to the occlusion of the arterial lumen. A build-up of fatty deposits inside the arteries (atherosclerosis) and an increased risk of blood clots are usually associated with cardiovascular diseases, a general term for conditions affecting the heart or blood vessels. In fact, in several cardiovascular diseases, a non-resolved acute inflammation [3] could cause chronic fibrotic disease [6,7]. Sometimes, plaque necrosis further increases the inflammatory response [8] and could break down the integrity of the fibrous cap, triggering acute occlusive thrombosis and tissue infarction [8,9].
In the past, it was assumed that the reduction or elimination of vascular inflammation was principally caused by the decay or cessation of lipid inducers; therefore, the use of LDL-lowering therapy has been revealed to be very useful. Data from basic and clinical studies indicates that anti-inflammatory treatment could represent an important approach to prevent coronary and cerebral atherosclerotic events. However, despite the availability of this safe therapy, the incidence of cardiovascular disease has been increasing [10].
In recent years, the importance of lipid mediators in resolving inflammation has been spotlighted [11,12,13,14,15]. Lipid mediators originate from PUFAs, which represent an important constituent of all the cell membranes in the body. In particular, AA, EPA, and DHA are precursors of several lipid mediators that display pro- or anti-inflammatory properties based on the PUFA, enzyme, and receptor to which they are related. Originally considered only as membrane and energy storage molecules, lipid mediators are now recognized as powerful signaling molecules that regulate a multitude of cellular responses, including growth, apoptosis, and inflammation/infection [16].
However, defects in the resolution of inflammation by lipid mediators have emerged as key causal factors of atherosclerosis [6,17,18]. Specifically, here, we will discuss the role of lipids derived from AA (prostanoids, leukotrienes, epoxyeicosatrienoic acids, and lipoxins), EPA (E-series resolvins), and DHA (D-series resolvins, protectins, and maresins) in relation to cardiovascular inflammation, with the goals of investigating obstacles in the path of vascular inflammation resolution; and identifying novel therapeutic targets in cardiovascular diseases.
Lipid mediators are grouped into three classes based on their structure and function [19].

2. Class 1: Eicosanoids

Class 1: Eicosanoids (Prostanoids, Leukotrienes, Epoxyeicosatrienoic acids, and Lipoxins). Eicosanoids derive from membrane phospholipid-released AA which is metabolized by cyclooxygenases (COXs), lipoxygenases (LOXs), and cytochrome P450 (CYP) enzymes to form prostanoids, leukotrienes (LTs), lipoxins (LXs), epoxyeicosatrienoic acids (EETs), and eicosatetraenoic acids (ETEs) [20,21,22,23].

2.1. Molecular and Biochemical Characteristic of PROSTANOIDS

The prostanoids comprise the prostaglandins (PGs), the prostacyclins (PGIs), and the thromboxanes (TXA2) (Figure 1).
Prostaglandins show the prefix ‘PG’ followed by a letter A to K, depending on the nature and position of the substituents on the ring, and on the presence or absence of double bonds or hydroxyl groups in various positions in the ring. TXA2 contains an unstable bicyclic oxygenated ring structure [24].
COX-1 is mainly a constitutive enzyme that maintains basal levels of PGs, while COX-2 is an inducible isoform that is regulated by growth factors, cytokines, shear stress, and mitogens. COX-2 is typically upregulated in atherosclerotic lesions [25]. Cyclooxygenases sequentially convert arachidonic acid to the endoperoxides PGG2 and PGH2. PGH2 is further isomerized to each terminal prostanoid by the corresponding synthases. Notably, mPGES (microsomal PGE synthase-1) catalyzes the isomerization of PGH2 into PGE2 biosynthesis. mPGES-1 can couple with both COX-1 and COX-2, and its induced expression increases PGE2, which often correlates with the induction of COX-2 [26].
Prostanoids exert a variety of actions through their specific G-protein-coupled receptors, which are differentially expressed in tissues and cells, triggering varied downstream signaling [27]. Among them, the IP, DP, EP2, and EP4 receptors mediate a cAMP rise, whereas EP3 induces a decline in cAMP levels. Finally, FP, and EP1 receptors induce calcium mobilization. However, the concentration or structure of the ligand could change their action [28]. For example, PGI2 stimulates a G-protein-coupled increase in cAMP and protein kinase A, resulting in a decreased intracellular Ca2+ concentration [29]. Instead, PGE2, which is produced by a variety of cell types [30], exerts different actions depending on the type of receptor; it increases intracellular cAMP when binding to EP2 and IP receptors, mainly coupled to Gs protein, while it inhibits intracellular cAMP production binding to EP3 receptors, coupled to Gi protein [26]. Finally, its interaction with the EP4 receptor involves PI3K (phosphatidylinositol 3-kinase)/Akt/ERK (extracellular signal-regulated kinase [31].
TXA2 is an arachidonic acid metabolite that is a potent vasoconstrictor released by platelet aggregation and involved in atherosclerosis [32,33]. The biological activity of TXA2 is mediated by thromboxane-prostanoid (TP) receptors. TP activation results in the production of inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG) via the activation of phosphatidylinositol (PI)-specific phospholipase C (PI-PLC), through Gq to induce the increase in intracellular Ca2+ concentration [33].

2.2. Molecular and Biochemical Characteristic of LTs and HETEs

There are six known functional LOX genes in humans (ALOX5, ALOX12, ALOX12B, ALOX15, ALOX15B, ALOXE3), but only three of them, 5-LOX, 12-LOX (platelet-type), and 15-LOX (reticulocyte-type), appear to be important for vascular disorders [34].
LTs are a family of eicosanoid inflammatory mediators produced in leukocytes, macrophages and mast cells by the oxidation of AA, and the essential fatty acid EPA by the cytosolic phospholipase A2 and 5-LOX enzymes. 5-LOX, with the aid of the accessory 5-LOX-activating protein (FLAP), catalyzes the conversion of AA to 5-hydroperoxyeicosatetraenoic acid (5-HETE) and then to Leukotriene A4 (LTA4) [35], an unstable intermediate, which can be either metabolized by LTA4 hydrolase to LTB4 (see structure in Figure 1), a potent chemoattractant, or conjugated to glutathione by LTC4 synthase (LTC4S), producing cysteinyl LTs [36]. The LTs exert their actions through interaction with specific 7-transmembrane G-protein-coupled cell surface receptors, BLT1 and BLT2, and the CysLT1 receptor [37].
12,15HETEs:12/15-LOX metabolizes arachidonic acid to form 15(S)-hydroperoxyeicosatetraenoic acid (15(S)-HPETE) and 12(S)-hydroperoxyeicosatetraenoic acid (12(S)-HPETE). Both 15(S)-HPETE and 12(S)-HPETE are further reduced by cellular glutathione peroxidase to their corresponding hydroxy analogs: 15-hydroxyicosatetraenoic acid (15(S)-HETE) and 12-hydroxyeicosatetraenoic acid (12(S)-HETE), respectively. Both 15(S)-HPETE and 15(S)-HETE bind to and activate LTB4 receptor 2 and peroxisome proliferator-activated receptor γ (PPAR) [38].

2.3. Molecular and Biochemical Characteristic of Lxs

Lipoxin A4 (LXA4) and its isomer Lipoxin B4 (LXB4)(see structure in Figure 1) are the principal LXs formed in mammals. They are enzymatically derived from the endogenous ω-6 fatty acid, AA, by 5-LOX and either 12-LOX or 15-LOX enzymes. Additionally, the aspirin-induced acetylation of COX2 can function through a lipoxygenase instead of an endoperoxidase, resulting in the generation of aspirin-triggered lipoxin A4 (15-epi-LXA4) and aspirin-triggered lipoxin B4 (15-epi-LXB4) [39,40].
LXA4 and LXB4 are produced principally in human mucosal tissues, in blood vessels, and in platelets [22], and are present in all tissues via transcellular interactions between platelets, neutrophils, and resident cells [41]. LXs were first isolated and identified as inhibitors of polymorphonuclear neutrophil infiltration and as stimulators of the non-phlogistic recruitment of macrophages [42,43]. LXA4 regulates cellular functions through the activation of specific receptors (LXA4 receptor/formyl peptide receptor 2 and G-protein-coupled receptor 32) expressed by neutrophils and monocytes [44].

2.4. Molecular and Biochemical Characteristic of EETs

EETs are metabolites of AA by cytochrome CYP epoxygenases [45,46] that show beneficial effects in cardiovascular diseases attributed to anti-inflammation, vasodilation, and natriuresis [47].

2.5. Role of Prostanoids, LTs, HETEs, LXs, EETs in Atherosclerotic Progression

Prostanoids: Studies performed in mice through genetically manipulated enzymes and receptors highlighted the many physiological and pathological functions of prostanoids in atherosclerosis (see Table 1).
For example, PGD2 is involved in sleep, in allergies, and adiposity [60]; PGF 2α in childbirth and fibrosis; and PGE2 is involved in many pathological events, such as inflammation, fever, pain, and cancer. The potent vasoconstrictor TxA2 seems to be involved in coronary ischemia.; in contrast, the potent vasodilator PGI2 is involved in antithrombosis [61] and regulates coronary blood flow in humans [62,63] (see Table 2).
In mice, TxA2 promotes, while PGI2 prevents, atherogenesis through the control of platelet activation and leukocyte–EC interaction [48]. At the same time, PGE2 can dilate or constrict large blood vessels, depending on the vessel type and on the receptor subtypes involved (EP2, EP4 or EP1, EP3) and receptor, respectively [77,78,79,80]. mPGES, which catalyzes the isomerization of PGH2 into PGE2, represents a possible target against inflammation and thrombotic risk, because its inhibition augments PGI2 levels. [77]. Studies show that male mice that are deficient in both LDL receptor and mPGES-1 delay atherosclerosis development [49] and reduce abdominal aneurysm events induced by angiotensin II [50,77]. The deletion of mPGES-1 attenuates injury-induced neointima formation in mice by the augmented levels of PGI2, a known restraint of the vascular response to injury, acting via the IP receptor. On the other hand, the global deletion of mPGES-1 exacerbates MI/R injury, despite the increased biosynthesis of PGI2 [81]. Mechanistically, it has been reported that mPGES-1–derived PGE2 and the endothelial EP4 receptor preserve microcirculatory perfusion in MI/R through their vasodilatory effect on arterioles [51].
COX-1 and -2 are mediators of pain, fever, and inflammation and the primary targets of nonsteroidal anti-inflammatory drugs (NSAIDs). However, studies in mice using COX-2 knockout or COX-2 derived PGI2 receptor deletion showed elevated blood pressure and thrombogenesis [82]. Moreover, COX-2-selective drugs such as coxibs, rofecoxib, celecoxib, and lumiracoxib NSAIDs, inhibit cyclooxygenase-2 (COX-2), thereby suppressing the biosynthesis of PGI2, and increasing the risk of thrombotic events such as in myocardial infarction and stroke [83,84].
LTs: Besides their principal role in asthma and in inflammation, LTs can also increase cardiovascular risk, being important mediators of the inflammatory process in cardiovascular pathologies [64,65]. In particular, cysteinyl-LTs seem to be a potent coronary artery vasoconstrictor that stimulates the proliferation of arterial smooth muscle cells and increases the P-selectin surface expression of endothelial cells [36,64]. Increased levels of LTs are present in patients with myocardial infarction (MI), stroke, atherosclerosis, and aortic aneurysms; and the urinary excretion of LTE4 has been reported to be increased in patients with cardiac ischemia [64,85]. Interestingly, two human genetic studies showed a relationship between polymorphism of the 5-LOX pathway and relative risk for MI, stroke, and atherosclerosis, focusing attention on LTs in the pathogenesis of these diseases [86].
HETEs: Increasing evidence highlights the controversial nature of 12/15-LOX in inflammation, as its metabolites possess both pro- and anti-inflammatory properties [87]. The 12/15-LOX metabolite 12(S)-HETE is a potent, pro-inflammatory chemoattractant for neutrophils and leukocytes [38,88]. The 12/15-LOX-derived metabolites 12(S)-HPETE and 15(S)-HPETE were reported to inhibit prostacyclin synthase activity, thereby decreasing the levels of vasodilator prostacyclin [89]. In addition, it was suggested that 12/15-LOX reduces the bioavailability of nitric oxide, thus promoting vasoconstriction [90,91]. At the same time, several reports demonstrate the anti-inflammatory properties of 12/15-LOX and its metabolites [92]. In fact, in human coronary arteries, 12(S)-HETE induced vascular smooth muscle cell hyperpolarization through the activation of large conductance KCa (BKCa) channels, resulting in relaxation [66].
Studies of 15-lipoxygenating LOX-isoforms (ALOX15, ALOX15B) indicate that their regulation of vascular tone plays an important role in the development of hypertension [66]. It has been reported that ALOX15-deficient mice exhibited higher resistance towards L-NAME- and high-salt-induced hypertension than wild type controls, although systolic blood pressures did not differ [52]. In support of this, ALOX15-deficient mice treated with wild-type peritoneal macrophages, which are a major source of ALOX15 in mice, lost their resistance toward L-NAME-induced hypertension. Analyzing human atherosclerotic plaques revealed a significant increase in 12/15-LOX expression [93]. It was also reported that ALOX5 and ALOX15B were expressed at high levels in advanced atherosclerotic lesions [94]. However, the role of 12/15-LOX in human atherosclerotic lesion development is still not yet clear. Other studies showed an increased 12/15-LOX expression with decreased lesion severity in human clinical samples. Specifically, a polymorphism in 12/15-LOX promoter (a C to T substitution at position −292), which promotes its increased expression and activity, was found to be associated with a reduction in the risk of atherosclerosis [95].
EETs: EETs are expressed in the heart, especially in the endothelium, and possess beneficial effects against inflammation, fibrosis, and apoptosis, which could combat cardiovascular diseases [96]. Some studies showed that EETs mediate the vasodilation of vascular smooth muscle by activating Ca2+-activated K+ channels [97], and that its vasodilator effect is independent of nitric oxide (NO) in response to bradykinin [98]. In animal models of hypertension, EETs synthesis was inhibited [99], while it was upregulated by soluble epoxide hydrolase (sEH) inhibitors or by CYP gene overexpression, which in turn decreased blood pressure [100,101]. Interestingly, in Ang II-induced hypertension, sEH expression was upregulated, while EETs were downregulated [53].
Other studies showed that sEH inhibition reduced cholesterol and low-density lipoprotein (LDL) levels, while it increased high-density lipoprotein (HDL) levels [102], preventing atherosclerosis in several models, including ApoE −/− mice and high-fat diet (HFD)-induced models [103]. Lower EET levels were observed in obstructed CAD patients compared with patients with no apparent CAD [67]. Furthermore, some authors provided evidence that treatments with sEH inhibitors could reduce the infarct size and chronic cardiac remodeling post MI [104] and that the elevation of EETs might represent a promising therapeutic strategy combating chronic cardiac remodeling post MI and heart failure [105,106]. Already in 2006, Xu et al. reported that treatment with sEH inhibitors prevented the development of cardiac hypertrophy after 3 weeks of the transverse aortic constriction in mice, and reversed the development of cardiac hypertrophy caused by chronic pressure overload [107].
Moreover, Althurwi et al. demonstrated that the elevation of EET levels by the overexpression of YP2J2 prevented the initiation of cardiac hypertrophy through the NF-κB-mediated mechanism [108,109]. In particular, in vitro studies showed that the overexpression of CYP2J2 or the increasing levels of 14,15-EET (see structure in Figure 1) ameliorated oxidative-stress-reducing apoptosis and inflammation [110]. However, despite the beneficial effect of EETs as anti-inflammatory mediators, because of their instability, it was impossible to develop corresponding exogenous drugs; several efforts with unremarkable results have been made to increase the level of endogenous EETs, including gene therapy, and EET analogs. Only sEH inhibitor and CYP2J2 gene upregulation seem to provide promising data.
LXs: LXA4 and LXB4 exert potent anti-inflammatory and resolution actions [111,112]. We will discuss these, together with the lipid mediators, in Class 3.

3. Class 2: Lysophospholipids

Lysophospholipids (LPLs) are bioactive signaling lipids that are generated from the phospholipase-mediated hydrolyzation of membrane phospholipids (PLs) and sphingolipids (SLs). Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (SP1) are two of the best-characterized LPLs which mediate a variety of cellular physiological responses via specific G-protein-coupled-receptor (GPCR)-mediated signaling pathways. Lysophospholipids include the platelet activating factor (PAF), and the LPA and the SP1 (Figure 2) [113,114,115,116].

3.1. Molecular and Biochemical Characteristics of PAF, LPA and SP1

PAF: PAF is 1-o-alkyl-2-acetyl-sn-glycero-3-phosphocholine, synthesized from 1-O-alkyl-lysophosphatidylcholine (LPC) by the phospholipase A2 (PLA 2) enzyme [68]. Acetylation by LPC acyltransferase 2 (LPCAT2) terminates PAF biosynthesis [117]. Inactivation of PAF occurs by plasma and cellular acetylhydrolase, a special group of the PLA 2 family [118]. PAF binds to a unique G-protein-coupled seven-transmembrane receptor [119,120] that is highly expressed in cells within the cardiovascular system and central nervous system [121]. It causes many inflammatory reactions and allergic responses, including increased platelet activation, leukocyte adhesion, chemotaxis, and vascular permeability [122,123].
LPA: LPA belongs to the glyceryl-based lysophospholipid family [124] and is produced externally to cells, specifically by LPC or autotaxin (ATX; lysophospholipase D) [125], and is degraded by lipid phosphatases [126].
SP1: SP1 is structurally similar to LPA, with the one difference of having a sphingosine backbone rather than glycerol. Sphingolipids are essential structural constituents of the plasma membrane of eukaryotic organisms and some bacteria, where they are highly enriched along with cholesterol in microdomains or lipid rafts. In response to a wide range of stressful stimuli, membrane sphingomyelin are rapidly metabolized to the bioactive sphingolipid intermediate, ceramide, and subsequently to sphingosine phosphorylation by two sphingosine kinases, resulting in the formation of SP1 [127].

3.2. Role of PAF, LPA, and SP1 in Atherosclerotic Progression

PAF: Experiments on animal models showed that PAF plays an important role in the pathogenesis of cardiovascular disorders, and some studies performed in humans reported elevated PAF levels in patients, with increased risk of ischemic events in those with coronary artery disease [68,69,70] (see Table 2). In particular, PAFs seems to stimulate reactive oxygen and nitrogen species, thus triggering the oxidative and nitrosative stress pathways, inducing vasoconstriction [128]. Various substances such as thrombin, angiotensin II, vasopressin, histamine, leukotrienes, and hydrogen peroxide cause the production of PAF in endothelial cells [129]. Other studies suggest that PAF plays a role in the evolution of myocardial injury observed during reperfusion [54] (see Table 1). In conclusion, the effects of PAF are closely associated with atherosclerosis, through various mechanisms such as inflammation, endothelial dysfunction, oxidative and nitrosative stress, and platelet reactivity.
LPA: LPA exerts several effects on vasculature [130,131] resulting in vasoconstriction and increased blood pressure [132]. This happens through the activation of at least six known G-protein-coupled LPA receptors (LPARs) [133] and also peroxisome proliferator-activated receptor γ (PPARγ) [134], and P2Y10 [135]. Compared to normal arteries, individual LPARs are differentially expressed in human atherosclerotic lesions suggesting a distinct role in the initiation and progression of atherosclerosis [136]. Pathological conditions that involve platelet activation, such as inflammation and atherosclerosis, seem to increase LPA production [137]. LPA is elevated in the serum of patients with acute heart attacks [71] and with acute coronary syndrome [72] and is found accumulated in atherosclerotic plaques [73].
In the vasculature, platelets are one of the major sources of LPA production [138], and LPA in turn can contribute to thrombosis, a critical pathogenic condition of vascular diseases. LPA seems also to increase the permeability of the endothelium. In fact in HUVECs LPA via LPAR6-Gα13-RhoA-ROCK pathway induces actin stress fiber formation and increases cell permeability [139,140]. Finally, LPA promotes the progression of atherosclerosis via its various effects on inflammatory cells; for example, promoting inflammatory cell migration [141], cytokine secretion [142], and macrophage transformation [143]. LPA also regulates macrophage-related processes, ox-LDL uptake [143], and matrix metalloproteinase (MMP) secretion [144]. MMPs, particularly MMP9, are principle mediators of extracellular matrix production and degradation, which contribute to atherosclerotic plaque instability [145]. Key proteins in LPA homeostasis are increasingly dysregulated in the plaque during atherogenesis, favoring intracellular LPA production. This might at least partly explain the observed progressive accumulation of this thrombogenic proinflammatory lipid in human and mouse plaques. Thus, intervention in enzymatic LPA production may be an attractive measure to lower intraplaque LPA content, thereby reducing plaque progression and thrombogenicity [55].
SP1: S1P has emerged as a potent signaling molecule that regulates diverse cellular processes including cell proliferation, survival, differentiation, and migration [146], maintaining vascular integrity. S1P stimulates COX 2, suggesting the existence of a functional crosstalk between the sphingolipid and eicosanoid pathways [147]. Moreover, many of the cardioprotective functions of HDL, such as vasodilation, angiogenesis, and the endothelial barrier function, may be attributable to its S1P cargo [148]. In a mouse model of acute myocardial infarction, treatment with an engineered S1P chaperone (ApoM-Fc) attenuated myocardial damage after ischemia/reperfusion injury [56].
Additionally, ceramides, that belong to sphingolipids, are grouped in a class of bioactive lipids which are present during vascular inflammation [149], but they have opposing effects on vascular endothelium compared to S1P [150,151]. Ceramide properties mainly involve the regulation of apoptosis, and their signaling is potentially linked to atherosclerosis progression and pathology. Growing evidence indicates that circulating ceramides may predict acute cardiovascular risk, and may potentially be more predictive than LDL cholesterol [152].

4. Class 3: ω-3 PUFA Derivatives

Anti-inflammatory lipid mediators derive from dietary omega-3 PUFA, specifically the ω-3 fatty acids found in fish oil. They include resolvins (Rvs), protectins(Ps), and maresins (MaRs) (Figure 3) [153,154].

4.1. Molecular and Biochemical Characteristics of Rvs, PDs and MaRs

Rvs: Rvs are divided into two groups: E-series derived from eicosapentaenoic acid and RvD-series from docosahexaenoic acid [155]. The biosynthesis of RvEs is initiated by vascular endothelial cells that convert EPA into 18R-hydroperoxyeicoapentaenoic acid and 18S-hydroperoxyeicoapentaenoic acid (18-HEPE), and proceeds via a transcellular biosynthesis via acetylated COX-2 and cytochrome P450 [156]. These intermediates are rapidly taken up by human neutrophils and metabolized to RvE1 and RvE2 by 5-lipoxygenase. 8-HEPE can directly be transformed into RvE3 through the action of 12/15-LOX by leukocytes [157].
In addition, another EPA–derived E-series resolvin, RvE4, is produced by human neutrophils and macrophages by 5-LOX, and successively by a second enzymatic lipoxygenation, to yield a hydroperoxyl group at the carbon C-5 position [158]. RvE4 activates G-protein-coupled receptors such as Chemokine-like receptor 1 (ChemR23) and antagonizes the BLT1 receptor, which usually mediates the proinflammatory actions of LTB4 [159,160,161]. Upon selective binding to the receptors, RvE1 attenuates nuclear factor-kappaB signaling and the production of pro-inflammatory cytokines, including tumor necrosis factor alpha. RvDs target G-protein-coupled receptor 32, LxA4 receptor/formyl peptide receptor 2 receptors, Formyl-Peptide Receptor 2/3/LXA4 Receptor, and CB2 receptors that are expressed on platelets and polymorphonuclear neutrophils. The activation of CB2 leads to the inhibition of P-selectin expression, thus decreasing polymorphonuclear neutrophil chemotaxis [44].
PD: Protectins are biosynthesized via a lipoxygenase-mediated pathway from Docosahexaenoic acid and it is converted into an intermediate that contains 17S-hydroxyperoxide, and successively, by leukocytes, into 10,17 Dihydroxy-docosahexaenoic acid, known as PD1 [162,163] or neuroprotection [164], for its protective role in the nervous and immune systems [165,166]. PD1 is also produced by human peripheral blood lymphocytes with a T-helper 2 phenotype; it reduces tumor necrosis factor alpha and interferon gamma secretion, blocks T-cell migration, and promotes T-cell apoptosis [167].
A novel protectin-synthesis pathway, called aspirin-triggered PD1, utilizes aspirin-triggered cyclooxygenase-2 to synthesize epimeric 17 R-hydroxyperoxide from docosahexaenoic acid [168,169]. It has shown a positive interaction with CB2 and peroxisome proliferator-activated receptor family receptors [164,170].
MaRs: Maresins form the third-largest family of pro-resolving lipid mediators (PRLMs) derived from DHA20. The biosynthesis of maresins occurs primarily in M2 macrophages and is initiated by a reaction involving human macrophage 12-lipoxygenase (12-LOX), which is a key enzyme in the synthesis of maresins [169,171].

4.2. Role of PRLMs in Atherosclerotic Progression

PRLMs such as RvE and RvD, PD, MaR, and LX induce a biochemical transition from inflammation to resolution, taking the place of prostaglandins and leukotrienes in exudates at the tissue level [8,169,172,173]. RvE1-E4 are more prominently involved in reducing inflammation by increasing the efferocytosis of apoptotic cells by macrophages, blocking leukocyte recruitment into inflamed tissues (Figure 4).
Increased plasma levels of RvE1 have been observed in individuals taking aspirin or eicosapentaenoic acid, resulting in the amelioration of the clinical signs of inflammation [74,75]. D-series have been shown to reduce inflammation by decreasing platelet–leukocyte adhesion [174].
Protectins reduce polymorphonuclear neutrophil transmigration through endothelial cells and enhance the clearance (efferocytosis) of apoptotic polymorphonuclear neutrophils by human macrophages [170].
Maresins were identified as molecules produced by macrophages with homeostatic functions in resolving inflammation. In fact, the macrophage phagocytosis of apoptotic cells triggers the biosynthesis of maresin-1 [175]. Maresin-1 stimulates efferocytosis with human cells and also has regenerative functions [176]. Finally, LXA4 and B4 exert potent anti-inflammatory and resolution actions [110].
Temporal changes in the expression, activity, and localization of enzymes such as 4- and 15-lipoxygenases (LOXs) appear to activate the “switch” responsible for the resolution actions [173,177,178]. Consequently, PRLMs coordinate crosstalk between leukocytes and local cell populations, promoting an M1–M2 phenotypic transition in macrophages, which is central to tissue repair. PRLMs further promote resolution by positive-feedback effects on LOX activity and their receptor expression in leukocytes [177,178,179,180]. Data from murine and rabbit models showed that pro-resolving lipid mediators decrease atheroprogression and promote plaque stability [18,172,180,181].
Alternatively a relative “resolution deficit” may contribute to disease progression or limit clinical responses to existing therapies and would therefore suggest opportunities for monitoring and/or treatments that incorporate lipid mediators [74,179,181,182,183]. In fact, circulating levels of PRLMs, in particular 15-epi-lipoxin A4 (15-epi-LXA 4), were significantly lower in individuals with peripheral arterial disease and correlated inversely with disease severity [74,76](see Table 2).
In situations such as restenosis, and other pathological reactions secondary to interventions, characterized by high levels of IL-6 and other inflammatory cytokines, augmented levels of PRLMs could limit harmful arterial reactions and restenosis. An imbalance between proinflammatory and resolving factors may decide the evolution of atherosclerotic disease [3].
There is also favorable data regarding the antithrombotic activity of Maresin 1. In fact, it promotes the hemostatic function of human platelets, but suppresses the inflammatory activity, suggesting its important role in the resolution of thrombotic events [184]. At sites of thrombosis or injury, platelet–neutrophil interaction gives rise to the biosynthesis of PRLMs such as MaR1 which induce resolution [185].
The activity of RvS in thrombosis is directed towards reducing platelet activation [180,186]. RvDs and Lxs also activate macrophages for the phagocytosis of the clot and apoptotic cells [187]. In addition, myocardial ischemia/reperfusion studies in the mouse animal model have established a protective effect of PRLMs with respect to tissue damage and the reduction of ROS [57,58,59] (see Table 1).
Although until recently lipid mediators have been overlooked as resolvers of inflammation, they are now being reevaluated because of their activities and biosynthesis in the vascular system. In fact, in vitro, RvD1, RvD2, MaR1, LXA4, and protectin D1 reduce the production of inflammatory cytokines, adhesion molecules, and the adhesive effect of leukocytes to endothelial cells during an inflammatory event [6,188,189,190]. These resolving effects result from the ability of these molecules to reduce NF-kB, NO, prostacyclin synthesis, and ROS production [191,192].
Reduced monocyte adhesion to VSMCs and the reduced expression of adhesion molecules have been also observed in vitro studies with inflammatory cytokines after treatment with RvD1, RvD2, and MaR1. Another resolving effect by LXA4, RvD1, RvD2, and RvE1 is to attenuate VSMC migration [193,194].

4.3. Protective Function of ω-3 Fatty Acid Diet in Atherosclerotic Progression

Individuals with a high consumption of fish oils show a greater proportion of EPA and DHA-containing phospholipids in particular cell types, compared to individuals consuming plant oil supplements [195]. EPA provides precursors for the production of the anti-inflammatory mediator family of series 3 prostanoids. The synthesis of prostanoids is catalyzed by cyclooxygenase enzymes. The cleavage of EPA by phospholipase A 2 allows EPA to be converted to anti-inflammatory mediators [196].
Human studies have shown the limited endogenous synthesis of docosahexaenoic acid (DHA) from ω-3 fatty acid precursor α-linolenic acid (ALA), therefore supporting the necessity of a diet containing ω -3 PUFAs for optimal health and development [197,198]. Circulating levels of the RvD1 or its precursor DHA are reduced in individuals with symptomatic carotid atherosclerosis [199].
Adults receiving EPA+DHA therapy demonstrated significantly greater reductions in circulating levels of proinflammatory cytokines, compared with those receiving placebo therapy [200]. EPA+DHA therapy may be an effective low-risk dietary intervention for assuaging the harmful effects of inflammation.
A large prospective randomized clinical trial of 11,324 patients with a recent myocardial infarction showed a relatively reduced risk of death in response to long chain polyunsaturated fatty acid dietary supplementation [201] (see Table 3).
In another clinical trial, 0.8–1.2 g omega-3 FA supplementation has shown a positive effect in reducing the incidence of major adverse cardiovascular events (MACE), cardiovascular death, and myocardial infarction [202]. A lower rate of myocardial infarction was also observed in people consuming an Inuit diet, characterized by the high consumption of marine mammals and fish [207]. Meta-analyses in adults have shown that the Mediterranean diet, enriched in ω-3 PUFA and oleic acid, exerts a protective effect against cardiovascular diseases [208,209,210,211]. This beneficial effect could be due to their anti-arrhythmic, anti-inflammatory, anti-thrombotic, and hypolipidemic effects, improving vascular function [212,213].
PUFA administration seems to be also protective against aneurysm, a condition characterized by chronic inflammation of the arterial wall that gradually loses its structural integrity by an incessant protease activity. In fact, in animal studies, PUFAs reduced aortic wall inflammation and impairment of the matrix [184]. A recently published trial by Bhatt et al., involving 8179 participants and a 5 year follow-up, reported that patients who received 4 g/d ω-3 PUFA showed a 30% reduction in cardiovascular events, compared with the control group [203]. The JELIS trial, conducted in Japan, recruited 18,645 patients with a total cholesterol of 6.5 mmol/L or greater, and demonstrated a 19% relative reduction in major coronary events after a 1800 mg dose of EPA daily [204].
However, some conflicting data exists [214,215,216]. Other clinical trials on PUFA administration have reported contradictory findings with regard to cardiovascular death and/or major cardiovascular events [205,214,217,218]. Recent studies by Xie et al. suggest that the population most at risk of suffering major cardiovascular events and deaths could benefit more by taking a higher dose of ω-3PUFA for a more prolonged intervention period (daily dose × intervention period > 8 g/day). Earlier treatment schedules of only 1 g/d for 1-year ω-3 PUFA supplementation showed no benefits regarding sudden cardiac death, and in fact revealed a small increase in major cardiovascular and cerebrovascular events [206]. Furthermore, a large-scale randomized controlled trial of the effects of marine ω-3 fatty acid supplementation on cardiovascular outcomes has reported increased risks of atrial fibrillation (AF), in particular in trials testing >1 g/d. [205]. Recent meta-analyses reflected these discrepancies [219,220].
Therefore, the potential reasons for disparate findings may be dose-related. In addition, isomeric forms of the mediators may show different capabilities or conflicting biological effects. More advanced age or smoking habits may also affect the biosynthesis of lipid mediators [221].
In conclusion, further studies on pro-resolving lipid-mediator biosynthesis and pathways and on diets, with related doses and periods, are needed to understand the best formulations for an effective resolution in relation to cardiovascular disease. Understanding these variables will be necessary for the development of effective drug therapies.

Funding

This research received no external funding.

Acknowledgments

The authors are grateful to Sherry Lubber for her English editing support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lusis, A.J. Atherosclerosis. Nature 2000, 407, 233–241. [Google Scholar] [CrossRef] [PubMed]
  2. Soehnlein, O.; Libby, P. Targeting inflammation in atherosclerosis—From experimental insights to the clinic. Nat. Rev. Drug Discov. 2021, 20, 589–610. [Google Scholar] [CrossRef] [PubMed]
  3. Kasikara, C.; Doran, A.C.; Cai, B.; Tabas, I. The role of non-resolving inflammation in atherosclerosis. J. Clin. Investig. 2018, 128, 2713–2723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Orekhov, A.N. The Role of Modified and Dysfunctional Lipoproteins in Atherogenesis. Curr. Med. Chem. 2019, 26, 1509–1511. [Google Scholar] [CrossRef] [PubMed]
  5. Moore, K.J.; Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 2011, 145, 341–355. [Google Scholar] [CrossRef] [Green Version]
  6. Merched, A.J.; Ko, K.; Gotlinger, K.H.; Serhan, C.N.; Chan, L. Atherosclerosis: Evidence for impairment of resolution of vascular inflammation governed by specific lipid mediators. FASEB J. 2008, 22, 3595–3606. [Google Scholar] [CrossRef] [Green Version]
  7. Pullen, A.B.; Jadapalli, J.K.; Rhourri-Frih, B.; Halade, G.V. Re-evaluating the causes and consequences of non-resolving inflammation in chronic cardiovascular disease. Heart Fail. Rev. 2020, 25, 381–391. [Google Scholar] [CrossRef]
  8. Hansson, G.K.; Libby, P.; Tabas, I. Inflammation and plaque vulnerability. J. Intern. Med. 2015, 278, 483–493. [Google Scholar] [CrossRef] [Green Version]
  9. Bentzon, J.F.; Otsuka, F.; Virmani, R.; Falk, E. Mechanisms of plaque formation and rupture. Circ. Res. 2014, 114, 1852–1866. [Google Scholar] [CrossRef]
  10. Ma, J.; Chen, X. Anti-inflammatory Therapy for Coronary Atherosclerotic Heart Disease: Unanswered questions behind existing successes. Front. Cardiovasc. Med. 2021, 7, 631398. [Google Scholar] [CrossRef]
  11. Bannenberg, G.L.; Chiang, N.; Ariel, A.; Arita, M.; Tjonahen, E.; Gotlinger, K.H.; Hong, S.; Serhan, C.N. Molecular circuits of resolution: Formation and actions of resolvins and protectins. J. Immunol. 2005, 174, 4345–4355. [Google Scholar] [CrossRef] [Green Version]
  12. Levy, B.D.; Clish, C.; Schmidt, B.A.; Gronert, K.; Serhan, C.N. Lipid mediator class switching during acute inflammation: Signals in resolution. Nat. Immunol. 2001, 2, 612–619. [Google Scholar] [CrossRef]
  13. Serhan, C.N.; Chiang, N. Lipid-Derived Mediators in Endogenous Anti-Inflammation and Resolution: Lipoxins and Aspirin-Triggered 15-epi-Lipoxins. Sci. World J. 2002, 2, 169–204. [Google Scholar] [CrossRef] [Green Version]
  14. Serhan, C.N. Resolution Phase of Inflammation: Novel Endogenous Anti-Inflammatory and Proresolving Lipid Mediators and Pathways. Annu. Rev. Immunol. 2007, 25, 101–137. [Google Scholar] [CrossRef] [Green Version]
  15. Serhan, C.N. Novel Lipid Mediators and Resolution Mechanisms in Acute Inflammation: To Resolve or Not? Am. J. Pathol. 2010, 177, 1576–1591. [Google Scholar] [CrossRef]
  16. Bennett, M.; Gilroy, D.W. Lipid Mediators in Inflammation. Microbiol. Spectr. 2016, 4, 6. [Google Scholar] [CrossRef]
  17. Fredman, G.; Hellmann, J.; Proto, J.D.; Kuriakose, G.; Colas, R.A.; Dorweiler, B.; Connolly, E.S.; Solomon, R.; Jones, D.M.; Heyer, E.J.; et al. An imbalance between specialized pro-resolving lipid mediators and pro-inflammatory leukotrienes promotes instability of atherosclerotic plaques. Nat. Commun. 2016, 7, 12859. [Google Scholar] [CrossRef] [Green Version]
  18. Viola, J.R.; Lemnitzer, P.; Jansen, Y.; Csaba, G.; Winter, C.; Neideck, C.; Silvestre-Roig, C.; Dittmar, G.; Döring, Y.; Drechsler, M.; et al. Resolving Lipid Mediators Maresin 1 and Resolvin D2 Prevent Atheroprogression in Mice. Circ. Res. 2016, 119, 1030–1038. [Google Scholar] [CrossRef] [Green Version]
  19. Schebb, N.H.; Kühn, H.; Kahnt, A.S.; Rund, K.M.; O’Donnell, V.B.; Flamand, N.; Peters-Golden, M.; Jakobsson, P.-J.; Weylandt, K.H.; Rohwer, N.; et al. Formation, Signaling and Occurrence of Specialized Pro-Resolving Lipid Mediators—What is the Evidence so far? Front. Pharmacol. 2022, 13, 838782. [Google Scholar] [CrossRef]
  20. Austen, K.F. The cysteinyl leukotrienes: Where do they come from? What are they? Where are they going? Nat. Immunol. 2008, 9, 113–115. [Google Scholar] [CrossRef]
  21. Narumiya, S.; Fitzgerald, G.A. Genetic and pharmacological analysis of prostanoid receptor function. J. Clin. Investig. 2001, 108, 25–30. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, B.; Wu, L.; Chen, J.; Dong, L.; Chen, C.; Wen, Z.; Hu, J.; Fleming, I.; Wang, D.W. Metabolism pathways of arachidonic acids: Mechanisms and potential therapeutic targets. Signal Transduct. Target. Ther. 2021, 6, 94. [Google Scholar] [CrossRef] [PubMed]
  23. Yamaguchi, A.; Botta, E.; Holinstat, M. Eicosanoids in inflammation in the blood and the vessel. Front. Pharmacol. 2022, 13, 997403. [Google Scholar] [CrossRef] [PubMed]
  24. Idborg, H.; Pawelzik, S.-C. Prostanoid Metabolites as Biomarkers in Human Disease. Metabolites 2022, 12, 721. [Google Scholar] [CrossRef] [PubMed]
  25. Smith, W.L.; DeWitt, D.L.; Garavito, R.M. Cyclooxygenases: Structural, Cellular, and Molecular Biology. Annu. Rev. Biochem. 2000, 69, 145–182. [Google Scholar] [CrossRef] [Green Version]
  26. Zhu, L.; Zhang, Y.; Guo, Z.; Wang, M. Cardiovascular Biology of Prostanoids and Drug Discovery. Arter. Thromb. Vasc. Biol. 2020, 40, 1454–1463. [Google Scholar] [CrossRef]
  27. Woodward, D.F.; Jones, R.L.; Narumiya, S. International Union of Basic and Clinical Pharmacology. LXXXIII: Classification of Prostanoid Receptors, Updating 15 Years of Progress. Pharmacol. Rev. 2011, 63, 471–538. [Google Scholar] [CrossRef] [Green Version]
  28. Smith, E.M.; Austin, S.C.; Reilly, M.P.; FitzGerald, G.A. Internalization and sequestration of the human prostacyclin receptor. J. Biol. Chem. 2000, 275, 32037–32045. [Google Scholar] [CrossRef] [Green Version]
  29. Miller, S.B. Prostaglandins in health and disease: An overview. In Seminars in Arthritis and Rheumatism; WB Saunders: Philadelphia, PA, USA, 2006; Volume 36, pp. 37–49. [Google Scholar]
  30. Sugimoto, Y.; Narumiya, S. Prostaglandin E Receptors. J. Biol. Chem. 2007, 282, 11613–11617. [Google Scholar] [CrossRef] [Green Version]
  31. Xu, S.; Zhou, W.; Ge, J.; Zhang, Z. Prostaglandin E2 receptor EP4 is involved in the cell growth and invasion of prostate cancer via the cAMP-PKA/PI3K-Akt signaling pathway. Mol. Med. Rep. 2018, 17, 4702–4712. [Google Scholar] [CrossRef]
  32. Rucker, D.; Dhamoon, A.S. Physiology, Thromboxane A2. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  33. Chen, H. Role of thromboxane A2 signaling in endothelium-dependent contractions of arteries. Prostaglandins Other Lipid Mediat. 2018, 134, 32–37. [Google Scholar] [CrossRef]
  34. Funk, C.D.; Chen, X.S.; Johnson, E.N.; Zhao, L. Lipoxygenase genes and their targeted disruption. Prostaglandins Other Lipid Mediat 2002, 68–69, 303–312. [Google Scholar] [CrossRef]
  35. Funk, C.D. Prostaglandins and Leukotrienes: Advances in Eicosanoid Biology. Science 2001, 294, 1871–1875. [Google Scholar] [CrossRef] [Green Version]
  36. Colazzo, F.; Gelosa, P.; Tremoli, E.; Sironi, L.; Castiglioni, L. Role of the Cysteinyl Leukotrienes in the Pathogenesis and Progression of Cardiovascular Diseases. Mediat. Inflamm. 2017, 2017, 2432958. [Google Scholar] [CrossRef] [Green Version]
  37. Maekawa, A.; Kanaoka, Y.; Xing, W.; Austen, K.F. Functional recognition of a distinct receptor preferential for leukotriene E 4 in mice lacking the cysteinyl leukotriene 1 and 2 receptors. Proc. Natl. Acad. Sci. USA 2008, 105, 16695–16700. [Google Scholar] [CrossRef] [Green Version]
  38. Singh, N.K.; Rao, G.N. Emerging role of 12/15-Lipoxygenase (ALOX15) in human pathologies. Prog. Lipid Res. 2019, 73, 28–45. [Google Scholar] [CrossRef]
  39. Serhan, C.N.; Chiang, N. Novel endogenous small molecules as the checkpoint controllers in inflammation and resolution: Entrée for resoleomics. Rheum. Dis. Clin. North Am. 2004, 30, 69–95. [Google Scholar] [CrossRef]
  40. Andrews, D.; Godson, C. Lipoxins and synthetic lipoxin mimetics: Therapeutic potential in renal diseases. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158940. [Google Scholar] [CrossRef]
  41. Papayianni, A.; Serhan, C.N.; Phillips, M.L.; Rennke, H.G.; Brady, H.R. Transcellular biosynthesis of lipoxin A4 during adhesion of platelets and neutrophils in experimental immune complex glomerulonephritis. Kidney Int. 1995, 47, 1295–1302. [Google Scholar] [CrossRef] [Green Version]
  42. Bannenberg, G.; Moussignac, R.-L.; Gronert, K.; Devchand, P.R.; Schmidt, B.A.; Guilford, W.J.; Bauman, J.G.; Subramanyam, B.; Perez, H.D.; Parkinson, J.F.; et al. Lipoxins and novel 15-epi-lipoxin analogs display potent anti-inflammatory actions after oral administration. Br. J. Pharmacol. 2004, 143, 43–52. [Google Scholar] [CrossRef]
  43. Maddox, J.F.; Colgan, S.P.; Clish, C.B.; Petasis, N.A.; Fokin, V.V.; Serhan, C.N. Lipoxin B4regulates human monocyte/neutrophil adherence and motility: Design of stable lipoxin B4analogs with increased biologic activity. FASEB J. 1998, 12, 487–494. [Google Scholar] [CrossRef] [PubMed]
  44. Vital, S.A.; Becker, F.; Holloway, P.M.; Russell, J.; Perretti, M.; Granger, D.N.; Gavins, F.N. Formyl-Peptide Receptor 2/3/Lipoxin A 4 Receptor Regulates Neutrophil-Platelet Aggregation and Attenuates Cerebral Inflammation: Impact for Therapy in Cardiovascular Disease. Circulation 2016, 133, 2169–2179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Capdevila, J.H.; Falck, J.R.; Harris, R.C. Cytochrome P450 and arachidonic acid bioactivation: Molecular and functional properties of the arachidonate monooxygenase. J. Lipid Res. 2000, 41, P163–P181. [Google Scholar] [CrossRef]
  46. Shi, Z.; He, Z.; Wang, D.W. CYP450 Epoxygenase Metabolites, Epoxyeicosatrienoic Acids, as Novel Anti-Inflammatory Mediators. Molecules 2022, 27, 3873. [Google Scholar] [CrossRef] [PubMed]
  47. Imig, J. Epoxyeicosanoids in Hypertension. Physiol. Res. 2019, 68, 695–704. [Google Scholar] [CrossRef]
  48. Kobayashi, T.; Tahara, Y.; Matsumoto, M.; Iguchi, M.; Sano, H.; Murayama, T.; Arai, H.; Oida, H.; Yurugi-Kobayashi, T.; Yamashita, J.K.; et al. Roles of thromboxane A(2) and prostacyclin in the development of atherosclerosis in apoE-deficient mice. J Clin Investig. 2004, 114, 784–794. [Google Scholar] [CrossRef] [Green Version]
  49. Wang, M.; Zukas, A.M.; Hui, Y.; Ricciotti, E.; Puré, E.; FitzGerald, G.A. Deletion of microsomal prostaglandin E synthase-1 augments prostacyclin and retards atherogenesis. Proc. Natl. Acad. Sci. 2006, 103, 14507–14512. [Google Scholar] [CrossRef] [Green Version]
  50. Wang, M.; Lee, E.; Song, W.; Ricciotti, E.; Rader, D.J.; Lawson, J.A.; Puré, E.; FitzGerald, G.A. Microsomal Prostaglandin E Synthase-1 Deletion Suppresses Oxidative Stress and Angiotensin II–Induced Abdominal Aortic Aneurysm Formation. Circulation 2008, 117, 1302–1309. [Google Scholar] [CrossRef] [Green Version]
  51. Hao, H.; Hu, S.; Wan, Q.; Xu, C.; Chen, H.; Zhu, L.; Xu, Z.; Meng, J.; Breyer, R.M.; Li, N.; et al. Protective Role of mPGES-1 (Microsomal Prostaglandin E Synthase-1)–Derived PGE2 (Prostaglandin E2) and the Endothelial EP4 (Prostaglandin E Receptor) in Vascular Responses to Injury. Arter. Thromb. Vasc. Biol. 2018, 38, 1115–1124. [Google Scholar] [CrossRef] [Green Version]
  52. Kriska, T.; Cepura, C.; Magier, D.; Siangjong, L.; Gauthier, K.M.; Campbell, W.B. Mice lacking macrophage 12/15-lipoxygenase are resistant to experimental hypertension. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H2428–H2438. [Google Scholar] [CrossRef]
  53. Pang, W.; Li, N.; Ai, D.; Niu, X.-L.; Guan, Y.-F.; Zhu, Y. Activation of peroxisome proliferator-activated receptor-γ downregulates soluble epoxide hydrolase in cardiomyocytes. Clin. Exp. Pharmacol. Physiol. 2011, 38, 358–364. [Google Scholar] [CrossRef] [PubMed]
  54. Montrucchio, G.; Alloatti, G.; Mariano, F.; De Paulis, R.; Comino, A.; Emanuelli, G.; Camussi, G. Role of platelet-activating factor in the reperfusion injury of rabbit ischemic heart. Am. J. Pathol. 1990, 137, 71–83. [Google Scholar] [PubMed]
  55. Bot, M.; Bot, I.; Lopez-Vales, R.; van de Lest, C.H.; Saulnier-Blache, J.S.; Helms, J.B.; David, S.; van Berkel, T.J.; Biessen, E.A. Atherosclerotic Lesion Progression Changes Lysophosphatidic Acid Homeostasis to Favor its Accumulation. Am. J. Pathol. 2010, 176, 3073–3084. [Google Scholar] [CrossRef] [Green Version]
  56. Swendeman, S.L.; Xiong, Y.; Cantalupo, A.; Yuan, H.; Burg, N.; Hisano, Y.; Cartier, A.; Liu, C.H.; Engelbrecht, E.; Blaho, V.; et al. An engineered S1P chaperone attenuates hypertension and ischemic injury. Sci. Signal. 2017, 10, eaal2722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Gilbert, K.; Bernier, J.; Godbout, R.; Rousseau, G. Resolvin D1, a Metabolite of Omega-3 Polyunsaturated Fatty Acid, Decreases Post-Myocardial Infarct Depression. Mar. Drugs 2014, 12, 5396–5407. [Google Scholar] [CrossRef] [Green Version]
  58. Keyes, K.T.; Ye, Y.; Lin, Y.; Zhang, C.; Perez-Polo, J.R.; Gjorstrup, P.; Birnbaum, Y. Resolvin E1 protects the rat heart against reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H153–H164. [Google Scholar] [CrossRef] [Green Version]
  59. Kain, V.; Ingle, K.A.; Colas, R.A.; Dalli, J.; Prabhu, S.D.; Serhan, C.N.; Joshi, M.D.; Halade, G.V. Resolvin D1 activates the inflammation resolving response at splenic and ventricular site following myocardial infarction leading to improved ventricular function. J. Mol. Cell. Cardiol. 2015, 84, 24–35. [Google Scholar] [CrossRef] [Green Version]
  60. Joo, M.; Sadikot, R.T. PGD Synthase and PGD2 in Immune Resposne. Mediat. Inflamm. 2012, 2012, 503128. [Google Scholar] [CrossRef] [Green Version]
  61. Li, K.; Zhao, J.; Wang, M.; Niu, L.; Wang, Y.; Li, Y.; Zheng, Y. The Roles of Various Prostaglandins in Fibrosis: A Review. Biomolecules 2021, 11, 789. [Google Scholar] [CrossRef]
  62. Smyth, E.M. Thromboxane and the thromboxane receptor in cardiovascular disease. Clin. Lipidol. 2010, 5, 209–219. [Google Scholar] [CrossRef]
  63. Gutterman, D.D.; Chabowski, D.S.; Kadlec, A.O.; Durand, M.J.; Freed, J.K.; Ait-Aissa, K.; Beyer, A.M. The human microcirculation: Regulation of flow and beyond. Circ. Res. 2016, 118, 157–172. [Google Scholar] [CrossRef] [Green Version]
  64. Hoxha, M.; Rovati, G.; Cavanillas, A.B. The leukotriene receptor antagonist montelukast and its possible role in the cardiovascular field. Eur. J. Clin. Pharmacol. 2017, 73, 799–809. [Google Scholar] [CrossRef]
  65. Lotzer, K.; Funk, C.D.; Habenicht, A.J. The 5-lipoxygenase pathway in arterial wall biology and atherosclerosis. Biochim. Biophys. Acta 2005, 1736, 30–37. [Google Scholar] [CrossRef]
  66. Chawengsub, Y.; Gauthier, K.M.; Campbell, W.B. Role of arachidonic acid lipoxygenase metabolites in the regulation of vascular tone. Am. J. Physiol. Heart Circ Physiol. 2009, 297, H495–H507. [Google Scholar] [CrossRef] [Green Version]
  67. Oni-Orisan, A.; Edin, M.L.; Lee, J.A.; Wells, M.A.; Christensen, E.S.; Vendrov, K.; Lih, F.B.; Tomer, K.B.; Bai, X.; Taylor, J.; et al. Cytochrome P450-derived epoxyeicosatrienoic acids and coronary artery disease in humans: A targeted metabolomics study. J. Lipid Res. 2016, 57, 109–119. [Google Scholar] [CrossRef] [Green Version]
  68. Ramakrishnan, A.V.K.P.; Varghese, T.P.; Vanapalli, S.; Nair, N.K.; Mingate, M.D. Platelet activating factor: A potential biomarker in acute coronary syndrome? Cardiovasc. Ther. 2017, 35, 64–70. [Google Scholar] [CrossRef]
  69. Gabbasov, Z.; Parfyonova, Y.; Popov, E.; Gavrilov, I.; Anuchin, V.; Dubov, P.; Djakonova, Y. Association of platelet function in hypertensive patients with left ventricular hypertrophy, transient myocardial ischemia, and coronary artery disease. Platelets 1998, 9, 191–195. [Google Scholar] [CrossRef]
  70. Chou, Y.I.; Kong, J.; Song, R.; Yan, L.; Zheng, L.; Zhang, Y. Correlation of platelet derived microparticles with thromboxane B2, platelet activating factor, endothelin 1 and neutrophil to lymphocyte ratio in patients with coronary intermediate lesion. Biomarkers 2014, 8, 684–692. [Google Scholar] [CrossRef]
  71. Chen, X.; Yang, X.Y.; Wang, N.D.; Ding, C.; Yang, Y.J.; You, Z.J.; Su, Q.; Chen, J.H. Serum lysophosphatidic acid concentrations measured by dot immunogold filtration assay in patients with acute myocardial infarction. Scand. J. Clin. Lab. Investig. 2003, 63, 497–504. [Google Scholar] [CrossRef]
  72. Dohi, T.; Miyauchi, K.; Ohkawa, R.; Nakamura, K.; Kishimoto, T.; Miyazaki, T.; Nishino, A.; Nakajima, N.; Yaginuma, K.; Tamura, H.; et al. Increased circulating plasma lysophosphatidic acid in patients with acute coronary syndrome. Clin. Chim. Acta 2012, 413, 207–212. [Google Scholar] [CrossRef]
  73. Rother, E.; Brandl, R.; Baker, D.L.; Goyal, P.; Gebhard, H.; Tigyi, G.; Siess, W. Subtype-Selective Antagonists of Lysophosphatidic Acid Receptors Inhibit Platelet Activation Triggered by the Lipid Core of Atherosclerotic Plaques. Circulation 2003, 108, 741–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ho, K.J.; Spite, M.; Owens, C.D.; Lancero, H.; Kroemer, A.H.; Pande, R.; Creager, M.A.; Serhan, C.N.; Conte, M.S. Aspirin-Triggered Lipoxin and Resolvin E1 Modulate Vascular Smooth Muscle Phenotype and Correlate with Peripheral Atherosclerosis. Am. J. Pathol. 2010, 177, 2116–2123. [Google Scholar] [CrossRef] [PubMed]
  75. Makriyannis, A.; Nikas, S.P. Aspirin-Triggered Metabolites of EFAs. Chem. Biol. 2011, 18, 1208–1209. [Google Scholar] [CrossRef] [Green Version]
  76. Conte, M.S.; Desai, T.A.; Wu, B.; Schaller, M.; Werlin, E. Pro-resolving lipid mediators in vascular disease. J. Clin. Investig. 2018, 128, 3727–3735. [Google Scholar] [CrossRef] [Green Version]
  77. Yang, G.; Chen, L. An Update of Microsomal Prostaglandin E Synthase-1 and PGE2Receptors in Cardiovascular Health and Diseases. Oxidative Med. Cell. Longev. 2016, 2016, 5249086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Foudi, N.; Kotelevets, L.; Gomez, I.; Louedec, L.; Longrois, D.; Chastre, E.; Norel, X. Differential reactivity of human mammary artery and saphenous vein to prostaglandin E2: Implication for cardiovascular grafts. Br. J. Pharmacol. 2011, 163, 826–834. [Google Scholar] [CrossRef] [Green Version]
  79. Chen, L.; Miao, Y.; Zhang, Y.; Dou, D.; Liu, L.; Tian, X.Y.; Yang, G.; Pu, D.; Zhang, X.; Kang, J.; et al. Inactivation of the E-Prostanoid 3 Receptor Attenuates the Angiotensin II Pressor Response via Decreasing Arterial Contractility. Arter. Thromb. Vasc. Biol. 2012, 32, 3024–3032. [Google Scholar] [CrossRef] [Green Version]
  80. Eskildsen, M.P.; Hansen, P.B.; Stubbe, J.; Toft, A.; Walter, S.; Marcussen, N.; Rasmussen, L.M.; Vanhoutte, P.M.; Jensen, B.L. Prostaglandin I2 and Prostaglandin E2 Modulate Human Intrarenal Artery Contractility Through Prostaglandin E2-EP4, Prostacyclin-IP, and Thromboxane A2-TP Receptors. Hypertension 2014, 64, 551–556. [Google Scholar] [CrossRef] [Green Version]
  81. Degousee, N.; Fazel, S.; Angoulvant, D.; Stefanski, E.; Pawelzik, S.-C.; Korotkova, M.; Arab, S.; Liu, P.; Lindsay, T.F.; Zhuo, S.; et al. Microsomal Prostaglandin E2 Synthase-1 Deletion Leads to Adverse Left Ventricular Remodeling After Myocardial Infarction. Circulation 2008, 117, 1701–1710. [Google Scholar] [CrossRef] [Green Version]
  82. Cathcart, M.-C.; Tamosiuniene, R.; Chen, G.; Neilan, B.; Bradford, A.; O’Byrne, K.J.; Fitzgerald, D.J.; Pidgeon, G.P. Cyclooxygenase-2-Linked Attenuation of Hypoxia-Induced Pulmonary Hypertension and Intravascular Thrombosis. J. Pharmacol. Exp. Ther. 2008, 326, 51–58. [Google Scholar] [CrossRef]
  83. Mitchell, J.A.; Kirkby, N.S.; Ahmetaj-Shala, B.; Armstrong, P.C.; Crescente, M.; Ferreira, P.; Pires, M.E.L.; Vaja, R.; Warner, T.D. Cyclooxygenases and the cardiovascular system. Pharmacol. Ther. 2021, 217, 107624. [Google Scholar] [CrossRef]
  84. Chen, W.; Yingjie, Z.; Nuan, F.; Zhu, G.; Shuai, W.; Dongming, X. New horizons in the roles and associations of COX-2 and novel natural inhibitors in cardiovascular diseases. Mol. Med. 2021, 27, 123. [Google Scholar] [CrossRef]
  85. De Caterina, R.; Giannessi, D.; Lazzerini, G.; Bernini, W.; Sicari, R.; Cupelli, F.; Lenzi, S.; Rugolotto, M.M.; Madonna, R.; Maclouf, J. Sulfido-peptide leukotrienes in coronary heart disease—Relationship with disease instability and myocardial ischaemia. Eur. J. Clin. Investig. 2010, 40, 258–272. [Google Scholar] [CrossRef]
  86. Helgadottir, A.; Manolescu, A.; Thorleifsson, G.; Gretarsdottir, S.; Jonsdottir, H.; Thorsteinsdottir, U.; Samani, N.J.; Gudmundsson, G.; Grant, S.F.; Thorgeirsson, G.; et al. The gene encoding 5-lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nat. Genet. 2004, 36, 233–239. [Google Scholar] [CrossRef]
  87. Dobrian, A.D.; Lieb, D.C.; Cole, B.K.; Taylor-Fishwick, D.A.; Chakrabarti, S.K.; Nadler, J.L. Functional and pathological roles of the 12- and 15-lipoxygenases. Prog. Lipid Res. 2011, 50, 115–131. [Google Scholar] [CrossRef] [Green Version]
  88. Kayama, Y.; Minamino, T.; Toko, H.; Sakamoto, M.; Shimizu, I.; Takahashi, H.; Okada, S.; Tateno, K.; Moriya, J.; Yokoyama, M. Cardiac 12/15 lipoxygenase-induced inflammation is involved in heart failure. J. Exp. Med. 2009, 206, 1565–1574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Stern, N.; Golub, M.; Nozawa, K.; Berger, M.; Knoll, E.; Yanagawa, N.; Natarajan, R.; Nadler, J.L.; Tuck, M.L. Selective inhibition of angiotensin II-mediated vasoconstriction by lipoxygenase blockade. Am. J. Physiol. Content 1989, 257, H434–H443. [Google Scholar] [CrossRef]
  90. Rubbo, H.; O’Donnell, V. Nitric oxide, peroxynitrite and lipoxygenase in atherogenesis: Mechanistic insights. Toxicology 2008, 208, 305–317. [Google Scholar] [CrossRef]
  91. Holzhütter, H.G.; Wiesner, R.; Rathmann, J.; Stösser, R.; Kühn, H. A kinetic model for the interaction of nitric oxide with a mammalian lipoxygenase. Eur. J. Biochem. 1997, 245, 608–616. [Google Scholar] [CrossRef] [Green Version]
  92. Kühn, H. Biosynthesis, metabolization and biological importance of the primary 15-lipoxygenase metabolites 15-hydro(pero)xy-5Z,8Z,11Z,13E-eicosatetraenoic acid and 13-hydro(pero)xy-9Z,11E-octadecadienoic acid. Prog. Lipid Res. 1996, 35, 203–226. [Google Scholar] [CrossRef]
  93. Kotla, S.; Singh, N.K.; Traylor, J.G.; Orr, A.W.; Rao, G.N. ROS-dependent Syk and Pyk2-mediated STAT1 activation is required for 15(S)-hydroxyeicosatetraenoic acid-induced CD36 expression and foam cell formation. Free Radic. Biol. Med. 2014, 76, 147–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Gertow, K.; Nobili, E.; Folkersen, L.; Newman, J.W.; Pedersen, T.L.; Ekstrand, J.; Swedenborg, J.; Kühn, H.; Wheelock, C.E.; Hansson, G.K.; et al. 12- and 15-lipoxygenases in human carotid atherosclerotic lesions: Associations with cerebrovascular symptoms. Atherosclerosis 2011, 215, 411–416. [Google Scholar] [CrossRef]
  95. Wittwer, J.; Marti-Jaun, J.; Hersberger, M. Functional polymorphism in ALOX15 results in increased allele-specific transcription in macrophages through binding of the transcription factor SPI1. Hum. Mutat. 2006, 27, 78–87. [Google Scholar] [CrossRef] [PubMed]
  96. Lai, J.; Chen, C. The Role of Epoxyeicosatrienoic Acids in Cardiac Remodeling. Front. Physiol. 2021, 12, 642470. [Google Scholar] [CrossRef] [PubMed]
  97. Campbell, W.B.; Fleming, I. Epoxyeicosatrienoic acids and endothelium-dependent responses. Pflug. Arch. 2010, 459, 881–895. [Google Scholar] [CrossRef] [Green Version]
  98. Imig, J.D.; Falck, J.R.; Wei, S.; Capdevila, J.H. Epoxygenase Metabolites Contribute to Nitric Oxide-Independent Afferent Arteriolar Vasodilation in Response to Bradykinin. J. Vasc. Res. 2001, 38, 247–255. [Google Scholar] [CrossRef]
  99. Capdevila, J.; Falck, J.; Imig, J. Roles of the cytochrome P450 arachidonic acid monooxygenases in the control of systemic blood pressure and experimental hypertension. Kidney Int. 2007, 72, 683–689. [Google Scholar] [CrossRef] [Green Version]
  100. Imig, J.D.; Zhao, X.; Capdevila, J.H.; Morisseau, C.; Hammock, B.D. Soluble epoxide hydrolase inhibition lowers arterial blood pressure in angiotensin II hypertension. Hypertension 2002, 39, 690–694. [Google Scholar] [CrossRef] [Green Version]
  101. Lee, J.; Dahl, M.; Grande, P.; Tybjaerg-Hansen, A.; Nordestgaard, B.G. Genetically Reduced Soluble Epoxide Hydrolase Activity and Risk of Stroke and Other Cardiovascular Disease. Stroke 2010, 41, 27–33. [Google Scholar] [CrossRef]
  102. Zhang, L.-N.; Vincelette, J.; Cheng, Y.; Mehra, U.; Chen, D.; Anandan, S.-K.; Gless, R.; Webb, H.K.; Wang, Y.-X. Inhibition of Soluble Epoxide Hydrolase Attenuated Atherosclerosis, Abdominal Aortic Aneurysm Formation, and Dyslipidemia. Arter. Thromb. Vasc. Biol. 2009, 29, 1265–1270. [Google Scholar] [CrossRef]
  103. Liu, W.; Wang, T.; He, X.; Liu, X.; Wang, B.; Liu, Y.; Li, Z.; Tan, R.; Ding, C.; Wang, H.; et al. CYP2J2 Overexpression Increases EETs and Protects Against HFD-Induced Atherosclerosis in ApoE−/− Mice. J. Cardiovasc. Pharmacol. 2016, 67, 491–502. [Google Scholar] [CrossRef]
  104. Li, N.; Liu, J.-Y.; Timofeyev, V.; Qiu, H.; Hwang, S.H.; Tuteja, D.; Lu, L.; Yang, J.; Mochida, H.; Low, R.; et al. Beneficial effects of soluble epoxide hydrolase inhibitors in myocardial infarction model: Insight gained using metabolomic approaches. J. Mol. Cell. Cardiol. 2009, 47, 835–845. [Google Scholar] [CrossRef] [Green Version]
  105. Qiu, H.; Li, N.; Liu, J.-Y.; Harris, T.R.; Hammock, B.D.; Chiamvimonvat, N. Soluble Epoxide Hydrolase Inhibitors and Heart Failure. Cardiovasc. Ther. 2011, 29, 99–111. [Google Scholar] [CrossRef]
  106. Romashko, M.; Schragenheim, J.; Abraham, N.G.; McClung, J.A. Epoxyeicosatrienoic Acid as Therapy for Diabetic and Ischemic Cardiomyopathy. Trends Pharmacol. Sci. 2016, 37, 945–962. [Google Scholar] [CrossRef]
  107. Xu, D.; Li, N.; He, Y.; Timofeyev, V.; Lu, L.; Tsai, H.-J.; Kim, I.-H.; Tuteja, D.; Mateo, R.K.P.; Singapuri, A.; et al. Prevention and reversal of cardiac hypertrophy by soluble epoxide hydrolase inhibitors. Proc. Natl. Acad. Sci. 2006, 103, 18733–18738. [Google Scholar] [CrossRef] [Green Version]
  108. Althurwi, H.N.; Maayah, Z.H.; Elshenawy, O.H.; El-Kadi, A.O. Early changes in cytochrome P450s and their associated arachidonic acid metabolites play a crucial role in the initiation of cardiac hypertrophy induced by isoproterenol. Drug Metab. Dispos. 2015, 43, 1254–1266. [Google Scholar] [CrossRef] [Green Version]
  109. Althurwi, H.N.; Tse, M.M.; Abdelhamid, G.; Zordoky, B.N.; Hammock, B.D.; El-Kadi, A.O. Soluble epoxide hydrolase inhibitor, TUPS, protects against isoprenaline-induced cardiac hypertrophy. Br. J. Pharmacol. 2013, 168, 1794–1807. [Google Scholar] [CrossRef] [Green Version]
  110. Ding, Y.; Tu, P.; Chen, Y.; Huang, Y.; Pan, X.; Chen, W. CYP2J2 and EETs protect against pulmonary arterial hypertension with lung ischemia–reperfusion injury in vivo and in vitro. Respir. Res. 2021, 22, 291. [Google Scholar] [CrossRef]
  111. Serhan, C.N.; Clish, C.; Brannon, J.; Colgan, S.P.; Chiang, N.; Gronert, K. Novel Functional Sets of Lipid-Derived Mediators with Antiinflammatory Actions Generated from Omega-3 Fatty Acids via Cyclooxygenase 2–Nonsteroidal Antiinflammatory Drugs and Transcellular Processing. J. Exp. Med. 2000, 192, 1197–1204. [Google Scholar] [CrossRef] [Green Version]
  112. Serhan, C.N.; Maddox, J.F.; Petasis, N.; Akritopoulou-Zanze, I.; Papayianni, A.; Brady, H.R.; Colgan, S.P.; Madara, J.L. Design of Lipoxin A4 Stable Analogs That Block Transmigration and Adhesion of Human Neutrophils. Biochemistry 1995, 34, 14609–14615. [Google Scholar] [CrossRef]
  113. Tan, S.T.; Ramesh, T.; Toh, X.R.; Nguyen, L.N. Emerging roles of lysophospholipids in health and disease. Prog. Lipid Res. 2020, 80, 101068. [Google Scholar] [CrossRef] [PubMed]
  114. Choi, J.W.; Herr, D.R.; Noguchi, K.; Yung, Y.C.; Lee, C.-W.; Mutoh, T.; Lin, M.E.; Teo, S.T.; Park, K.E.; Mosley, A.N.; et al. LPA Receptors: Subtypes and Biological Actions. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 157–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Ishii, S.; Nagase, T.; Shimizu, T. Platelet-activating factor receptor. Prostaglandins Other Lipid Mediat. 2002, 68–69, 599–609. [Google Scholar] [CrossRef] [PubMed]
  116. Takabe, K.; Paugh, S.W.; Milstien, S.; Spiegel, S. “Inside-Out” Signaling of Sphingosine-1-Phosphate: Therapeutic Targets. Pharmacol. Rev. 2008, 60, 181–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Shindou, H.; Hishikawa, D.; Nakanishi, H.; Harayama, T.; Ishii, S.; Taguchi, R.; Shimizu, T. A single enzyme catalyzes both platelet-activating factor production and membrane biogenesis of inflammatory cells: Cloning and characterization of acetyl-coa:lyso-paf acetyltransferase. J. Biol. Chem. 2007, 282, 6532–6539. [Google Scholar] [CrossRef] [Green Version]
  118. Arai, H. Platelet-activating factor acetylhydrolase. Prostaglandins Other Lipid Mediat. 2002, 69, 83–94. [Google Scholar] [CrossRef] [PubMed]
  119. Honda, Z.-I.; Ishii, S.; Shimizu, T. Platelet-Activating Factor Receptor. J. Biochem. 2002, 131, 773–779. [Google Scholar] [CrossRef]
  120. Shimizu, T.; Mutoh, H.; Kato, S. Platelet-activating factor receptor. Gene structure and tissue-specific regulation. Adv. Exp. Med. Biol. 1996, 416, 479–484. [Google Scholar]
  121. Montrucchio, G.; Lupia, E.; Battaglia, E.; Passerini, G.; Bussolino, F.; Emanuelli, G.; Camussi, G. Tumor necrosis factor alpha-induced angiogenesis depends on in situ platelet-activating factor biosynthesis. J. Exp. Med. 1994, 180, 377–382. [Google Scholar] [CrossRef]
  122. Reznichenko, A.; Korstanje, R. The role of platelet activating factor in Mesangial Pathophysiology. Am. J. Pathol. 2015, 185, 887–896. [Google Scholar] [CrossRef] [Green Version]
  123. Braquet, P.; Hosford, D.; Braquet, M.; Bourgain, R.; Bussolino, F. Role of cytokines and platelet activating factor in microvascular immune injury. Int. Arch. Allergy Immunol. 1989, 88, 88–100. [Google Scholar] [CrossRef]
  124. Yung, Y.C.; Stoddard, N.C.; Mirendil, H.; Chun, J. Lysophosphatidic Acid Signaling in the Nervous System. Neuron 2015, 85, 669–682. [Google Scholar] [CrossRef] [Green Version]
  125. Knowlden, S.; Georas, S.N. The Autotaxin–LPA Axis Emerges as a Novel Regulator of Lymphocyte Homing and Inflammation. J. Immunol. 2014, 192, 851–857. [Google Scholar] [CrossRef] [Green Version]
  126. Umezu-Goto, M.; Kishi, Y.; Taira, A.; Hama, K.; Dohmae, N.; Takio, K.; Yamori, T.; Mills, G.B.; Inoue, K.; Aoki, J.; et al. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J. Cell Biol. 2002, 158, 227–233. [Google Scholar] [CrossRef]
  127. Hannun, Y.A.; Obeid, L.M. Many Ceramides. J. Biol. Chem. 2011, 286, 27855–27862. [Google Scholar] [CrossRef] [Green Version]
  128. Sandoo, A.; van Zanten, J.J.; Metsios GS: Carrol, D.; Kitas, G.D. The endothelium and its role in regulating vascular tone. Open Cardiovasc. Med. J. 2010, 4, 302–312. [Google Scholar] [CrossRef] [Green Version]
  129. McIntyre, T.; Zimmerman, G.A.; Satoh, K. Cultured endothelial cells synthesize both PAF and prostacyclin in response to histamine, bradykinin and adenosine tri phosphate. J. Clin. Investig. 1985, 76271–76280. [Google Scholar]
  130. Zhou, Y.; Little, P.J.; Ta, H.T.; Xu, S.; Kamato, D. Lysophosphatidic acid and its recep tors: Pharmacology and therapeutic potential in atherosclerosis and vascular disease. Pharmacol. Ther. 2019, 204, 107404. [Google Scholar]
  131. Smyth, S.S.; Cheng, H.-Y.; Miriyala, S.; Panchatcharam, M.; Morris, A.J. Roles of lysophosphatidic acid in cardiovascular physiology and disease. Biochim. Biophys. Acta 2008, 1781, 563–570. [Google Scholar] [CrossRef] [Green Version]
  132. Tokumura, A.; Yotsumoto, T.; Masuda, Y.; Tanaka, S. Vasopressor effect of lysophosphatidic acid on spontaneously hypertensive rats and Wistar Kyoto rats. Res. Commun. Mol. Pathol. Pharmacol. 1995, 90, 96–102. [Google Scholar] [PubMed]
  133. Kihara, Y.; Maceyka, M.; Spiegel, S.; Chun, J. Lysophospholipid receptor nomenclature review: IUPHAR Review 8. Br. J. Pharmacol. 2014, 171, 3575–3594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zhang, C.; Baker, D.L.; Yasuda, S.; Makarova, N.; Balazs, L.; Johnson, L.R.; Marathe, G.K.; McIntyre, T.M.; Xu, Y.; Prestwich, G.D.; et al. Lysophosphatidic Acid Induces Neointima Formation Through PPARγ Activation. J. Exp. Med. 2004, 199, 763–774. [Google Scholar] [CrossRef] [Green Version]
  135. Murakami, M.; Shiraishi, A.; Tabata, K.; Fujita, N. Identification of the orphan GPCR, P2Y10 receptor as the sphingosine-1-phosphate and lysophosphatidic acid receptor. Biochem. Biophys. Res. Commun. 2008, 371, 707–712. [Google Scholar] [CrossRef]
  136. Aldi, S.; Matic, L.P.; Hamm, G.; van Keulen, D.; Tempel, D.; Holmstrøm, K.; Szwajda, A.; Nielsen, B.S.; Emilsson, V.; Ait-Belkacem, R.; et al. Integrated Human Evaluation of the Lysophosphatidic Acid Pathway as a Novel Therapeutic Target in Atherosclerosis. Mol. Ther.-Methods Clin. Dev. 2018, 10, 17–28. [Google Scholar] [CrossRef] [Green Version]
  137. Eichholtz, T.; Jalink, K.; Fahrenfort, I.; Moolenaar, W.H. The bioactive phospholipid lysophosphatidic acid is released from activated platelets. Biochem. J. 1993, 291, 677–680. [Google Scholar] [CrossRef] [Green Version]
  138. Aoki, J.; Inoue, A.; Okudaira, S. Two pathways for lysophosphatidic acid production. Biochim. Biophys. Acta 2008, 1781, 513–518. [Google Scholar] [CrossRef]
  139. Yukiura, H.; Kano, K.; Kise, R.; Inoue, A.; Aoki, J. Autotaxin Overexpression Causes Embryonic Lethality and Vascular Defects. PLoS ONE 2015, 10, e0126734. [Google Scholar] [CrossRef] [Green Version]
  140. Yukiura, H.; Kano, K.; Kise, R.; Inoue, A.; Aoki, J. LPP3 localizes LPA6 signalling to non-contact site in endothelial cells. J. Cell Sci. 2015, 128, 3871–3877. [Google Scholar] [CrossRef] [Green Version]
  141. Takeda, A.; Kobayashi, D.; Aoi, K.; Sasaki, N.; Sugiura, Y.; Igarashi, H.; Tohya, K.; Inoue, A.; Hata, E.; Akahoshi, N.; et al. Fibroblastic reticular cell-derived lysophosphatidic acid regulates confined intranodal T-cell motility. Elife 2016, 5, e10561. [Google Scholar] [CrossRef] [PubMed]
  142. Kozian, D.H.; von Haeften, E.; Joho, S.; Czechtizky, W.; Anumala, U.R.; Roux, P.; Dudda, A.; Evers, A.; Nazare, M. Modulation of Hexadecyl-LPA-Mediated Activation of Mast Cells and Microglia by a Chemical Probe for LPA5. Chem. Biochem. 2016, 17, 861–865. [Google Scholar] [CrossRef]
  143. Chen, L.; Zhang, J.; Deng, X.; Liu, Y.; Yang, X.; Wu, Q.; Yu, C. Lysophosphatidic acid directly induces macrophage-derived foam cell formation by blocking the expression of SRBI. Biochem. Biophys. Res. Commun. 2017, 491, 587–594. [Google Scholar] [CrossRef] [PubMed]
  144. Gu, C.; Wang, F.; Zhao, Z.; Wang, H.; Cong, X.; Chen, X. Lysophosphatidic Acid Is Associated with Atherosclerotic Plaque Instability by Regulating NF-κB Dependent Matrix Metalloproteinase-9 Expression via LPA2 in Macrophages. Front. Physiol. 2017, 8, 266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Ketelhuth, D.F.J.; Bäck, M. The Role of Matrix Metalloproteinases in Atherothrombosis. Curr. Atheroscler. Rep. 2011, 13, 162–169. [Google Scholar] [CrossRef] [PubMed]
  146. Cartier, A.; Hla, T. Sphingosine 1-phosphate: Lipid signaling in pathology and therapy. Science 2019, 366, eaar5551. [Google Scholar] [CrossRef] [PubMed]
  147. Lamour, N.F.; Subramanian, P.; Wijesinghe, D.S.; Stahelin, R.V.; Bonventre, J.V.; Chalfant, C.E. Ceramide 1-Phosphate Is Required for the Translocation of Group IVA Cytosolic Phospholipase A2 and Prostaglandin Synthesis. J. Biol. Chem. 2009, 284, 26897–26907. [Google Scholar] [CrossRef] [Green Version]
  148. Egom, E.E.; Mamas, M.A.; Soran, H. HDL quality or cholesterol cargo: What really matters—Spotlight on sphingosine-1-phosphate-rich HDL. Curr. Opin. Infect. Dis. 2013, 24, 351–356. [Google Scholar] [CrossRef]
  149. De Mello, V.D.F.; Lankinen, M.; Schwab, U.; Kolehmainen, M.; Lehto, S.; Seppänen-Laakso, T.; Orešič, M.; Pulkkinen, L.; Uusitupa, M.; Erkkilä, A.T. Link between plasma ceramides, inflammation and insulin resistance: Association with serum IL-6 concentration in patients with coronary heart disease. Diabetologia 2009, 52, 2612–2615. [Google Scholar] [CrossRef] [Green Version]
  150. Maceyka, M.; Spiegel, S. Sphingolipid metabolites in inflammatory disease. Nature 2014, 510, 58–67. [Google Scholar] [CrossRef] [Green Version]
  151. Chatterjee, M.; Rath, D.; Schlotterbeck, J.; Rheinlaender, J.; Walker-Allgaier, B.; Alnaggar, N.; Zdanyte, M.; Müller, I.; Borst, O.; Geisler, T.; et al. Regulation of oxidized platelet lipidome: Implications for coronary artery disease. Eur. Heart J. 2017, 38, 1993–2005. [Google Scholar] [CrossRef] [Green Version]
  152. Cheng, J.M.; Suoniemi, M.; Kardys, I.; Vihervaara, T.; de Boer, S.P.M.; Akkerhuis, K.M.; Sysi-Aho, M.; Ekroos, K.; Garcia-Garcia, H.M.; Oemrawsingh, R.M.; et al. Plasma concentrations of molecular lipid species in relation to coronary plaque characteristics and cardiovascular outcome: Results of the ATHEROREMO-IVUS study. Atherosclerosis 2015, 243, 560–566. [Google Scholar] [CrossRef]
  153. Ariel, A.; Serhan, C.N. Resolvins and protectins in the termination program of acute inflammation. Trends Immunol. 2007, 28, 176–183. [Google Scholar] [CrossRef]
  154. Seki, H.; Tani, Y.; Arita, M. Omega-3 PUFA derived anti-inflammatory lipid mediator resolvin E1. Prostaglandins Other Lipid Mediat. 2009, 89, 126–130. [Google Scholar] [CrossRef]
  155. Vik, A.; Hansen, T.V. Stereoselective syntheses and biological activities of E-series resolvins. Org. Biomol. Chem. 2021, 19, 705–721. [Google Scholar] [CrossRef]
  156. Freire, M.O.; Van Dyke, T.E. Natural resolution of inflammation. Periodontology 2000 2013, 63, 149–164. [Google Scholar] [CrossRef] [Green Version]
  157. Isobe, Y.; Arita, M.; Matsueda, S.; Iwamoto, R.; Fujihara, T.; Nakanishi, H.; Taguchi, R.; Masuda, K.; Sasaki, K.; Urabe, D.; et al. Identification and structure determination of novel anti-inflammatory mediator resolvin E3, 17,18-dihydroxyeicosapentaenoic acid. J. Biol. Chem. 2012, 287, 10525–10534. [Google Scholar] [CrossRef] [Green Version]
  158. Libreros, S.; Shay, A.E.; Nshimiyimana, R.; Fichtner, D.; Martin, M.J.; Wourms, N.; Serhan, C.N. A New E-Series Resolvin: RvE4 Stereochemistry and Function in Efferocytosis of Inflammation-Resolution. Front. Immunol. 2021, 11, 631319. [Google Scholar] [CrossRef]
  159. Chiang, N.; Serhan, C.N. Structural elucidation and physiologic functions of specialized pro-resolving mediators and their receptors. Mol. Asp. Med. 2019, 58, 114–129. [Google Scholar] [CrossRef]
  160. Arita, M.; Bianchini, F.; Aliberti, J.; Sher, A.; Chiang, N.; Hong, S.; Yang, R.; Petasis, N.A.; Serhan, C.N. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J. Exp. Med. 2005, 201, 713–722. [Google Scholar] [CrossRef]
  161. Arita, M.; Ohira, T.; Sun, Y.-P.; Elangovan, S.; Chiang, N.; Serhan, C.N. Resolvin E1 Selectively Interacts with Leukotriene B4 Receptor BLT1 and ChemR23 to Regulate Inflammation. J. Immunol. 2007, 178, 3912–3917. [Google Scholar] [CrossRef] [Green Version]
  162. Schwab, J.M.; Chiang, N.; Arita, M.; Serhan, C.N. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 2007, 447, 869–874. [Google Scholar] [CrossRef] [Green Version]
  163. Serhan, C.N.; Gotlinger, K.; Hong, S.; Lu, Y.; Siegelman, J.; Baer, T.; Yang, R.; Colgan, S.P.; Petasis, N.A. Anti-Inflammatory Actions of Neuroprotectin D1/Protectin D1 and Its Natural Stereoisomers: Assignments of Dihydroxy-Containing Docosatrienes. J. Immunol. 2006, 176, 1848–1859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Bento, A.F.; Claudino, R.F.; Dutra, R.C.; Marcon, R.; Calixto, J.B. Omega-3 Fatty Acid-Derived Mediators 17(R)-Hydroxy Docosahexaenoic Acid, Aspirin-Triggered Resolvin D1 and Resolvin D2 Prevent Experimental Colitis in Mice. J. Immunol. 2011, 187, 1957–1969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Lukiw, W.J.; Cui, J.-G.; Marcheselli, V.L.; Bodker, M.; Botkjaer, A.; Gotlinger, K.; Serhan, C.N.; Bazan, N.G. A role for docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and Alzheimer disease. J. Clin. Investig. 2005, 115, 2774–2783. [Google Scholar] [CrossRef]
  166. Mukherjee, P.K.; Marcheselli, V.L.; Serhan, C.N.; Bazan, N.G. Neuroprotectin D1: A docosahexaenoic acid-derived docosatriene protects human retinal pigment epithelial cells from oxidative stress. Proc. Natl. Acad. Sci. USA 2004, 101, 8491–8496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Anderson, P.; Delgado, M. Endogenous anti-inflammatory neuropeptides and pro-resolving lipid mediators: A new therapeutic approach for immune disorders. J. Cell. Mol. Med. 2008, 12, 1830–1847. [Google Scholar] [CrossRef] [Green Version]
  168. Serhan, C.N.; Chiang, N. Endogenous pro-resolving and anti-inflammatory lipid mediators: A new pharmacologic genus. Br. J. Pharmacol. 2008, 153, S200–S215. [Google Scholar] [CrossRef] [Green Version]
  169. Serhan, C.N.; Dalli, J.; Colas, R.A.; Winkler, J.W.; Chiang, N. Protectins and maresins: New pro-resolving families of mediators in acute inflammation and resolution bioactive metabolome. Biochim. Biophys. Acta 2015, 1851, 397–413. [Google Scholar] [CrossRef] [Green Version]
  170. Shinohara, M.; Mirakaj, V.; Serhan, C.N. Functional Metabolomics Reveals Novel Active Products in the DHA Metabolome. Front. Immunol. 2012, 3, 81. [Google Scholar] [CrossRef] [Green Version]
  171. Li, Q.F.; Hao, H.; Tu, W.S.; Guo, N.; Zhou, X.Y. Maresins: Anti-inflammatory pro-resolving mediators with therapeutic potential. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7442–7453. [Google Scholar] [CrossRef]
  172. Hasturk, H.; Kantarci, A.; Goguet-Surmenian, E.; Blackwood, A.; Andry, C.; Serhan, C.N.; Van Dyke, T.E. Resolvin E1 Regulates Inflammation at the Cellular and Tissue Level and Restores Tissue Homeostasis In Vivo. J. Immunol. 2007, 179, 7021–7029. [Google Scholar] [CrossRef] [Green Version]
  173. Reátegui, E.; Jalali, F.; Khankhel, A.H.; Wong, E.; Cho, H.; Lee, J.; Serhan, C.N.; Dalli, J.; Elliott, H.; Irimia, D. Microscale arrays for the profiling of start and stop signals coordinating human-neutrophil swarming. Nat. Biomed. Eng. 2017, 1, 0094. [Google Scholar] [CrossRef] [Green Version]
  174. Oh, S.F.; Pillai, P.S.; Recchiuti, A.; Yang, R.; Serhan, C.N. Pro-resolving actions and stereoselective biosynthesis of 18S E-series resolvins in human leukocytes and murine inflammation. J. Clin. Investig. 2011, 121, 569–581. [Google Scholar] [CrossRef] [Green Version]
  175. Norling, L.V.; Serhan, C.N. Profiling in resolving inflammatory exudates identifies novel anti-inflammatory and pro-resolving mediators and signals for termination. J. Intern. Med. 2010, 268, 15–24. [Google Scholar] [CrossRef]
  176. Serhan, C.N.; Yang, R.; Martinod, K.; Kasuga, K.; Pillai, P.S.; Porter, T.F.; Oh, S.F.; Spite, M. Maresins: Novel macrophage mediators with potent antiinflammatory and proresolving actions. J. Exp. Med. 2009, 206, 15–23. [Google Scholar] [CrossRef] [Green Version]
  177. Werz, O.; Gerstmeier, J.; Libreros, S.; De la Rosa, X.; Werner, M.; Norris, P.C.; Chiang, N.; Serhan, C.N. Human macrophages differentially produce specific resolvin or leukotriene signals that depend on bacterial pathogenicity. Nat. Commun. 2018, 9, 59. [Google Scholar] [CrossRef] [Green Version]
  178. Motwani, M.P.; Colas, R.A.; George, M.J.; Flint, J.D.; Dalli, J.; Richard-Loendt, A.; De Maeyer, R.P.; Serhan, C.N.; Gilroy, D.W. Pro-resolving mediators promote resolution in a human skin model of UV-killed Escherichia coli–driven acute inflammation. J. Clin. Investig. 2018, 3, e94463. [Google Scholar] [CrossRef] [Green Version]
  179. Dalli, J.; Serhan, C.N. Pro-Resolving Mediators in Regulating and Conferring Macrophage Function. Front. Immunol. 2017, 8, 1400. [Google Scholar] [CrossRef] [Green Version]
  180. Fredman, G.; Van Dyke, T.E.; Serhan, C.N. Resolvin E1 Regulates Adenosine Diphosphate Activation of Human Platelets. Arter. Thromb. Vasc. Biol. 2010, 30, 2005–2013. [Google Scholar] [CrossRef] [Green Version]
  181. Petri, M.H.; Laguna-Fernandez, A.; Arnardottir, H.; Wheelock, C.E.; Perretti, M.; Hansson, G.K.; Bäck, M. Aspirin-triggered lipoxin A4 inhibits atherosclerosis progression in apolipoprotein E−/−mice. Br. J. Pharmacol. 2017, 174, 4043–4054. [Google Scholar] [CrossRef] [Green Version]
  182. Serhan, C.N.; Chiang, N.; Dalli, J. New pro-resolving n-3 mediators bridge resolution of infectious inflammation to tissue regeneration. Mol. Asp. Med. 2018, 64, 1–17. [Google Scholar] [CrossRef]
  183. Kozłowska, H.; Baranowska-Kuczko, M.; Schlicker, E.; Kozłowski, M.; Zakrzeska, A.; Grzęda, E.; Malinowska, B. EP3 receptor-mediated contraction of human pulmonary arteries and inhibition of neurogenic tachycardia in pithed rats. Pharmacol. Rep. 2012, 64, 1526–1536. [Google Scholar] [CrossRef] [PubMed]
  184. Lannan, K.L.; Spinelli, S.L.; Blumberg, N.; Phipps, R.P. Maresin 1 induces a novel pro-resolving phenotype in human platelets. J. Thromb. Haemost. 2017, 15, 802–813. [Google Scholar] [CrossRef] [Green Version]
  185. Abdulnour, R.-E.E.; Dalli, J.; Colby, J.K.; Krishnamoorthy, N.; Timmons, J.Y.; Tan, S.H.; Colas, R.A.; Petasis, N.A.; Serhan, C.N.; Levy, B.D. Maresin 1 biosynthesis during platelet–neutrophil interactions is organ-protective. Proc. Natl. Acad. Sci. USA 2014, 111, 16526–16531. [Google Scholar] [CrossRef] [PubMed]
  186. Dona, M.; Fredman, G.; Schwab, J.M.; Chiang, N.; Arita, M.; Goodarzi, A.; Cheng, G.; von Andrian, U.H.; Serhan, C.N. Resolvin E1, an EPA-derived mediator in whole blood, selectively counterregulates leukocytes and platelets. Blood 2008, 112, 848–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Elajami, T.K.; Colas, R.A.; Dalli, J.; Chiang, N.; Serhan, C.N.; Welty, F.K. Specialized proresolving lipid mediators in patients with coronary artery disease and their potential for clot remodeling. FASEB J. 2016, 30, 2792–2801. [Google Scholar] [CrossRef] [Green Version]
  188. Sansbury, B.E.; Spite, M. Resolution of Acute Inflammation and the Role of Resolvins in Immunity, Thrombosis, and Vascular Biology. Circ. Res. 2016, 119, 113–130. [Google Scholar] [CrossRef]
  189. Filep, J.G.; Zouki, C.; Petasis, N.A.; Hachicha, M.; Serhan, C.N. Anti-inflammatory actions of lipoxin A(4) stable analogs are demonstrable in human whole blood: Modulation of leukocyte adhesion molecules and inhibition of neutrophil-endothelial interactions. Blood 1999, 94, 4132–4142. [Google Scholar] [CrossRef]
  190. Chattopadhyay, R.; Mani, A.M.; Singh, N.K.; Rao, G.N. Resolvin D1 blocks H2O2-mediated inhibitory crosstalk between SHP2 and PP2A and suppresses endothelial-monocyte interactions. Free Radic. Biol. Med. 2018, 117, 119–131. [Google Scholar] [CrossRef]
  191. Chatterjee, A.; Sharma, A.; Chen, M.; Toy, R.; Mottola, G.; Conte, M.S. The Pro-Resolving Lipid Mediator Maresin 1 (MaR1) Attenuates Inflammatory Signaling Pathways in Vascular Smooth Muscle and Endothelial Cells. PLoS ONE 2014, 9, e113480. [Google Scholar] [CrossRef] [Green Version]
  192. Chattopadhyay, R.; Raghavan, S.; Rao, G.N. Resolvin D1 via prevention of ROS-mediated SHP2 inactivation protects endothelial adherens junction integrity and barrier function. Redox Biol. 2017, 12, 438–455. [Google Scholar] [CrossRef]
  193. Miyahara, T.; Runge, S.; Chatterjee, A.; Chen, M.; Mottola, G.; Fitzgerald, J.M.; Serhan, C.N.; Conte, M.S. D-series resolvin attenuates vascular smooth muscle cell activation and neointimal hyperplasia following vascular injury. FASEB J. 2013, 27, 2220–2232. [Google Scholar] [CrossRef] [Green Version]
  194. Wu, B.; Mottola, G.; Chatterjee, A.; Lance, K.D.; Chen, M.; Siguenza, I.O.; Desai, T.A.; Conte, M.S. Perivascular delivery of resolvin D1 inhibits neointimal hyperplasia in a rat model of arterial injury. J. Vasc. Surg. 2017, 65, 207–217.e3. [Google Scholar] [CrossRef] [Green Version]
  195. Egert, S.; Somoza, V.; Kannenberg, F.; Fobker, M.; Krome, K.; Erbersdobler, H.F.; Wahrburg, U. Influence of three rapeseed oil-rich diets, fortified with α-linolenic acid, eicosapentaenoic acid or docosahexaenoic acid on the composition and oxidizability of low-density lipoproteins: Results of a controlled study in healthy volunteers. Eur. J. Clin. Nutr. 2007, 61, 314–325. [Google Scholar] [CrossRef]
  196. Calder, P.C. Polyunsaturated fatty acids, inflammation, and immunity. Lipids 2000, 36, 1007–1024. [Google Scholar] [CrossRef]
  197. Zárate, R.; El Jaber-Vazdekis, N.; Tejera, N.; Pérez, J.A.P.; Rodríguez, C. Significance of long chain polyunsaturated fatty acids in human health. Clin. Transl. Med. 2017, 6, 25. [Google Scholar] [CrossRef] [Green Version]
  198. Brenna, J.T.; Salem, N.; Sinclair, A.J.; Cunnane, S.C. International Society for the Study of Fatty Acids and Lipids, ISSFAL. α-Linolenic acid supplementation and conversion to n-3 long-chain polyunsaturated fatty acids in humans. Prostaglandins Leukot. Essent. Fat. Acids 2009, 80, 85–91. [Google Scholar] [CrossRef]
  199. Bazan, H.A.; Lu, Y.; Jun, B.; Fang, Z.; Woods, T.C.; Hong, S. Circulating inflammation-resolving lipid mediators RvD1 and DHA are decreased in patients with acutely symptomatic carotid disease. Prostaglandins, Leukot. Essent. Fat. Acids 2017, 125, 43–47. [Google Scholar] [CrossRef]
  200. Tan, A.; Sullenbarger, B.; Prakash, R.; McDaniel, J.C. Supplementation with eicosapentaenoic acid and docosahexaenoic acid reduces high levels of circulating proinflammatory cytokines in aging adults: A randomized, controlled study. Prostaglandins Leukot. Essent. Fat. Acids 2018, 132, 23–29. [Google Scholar] [CrossRef]
  201. Jialal, I.; Devaraj, S.; Huet, B.A.; Traber, M. GISSI-Prevenzione trial. GISSI-Prevenzione trial. Lancet 1999, 354, 1554. [Google Scholar] [CrossRef]
  202. Shen, S.; Gong, C.; Jin, K.; Zhou, L.; Xiao, Y.; Ma, L. Omega-3 Fatty Acid Supplementation and Coronary Heart Disease Risks: A Meta-Analysis of Randomized Controlled Clinical Trials. Front. Nutr. 2022, 9, 809311. [Google Scholar] [CrossRef]
  203. Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. REDUCE-IT Investigators. Effects of icosapent ethyl on total ischemic events: From REDUCE-IT. J. Am. Coll. Cardiol. 2019, 73, 2791–2802. [Google Scholar] [CrossRef] [PubMed]
  204. Yokoyama, M.; Origasa, H.; Matsuzaki, M.; Matsuzawa, Y.; Saito, Y.; Ishikawa, Y.; Oikawa, S.; Sasaki, J.; Hishida, H.; Itakura, H.; et al. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): A randomised open-label, blinded endpoint analysis. Lancet 2007, 369, 1090–1098. [Google Scholar] [CrossRef] [PubMed]
  205. Gencer, B.; Djousse, L.; Al-Ramady, O.T.; Cook, N.R.; Manson, J.E.; Albert, C.M. Effect of Long-Term Marine ɷ-3 Fatty Acids Supplementation on the Risk of Atrial Fibrillation in Randomized Controlled Trials of Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. Circulation 2021, 144, 1981–1990. [Google Scholar] [CrossRef] [PubMed]
  206. Xie, L.; Zhen, P.; Wei, Q.; Yu, F.; Song, S.; Tong, J. Effects of omega-3 polyunsaturated fatty acids supplementation for patients with cardiovascular disease risks: A dose-response meta-analysis. Am. J. Transl. Res. 2021, 13, 8526–8539. [Google Scholar] [PubMed]
  207. Hu, X.F.; Kenny, T.-A.; Chan, H.M. Inuit Country Food Diet Pattern Is Associated with Lower Risk of Coronary Heart Disease. J. Acad. Nutr. Diet. 2018, 118, 1237–1248.e1. [Google Scholar] [CrossRef] [PubMed]
  208. Sales-Campos, H.; Souza, P.R.; Peghini, B.C.; da Silva, J.S.; Cardoso, C.R. An overview of the modulatory effects of oleic acid in health and disease. Mini Rev. Med. Chem. 2013, 13, 201–210. [Google Scholar]
  209. Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean Diet, its Components, and Cardiovascular Disease. Am. J. Med. 2015, 128, 229–238. [Google Scholar] [CrossRef] [Green Version]
  210. Liyanage, T.; Ninomiya, T.; Wang, A.; Neal, B.; Jun, M.; Wong, M.G.; Jardine, M.; Hillis, G.S.; Perkovic, V. Effects of the Mediterranean Diet on Cardiovascular Outcomes—A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0159252. [Google Scholar] [CrossRef] [Green Version]
  211. Román, G.C.; Jackson, R.E.; Gadhia, R.; Román, A.N.; Reis, J. Mediterranean diet: The role of long-chain ω-3 fatty acids in fish; polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine; probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Rev. Neurol. 2019, 175, 724–741. [Google Scholar] [CrossRef]
  212. Lavie, C.J.; Milani, R.V.; Mehra, M.R.; Ventura, H.O. Omega-3 Polyunsaturated Fatty Acids and Cardiovascular Diseases. J. Am. Coll. Cardiol. 2009, 54, 585–594. [Google Scholar] [CrossRef] [Green Version]
  213. Mozaffarian, D. Fish and n−3 fatty acids for the prevention of fatal coronary heart disease and sudden cardiac death. Am. J. Clin. Nutr. 2008, 87, 1991S–1996S. [Google Scholar] [CrossRef] [Green Version]
  214. Rauch, B.; Schiele, R.; Schneider, S.; Diller, F.; Victor, N.; Gohlke, H.; Gottwik, M.; Steinbeck, G.; Del Castillo, U.; Sack, R.; et al. OMEGA Study Group. OMEGA, a randomized, placebo-controlled trial to test the effect of highly purified omega-3 fatty acids on top of modern guideline-adjusted therapy after myocardial infarction. Circulation 2010, 122, 2152–2159. [Google Scholar] [CrossRef] [Green Version]
  215. Martino, A.; Pezzi, L.; Magnano, R.; Salustri, E.; Penco, M.; Calo’, L. Omega 3 and atrial fibrillation: Where are we? World J. Cardiol. 2016, 8, 114–119. [Google Scholar] [CrossRef]
  216. Rizos, E.C.; Ntzani, E.E.; Bika, E.; Kostapanos, M.S.; Elisaf, M.S. Association Between Omega-3 Fatty Acid Supplementation and Risk of Major Cardiovascular Disease Events: A systematic review and meta-analysis. JAMA 2012, 308, 1024–1033. [Google Scholar] [CrossRef]
  217. He, K.; Song, Y.; Daviglus, M.L.; Liu, K.; Van Horn, L.; Dyer, A.R.; Greenland, P. Accumulated Evidence on Fish Consumption and Coronary Heart Disease Mortality. Circulation 2004, 109, 2705–2711. [Google Scholar] [CrossRef] [Green Version]
  218. Bosch, J.; Gerstein, H.C.; Dagenais, G.R.; Diaz, R.; Dyal, L.; Jung, H.; Maggiono, A.P.; Probstfield, J.; Ramachandran, A.; Riddle, M.C.; et al. n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N. Engl. J. Med. 2012, 367, 309–318. [Google Scholar]
  219. Alexander, D.D.; Miller, P.E.; Van Elswyk, M.E.; Kuratko, C.N.; Bylsma, L.C. A meta-analysis of randomized controlled trials and prospective cohort studies of eicosapentaenoic and docosahexaenoic long-chain omega-3 fatty acids and coronary heart disease risk. In Mayo Clinic Proceedings; Elsevier: Amsterdam, The Netherlands, 2017; Volume 92, pp. 15–29. [Google Scholar]
  220. Aung, T.; Halsey, J.; Kromhout, D.; Gerstein, H.C.; Marchioli, R.; Tavazzi, L.; Geleijnse, J.M.; Rauch, B.; Ness, A.; Galan, P.; et al. Omega-3 Treatment Trialists’ Collaboration. Associations of omega-3 fatty acid supplement use with cardiovascular disease risks: Meta-analysis of 10 trials involving 77917 individuals. JAMA Cardiol. 2018, 3, 225–234. [Google Scholar] [CrossRef] [Green Version]
  221. Poorani, R.; Bhatt, A.N.; Dwarakanath, B.; Das, U.N. COX-2, aspirin and metabolism of arachidonic, eicosapentaenoic and docosahexaenoic acids and their physiological and clinical significance. Eur. J. Pharmacol. 2016, 785, 116–132. [Google Scholar] [CrossRef]
Figure 1. Molecular structure of the principal class 1 lipid mediators (on the left). On the right, the class 1 lipid mediators are divided according to their anti and pro cardiovascular risk properties.PGE2: prostaglandin E2; TXA2: thromboxane A2; 14-15 EET: 14-15 epoxyeicosatrienoic acid; LTB4: Leukotriene B4; LXA4: Lipoxin A4; 12(S)-HETE:12-hydroxyeicosatetraenoic acid; EP1,2,3, 4: prostaglandin Receptors.
Figure 1. Molecular structure of the principal class 1 lipid mediators (on the left). On the right, the class 1 lipid mediators are divided according to their anti and pro cardiovascular risk properties.PGE2: prostaglandin E2; TXA2: thromboxane A2; 14-15 EET: 14-15 epoxyeicosatrienoic acid; LTB4: Leukotriene B4; LXA4: Lipoxin A4; 12(S)-HETE:12-hydroxyeicosatetraenoic acid; EP1,2,3, 4: prostaglandin Receptors.
Ijms 24 01637 g001
Figure 2. Molecular structure of the principal class 2 lipid mediators (on the left). On the right, the class 2 lipid mediators are divided according to their anti- and pro-cardiovascular risk properties. PAF: Platelet activating factor; LPA: Lysophosphatidic acid; SP1: sphingosine-1-phosphate.
Figure 2. Molecular structure of the principal class 2 lipid mediators (on the left). On the right, the class 2 lipid mediators are divided according to their anti- and pro-cardiovascular risk properties. PAF: Platelet activating factor; LPA: Lysophosphatidic acid; SP1: sphingosine-1-phosphate.
Ijms 24 01637 g002
Figure 3. Molecular structure of the principal class 3 lipid mediators (on the left). On the right, the class 3 lipid mediators are divided according to their anti- and pro-cardiovascular risk properties. RvE: Resolvin E; RvD: Resolvin D; PD: Protectin; Maresin: MaR.
Figure 3. Molecular structure of the principal class 3 lipid mediators (on the left). On the right, the class 3 lipid mediators are divided according to their anti- and pro-cardiovascular risk properties. RvE: Resolvin E; RvD: Resolvin D; PD: Protectin; Maresin: MaR.
Ijms 24 01637 g003
Figure 4. Simplified scheme of biochemical transition from inflammation to resolution. Pro-resolving lipid mediators enhance the clearance (efferocytosis) of apoptotic PNM by macrophages, blocking leukocyte recruitment and favoring tissue repair. PNM (circle in red colour): polymorphonuclear neutrophils; PNM (in gray colour):apoptotic polymorphonuclear neutrophils; stars in blue color: pro-resolving monocytes.
Figure 4. Simplified scheme of biochemical transition from inflammation to resolution. Pro-resolving lipid mediators enhance the clearance (efferocytosis) of apoptotic PNM by macrophages, blocking leukocyte recruitment and favoring tissue repair. PNM (circle in red colour): polymorphonuclear neutrophils; PNM (in gray colour):apoptotic polymorphonuclear neutrophils; stars in blue color: pro-resolving monocytes.
Ijms 24 01637 g004
Table 1. Studies of Class 1,2,3 lipid mediators in animal models.
Table 1. Studies of Class 1,2,3 lipid mediators in animal models.
Studies in Animal Models Lipid MediatorsEffectsRef.
ApoE-deficient mice↑ TXA2↑ Atherogenesis[48]
↓ PGI2↑ Atherogenesis[48]
Mice deficient in LDL receptor/mPGEs 1↑ PGI2↓ Atherosclerosis[49,50]
Mice COX-2 knockout ↑ HBP, Trombosis[51]
ALOX15-deficient mice↓ HETES↑ resistance L-NAME-induced hypertension[52]
Mice Ang II-induced hypertension↓EETS↑ sEH↑ AngII-induced cardiac hypertrophy[53]
Administration of PAF receptor antagonist (SDZ 63.675) before reperfusion of the ischemic-isolated rabbit heart↓ PAF↓ Myocardial injury during reperfusion[54]
Male LDLr−/− mice (two weeks cholesterol + cacao butter diet) before surgery of carotid artery↑ LPA in atherosclerotic tissueAtherosclerotic Lesion Progression[55]
WT mice administered with soluble carrier for S1P (ApoM-Fc) after I/R injury in heart↑ SP1 Receptor↓ Myocardial damage after I/R injury[56]
Myocardial I/R in mouse ↑ RvE, RvD, PD, MaR↓ ROS, Inflammation[57,58,59]
Table 2. Studies of Class 1,2,3 lipid mediators in humans.
Table 2. Studies of Class 1,2,3 lipid mediators in humans.
Lipid MediatorsEffectsRef
↑ TXA2Systemic inflammation, myocardial ischemia[61]
↑ LTsInflammation, CV risk, MI[64,65]
↑ 12(S)-HETEVSMC relaxation[66]
↓ EETsObstructed CAD[67]
↑ PAFIschemic events[68,69,70]
↑ LPAAcute heart attacks and coronary syndrome[71,72,73]
↓ RvE1Vascular inflammation[74,75]
↓ 15-epi-LXA 4Disease severity[74,76]
Table 3. Trials on ω-3 fatty acid diet in humans.
Table 3. Trials on ω-3 fatty acid diet in humans.
StudyDesignSupplementationEnd PointRef.
GISSI-Prevenzione TrialsClinical trial of 11,324 post-MI patientsFish oil 850–882 mg EPA and DHAReduction Death Risk[201]
Meta analysis of 14 clinical Trials 135,291 subjects 0.8-1.2 g omega-3 FA(EPA, DHA)MACE, CVD, and MI[202]
Trial8179 participants and 5 years follow-up4 g/d ω-3 PUFA 30% reduction CVE/ control [203]
JELIS trial in Japan18,645 patients with a total cholesterol of 6.5 mmol/L1800 mg of EPA daily 19% reduction of coronary events[204]
TrialPatients suffering of cardiovascular diseases>1 g/d marine ɷ-3 fatty Increased AF Risk[205]
TrialPartecipants suffering major CVE and deathPUFA daily dose × period > 8 grams/day × yearsMore benefit/1 g/d for 1-year ω-3 PUFA[206]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lubrano, V.; Ndreu, R.; Balzan, S. Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis. Int. J. Mol. Sci. 2023, 24, 1637. https://doi.org/10.3390/ijms24021637

AMA Style

Lubrano V, Ndreu R, Balzan S. Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis. International Journal of Molecular Sciences. 2023; 24(2):1637. https://doi.org/10.3390/ijms24021637

Chicago/Turabian Style

Lubrano, Valter, Rudina Ndreu, and Silvana Balzan. 2023. "Classes of Lipid Mediators and Their Effects on Vascular Inflammation in Atherosclerosis" International Journal of Molecular Sciences 24, no. 2: 1637. https://doi.org/10.3390/ijms24021637

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