You are on page 1of 6

Journal of Cardiology 67 (2016) 22–27

Contents lists available at ScienceDirect

Journal of Cardiology
journal homepage: www.elsevier.com/locate/jjcc

Review

Cardioprotective mechanism of omega-3 polyunsaturated fatty acids


Jin Endo (MD, PhD)a,*, Makoto Arita (PhD)b,c,**
a
Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
b
Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
c
Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan

A R T I C L E I N F O A B S T R A C T

Article history: Omega-3 polyunsaturated fatty acids (PUFAs), such as eicosapentaenoic acid and docosahexaenoic acid,
Received 31 July 2015 are widely regarded as cardioprotective. Several large-scale, randomized clinical trials have shown that
Accepted 31 July 2015 dietary intake of omega-3 PUFAs improves the prognosis of patients with symptomatic heart failure or
Available online 8 September 2015
recent myocardial infarction. Therefore, dietary consumption of omega-3 PUFA is recommended in
international guidelines for the general population to prevent the occurrence of cardiovascular diseases
Keywords: (CVDs). However, the precise mechanisms underlying the cardioprotective effects of omega-3 PUFAs are
Omega-3 polyunsaturated fatty acids
not fully understood. Omega-3 PUFAs can be incorporated into the phospholipid bilayer of cell
Cardiovascular disease
Anti-inflammation
membranes and can affect membrane fluidity, lipid microdomain formation, and signaling across
Specialized proresolving mediators membranes. Omega-3 PUFAs also modulate the function of membrane ion channels, such as Na and
18-Hydroxyeicosapentaenoic acid L-type Ca channels, to prevent lethal arrhythmias. Moreover, omega-3 PUFAs also prevent the
conversion of arachidonic acid into pro-inflammatory eicosanoids by serving as an alternative substrate
for cyclooxygenase or lipoxygenase, resulting in the production of less potent products. In addition, a
number of enzymatically oxygenated metabolites derived from omega-3 PUFAs were recently identified
as anti-inflammatory mediators. These omega-3 metabolites may contribute to the beneficial effects
against CVDs that are attributed to omega-3 PUFAs.
ß 2015 Japanese College of Cardiology. Published by Elsevier Ltd. All rights reserved.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Modification of the cell membrane milieu by incorporation of omega-3 PUFAs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Anti-arrhythmic effects of omega-3 PUFAs due to ion channel modulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
The anti-inflammatory effects of omega-3 PUFAs mediated by nuclear receptors, G-protein coupled receptors, and other mechanisms . . . 24
Novel bioactive lipid mediators of omega-3 PUFAs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Novel beneficial effects of a macrophage-derived EPA metabolite, 18-HEPE, on cardiac remodeling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

Introduction
* Corresponding author at: Department of Cardiology, Keio University School of
Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan. The beneficial effects of n-3 polyunsaturated fatty acids
Tel.: +81 3 5363 3874; fax: +81 3 5363 3875. (PUFAs), primarily eicosapentaenoic acid (EPA) and docosahex-
** Corresponding author at: Laboratory for Metabolomics, RIKEN Center for aenoic acid (DHA), were first recognized in the late 1960s when
Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama
epidemiological evidence from the Inuit population, who consume
City, Kanagawa 230-0045, Japan. Tel.: +81 45 503 7055; fax: +045 503 7054.
E-mail addresses: jinendo@keio.jp (J. Endo), makoto.arita@riken.jp an n-3 PUFA-rich diet, showed that they have a low incidence
(M. Arita). of myocardial infarction [1]. Subsequently, a large number

http://dx.doi.org/10.1016/j.jjcc.2015.08.002
0914-5087/ß 2015 Japanese College of Cardiology. Published by Elsevier Ltd. All rights reserved.
J. Endo, M. Arita / Journal of Cardiology 67 (2016) 22–27 23

of randomized clinical trials have investigated the efficacy of Modification of the cell membrane milieu by incorporation of
supplementation with fish oil or omega-3 PUFAs. The GISSI- omega-3 PUFAs
Prevenzione trial [2] was the first large-scale, randomized trial to
provide evidence that dietary supplementation with omega-3 The cell membrane is composed of phospholipids (PLs) that
PUFAs had favorable effects on hard clinical end-points in post- contain various types of fatty acids. The length and saturation of
myocardial infarction patients. The GISSI-HF trial [3] also showed the fatty acids in these PLs is thought to affect the properties of cell
that omega-3 PUFAs could reduce morbidity and mortality in membranes by altering the microdomain ‘‘rafts’’ and ‘‘caveolae’’
patients with symptomatic chronic heart failure who were that concentrate membrane proteins and lipids and function as
receiving standard treatments, including aspirin, beta-blockers, signaling platforms. Since omega-3 PUFAs have many double
angiotensin-converting enzyme inhibitor/angiotensin receptor bonds and long-chain carbons, their incorporation into the PLs
blockers, and aldosterone receptor blockers. Further, the JELIS within a membrane can alter its properties and influence the
study [4], a randomized large trial conducted in Japan, revealed function of various membrane proteins (Fig. 1), including the
that treatment with a pharmaceutical preparation of highly suppression of protein kinase C theta signaling and interleukin (IL)-
purified EPA in addition to statin significantly prevented cardio- 2 production [7], and the disruption of dimerization and
vascular events in patients with recent myocardial infarction recruitment of toll-like receptor 4 [8]. Of note, alteration of the
through an apparently cholesterol-independent mechanism. The lipid microenvironment in cardiomyocytes through the inclusion
results of the above-mentioned studies showed that omega-3 of omega-3 PUFAs can modulate ion channel function, leading to
PUFA treatment is safe and well tolerated, and the clinical anti-arrhythmic effects [6].
improvements observed were additive to those of other well-
established therapies. Therefore, in a scientific statement from the Anti-arrhythmic effects of omega-3 PUFAs due to ion channel
American Heart Association, the consumption of >1 g/day of modulation
omega-3 PUFAs (EPA+DHA; this dose was largely determined from
the results of the GISSI-Prevenzione study) was recommended for The clinical outcomes of several trials, including GISSI-
patients with coronary artery disease to reduce triglyceride (TG) Prevenzione, have suggested that omega-3 PUFAs might prevent
levels, maintain cardiac function, and reduce the risk of coronary the occurrence of sudden cardiac death triggered by lethal
heart disease [5]. In addition, many animal studies have arrhythmias. Accumulating evidence from in vivo and in vitro
demonstrated that omega-3 PUFAs have pleiotropic beneficial experiments has demonstrated that omega-3 PUFAs exert anti-
effects in the cardiovascular system, including anti-arrhythmic, arrhythmic effects through modulation of myocyte electrophysi-
plasma TG-lowering, anti-thrombotic, anti-atherosclerotic, endo- ology. Omega-3 PUFAs reduce the activity of membrane sodium
thelial relaxation, blood pressure-lowering, anti-inflammatory, channels in cardiomyocytes, thus increasing the threshold for
and anti-fibrotic effects [6]. In this review, we present recent membrane potential depolarization [9]. EPA and DHA also
advances in our understanding of the molecular mechanisms modulate the activity of L-type calcium channels, leading to a
underlying the cardioprotective effects of omega-3 PUFAs. reduction in free cytosolic calcium ion, which stabilizes myocyte

[(Fig._1)TD$IG]

Fig. 1. The proposed molecular mechanism of cardioprotection by omega-3 PUFAs. Omega-3 PUFAs modulate cell membrane property when incorporated into the
phospholipid bilayer and control membrane ion channels to prevent lethal arrhythmia. Also omega-3 PUFAs exert anti-inflammatory and anti-fibrotic effects by modifying
NF-kB signaling, the NLRP3 inflammasome, PPARa/g, GPR120, and TGF-b signaling.
NF-kB: nuclear factor-kB; NLRP3: NOD-like receptor family, pyrin domain containing 3; PPARa/g: peroxisome proliferator-activated receptor a/g; GPR120: G protein-
coupled receptor 120; TGF-b: transforming growth factor-b.
24 J. Endo, M. Arita / Journal of Cardiology 67 (2016) 22–27

electrical excitability to prevent fatal arrhythmia [10]. EPA blocks IkB phosphorylation [14] (Fig. 1) or through the nuclear receptor
the sodium-calcium channel; however, a single amino-acid point PPARa/g [15] (Fig. 1). In addition, omega-3 PUFA is a ligand for
mutation in this channel attenuated the inhibitory effect of EPA GPR120, which attenuates both toll-like receptor 4- and TNF-a-
[11]. These findings suggested that the cardioprotective effect of n- mediated proinflammatory signaling in macrophages [16] (Fig. 1).
3 PUFAs is mediated by direct interaction with membrane ion GPR120, a member of the GPCR family, is also highly expressed in
channels (Fig. 1). mature adipocytes, and GPR120 stimulation with omega-3 PUFAs
The anti-arrhythmic effects of omega-3 PUFAs, which occur by was shown to augment GLUT4 glucose transporter expression to
blocking various ion channels, are encouraging. In fact, the results promote glucose uptake in adipocytes [16]. Chronic tissue
of several trials have suggested that dietary supplementation with inflammation is a well-known cause of insulin resistance. In a
omega-3 PUFAs might be an effective optional therapy for mouse obesity model, the anti-inflammatory and insulin-sensitiz-
arrhythmias. However, the FORWARD trial, a prospective, ran- ing effects of omega-3 PUFAs were shown to be dependent on the
domized, clinical trial, found that treatment with omega-3 PUFAs expression of GPR120, and administration of a selective agonist
over a 6-month period did not reduce recurrent symptomatic atrial improved insulin resistance [17]. NOD-like receptor family, pyrin
fibrillation [12]. In the specific study population with paroxysmal domain-containing 3 (NLRP3) senses non-microbial danger signals
atrial fibrillation (AF), it was concluded that omega-3 PUFAs are and forms an inflammasome, leading to a sterile inflammatory
not a useful treatment. These results do not exclude the potential response in myocardial infarction, ischemia reperfusion injury, and
anti-arrhythmic effects of prescription omega-3 PUFAs in combi- pressure overload-induced cardiac remodeling. Omega-3 PUFAs
nation with antiarrhythmic drugs in different populations, such as prevented NLRP3 inflammasome-dependent inflammation and
patients with heart failure; however, validation in prospective metabolic disorder in HFD-induced diabetic mice [18] (Fig. 1).
trials is required. Moreover, EPA and DHA directly reduced cardiac fibrosis under
pressure overload by inhibiting TGF-b1-induced smad2/3 nuclear
The anti-inflammatory effects of omega-3 PUFAs mediated by translocation, a major pathway involved in the development of
nuclear receptors, G-protein coupled receptors, and other cardiac remodeling [19] (Fig. 1). EPA and DHA increased nitric
mechanisms oxide production and promoted subsequent activation of the cyclic
GMP/PKG pathway in cardiac fibroblasts. Taken together, these
Acute and chronic inflammation underlie the pathogenesis and findings suggest that omega-3 PUFAs may have both anti-
progression of various cardiovascular diseases (CVDs), including inflammatory and anti-fibrotic properties.
myocarditis, myocardial infarction, aortic dissection, atheroscle-
rosis, and cardiac remodeling. It is widely believed that the anti- Novel bioactive lipid mediators of omega-3 PUFAs
inflammatory properties of omega-3 PUFAs contribute to their
cardioprotective effects. In fact, dietary intake of omega-3 PUFAs Since the 1970s, the beneficial effects of omega-3 PUFAs have
was reported to decrease the circulating concentrations of been commonly explained to be attributed to either prevention of
inflammatory cytokines such as tumor necrosis factor (TNF), IL- the conversion of the omega-6 PUFA arachidonic acid (AA) into
1b, and IL-6, and ameliorate left ventricular functional capacity in pro-inflammatory prostaglandins (PGs) and leukotrienes (LTs), or
non-ischemic dilated cardiomyopathy [13]. its ability to serve as an alternative substrate, producing less potent
EPA and DHA downregulate the expression of inflammation- mediators, such as 3-series PGs and thromboxanes (TXs) and
[(Fig._2)TD$IG]
related genes through inhibition of NF-kB signaling by blocking 5-series LTs (Fig. 2). For instance, the lower incidence of myocardial

Fig. 2. Anti-inflammatory effects of omega-3 PUFAs through mediator balance. The anti-inflammatory effects of omega-3 PUFAs are attributed to (1) prevention of the
conversion of AA into PGs and LTs or (2) its ability to function as an alternative substrate to produce less potent mediators. (3) Resolvins, protectins, and maresins, distinct
anti-inflammatory and pro-resolving lipid mediators derived from omega-3 PUFAs.
AA: arachidonic acid; PGs: prostaglandins; LTs: leukotrienes.
J. Endo, M. Arita / Journal of Cardiology 67 (2016) 22–27 25

infarction in a population that consumed a diet rich in omega-3 changes. One study showed that 12/15-LOX is protective against
PUFAs could be due, in part, to a reduction in the formation of the atherosclerosis through the production of SPMs including RvD1
pro-thrombotic prostanoid TXA2 from AA. Moreover, omega-3 and PD1, which could attenuate the local inflammatory response in
PUFAs are metabolized to PGI3, which possesses anti-platelet macrophages and endothelial cells [23]. Hasturk et al. found that
effects, and TXA3, which does not induce platelet aggregation [20]. oral-topical application of RvE1 reduced atherogenesis induced by
Liquid chromatography–mass spectrometry (LC–MS)-based both diet and periodontal inflammation and could prevent the
lipidomic analyses of murine inflammatory exudates or activated systemic inflammatory response and aortic atherosclerosis in the
cell supernatants have identified distinct omega-3 PUFA-derived absence of periodontitis [24].
pro-resolving mediators, such as resolvins, protectins, and There is accumulating evidence demonstrating that SPMs
maresins [21] (Fig. 2). They possess distinct chemical structures directly exert cardioprotective actions in vivo. Keyes et al. [25]
and exert their anti-inflammatory effects in a stereospecific reported that administration of RvE1 attenuated the infarct size in
manner. These mediators are now generally termed specialized rats subjected to ischemia/reperfusion injury. The results of this
pro-resolving mediators (SPMs). Resolvin E series are synthesized study suggested that RvE1 directly affected cardiomyocytes and
from EPA through the conversion of 18-hydroxyeicosapentaenoic protected against cardiac injury. In addition, Kain et al. [26]
acid (18-HEPE) by aspirin-acetylated COX2 or CYP450 monoox- showed that treatment with RvD1 accelerated the resolution of
ygenase. RvE1 actively switches off leukocyte trafficking to the acute inflammation following myocardial infarction by promoting
inflamed site, promotes the clearance of inflammatory cells and the production of SPM in the spleen and switching to anti-
debris, and suppresses cytokine production, thereby leading to inflammatory M2 macrophages in the left ventricle, which
resolution of acute inflammation [21]. In addition, RvE1 can inhibit prevented cardiac fibrosis and preserved cardiac function.
platelet aggregation by ADP or TXA2 receptor activation [21]. PUFAs are oxidized by CYP450 monooxygenase to epoxides,
DHA-derived mediators, such as protectins, resolvin D-series, which function as potent lipid mediators in the cardiovascular
and maresins, are generated by 15-LOX in humans or by 12/15- system. The epoxyeicosatrienoic acids (EETs) generated from AA
LOX in mice. PD1 exhibits protective effects against brain induce vasodilation, stimulate angiogenesis, and protect the heart
ischemia, resistance to retinal oxidative injury, and protection from ischemia/reperfusion injury. CYP450 monooxygenase also
against renal ischemia/reperfusion injury [21]. RvD1 is shown to converts EPA and DHA to epoxyeicosatetraenoic acids (EpETEs)
be protective in various disease models, such as insulin resistance, and epoxydocosapentaenoic acids (EpDPAs), respectively, which
atherosclerosis, ischemia reperfusion, and others [21]. In addition, function as lipid mediators similar to EET. By activating BK
RvD2 could reduce the damage and subsequent scarring of channels, 17(18)-EpETE dilates human pulmonary arteries and
kidneys after ischemia/reperfusion [21]. PD1 and RvD1 have been 16(17)-EpDPA activates rat coronary smooth muscle [27]. Omega-
shown to reduce pathogenic neovascularization in murine 3 PUFA-derived epoxides possess anti-inflammatory properties in
oxygen-induced retinopathy [21]. Furthermore, a recent report the setting of CVD. The increased EpETE and EpDPA levels
showed that administration of PD1 isomer (PDX) improved following dietary administration of omega-3 PUFAs reduced the
insulin sensitivity in obese diabetic db/db mice, suggesting that renal markers of inflammation in angiotensin II-dependent
this SPM possesses therapeutic potential for the treatment of hypertension [27]. EpETEs and EpDPAs protect the heart through
metabolic disorders [22]. their anti-arrhythmic actions. Recent studies are focusing on the
Atherosclerosis is now commonly recognized as an unresolved development of stable epoxide bioisosteric analogs and soluble
inflammatory disease involving the vascular wall. Therefore, it is epoxide hydrolase inhibitors that can stabilize epoxides by
assumed that the pathogenesis of atherosclerosis involves a inhibiting their hydrolysis to the corresponding diols in order
decrease in SPM production; thus, SPM replacement could control to potentiate their functional effects and for use as novel
[(Fig._3)TD$IG] local inflammatory response and improve the atherosclerotic
the therapeutics against CVD [28].

Fig. 3. An EPA metabolite, 18-HEPE, from cardiac macrophages rich in omega-3 PUFAs prevents cardiac remodeling under pressure overload. Activated cardiac
fibroblasts, namely myofibroblasts, produce pro-inflammatory mediators that facilitate cardiac macrophage activation. Cardiac fibroblasts can be activated directly by
pressure overload or secondarily by inflammatory mediators released from activated inflammatory cells. EPA-enriched macrophages generate an 18-HEPE-rich milieu in the
heart, thereby discontinue the profibrotic feed-forward loop that is involved in the development of cardiac fibrosis under pressure overload.
EPA: eicosapentaenoic acid; 18-HEPE: 18-hydroxy eicosapentaenoic acid.
26 J. Endo, M. Arita / Journal of Cardiology 67 (2016) 22–27

Novel beneficial effects of a macrophage-derived EPA References


metabolite, 18-HEPE, on cardiac remodeling
[1] Mozaffarian D, Wu JH. Omega-3 fatty acids and cardiovascular disease: effects
on risk factors, molecular pathways, and clinical events. J Am Coll Cardiol
Recently, we identified a novel anti-inflammatory and anti- 2011;58:2047–67.
fibrotic EPA metabolite, 18-HEPE, and elucidated the mechanism [2] GISSI-Prevenzione Investigators. (Gruppo Italiano per lo Studio della Soprav-
vivenza nell’Infarto miocardico) Dietary supplementation with n-3 polyun-
underlying omega-3 PUFA-mediated prevention of cardiac remo- saturated fatty acids and vitamin E after myocardial infarction: results of the
deling under pressure overload [29] (Fig. 3). Kang et al. developed a GISSI-Prevenzione trial. Gruppo Italiano per lo Studio della Sopravvivenza
transgenic mouse expressing the Caenorhabditi elegans fat-1 gene, nell’Infarto miocardico. Lancet 1999;354:447–55.
[3] Tavazzi L, Maggioni AP, Marchioli R, Barlera S, Franzosi MG, Latini R, Lucci D,
which encodes an omega-3 desaturase that converts omega-6 Nicolosi GL, Porcu M, Tognoni G. Effect of n-3 polyunsaturated fatty acids in
PUFAs to omega-3 PUFAs [30]. Fat-1 mice showed enrichment of patients with chronic heart failure (the GISSI-HF trial): a randomised, double-
omega-3 PUFAs in almost all cells and tissues, and displayed blind, placebo-controlled trial. Lancet 2008;372:1223–30.
[4] Yokoyama M, Origasa H, Matsuzaki M, Matsuzawa Y, Saito Y, Ishikawa Y,
resistance to numerous inflammatory diseases, including colitis,
Oikawa S, Sasaki T, Hishida H, Itakura H, Kita T, Kitabatake A, Nakaya N, Sakata
pancreatitis, osteoarthritis, atherosclerosis, obesity-linked insulin T, Shimada K, et al. Effects of eicosapentaenoic acid on major coronary events
resistance, and some cancers [31]. Compared to wild-type mice, in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded
fat-1 mice subjected to pressure overload showed sustained heart endpoint analysis. Lancet 2007;369:1090–8.
[5] Kris-Etherton PM, Harris WS, Appel LJ. American Heart Association. Nutrition
function and reduced cardiac remodeling (fibrosis) without any Committee. Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular
difference in cardiac hypertrophy. Examination of bone marrow disease. Circulation 2002;106:2747–57.
transplantation revealed that enrichment of omega-3 PUFAs in [6] Mozaffarian D, Wu JH. Omega-3 fatty acids and cardiovascular disease: effects
on risk factors, molecular pathways, and clinical events. J Am Coll Cardiol
bone marrow-derived cells (mostly macrophages), but not in 2011;58:2047–67.
cardiac cells, was responsible for the anti-fibrotic phenotype [7] Fan YY, Ly LH, Barhoumi R, McMurray DN, Chapkin RS. Dietary docosahex-
observed in fat-1 mice subjected to pressure overload. LC–MS/MS- aenoic acid suppresses T cell protein kinase C theta lipid raft recruitment and
IL-2 production. J Immunol 2004;173:6151–60.
based lipidomics revealed that high levels of 18-HEPE, an EPA [8] Wong SW, Kwon MJ, Choi AM, Kim HP, Nakahira K, Hwang DH. Fatty acids
metabolite, were produced by fat-1 transgenic macrophages, and modulate Toll-like receptor 4 activation through regulation of receptor dimer-
18-HEPE suppressed IL-6 production from cardiac fibroblasts in ization and recruitment into lipid rafts in a reactive oxygen species-dependent
manner. J Biol Chem 2009;284:27384–92.
nanomolar range (Fig. 3). Of note, dietary intake of EPA ethyl ester
[9] Xiao YF, Kang JX, Morgan JP, Leaf A. Blocking effects of polyunsaturated fatty
(2700 mg/day) significantly increased the plasma concentration of acids on Na+ channels of neonatal rat ventricular myocytes. Proc Natl Acad Sci
18-HEPE. Furthermore, administration of 18-HEPE prevented U S A 1995;92:11000–04.
[10] Hallaq H, Smith TW, Leaf A. Modulation of dihydropyridine-sensitive calci-
cardiac dysfunction, macrophage infiltration, and cardiac fibrosis
um channels in heart cells by fish oil fatty acids. Proc Natl Acad Sci U S A
after transverse aortic constriction (TAC), indicating the therapeu- 1992;89:1760–4.
tic potential of 18-HEPE for cardiac remodeling under pressure [11] Xiao YF, Ke Q, Wang SY, Auktor K, Yang Y, Wang GK, Morgan JP, Leaf A. Single
overload [29]. point mutations affect fatty acid block of human myocardial sodium channel
alpha subunit Na+ channels. Proc Natl Acad Sci U S A 2001;98:3606–11.
[12] Macchia A, Grancelli H, Varini S, Nul D, Laffaye N, Mariani J, Ferrante D, Badra R,
Conclusions Figal J, Ramos S, Tognoni G, Doval HC. GESICA Investigators. Omega-3 fatty
acids for the prevention of recurrent symptomatic atrial fibrillation: results of
the FORWARD (Randomized Trial to Assess Efficacy of PUFA for the Mainte-
Not all omega-3 PUFAs trials have shown reductions in CVD; nance of Sinus Rhythm in Persistent Atrial Fibrillation) trial. J Am Coll Cardiol
however, several adequately powered observation and inter- 2013;61:463–8.
vention trials have strongly supported the efficacy of omega-3 [13] Nodari S, Triggiani M, Campia U, Manerba A, Milesi G, Cesana BM, Gheorghiade
M, Dei Cas L. Effects of n-3 polyunsaturated fatty acids on left ventricular
PUFAs for the prevention of CVD. Furthermore, experimental function and functional capacity in patients with dilated cardiomyopathy. J
studies have revealed multiple underlying molecular mecha- Am Coll Cardiol 2011;57:870–9.
nisms, including membrane modification, attenuation of ion [14] Zhao Y, Joshi-Barve S, Barve S, Chen LH. Eicosapentaenoic acid prevents LPS-
induced TNF-alpha expression by preventing NF-kappaB activation. J Am Coll
channels, regulation of pro-inflammatory gene expression, Nutr 2004;23:71–8.
and production of lipid mediators. It remains unclear which [15] Gani OA, Sylte I. Molecular recognition of docosahexaenoic acid by peroxisome
mechanism contributes the most to the cardioprotective effects proliferator-activated receptors and retinoid-X receptor alpha. J Mol Graph
Model 2008;27:217–24.
of omega-3 PUFAs observed in vivo; however, the pleiotropic
[16] Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins
anti-inflammatory effects of omega-3 PUFAs could be valuable, SM, Olefsky JM. GPR120 is an omega-3 fatty acid receptor mediating potent
especially in the setting of atherosclerosis and cardiac remodel- anti-inflammatory and insulin-sensitizing effects. Cell 2010;142:687–98.
ing. Although further work is needed to clarify the molecular [17] Oh DY, Walenta E, Akiyama TE, Lagakos WS, Lackey D, Pessentheiner AR, Sasik
R, Hah N, Chi TJ, Cox JM, Powels MA, Di Salvo J, Sinz C, Watkins SM, Armando
relationship between omega-3 PUFAs and cardiac physiology/ AM, et al. A Gpr120-selective agonist improves insulin resistance and chronic
pathology, it might be useful to consider bioactive omega-3 inflammation in obese mice. Nat Med 2014;20:942–7.
PUFA-derived metabolites, such as 18-HEPE, as endogenous [18] Yan Y, Jiang W, Spinetti T, Tardivel A, Castillo R, Bourquin C, Guarda G, Tian Z,
Tschopp J, Zhou R. Omega-3 fatty acids prevent inflammation and metabolic
anti-inflammatory molecules and potential new therapeutic disorder through inhibition of NLRP3 inflammasome activation. Immunity
targets for CVD. 2013;38:1154–63.
[19] Chen J, Shearer GC, Chen Q, Healy CL, Beyer AJ, Nareddy VB, Gerdes AM, Harris
WS, O’Connell TD, Wang D. Omega-3 fatty acids prevent pressure overload-
Funding induced cardiac fibrosis through activation of cyclic GMP/protein kinase G
signaling in cardiac fibroblasts. Circulation 2011;123:584–93.
This work was supported by funding from the Japan Science and [20] Dyerberg J, Bang HO, Stoffersen E, Moncada S, Vane JR. Eicosapentaenoic acid
and prevention of thrombosis and atherosclerosis? Lancet 1978;2:117–9.
Technology Agency Precursory Research for Embryonic Science [21] Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology.
and Technology (PRESTO) [to M.A.], a Grant-in-aid for Scientific Nature 2014;510:92–101.
Research from the Ministry of Education, Culture, Sports, Science, [22] White PJ, St-Pierre P, Charbonneau A, Mitchell PL, St-Amand E, Marcotte B,
Marette A. Protectin DX alleviates insulin resistance by activating a myokine-
and Technology of Japan [to M.A.], and a Research Fellowship from
liver glucoregulatory axis. Nat Med 2014;20:664–9.
the Japan Society for the Promotion of Science for Young Scientists [23] Merched AJ, Ko K, Gotlinger KH, Serhan CN, Chan L. Atherosclerosis: evidence
[to J.E.] for impairment of resolution of vascular inflammation governed by specific
lipid mediators. FASEB J 2008;22:3595–606.
[24] Hasturk H, Abdallah R, Kantarci A, Nguyen D, Giordano N, Hamilton J, Van
Conflict of interest Dyke TE. Resolvin E1 attenuates atherosclerotic plaque formation in diet
and inflammation-induced atherogenesis. Arterioscler Thromb Vasc Biol
The authors have no conflict of interest to disclose. 2015;35:1123–33.
J. Endo, M. Arita / Journal of Cardiology 67 (2016) 22–27 27

[25] Keyes KT, Ye Y, Lin Y, Zhang C, Perez-Polo JR, Gjorstrup P, Birnbaum Y. Resolvin [28] Liu Y1, Wang R, Li J, Rao J, Li W, Falck JR, Manthati VL, Medhora M, Jacobs ER,
E1 protects the rat heart against reperfusion injury. Am J Physiol Heart Circ Zhu D. Stable EET urea agonist and soluble epoxide hydrolase inhibitor regulate
Physiol 2010;299:153–64. rat pulmonary arteries through TRPCs. Hypertens Res 2011;34:630–9.
[26] Kain V, Ingle KA, Colas RA, Dalli J, Prabhu SD, Serhan CN, Joshi M, Halade GV. [29] Endo J, Sano M, Isobe Y, Fukuda K, Kang JX, Arai H, Arita M. 18-HEPE, an n-3
Resolvin D1 activates the inflammation resolving response at splenic and fatty acid metabolite released by macrophages, prevents pressure overload-
ventricular site following myocardial infarction leading to improved ventric- induced maladaptive cardiac remodeling. J Exp Med 2014;211:1673–87.
ular function. J Mol Cell Cardiol 2015;84:24–35. [30] Kang JX, Wang J, Wu L, Kang ZB. Transgenic mice: fat-1 mice convert n-6 to n-3
[27] Spector AA, Kim HY. Cytochrome P450 epoxygenase pathway of polyun- fatty acids. Nature 2004;427:504.
saturated fatty acid metabolism. Biochim Biophys Acta 2015;1851: [31] Kang JX. A transgenic mouse model for gene-nutrient interactions. J Nutri-
356–65. genet Nutrigenomics 2008;1:172–7.

You might also like