Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Critical Reviews in Food Science and Nutrition

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/bfsn20

Omega-3 pleiad: The multipoint anti-inflammatory


strategy

Ellencristina da Silva Batista, Susana Castelo Branco Ramos Nakandakari,


Adelino Sanchez Ramos da Silva, José Rodrigo Pauli, Leandro Pereira de
Moura, Eduardo Rochete Ropelle, Enilton A. Camargo & Dennys Esper Cintra

To cite this article: Ellencristina da Silva Batista, Susana Castelo Branco Ramos Nakandakari,
Adelino Sanchez Ramos da Silva, José Rodrigo Pauli, Leandro Pereira de Moura, Eduardo
Rochete Ropelle, Enilton A. Camargo & Dennys Esper Cintra (2022): Omega-3 pleiad: The
multipoint anti-inflammatory strategy, Critical Reviews in Food Science and Nutrition, DOI:
10.1080/10408398.2022.2146044

To link to this article: https://doi.org/10.1080/10408398.2022.2146044

Published online: 16 Nov 2022.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=bfsn20
Critical Reviews in Food Science and Nutrition
https://doi.org/10.1080/10408398.2022.2146044

Review

Omega-3 pleiad: The multipoint anti-inflammatory strategy


Ellencristina da Silva Batistaa,b,c,d*, Susana Castelo Branco Ramos Nakandakarib,c*, Adelino Sanchez
Ramos da Silvae, José Rodrigo Paulic,f, Leandro Pereira de Mouraf, Eduardo Rochete Ropellec,f, Enilton A.
Camargoa,g and Dennys Esper Cintrab,c,h
a
Graduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Aracaju, Sergipe, Brazil; bNutritional Genomics Laboratory,
LabGeN, School of Applied Sciences, UNICAMP, São Paulo, Brazil; cNutrigenomics and Lipids Research Center, CELN, School of Applied
Sciences, UNICAMP, São Paulo, Brazil; dNutrition Department, Federal University of Sergipe, Lagarto, Sergipe, Brazil; eSchool of Physical
Education and Sport, University of São Paulo – USP, Ribeirão Preto, São Paulo, Brazil; fLaboratory of Molecular Biology of Exercise, School of
Applied Sciences, UNICAMP, São Paulo, Brazil; gDepartment of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil; hOCRC
– Obesity and Comorbidities Research Center, UNICAMP, São Paulo, Brazil

ABSTRACT KEYWORDS
Omega 3 (ω3) fatty acids have been described since the 1980s as promising anti-inflammatory Omega 3; inflammation;
substances. Prostaglandin and leukotriene modulation were exhaustively explored as the main chronic diseases; GPR120;
reason for ω3 beneficial outcomes. However, during the early 2000s, after the human genome cellular signaling; nutrigenomics
decoding advent, the nutrigenomic approaches exhibited an impressive plethora of ω3 targets,
now under the molecular point of view. Different G protein-coupled receptors (GPCRs) recognizing
ω3 and its derivatives appear to be responsible for blocking inflammation and insulin-sensitizing
effects. A new class of ω3-derived substances, such as maresins, resolvins, and protectins, increases
ω3 actions. Inflammasome disruption, the presence of GPR120 on immune cell surfaces, and
intracellular crosstalk signaling mediated by PPARγ compose the last discoveries regarding the
multipoint anti-inflammatory targets for this nutrient. This review shows a detailed mechanistic
proposal to understand ω3 fatty acid action over the inflammatory environment in the background
of several chronic diseases.

Introduction receptor (Hirasawa et al. 2005) their homologs, together


with nodal intracellular proteins, responsive structures,
During the 1970s, Dyerberg and Bang (1979) noticed the
nuclear receptors, genes, metabolites, particular conditions,
existing paradox between Danish and Greenlandic popula-
and other determinants involved in ω3 anti-inflammatory
tions related to cardiovascular disease incidence. Although
abilities or even their side effects (Bagger et al. 2020; Downie
the total fat consumption was the same among these pop-
et al. 2019; Hirasawa et al. 2005; Ishitani et al. 2003; Oh
ulations, coronary disease was less significant among
et al. 2010; Poreba et al. 2017; Yang, 2014).
Greenlandic Inuit Eskimos. For the first time, they showed
Thus, this review focuses on the multitude of
the influence of marine sources of omega-3 (ω3) fatty acids
anti-inflammatory molecular mechanisms targeted by ω3
on hemostatic function and platelets of Inuits and the par-
fatty acids and their new potential for use in experimental
ticipation of thromboxane A3, derived from ω3 after cyclo-
and clinical studies related to chronic diseases with the
oxygenase action, increasing the time of bleeding (Bang,
low-grade proinflammatory state as background.
Dyerberg, and Hjøorne 1976; Dyerberg et al. 1978; Dyerberg
and Bang 1979).
Throughout the 1980s, the hypothesis for eicosanoid ω3 and prostaglandins: an old relationship
modulation by ω3 was maintained and reinforced in vitro
(Croset et al. 1988) and in vivo, with animals (Adan et al. The ratio between ω3 and ω6 fatty acid intake needs to be
1999; Tashijan et al. 1984) and humans (Payan et al. 1986; considered in the context of inflammation, as ω6 intake is
Singer et al. 1990), until the 2000s. With the advent of considered pro-inflammatory when it is ten times higher
molecular approaches after human genome decoding, several than ω3 (10:1) due to the ability of ω6 (linoleic [C18:2] and
new signaling pathways were established for biological com- arachidonic [C20:4]) to be efficiently converted to prosta-
pounds, natural substances, or xenobiotics. However, the glandins from even series (i.e., PGE2, PGI2, and TXA2).
molecular mechanisms of action of ω3 stagnated for a long Interestingly, eicosapentaenoic acid (EPA – C20:5) competes
time. Currently, science has deciphered the specific ω3 with arachidonic for cyclo-oxygenases reducing its action

CONTACT Dennys Esper Cintra dcintra@yahoo.com


*These authors work equally.
© 2022 Taylor & Francis Group, LLC
2 E. DA SILVA BATISTA ET AL.

on arachidonic (DiNicolantonio, Okeefe 2019). Despite it, these enzymes works in a tissue-dependent manner (Figure
and considering the optimal ω6:ω3 ratio (5:1), Simopoulos 6B). As a confounding factor, delta-5 and delta-6 enzymes
(2016) described a very discrepant ratio for western coun- also act in the linoleic fatty acid (C18:2 [ω6]) bioconversion
tries, approximately 20:1. Nonetheless, some societies far route, becoming a competitive process between these fatty
exceed this value, reaching more than 50:1 (Udipi et al. 2006). acids, which needs to be considered (Picklo, Murphy 2016).
ω3 alters eicosanoid profile generation in a class of less Reductions in dietary ω3 result in an increased ability of
reactive prostaglandins and more effective leukotrienes. the liver to elongate and desaturate ω3 fatty acids, whereas
Prostaglandins such as PGE2 (Prostaglandin E2) and PGI2 the brain is not responsive to dietary levels (Demar et al.
(Prostacyclin I2) as well as TXA2 (Thromboxane A2), are 2005). This evidences that ω3 and ω6 fatty acid metabolism
the classical mediators of cardinal symptoms of inflamma- regulation is more complex than a trivial competition by
tion, such as pain, heat, rubor, and tumescence, which the substrates, once this process is regulated by the genetic
increase vasodilatation, bronchoconstriction, platelet aggre- background as well (Barceló-Coblijn, 2009; Demar et al.
gation, polymorphonuclear cell recruitment, and other effects 2005; Zhang, Kothapalli, and Brenna 2016).
(Esser-von Bieren 2019; Mastalerz et al. 2019). These medi- The metabolic effects of derived products from ALA or
ators are generated predominantly from arachidonic acid, other ω3 fatty acids are essential to be noticed. For example,
an ω6 fatty acid, after COX1 (Cyclooxygenase 1) action – a in a randomized double-blind crossover study, Gabbs et al.
constitutive enzyme – or mainly after COX2 (Cyclooxygenase (2021) treated twelve healthy volunteers with 4 g of ALA or
2) – an enzyme that rapidly emerges after tissue aggression DHA capsules for 28 days to measure oxylipin production.
(Esser-von Bieren 2019; Kain wt al. 2018). However, from Oxylipins are biologically active lipids formed from PUFAs
ω3, a mild alteration in the structure of these eicosanoids released from membrane phospholipids by phospholipase
can enormously change their functions. PGE3 (Prostaglandin A2 and oxygenated by COX (Cycloxygenases), ALOX
E3) and PGI3 (Prostaglandin I3) diminish vasodilatation, (Lipoxygenases), and cytochrome P450 enzymes (An, Kim,
while TXA3 (Thromboxane A3) markedly increases platelet and Oh 2021). After 28 days, ALA supplementation doubled
antiaggregant effects, leading to an increased time of bleed- ALA blood concentrations; however, without effects on ALA
ing (Dyerberg, Bang 1979). These effects, shown in several
oxylipins. Also, DHA supplementation tripled both DHA
studies, encouraged medical societies to prescribe ω3 fatty
and its oxylipins, showing a more active cytochrome metab-
acids mainly in poststroke and heart attacks (Bang, Dyerberg,
olism associated with DHA (Gabbs et al. 2021). This data
and Hjøorne 1976; Bhatt et al. 2019a; Bhatt et al. 2019b;
demonstrates that each nutrient has a specific role.
Croset et al. 1988; Dyerberg, Bang 1979; Dyerberg et al.
Nutrigenomic approaches have been decisive enlarging the
1978; Payan et al. 1986; Singer et al. 1990).
understanding of each nutrient. EPA and DHA were always
referenced as the main ω3 species associated with increased
bleeding time.
The neglected role of alpha-linolenic (C18:3) ω3 Recently, the same role was shown for ALA; however,
fatty acid using a different mechanism. While EPA and DHA influ-
The most relevant ω3 members considered over the years ence the time of bleeding through TXA3 prostaglandin
in the classical nutritional sciences literature were EPA and modulation, ALA changes the gene expression of adhesion
docosahexaenoic acid (DHA – C22:6), while alpha-linolenic molecules such as intercellular adhesion molecule-1 (ICAM),
acid (ALA – C18:3) was practically neglected (Arterburn, vascular cell adhesion molecule-1 (VCAM), and von
Hall, and Oken 2006). This fact could be attributed to the Willebrand Factor (vWF) (Stivala et al. 2022). The gene
low grade of C18:3 bioconversion to longer fatty acids (C20:5 expression of ICAM and VCAM is coordinated by NFkB
and C22:6) in mammals (Arterburn, Hall, and Oken 2006), (nuclear factor-kappa B), a transcription factor considered
and it has few biological activities compared to EPA and a masterpiece of proinflammatory genes. Holy et al. (2011)
DHA. In baboon neonates receiving oral doses of 13 C-labeled showed that dietary ALA could reduce endothelial cell
ALA or DHA for two weeks, the bioconversion from ALA activation by downregulating MAPK (Mitogen-Activated
to DHA was different for each tissue tested. In the brain, Protein Kinase) pathways and the nuclear translocation of
only 0.23% of DHA was preformed from ALA compared to NFkB. A more detailed ALA role associated with inflam-
1.71% of those treated directly with DHA, while in tissues mation was understood after discovering the GPR120 and
such as the liver or erythrocytes, this ratio reached up to GPR40 receptors, which recognize several unsaturated fatty
60% (Su et al. 1999). Demar Jr et al. (2006) found a low acids and ALA (Cintra et al. 2012; Oliveira et al. 2015).
rate of DHA bioconversion in the brains of rats receiving Thus, the demonstration of ALA influencing health and
ALA. In contrast, Sinclair, Guo, and Abedin (2022) showed disease was only at the beginning; however, its potential
DHA incorporation in the retina of guinea pigs proportion- for use needs to be reconsidered, once its natural sources
ally to the ALA content in the diet. The difference is that are abundant. The oils from chia seed and flaxseed have
retina cells are essentially composed of neuron cells (Dátilo approximately 50–60% ALA in their composition, making
et al. 2018). these oils or seeds a very attractive food due to the low
If the ALA bioconversion to elongated fatty acids is gov- cost compared to EPA or DHA capsules (Cintra et al. 2006;
erned by delta-5/6 desaturases (Zhang, Kothapalli, and Knez et al. 2019). The molecular mechanism by which ALA
Brenna 2016), it is plausible that the gene expression of coordinates the inflammatory process will be shown ahead.
Critical Reviews in Food Science and Nutrition 3

The interpretative transition from prostaglandins during the hypermetabolic context, followed by high amounts
to NFkB pathway as ω3-molecular targets of saturated free fatty acids released from adipose tissue
directly to the bloodstream. Although these mentioned con-
The lipid influence debates related to prostaglandins and ditions are commonly associated with an “obesogenic sce-
inflammation have crossed the decades. In 2003, some dif- nario,” different circumstances can interfere with the fatty
ferent hypotheses were proposed for the anti-inflammatory acids released from adipose or other tissues. For instance,
effects of ω3 once COX2 gene expression was discovered to cold exposition (Chen et al. 2022), gender (Hames et al.
be regulated by transcription factors such as NFkB, CREB 2015), exercise (de Melo et al. 2022). severe fasting (Marks
(Cyclic-AMP Response Element-Binding Protein), and C/EBP et al. 2015; Raclot 2003), parenteral nutrition (Guthrie 2022),
(CCAAT Enhancer-Binding Protein) (Park et al. 2005). drugs (Yu et al. 2022), burning injury, lipodystrophy, and
Posttranscriptionally, the COX2 protein structure can be cachexia (Vegiopoulos, Rohm, and Herzig 2017) are distinct
directly affected by the iNOS (inducible Nitric Oxide examples of fatty acids release.
Synthase) protein, which nitrosylates and intensifies COX2 Saturated fats bind and activate TLR4 (Huang et al. 2012),
activation (Kim, Huri, and Snyder 2005). Each step of COX2 a transmembrane innate immunity receptor that recruits
regulation can be used as a potential ω3-therapeutic target. MyD88 (Myeloid Differentiation Primary Response 88 Protein)
In 2003, Komatsu et al. (2003) showed a reduction in iNOS in juxtaposition. These connected proteins attract IRAK4
expression and its main product, nitric oxide (NO), after (interleukin-1 receptor-associated kinase 4), which associates
macrophage cell line (RAW264) DHA incubation. Then, ω3 with and phosphorylates a similar protein, IRAK1, inducing
fatty acids probably decrease prostaglandins due to iNOS TRAF6 (TNF Receptor-Associated Factor 6) recruitment and
inactivation. However, if iNOS gene expression is partially activation. The activated IRAK1 and TRAF6 complex is dis-
encoded by NFkB (Park et al. 2005), iNOS inhibition by ω3 sociated from this large protein conglomerate and migrates
may be an indirect activity. until the inoperant TAK1 (Transforming Growth Factor-β-
Different studies have shown the disruption of NFkB Activated Kinase 1) activates it (Adhikari, Xu, and Chen 2007).
signaling after ω3 treatment in several conditions, such as Juxtaposed to TAK1, IRAK1 and TRAF6 recruit the proteins
leukemia (Fahrmann et al. 2013), neuronal damage (Yang TAB1/2 (Transforming Growth Factor-Beta Activated Kinase
et al. 2014), and sickle cell disease (Daak et al. 2015), among 1 binding protein 1/2) to TAK1. TAB proteins induce TAK1
others. Sinha et al. (2004) postulated preliminary evidence ubiquitination through the activation of UBC3 and UEV1A
that ω3 could induce Ikk (Inhibitor Kappa Kinase) inhibi- (Ubiquitin Ligases), which allows TAK1 phosphorylation and
tion, an upstream NFkB protein (Spenser et al. 2013). activation (Adhikari, Xu, and Chen 2007; Shi, Kehrl 2010;
Mihaly, Morioka 2014) (Figure 1A). As shown above, the
Low-grade inflammation modulated by fats relative novelty in the ω3 anti-inflammatory mechanism is
related specifically to this molecular nodus. Once after TAK1
The NFkB proinflammatory end-products are controlled by phosphorylation, it phosphorylates and activates IKK (Inhibitor
both TLR2/4 (Toll-Like Receptors 2 and 4) and cytokine of Kappa Kinase), activating the canonical NFkB pathway (Oh
receptors such as TNFα (Tumor Necrosis Factor-α), IL et al. 2010; Figure 1B and C).
(Interleukin) 1β, and IL6 pathways. In the context of car- Parallel to TLR signaling, signals from cytokine receptors
diometabolic risk, proinflammatory stimuli are triggered by can enlarge the proinflammatory stimulus. When activated
the connection between saturated fatty acids and TLRs, by its ligands, at the base of cytokine receptors, TRAF2/6
mainly TLR4 (Huang et al. 2012). Despite the TLR-saturated (TNF Receptor-Associated Factor 2/6) is attached and acti-
fat binding has become a classical system explaining inflam- vated, transducing the signaling to TAK1 (Ishitani et al.
matory signaling beginning, it might be questionable. 2003; Figure 1B). Therefore, the signals from TLR or cyto-
Lancaster et al. (2018) showed that saturated fatty acids kine receptors culminate at the same point. The most evi-
could trigger inflammation, not precisely through TLR4, but denced ω3 action is attributed to anti-inflammatory ability
through its downstream pathway. Using bone marrow-derived more than antihypertensive, anti-thrombogenic, or hypoli-
macrophages, they showed that saturated fatty acids inter- pemiant actions. However, part of this ability is due to its
nally “pre-prepare” the cell for inflammation using recently deorphanized receptor, GPR120.
TLR4-associated proteins (MyD88 and others). The inflam-
mation was induced by saturated fatty acid even in the
absence of TLR4 (Tlr4-KO-/- cells). On the opposite, Sutter G protein-coupled receptors recognize and
et al. (2016) showed the ablation of pro-inflammatory stimuli mediate anti-inflammatory ω3 actions
induced by saturated fat in the liver of Tlr4 KO mice. Using
biophysical approaches, Kang and Lee (2011) demonstrated In 2005, the GPR120, a GPCR (G protein-coupled receptor),
the exact position of the structural binding site for saturated was partially deorphanized as a long-chain fatty acid recep-
lipids. For a while, all scientific evidence strongly suggests tor. However, there are many ligands as possibilities, with
the TLR4 or its surrounding proteins system is the primary an extensive range of fatty acids, among 16 to 24 carbons
conductor of inflammation induced by saturated fats. (Hirasawa et al. 2005). In elegant work, despite the vast
The pernicious amount of saturated fatty acids can be range among GPR120 activators, Oh et al. (2010) established
provided from its excessive intake during de novo lipogenesis a binding rank associated with GPR120 activation. DHA is
converting the excess of carbohydrates and proteins or the most significant fatty acid bound, followed by palmitoleic
4 E. DA SILVA BATISTA ET AL.

Figure 1. Pro-inflammatory signaling transduction disrupted by GPR120 receptor. (A) High amounts of saturated fatty acids activate TLR4 which transduces its
signaling to the intracellular medium coupling the accessory protein MyD88, attracting IRAK1/4 and TRAF6 proteins to form a structural conglomerate. After,
IRAK1 and TRAF6 migrate to TAK1, recruiting Tab1/2 proteins, which phosphorylate TAK1 through ubiquitinases support. (B) Cytokines such as TNFα and IL1β
activate their receptors and intracellular TRAF2 protein conduct the receptor signals until to TAK1 protein, reinforcing the proinflammatory signaling. (C) The
confluence between TLR and cytokine receptors culminates in the canonical signaling mediated by Ikk until NFkB which induces the gene transcription of
several pro-inflammatory targets. (D) The GPR120 receptor recognizes all species of ω3 recruiting the βArr2. The βArr2 trajectory removes both Tab1/2 (dashed
red arrows) from TLR4 and cytokine cascades, disrupting the multiple proinflammatory signalings by blocking the TAK1 phosphorylation (continuous red line).
All 3D molecules are originals from Homo sapiens.

(C16:1 [ω7]), oleic (C18:1 [ω9]), and EPA fatty acids (Oh et al. 2015), adipose tissue (Oliveira et al. 2015), retina
et al. 2010). The demonstration of the inability of saturated (Dátilo et al. 2018), aorta (Moura-Assis et al. 2018), and
fatty acids and ω6 family members to activate GPR120 was hypothalamus (Cintra et al. 2012). In the liver, muscle, and
as important as the discovery itself. The significance of this adipose tissue of obese mice treated with a diet containing
work goes further once they showed detailed downstream ALA, the improvement in metaflammation status was
anti-inflammatory mechanisms coordinated by ω3. In attributed to GPR120 action in all of these tissues (Oliveira
HEK293 cells treated with DHA, the βarr2 (beta-Arrestin et al. 2015). Additionally, there was a significant amelioration
2) protein was immediately attracted to GPR120. During in global glucose homeostasis, followed by an improvement
the βArr2 trajectory to GPR120, βArr2 “arrests” TAB1 and in fasting serum glucose and insulin (Oliveira et al. 2015).
TAB2 proteins from the TAK1 node, disrupting TLR, TNF, In practical terms, due to the similar ability to provide
and IL1 cascades (Oh et al. 2010; Figure 1D). GPR120 activation mediated by both ω3 members (ALA,
Interestingly, ω3 from vegetable oils (ALA [C18:3]) was EPA, and DHA), interest in ALA fatty acid therapeutic use
also able to activate the GPR120 receptor and its down- has been growing (Stivala et al. 2022).
stream signaling (Figure 1D). Cintra et al. (2012) treated GPR40 was deorphanized by Itoh et al. (2003), showing
obese mice for two months with diets containing ALA from several fatty acids as possible ligands, including oleic fatty
flaxseed oil. Systemic low-grade inflammation was controlled acid. Oh et al. (2010) tested a synthetic and specific GPR120
in the central nervous system. The ALA crossed the agonist, GW9508, which also can activate GPR40 due to
blood-brain barrier and reached the hypothalamus. The the 10% shared homology between these receptors. Obese
authors acutely infused purified ALA directly into the rodent mice treated with ω3 or ω9 diets showed a cross-activation
hypothalamus to guarantee that the observed phenomenon between GPR120 and GPR40, probably due to their homol-
was carried out by ALA, but not by some other diet com- ogy. The GPR40 downstream mechanism associated with
pound or metabolic interference. Using immunoprecipitation blocking inflammation is also partially mediated by βArr2
approaches, the authors attributed the effects to the con- (Oliveira et al. 2015). Nevertheless, the action or potency
nection between GPR120 and βarr2 followed by a relation- of these receptors appears to be tissue-specific. Oliveira
ship between βarr2 and TAB1/2 proteins (Cintra et al. 2012). et al. (2015) showed a more pronounced activity of both
The presence and functionality of GPR120 have already GPR120 and GPR40 receptors in skeletal muscle than in
been demonstrated in several tissues, such as the liver (Oh liver or adipose tissue. It appears to be a relevant open
et al. 2010; Oliveira et al. 2015), skeletal muscle (Oliveira avenue for further explorations related to controlling
Critical Reviews in Food Science and Nutrition 5

glucose metabolism and insulin resistance. These receptors the C-terminal GPR120, the inflammatory cascade is dis-
are also relevantly expressed in beta-pancreatic cells (Wu rupted (Figure 1E). Then, βArr2 internalizes GPR120 and
et al. 2021), influencing insulin secretion. Synthetic GPR120 its consecutive agonists (Figure 2A). From now on, ω3 fatty
and GPR40 agonists are currently being tested in clinical acids can follow their classical attributed physiological
randomized controlled trials, together with the actions (Figure 2B), such as retina and myelin sheath coat-
anti-inflammatory exploration hypothesis (Marcinak et al. ing, cell membrane constitution and fluidity, prostaglandin
2017; Mauricio et al. 2017). formation, and many other essentialities (Miles et al. 2004).
Notably, the anti-inflammatory GPR120-dependent mech- Still using GPCR-βarr2 protein aggregation, ω3 fatty acids
anism begins when ω3 is connected to GPR120, which is can interfere with inflammatory signaling by disrupting a
still outside the cell (Oh et al. 2010). When βArr2 binds to system known as the inflammasome (Figure 3).

Figure 2. The ω3 internalization mediated by GPR120 receptor. (A) After binding to GPR120, the ω3 fatty acids are internalized together with their receptor.
This process is mediated by βArr2 which drags the receptor and its ligand into the cell. (B) Once inside the cell, ω3 can assume distinct routes, many of them
also involved in inflammation interruption. For example, the generation of odd series prostaglandins, which counteractive the proinflammatory ω6 prostaglandins
from even series. ω3 molecules could be bioconverted in SPMs mediators such as resolvins, protectins, and maresins that show a powerful action blocking
inflammation. (C) The last pathway shows the ω3 ability in to directly activate the PPARγ, which inactivates the NFkB protein, avoiding its migration to the
nucleus. All 3D molecules are originals from Homo sapiens.

Figure 3. Inflammasome activation. (A) NLRP3 molecules produced from TLR or cytokine cascades are connected to ASC and Casp1 proteins to form the first
part of the inflammasome. (B) These three proteins are oligomerized multiple times until to form a nodal structure. (C) The inflammasome. (D) Once structured,
the inflammasome cleavages the immature IL1 β and IL18 to mature proteins, increasing the inflammatory tonus. All 3D molecules are originals from Homo
sapiens.
6 E. DA SILVA BATISTA ET AL.

The inflammasome disruption mediated by ω3


Inflammasomes are cytoplasmic multiprotein complexes that
respond to PAMPs or DAMPs (pathogen-associated or
damage-associated molecular patterns). Inflammasome struc-
ture formation is essential to maintain or increase inflam-
mation and is responsible for making interleukins IL1β and
IL18 mature, which are pre-translated from NFkB signaling
(Figure 1C). Once mature, IL1β and IL18 are secreted out-
side the cell. Studies have offered a glimpse into the sophis-
ticated mechanisms by which inflammasomes respond to
danger and secretion of IL1β and IL18 and the therapeutic
ω3 capacity (Martínez-Micaelo et al. 2016; Mortimer et al.
2016; Shen et al. 2017; Williams-Bey et al. 2014; Yan et al.
2013). PAMPS, such as LPS (lipopolysaccharides), activates
TLR4 and NFkB canonical signaling, which transcribes sev-
eral proinflammatory-related genes, such as TNFα, IL1β,
IL6, IL18, iNOS, COX2, NLRP3 (NACHT, LRR, and PYD
Domains-Containing Protein 3), and the anti-inflammatory
IL10 (Liu et al. 2017; Figure 1C). When the inflammatory
tonus is increased, NLRP3 is recruited to bind to ASC
(apoptosis-associated speck-like CARD-domain containing
protein) and Casp1 (Caspase 1) proteins, assembling the Figure 4. GPR120 activation elicits the inflammasome disruption. (A) GPR120
anti-inflammatory response involves βArr2 activation which sequesters NLRP3
first inflammasome masterpiece (Agostini et al. 2004; proteins (red arrows) disrupting inflammasome structure, reducing a matured
Compan et al. 2015; Figure 3A). The NLRP3-ASC-Casp1 form of IL1β and IL18 (B). (C) GPR120/βArr2 axis blocks NFkB activity (dashed
aggregate undergoes subsequent oligomerization (Figure 3B) red line - βArr2 blocks NFkB in an indirect form) which is the transcriptional
to form the final inflammasome complex (Figure 3C), which factor responsible for NLRP3 gene expression. All 3D molecules are originals
begins the IL1β and IL18 cleavages (Figure 3D). Both inter- from Homo sapiens.
leukins can reach the bloodstream or through autocrine
signaling, contributing to redundant or perpetual inflamma- hepatic NLRP3 inflammasome components with insulin resis-
tory signaling (Agostini et al. 2004; Compan et al. 2015). tance, inflammation, or morphological damage in the liver of
The ω3 fatty acids partially inhibit inflammasome acti- patients (Quezada et al. 2021). Notably, these authors carried
vation by disarming the original signaling from the saturated out an indirect inflammasome activity determination. In indi-
fat/LPS-TLR4 cascade, which decreases NFkB and its prod- viduals severely obese (BMI > 50 kg/m2) and with NASH
uct, NLRP3 (Martínez-Micaelo et al. 2016; Figure 4). (nonalcoholic steatohepatitis), NLRP3 inflammasome activation
Interestingly, ω3 could interfere differently with the inflam- was observed in hepatic tissue (Wree et al. 2014).
masome structure. Inhibition of NLRP3 and consequently Increasing evidence suggests that ω3 PUFAs can poten-
Casp1 activation was shown in different models through tially decrease the activity of the NLRP3 inflammasome in
GPR120/GPR40-βArr2 binding to NLRP3 (Mo et al. 2020; many types of cells, such as macrophages, hepatocytes, and
Williams-Bey et al. 2014; Yan et al. 2013). βArr2 sequesters adipocytes (Martínez-Micaelo et al. 2016; Mortimer et al.
NLRP3 from the inflammasome structure and disassembles 2016; Shen et al. 2017; Yan et al. 2013). The reduction in
the entire structure (Figure 4A). Willians-Bey et al. (2014) inflammasome signaling was shown in a recent study in
demonstrated that GPR120 rather than GPR40 is the relevant which Lovaza®(3,6 g of EPA and DHA) treatment for 8 weeks
ω3 receptor involved in limiting inflammation and likely in obese humans reduced NLRP3 inflammasome and Casp1,
inflammasome activation in mouse (bone marrow-derived IL1β, and IL18 gene expression in adipose tissue and cir-
macrophages) and human (THP1) macrophages. In addition, culating levels of IL18 (Lee, Midgette, and Shah 2019). In
the deletion of βArr2 partially inhibited ω3 effects, suggest- a randomized controlled trial (Satoh et al. 2014), 60 patients
ing that other mechanisms independent of GPR120/40 and with coronary artery disease (CAD) showed NLRP3 inflam-
βArr2 might be involved in inhibiting the activation of the masome levels (gene expression measured in mononuclear
NLRP3 inflammasome (Yan et al. 2013). PBMC cells) higher than in the non-CAD group, with a
The NLRP3 inflammasome and IL1β have been linked to higher correlation between NLRP3 and IL1β and IL18 in
obesity and its metabolic complications in murine models the bloodstream. Patients who randomly received 8 months
induced by a high-fat diet (Vandanmagsar et al. 2011; Youm of treatment with atorvastatin or rosuvastatin, showed sig-
et al. 2011) and obese humans (Quezada et al. 2021; Unamuno nificantly reduced NLRP3 levels. Despite pharmacogenomics
et al. 2021). Recently, Unamuno et al. (2021) found that the showing new molecular pathways for FDA-approved drugs,
NLRP3 inflammasome was strikingly involved in IL1β pro- the nutrigenomics perspective of disrupting the inflammaso-
duction in adipose tissue. Quezada et al. (2021) did not support mal structure mediated by a nutrient is an exciting and
an increase in the hepatic levels of NLRP3 components in novel possibility to be tested in several chronic diseases with
patients with cardiometabolic risk. They did not correlate low-grade inflammation as a background (Satoh et al. 2014).
Critical Reviews in Food Science and Nutrition 7

Intracellular ω3 anti-inflammatory signaling homeostasis control (Tanaka et al. 2020), cardiovascular


functions (Tanaka et al. 2020), energy expenditure (Risérus
The anti-inflammatory mechanism mediated by GPCRs et al. 2008), liver lipid metabolism in nonalcoholic steato-
induced by ω3 is strongly involved in the ability of βArr2 hepatitis (Francque et al. 2021, Lefere et al. 2020), modu-
to sequester TAB1/2 and NLRP3 with ω3 still outside the lation of innate immunity (Lefere et al. 2020), and NFkB
cell. GPCRs (GPR120 and GPR40) contribute to ω3 inter- signaling blockade (Zhao, Wang, and Guo 2016; Figures 2C
nalization, although there are many other mechanisms of and 6A), is not surprising. It is also attributed to fatty acid
ω3 uptake (Oh et al. 2010; Oliveira et al. 2015). Different PPAR modulation, which is widely proven to affect gene
membrane receptors and mechanisms (Figure 5) are involved expression and protein content; however, it is early to alle-
in ω3 cell absorption, such as the flip-flop hypothesis viate direct PPAR agonism by fatty acids.
(Gurtovenko, Vattulainen 2007), FABP (Fatty Acid-Binding In the studies from Kliewer et al. (1997), Magee et al.
Protein) (Liu et al. 2010), FATP (Fatty Acid Transporter (2012), and Yamada et al. (2014), luciferase assays were
Protein-1) (Ochiai et al. 2017), CD36 (Cluster of carried out to suggest the ω3-PPARγ connection and its
Differentiation 36) or synonymously SR-B2 (Scavenger downstream effects. Magee et al. (2012) tested EPA
Receptor B2) (29), and MFSD2A (Major Facilitator anti-inflammatory action in TNFα-injured C2C12 muscle
Superfamily Domain-Containing 2 A) (Duc, Kim, and Chung cells, suggesting a direct EPA effect on PPARγ blocking
2015.). Both FABP and FATP are superfamily receptors; NFkB signaling. Interestingly, Yamada et al. (2014) showed
however, the high ω3 affinity/specificity is related to FABP2 that both EPA and its metabolites are PPARγ agonists, rein-
and FATP1, respectively (Liu et al. 2010, Pishva et al. 2012). forcing the previous data that PPARγ is a relevant NFkB
Once inside the cell, ω3 could exhibit different ways to blocker. Under ALOX enzyme action (ALOX5, 12, and 15),
interfere with proinflammatory signaling through its protein EPA is derived in several leukotrienes from the HEPE
interaction or after its metabolic derivation. (Hydroxy Eicosapentaenoic Acid) family (Schulze et al.
Unlike protein-protein interactions, understanding the 2020). In this work, all tested HEPEs (HEPE5, HEPE8,
lipid-protein connection remains methodologically challeng- HEPE9, HEPE12, and HEPE18) were able to induce PPARγ
ing, relying only on indirect tests. The “lipidation” is chal- activation; however, HEPE8 and HEPE9 were the most
lenging to visualize, and all tests are estimative or potent EPA-derived metabolite PPARγ activators (Yamada
circumcircles. It is attributed to ω3 fatty acids and direct et al. 2014; Figure 6A).
protein connections, which are responsible for its intracel- The ω3 fatty acids can follow different intracellular met-
lular anti-inflammatory activities, representing ω3 nonge- abolic routes tracking specific enzyme systems. For example,
nomic signaling. the hydroxy-lipidic mediators derived from EPA and DHA
after oxidative metabolisms in the P450 system, such as
17S-hydroxyDHA (17-HDHA) (González‐Périz et al. 2006;
The anti-inflammatory ω3 actions mediated by Neuhofer et al. 2013) and protectin D1, can also act as
PPARγ PPARγ ligands (González‐Périz et al. 2006). However, other
metabolites derived from ω3 fatty acids can use different
Peroxisome proliferator-activated receptors (PPARs) are anti-inflammatory strategies independently of PPARγ acti-
ligand-activated transcription factors of the nuclear hormone vation. From the P450 system or ALOX enzymes, resolvins,
receptor superfamily, and PPAR-gamma (PPARγ) is the cen- protectins, and maresins configure a new species of potent
tral transcriptional regulator of lipid metabolism. Thus, the ω3-derived anti-inflammatory metabolites using different
broad spectrum of PPARγ action, such as glucose mechanisms (Figure 6B).

Figure 5. Intracellular ω3 uptake. Several ω3 receptors were described to promote its absorption. (A) GPCRs (GPR120 and GPR40), (B) FATP1 (fatty acid trans-
porter protein-1), (C) FABP2 (fatty acid-binding protein-2). ω3 internalization involves different membrane receptors and mechanisms, including (D) CD36 (cluster
of differentiation 36), (E) MFSD2A (major facilitator superfamily domain-containing 2A), and the flip-flop mechanism.
8 E. DA SILVA BATISTA ET AL.

Figure 6. The anti-inflammatory action of ω3-derived compounds. (A) EPA could be derived in several HEPE (hydroxy eicosapentaenoic acid) compounds. All
of them appear to have anti-inflammatory potentials, however, the HEPE8 and HEPE9 show described mechanisms of action, which activate PPARγ, direct (red
line) blocking NFkB, including EPA proper. (B) During the ω3 elongation and desaturation cascade, from EPA and DHA several new molecules can be derived.
EPA could be derived in odd series prostaglandins from the COX2 (Cyclooxygenase 2) enzyme. From P450 proteins (Cyp450), EPA can be derived in 18-hydroxy
eicosapentaenoic acid and resolvins (RvE1/E2). Also, three lipoxygenases could derivates EPA in active metabolites such as lipoxins, from lipoxygenases 12 and
15 (ALOX12/15), and leukotrienes of odd series, from lipoxygenase 5 (ALOX5). DHA could be derived in 14-HpDHA (14-hydroperoxide intermediate DHA) and
maresins MaR1/2 by lipoxygenase 12 (ALOX12) and 17-HpDHA (17-hydroperoxide intermediate DHA) and protectins (PD1 and NPD1) from lipoxygenase 15
(ALOX15). 17-hydroperoxide intermediate DHA could be converted into hydroxy docosahexaenoic acid by ALOX5. (C) Membrane receptor FPR2 recognizes RvD1
which blocks (dashed red lines) the pro-inflammatory signaling in the three hypothesized targets: IRAK1, TRAF6, and IkBα. (D) A GPCR receptor GPR32 which
also recognizes the RvD1, recruiting βArr2 that disarms proinflammatory cascades mediated by TLR4 and cytokine receptors. All 3D molecules are originals
from Homo sapiens.

Resolvins, protectins, and maresins anti-inflammatory role. From ALOX5 metabolism, resolvins
can also be generated from DHA (RvD1 to RvD6) (Schulze
After internalization, some ω3 molecules are metabolized et al. 2020). Serhan et al. (2002) first described the exis-
into an extensive range of substances, as previously tence and activity of resolvins. Since this, different ω3
described, such as prostaglandins and leukotrienes, derived metabolites have been described by Serhan’s group. In
from COX and ALOX enzymes (Schulze et al. 2020; 2003, they identified the first member of a new category
Figures 2B and 6A). Other ω3-EPA molecules are derived of substances derived from DHA, the “protectins,” repre-
in epoxyns by the P450 system, called E-resolvins 1/2 sented by the 10,17S-docosatrienoic acid, which was later
(RvE1 and RvE2) by hydroxylases (CYP4A and CYP4F) renamed neuroprotectin D1 (NPD1) (Mukherjee et al.
(Schulze et al. 2020), which shows a powerful 2004). In 2004, NPD1 was isolated, identified, and
Critical Reviews in Food Science and Nutrition 9

characterized as derived from DHA after ALOX15 metab-


olism, including protectin D1 (PD1), another essential
member of the protectin class (Hansen, Dalli, and Serhan
2017). Last, in 2009, maresins (MaR) derived from DHA
under ALOX12 metabolism were identified to have sig-
nificant anti-inflammatory properties in macrophages
(Serhan et al. 2009). This metabolic route was evidenced
in macrophages, which gave meaning to the maresin
nomenclature “macrophages mediators in resolving inflam-
mation” (Serhan et al. 2009). Together, all resolvins, pro-
tectins, and maresins are collectively termed specialized
pro-resolving mediators (Serhan 2017) (Figure 6B).
Several mechanisms of action have been proposed to
explain the anti-inflammatory role attributed to each
pro-resolving mediator. Until now, the primary highlighted
mechanism involves the interaction between resolvins and
the ALX/FPR2 (Lipoxin A4 Receptor/Formyl Peptide
Receptor 2) receptor, a GPCR member, at the 3rd extracel-
Figure 7. The macrophage profile is controlled by ω3 and its derived com-
lular loop (Bena et al. 2012). Recently, after FPR2 receptor pounds. During the proinflammatory stage, the immune cell release several
activation by RvD1, the receptor probably interacts with proteins such as TNFα, IL1β, iNOS, NLRP3, and many others, including the
IRAK1 or TRAF6 proteins, blocking the signal before reach- inflammasome assembling. (A) Protectins such as NPD1 (neuroprotection D1)
ing the NFkB transcription factor (Liu et al. 2020; Figure bind in the GPR37 receptor, which induces the gene transcription of the potent
anti-inflammatory interleukin, IL10. (B) Once inside the cell, the maresin “MaR1”
6C). It is very fascinating and biologically relevant to the
migrates to the nucleus binding to the nuclear receptor RORE (ROR-[retinoic
explicit demonstration of the mechanism of action. Although acid-related orphan receptor alpha (RORα)] Response Element) that also induces
in this work (Liu et al. 2020) the authors were relatively in the IL10 gene transcription. (C) Once activated by ω3, βArr2 recruited and
doubt between IRAK1 and TRAF6 proteins as a FPR2 target, activated by GPR120 block several proinflammatory proteins (dash red line –
IRAK1/TRAF6 work side-by-side, with the spectrum of indirectly through TAK1 inhibition), direct (continuous red line) disarm inflam-
masome, indirect (blue dashed lines) activate the anti-inflammatory proteins
action being very delimited. A specific nutrient can act on
such as arginase 1 Arg1), chemokine 13 (CXCL13), transforming growth
multiple protein targets. Thus, Wang et al. (Wang et al. factor-beta (TGFβ) and IL10. All these actions induce the anti-inflammatory
2011) showed the ability of RvD1 to reduce IkBα degrada- profile adopted by macrophages, resolving the inflammation. All 3D molecules
tion. IkBα acts as an anchor for the P50/P65 (NFkB) protein, are originals from Homo sapiens.
and its degradation allows continuous proinflammatory sig-
naling, releasing NFkB to the nucleus (Figure 6C). Thus, prognosis characterized by seizures (Frigerio et al. 2018).
RvD1 appears to act as an anti-inflammatory substance Despite PD1 controlling the inflammation and IL1β protein
working at different points in the same pathway, highlighting content (Frigerio et al. 2018), the authors (Mikroulis et al.
its potential. To reinforce this point of view, Krishnamoorthy 2022) noticed that the seizures were contained in an inde-
et al. (2010) identified GPR32 as another RvD1 recognizer pendent form of inflammation, suggesting different signalings
that uses the intracellular βArr2 protein to transduce part coordinating different phenomena. There are no identified
of its signal (34) (Figure 6D). As mentioned before, βArr2 PD1 receptors, but in this work, treatment with a Gi inhibitor
mediates the anti-inflammatory response induced by GPR120, completely abolished the neuroprotective PD1 action, sug-
an ω3 receptor, amplifying the RvD1 anti-inflammatory gesting a GPCR as its receptor (Mikroulis et al. 2022).
specter (DeMar et al. 2006). Although maresins appear to act as potent protective medi-
Receptors for protectins are still being described. GPR37 ators of macrophage function, promoting acute inflammation
is considered an orphan receptor; however, its affinity for resolution and tissue regeneration (Hwang et al. 2019), there
protectin NPD1 was recently postulated as a possible medi- are no well-established mechanisms. Recent and exciting dis-
ator of several NPD1 properties, mainly anti-inflammation coveries shine a light on MaR1 pathway possibilities. After
(Bang et al. 2014) (Figure 7A). Bang and colleagues reported intracellular generation, some MaR1 molecules migrate to the
that GPR37, expressed by macrophages but not microglia, nucleus and activate a nuclear retinoic acid-related orphan
contributes to the resolution of inflammatory pain. The receptor alpha (RORα), binding to the RORE (ROR-Response
absence of GPR37 makes macrophages unable to produce Element), which drives the complex (MaR1-RORα) to gene
IL10, even under NPD1 stimuli, and the lagging macrophagic target regions (Han et al. 2019; Figure 7B). The transcript
transition from a pro- to anti-inflammatory profile delays products are related to anti-inflammatory proteins such as
tissue repair (Bang et al. 2014). In the recent work published IL10 and TGFβ (transforming growth factor-beta) (Han et al.
by Mikroulis et al. (2022), a gripping PD1 action was 2019). MaR1 molecules are also released from cells, and LGR6
described as regulating the seizures induced by high GABA (Leucine-Rich Repeat-Containing G Protein-Coupled Receptor
neuron excitability (Mikroulis et al. 2022). During epilepsy 6) was recently described as its receptor. LGR6 is widely
pathogenesis, neuroinflammation is increased, and IL1β and expressed in various mouse and human tissues and induces
TNFα are the ictogenic cytokines, leading to the worst phagocytosis, efferocytosis, chemotaxis, and a distinct
10 E. DA SILVA BATISTA ET AL.

phosphorylation pathway coordinated by ERK (Extracellular the ablation of ω3 action after treatment, marked by non-
Signal-Regulated Kinase) (Chiang et al. 2019). Chiang et al. transition between pro- and anti-inflammatory macrophage
also demonstrated the ability of MaR1 to recruit the βArr2 profiles (Raptis et al. 2014)). A similar observation was
protein to the LGR6 base, evoking possible anti-inflammatory noticed by Oliveira et al. (2015) when liver steatohepatitis
MaR1 effects through the βArr2 downstream cascade (Chiang was restored in obese and insulin-resistant mice after flax-
et al. 2019). seed oil diet treatment. GPR120 was identified on the
In a proinflammatory context, MaR1 appears to firmly hepatocyte and Kupffer cell surfaces and bound to the βArr2
resolve it due to induced macrophage polarization to a more protein after treatment (Oliveira et al. 2015). In another
anti-inflammatory profile (CD11c-CD206+). Thus, MaR1 is experiment, both human macrophages (THP1) derived from
an indirect IL10- and TGFβ-inducible factor secreted by foam cells and lineage (Raw 264.7) were treated with a
CD206+ macrophages (Chiang et al. 2019). In a NASH GPR120-synthetic agonist (GW9508), and showed increased
model, Han et al. (2019) showed that the improvement in expression of ABCA1 and ABCG1 (ATP-binding cassette
liver status was not provided by DHA activating GPR120 transporters), increasing cholesterol efflux after GPR120
but through its derived, MaR1, preventing the progression activation (Ann et al. 2020). In obese and knockout LDLr
of high-fat diet-induced NASH in a RORα-dependent man- (low-density lipoprotein receptor) mouse models, Moura-Assis
ner. Thus, the confluence between extra and intracellular et al. (2018) identified GPR120 in endothelial cells and on
signaling mediated by MaR1 composes significant the surface of infiltrating macrophages in the intimal layer
anti-inflammatory resources. of the aortic wall (Moura-Assis et al. 2018). This last dis-
The evidence of these lipid mediators in humans is still covery is fascinating once the cardiovascular benefits attrib-
scarce, considering the methodological difficulties in access- utable to ω3 fatty acids, since its first demonstration
ing the measurements of resolvins, protectins, and maresins. (Dyerberg, Bang 1979), could be induced by different mech-
Some randomized and controlled studies have shown the anisms, such as switching the macrophage profile (Figure 7).
bioconversion process from ω3 food sources. Polus et al. There is still a wide range of miscellaneous signaling
(2016) observed that a three-month supplementation with pathways associated with the anti-inflammatory effects of
1.8 g ω3 (EPA and DHA) in obese women significantly ω3 in an attempt to orchestrate macrophages. Interestingly,
increased plasma RvD1 and RvD2. Mas et al. (2016) showed many of the above-described mechanisms could also interact
in a double-blind, placebo-controlled intervention that the and be counteractive in the cells. For example, GPR120 has
8 weeks of 4 g/d (1840 mg EPA and 1520 mg DHA) ω3 sup- a close relationship with PPARγ, and knowledge of this
plementation in patients with chronic kidney disease interaction is another crucial issue for better understanding
increased the plasma levels of RvD1 and the pathway pre- the complex metabolic reactions that appear to modulate.
cursors 18-HEPE (E-series resolvin from EPA) and 17-HDHA GPR120 is a direct PPARγ target gene in adipocytes, but
(D-series resolvin from DHA). In obese and insulin-resistant not in macrophages, and its induction by rosiglitazone
mice supplemented with flaxseed oil (ALA – C18:3 source), enables thiazolidinediones to potentiate the effects of the
the EPA, DHA, resolvin RvE1, RvE2, RvD2, and RvD6 levels GPR120 agonist. In addition, PPARγ activity is maintained
were increased, regulating the proinflammatory IL2, TNFα by activated GPR120, which increases the production of
and IFNγ cytokines and upregulating IL4 and IL10 produc- 15dPGJ2 and blocks ERK, a PPARγ inhibitor (Paschoal et al.
tion by macrophages from adipose tissue (Bashir et al. 2019). 2020). Not only marine ω3 species (EPA and DHA) act at
these proposed molecular points. Kumar et al. (2016) showed
that the ALOX15 ALA metabolites (13-S-HPOTrE and
The macrophage as the conductor 13-S-HOTrE) are natural PPARγ ligands, leading to the
From the nutritional point of view, ω3 fatty acids have been blocking of NLRP3 and the inflammasome, followed by its
described for a long time as the leading macrophage action downstream signaling molecules, such as Casp1 and IL1β
influencer (Leslie et al. 1985; Spencer et al. 2013). Several (Kumar et al. 2016).
reasons are attributable to this, such as the individual nutri-
tional status related to the ω6:ω3 ratio, the intracellular ω3 is not a panacea: adverse effects of ω3
macrophage signaling mediated by PPARγ among other
supplementation
molecules, and pro-resolvins, as mentioned above. However,
during the last decade, GPR120 has been described in sev- There is a high interest of all members of society (citizens,
eral studies as being expressed on the surface of macro- public health administrators, pharmaceutical companies, etc.)
phages (Figure 7C). Interestingly, GPR120 is expressed on in finding strategies to control inflammation. Both levels (high
the surface of tissue cells and immune cells (macrophages or low) of inflammation are the basis for almost all disease
and dendritic cells) (Feng et al. 2021). development. As shown, ω3 fatty acids have many actions,
For the first time, Raptis et al. (2014) showed GPR120 counteracting inflammation in a multipoint strategy. Then, if
expressed on the Kupffer cell surface in mice under hepatic ω3 is so potent, why is it still not considered in the leading
ischemia-reperfusion injury, and an exaggerated inflamma- international guidelines? Many questions could be detailed,
tory response was attenuated after Omegaven® (parenteral such as the absence of recommended dose of consumption,
ω3 nutrition) treatment. Once GPR120 was identified as a the quality of the oil from capsules, the ideal blend between
possible effector protein, its gene silencing (siRNA) showed EPA: DHA or its isolated consumption, the ALA bioconversion
Critical Reviews in Food Science and Nutrition 11

to longer ω3 species, the FADS (fatty acid desaturase) muta- this possibility, as ω3 is generally safe and well-tolerated but
tion dispersion among populations, the quality of experimental not free of adverse effects.
and clinical studies design, the compliance of the study
funders, and because it is a nutrient, not a medication.
Notwithstanding, some side effects need to be considered. Limitations
There is no established UL (Upper Level) for ω3 intake.
Still are several limitations related to the ω3 mechanisms
It is assumed that supplementation in clinical practice is safe
of action and the applicability of findings in humans. Lipids
due to the literature’s scarce evidence of toxic effects. On the
are non-crystallizable substances, which turns impossible
other hand, not all intervention studies describe adverse or
until now, the optical methods to binding studies. The cur-
side effects. Most clinical trials do not prepare concise or
rent methods in the literature only surround lipid action
reasonable reports showing adverse events (Abdelhamid et al.
measurements. The incorrect placebo adoption in studies
2018, Alvarez Campano et al. 2019). The most commonly
with cells, mice, and humans promote several biases. The
reported adverse event was gastrointestinal upset (Downie
results found in studies using ω6 sources (safflower, sun-
et al. 2019; Poreba et al. 2017). A significant concern with
flower, and corn oil) can overestimate the ω3 benefits, and
ω3 supplementation would be the bleeding risk (Dyerberg,
ω9 (olive oil) can underestimate it. Undisputedly, the wrong
Bang 1979). However, it is still unknown whether the anti-
dose definitions are a bottleneck in this area. Many studies,
platelet effect per se could be transposed into increased clin-
using large cohorts claim the benefits of using, for example,
ical risk for patients (Bagger et al. 2020; Jeansen et al. 2018).
4 grams of ω3. However, they used 4 capsules with 1 gram
In the clinical trials with large sample sizes such as JELIS
of oil each, but less than 2 grams of ω3 fatty acid in the
(Yokoyama et al. 2007) and REDUCE-IT (Bhatt et al. 2019b)
total. The fatty acid profile determination previously to the
studies, increased bleeding was observed using EPA alone
experimentations needs to be obligatory. When the study
over approximately 5 years. In JELIS, 1.8 g of EPA was tested
hits this aim, it might fail on non-checked adulterated prod-
in Japanese patients with hypercholesterolemia using statins
ucts; frequently found on the market. Science must overcome
(Yokoyama et al. 2007). In the REDUCE-IT study, patients
these questions to promote more realistic practice applica-
with elevated triglycerides on statins received 4 g/d of highly
tions based on these interesting molecular mechanisms find-
purified EPA ethyl ester (icosapent ethyl) (Bhatt et al.
ings. Finally, the gene variations associated with GPR120,
2019a). In the REDUCE-IT study (Bhatt et al. 2019a), there
FADS, and other genes participating in omega-3 metabolism
are reports that ω3 supplementation could cause atrial fibril-
will need to be investigated to understand if there are prone
lation, which was also achieved in the STRENGTH trial
or less responsive people to omega-3 fatty acids usage.
(Nicholls et al. 2020). STRENGTH was conducted in 13.078
patients receiving statins and 4 g/d of ω3 carboxylic acid
(75% EPA and DHA) over a 3.2-year average follow-up Conclusion
(Nicholls et al. 2020). Unexpectedly, no change in the risk
of atrial fibrillation was observed in the VITAL-AF study, The molecular sciences provided new exploratory instru-
which used 840 mg/d of marine ω3 fatty acids (EPA-DHA; mentals that enabled inside-the-cell pathway understanding.
1.2:1 ratio) for 5.3 years, carried out in patients with no The most explored nutrient in nutritional sciences now has
prior history of cardiovascular disease (Albert et al. 2021). a plethora of mechanistic possibilities. By targeting receptors
These findings suggest that the risk of atrial fibrillation is on the surface cells, managing macrophage polarization,
higher with increasing ω3 dose, especially EPA. disrupting the multiple proinflammatory intracellular cas-
Given the high natural amount of DHA in neurons, it is cades, or disassembling the inflammasome structure on both
hypothesized that its supplementation would have a neuro- tissue and immune cells, omega-3 fatty acids show their
protective effect. However, the maxim of “the greater, the impressive versatile action. Nonetheless, it is necessary to
better” does not apply in this case either. The opposite has understand why this enormous anti-inflammatory signal
been observed as a worsening cognitive function in the convergence is not translated into clinical significance. The
elderly (Danthiir et al. 2018; Naderali E, Naderali M-M, nutrigenomic approaches still have a long way to determine
Abubakari 2014) and throughout the life of animals’ off- the omega-3 ideal dose, treatment duration, the balance
spring supplemented with ω3 in pregnancy and lactation, between omega-3 and omega-6 ingestion, and tissue-specific
as well as a reduced life expectancy (Church et al. 2008; bioconversion from short (ALA) to long-chain species (EPA
Church et al. 2010). In an animal model for amyotrophic and DHA). Regardless, the science surrounding omega-3
lateral sclerosis, worsening of brain cell damage was observed anti-inflammatory investigation leaves a legacy with new
with EPA supplementation (Yip et al. 2013). The association proposed targets that enable other nutrients or even drugs
between ω3 supplementation and ethanol intake in animals to be intelligently designed.
worsened nonalcoholic fatty liver disease through an
increased inflammatory response by reducing IL4 and
anti-inflammatory macrophages (Feng et al. 2021; Li et al. Acknowledgments
2017). Although still not established, it seems necessary to We thank the Research Collaboratory for Structural Bioinformatics –
define an ω3 intake limit beyond which the beneficial capac- Protein Data Bank (RCSB-PDB) and AlphaFold Protein Structure
ity of these fatty acids would be extrapolated, leading to Database for the free use of tridimensional protein structures. We also
adverse effects. Further clinical trials are needed to address thank all the members of the Nutritional Genomics Lab for their helpful
12 E. DA SILVA BATISTA ET AL.

discussions. EAC thanks to CNPq (Brazilian National Council for pain. The Journal of Clinical Investigation 128 (8):3568–82. doi:
Scientific and Technological Development) for the scientific productivity 10.1172/JCI99888.
fellowship (CNPq: 315369/2021-3). S.C.B.R.N and D.E.C thanks to “São Barceló-Coblijn, G, and E. J. Murphy. 2009. Alpha-linolenic acid and
Paulo Research Foundation – FAPESP,” by grants 2020/13443-1, its conversion to longer chain n-3 fatty acids: Benefits for human
2019/13168-3 and 2019/13210-0, and CNPq: 312970/2022-6, which sup- health and a role in maintaining tissue n-3 fatty acid levels. Progress
port all scientific investigations at the laboratory. in Lipid Research 48 (6):355–74. doi: 10.1016/j.plipres.2009.07.002.
Bashir, S., Y. Sharma, D. Jairajpuri, F. Rashid, M. Nematullah, and F.
Khan. 2019. Alteration of adipose tissue immune cell milieu towards
the suppression of inflammation in high fat diet fed mice by flax-
Disclosure statement seed oil supplementation. PLoS One 14 (10):e0223070. doi: 10.1371/
The authors are not aware of any affiliations, memberships, funding, journal.pone.0223070.
or financial holdings that might be perceived as affecting the objectivity Bena, S., V. Brancaleone, J. M. Wang, M. Perretti, and R. J. Flower.
of this review. 2012. Annexin A1 interaction with the FPR2/ALX receptor:
Identification of distinct domains and downstream associated sig-
naling. The Journal of Biological Chemistry 287 (29):24690–7. doi:
10.1074/jbc.M112.377101.
Funding Bhatt, D. L., P. G. Steg, M. Miller, E. A. Brinton, T. A. Jacobson, S.
B. Ketchum, R. T. Doyle, Jr, R. A. Juliano, L. Jiao, C. Granowitz,
The author(s) reported there is no funding associated with the work
et al. 2019a. Cardiovascular risk reduction with icosapent ethyl for
featured in this article.
hypertriglyceridemia. The New England Journal of Medicine 380
(1):11–22. doi: 10.1056/NEJMoa1812792.
Bhatt, D. L., P. G. Steg, M. Miller, E. A. Brinton, T. A. Jacobson, S.
References B. Ketchum, R. T. Doyle, Jr, R. A. Juliano, L. Jiao, C. Granowitz,
REDUCE-IT Investigators, et al. 2019b. Effects of icosapent ethyl
Abdelhamid, A. S., T. J. Brown, J. S. Brainard, P. Biswas, G. C. Thorpe, on total ischemic events: From REDUCE-IT. Journal of the American
H. J. Moore, K. H. Deane, F. K. AlAbdulghafoor, C. D. Summerbell, College of Cardiology 73 (22):2791–802. doi: 10.1016/j.jacc.2019.02.032.
H. V. Worthington, et al. 2018. Omega-3 fatty acids for the prima- Chen, Y., Z. Wu, S. Huang, X. Wang, S. He, L. Liu, Y. Hu, L. Chen,
ry and secondary prevention of cardiovascular disease. Cochrane P. Chen, S. Liu, et al. 2022. Adipocyte IRE1α promotes PGC1α
Database of Systematic Reviews 7 (7):CD003177. doi: mRNA decay and restrains adaptive thermogenesis. Nature
10.1002/14651858.CD003177.pub4. Metabolism 4 (9):1166–84. doi: 10.1038/s42255-022-00631-8.
Adan, Y., K. Shibata, M. Sato, I. Ikeda, and K. Imaizum. 1999. Effects Chiang, N., S. Libreros, P. C. Norris, X. D. Rosa, and C. N. Serhan.
of docosahexaenoic and eicosapentaenoic acid on lipid metabolism, 2019. Maresin 1 activates LGR6 receptor promoting phagocyte im-
eicosanoid production, platelet aggregation and atherosclerosis in munoresolvent functions. The Journal of Clinical Investigation 129
hypercholesterolemic rats. Bioscience, Biotechnology, and Biochemistry (12):5294–311. doi: 10.1172/JCI129448.
63 (1):111–9. doi: 10.1271/bbb.63.111. Church, M. W., K.-L. C. Jen, J. I. Anumba, D. A. Jackson, B. R. Adams,
Adhikari, A., M. Xu, and Z. J. Chen. 2007. Ubiquitin-mediated acti- and J. W. Hotra. 2010. Excess omega-3 fatty acid consumption by
vation of TAK1 and IKK. Oncogene. 26 (22):3214–26. doi: 10.1038/ mothers during pregnancy and lactation caused shorter life span
sj.onc.1210413. and abnormal ABRs in old adult offspring. Neurotoxicology and
Agostini, L., F. Martinon, K. Burns, M. F. McDermott, P. N. Hawkins, Teratology 32 (2):171–81. doi: 10.1016/j.ntt.2009.09.006.
and J. Tschopp. 2004. NALP3 forms an IL-1β-processing inflam- Cintra, D. E. C., A. V. Costa, C. G. Peluzio M do, S. L. P. Matta, M. T.
masome with increased activity in muckle-wells autoinflammatory. C. Silva, and N. M. B. Costa. 2006. Lipid profile of rats fed high-fat
Immunity. 20 (3):319–25. doi: 10.1016/s1074-7613(04)00046-9. diets based on flaxseed, peanut, trout, or chicken skin. Nutrition
Albert, C. M., N. R. Cook, J. Pester, M. V. Moorthy, C. Ridge, J. S. (Burbank, Los Angeles County, Calif.) 22 (2):197–205. doi: 10.1016/j.
Danik, B. Gencer, H. K. Siddiqi, C. Ng, H. Gibson, et al. 2021. nut.2005.09.003.
Effect of marine omega-3 fatty acid and vitamin D supplementation Cintra, D. E., E. R. Ropelle, J. C. Moraes, J. R. Pauli, J. Morari, C. T.
on incident atrial fibrillation: A randomized clinical trial. JAMA Souza, R. Grimaldi, M. Stahl, J. B. Carvalheira, M. J. Saad, et al. 2012.
325 (11):1061–73. doi: 10.1001/jama.2021.1489. Unsaturated fatty acids revert diet-induced hypothalamic inflammation
Alvarez Campano, C. G., M. J. Macleod, L. Aucott, and F. Thies. 2019. in obesity. PloS One 7 (1):e30571. doi: 10.1371/journal.pone.0030571.
Marine-derived n-3 fatty acids therapy for stroke. Cochrane Database Compan, V., F. Martín-Sánchez, A. Baroja-Mazo, G. López-Castejón,
Systematic Reviews 26 (6):CD012815. A. I. Gomez, A. Verkhratsky, D. Brough, and P. Pelegrín. 2015.
An, J. U., S. E. Kim, and D. K. Oh. 2021. Molecular insights into lip- Apoptosis-associated speck-like protein containing a CARD forms
oxygenases for biocatalytic synthesis of diverse lipid mediators. specks but does not activate caspase-1 in the absence of NLRP3
Progress in Lipid Research 83:101110. doi: 10.1016/j.plipres.2021.101110. during macrophage swelling. Journal of Immunology (Baltimore, Md.:
An, T., X. Zhang, H. Li, L. Dou, X. Huang, Y. Man, X. Zhang, T. 1950) 194 (3):1261–73. doi: 10.4049/jimmunol.1301676.
Shen, G. Li, J. Li, et al. 2020. GPR120 facilitates cholesterol efflux Croset, M., A. Sala, G. Folco, and M. Lagarde. 1988. Inhibition by
in macrophages through activation of AMPK signaling pathway. The lipoxygenase products of TXA2-like responses of platelets and vas-
FEBS Journal 287 (23):5080–95. doi: 10.1111/febs.15310. cular smooth muscle. Biochemical Pharmacology 37 (7):1275–80.
Arterburn, L. M., E. B. Hall, and H. Oken. 2006. Distribution, inter- doi: 10.1016/0006-2952(88)90782-4.
conversion, and dose response of n − 3 fatty acids in humans. The Daak, A. A., A. Y. Elderdery, L. M. Elbashir, K. Mariniello, J. Mills,
American Journal of Clinical Nutrition 83 (Suppl 6):1467S–76S. doi: G. Scarlett, M. I. Elbashir, and K. Ghebremeskel. 2015. Omega 3
10.1093/ajcn/83.6.1467S. (n-3) fatty acids down-regulate nuclear factor-kappa B (NF-κB) gene
Bagger, H., M. Hansson, T. Kander, and U. Schött. 2020. Synergistic and blood cell adhesion molecule expression in patients with ho-
platelet inhibition between Omega-3 and acetylsalicylic acid dose mozygous sickle cell disease. Blood Cells, Molecules & Diseases 55
titration; an observational study. BMC Complementary Medicine and (1):48–55. doi: 10.1016/j.bcmd.2015.03.014.
Therapies 20 (1):1–9. doi: 10.1186/s12906-020-02990-9. Danthiir, V., D. E. Hosking, T. Nettelbeck, A. D. Vincent, C. Wilson,
Bang, H. O., J. Dyerberg, and N. Hjøorne. 1976. The composition of N. O’Callaghan, E. Calvaresi, P. Clifton, and G. A. Wittert. 2018.
food consumed by Greenland Eskimos. Acta Medica Scandinavica An 18-mo randomized, double-blind, placebo-controlled trial of
200 (1-2):69–73. doi: 10.1111/j.0954-6820.1976.tb08198.x. DHA-rich fish oil to prevent age-related cognitive decline in cog-
Bang, S., Y. Xie, Z. Zhang, Z. Wang, Z. Xu, and R. Ji. 2018. GPR37 nitively normal older adults. The American Journal of Clinical
regulates macrophage phagocytosis and resolution of inflammatory Nutrition 107 (5):754–62. doi: 10.1093/ajcn/nqx077.
Critical Reviews in Food Science and Nutrition 13

Dátilo, M. N., M. R. Sant’Ana, G. P. Formigari, P. B. Rodrigues, L. P. s­ upplements on human plasma oxylipins: A randomized double-blind
de Moura, A. S. R. da Silva, E. R. Ropelle, J. R. Pauli, and D. E. crossover trial. The Journal of Nutrition 151 (3):513–22. doi: 10.1093/
Cintra. 2018. Omega-3 from flaxseed oil protects obese mice against jn/nxaa294.
diabetic retinopathy through GPR120 receptor. Scientific Report 8 González-Périz, A., A. Planagumà, K. Gronert, R. Miquel, M.
(1):14318. López-Parra, E. Titos, R. Horrillo, N. Ferré, R. Deulofeu, V. Arroyo,
de Melo, D. G., C. P. Anaruma, K. C. da Cruz Rodrigues, R. M. et al. 2006. Docosahexaenoic acid (DHA) blunts liver injury by
Pereira, T. D. P. de Campos, R. S. Canciglieri, C. O. Ramos, D. E. conversion to protective lipid mediators: Protectin D1 and 17S‐hy-
Cintra, E. R. Ropelle, A. S. R. da Silva, et al. 2022. Strength train- droxy‐DHA. FASEB Journal: Official Publication of the Federation
ing alters the tissue fatty acids profile and slightly improves the of American Societies for Experimental Biology 20 (14):2537–9. doi:
thermogenic pathway in the adipose tissue of obese mice. Scientific 10.1096/fj.06-6250fje.
Reports 2812 (1):6913. doi: 10.1038/s41598-022-10688-w. Gurtovenko, A. A, and I. Vattulainen. 2007. Molecular mechanism for
Demar, J. C., Jr, K. Ma, L. Chang, J. M. Bell, and S. I. Rapoport. 2005. lipid flip-flops. The Journal of Physical Chemistry. B 111 (48):13554–
Alpha-Linolenic acid does not contribute appreciably to docosahex- 9. doi: 10.1021/jp077094k.
aenoic acid within brain phospholipids of adult rats fed a diet Guthrie, G. 2022. Parenteral nutrition associated hepatic steatosis and
enriched in docosahexaenoic acid. Journal of Neurochemistry 94 NAFLD intersect at AMPK. Cellular and Molecular Gastroenterology
(4):1063–76. doi: 10.1111/j.1471-4159.2005.03258.x. and Hepatology 14 (3):724–5. doi: 10.1016/j.jcmgh.2022.06.005.
DeMar, J. C., K. Ma, J. M. Bell, M. Igarashi, D. Greenstein, and S. I. Hames, K. C., C. Koutsari, S. Santosa, N. C. Bush, and M. D. Jensen.
Rapoport. 2006. One generation of n-3 polyunsaturated fatty acid 2015. Adipose tissue fatty acid storage factors: Effects of depot, sex
deprivation increases depression and aggression test scores in rats. and fat cell size. International Journal of Obesity (2005) 39 (6):884–
Journal of Lipid Research 47 (1):172–80. doi: 10.1194/jlr.M500362-JLR200. 7. doi: 10.1038/ijo.2015.10.
DiNicolantonio, J. J, and J. OKeefe. 2019. Importance of maintaining Han, Y. H., K. O. Shin, J. Y. Kim, D. B. Khadka, H. J. Kim, Y. M.
a low omega-6/omega-3 ratio for reducing platelet aggregation, Lee, W. J. Cho, J. Y. Cha, B. J. Lee, and M. O. Lee. 2019. A ma-
coagulation and thrombosis. Open Heart 6 (1):e001011. doi: 10.1136/ resin 1/ROR α/12-lipoxygenase autoregulatory circuit prevents in-
openhrt-2019-001011. flammation and progression of nonalcoholic steatohepatitis. The
Doege, H, and A. Stahl. 2006. Protein-mediated fatty acid uptake: Journal of Clinical Investigation 129 (4):1684–98. doi: 10.1172/
Novel insights from in vivo models. Physiology (Bethesda, Md.) JCI124219.
21:259–68. doi: 10.1152/physiol.00014.2006. Hansen, T. V., J. Dalli, and C. N. Serhan. 2017. The novel lipid me-
Downie, L. E., S. M. Ng, K. B. Lindsley, and E. K. Akpek. 2019. diator PD1n-3 DPA: An overview of the structural elucidation,
Omega-3 and omega-6 polyunsaturated fatty acids for dry eye dis- synthesis, biosynthesis and bioactions. Prostaglandins & Other Lipid
ease. The Cochrane Database of Systematic Reviews 12 (12):CD011016. Mediators 133:103–10. doi: 10.1016/j.prostaglandins.2017.06.003.
doi: 10.1002/14651858.CD011016.pub2. Hirasawa, A., T. Hara, S. Katsuma, T. Adachi, and G. Tsujimoto. 2008.
Duc, N. M., H. R. Kim, and K. Y. Chung. 2015. Structural mechanism Free fatty acid receptors and drug discovery. Biological &
of G protein activation by G protein-coupled receptor. European Pharmaceutical Bulletin 31 (10):1847–51. doi: 10.1248/bpb.31.1847.
Journal of Pharmacology 763 (Pt B):214–22. doi: 10.1016/j.ej- Hirasawa, A., K. Tsumaya, T. Awaji, S. Katsuma, T. Adachi, M. Yamada,
phar.2015.05.016. Y. Sugimoto, S. Miyazaki, and G. Tsujimoto. 2005. Free fatty acids
Dyerberg, J, and H. O. Bang. 1979. Haemostatic function and platelet regulate gut incretin glucagon-like peptide-1 secretion through
polyunsaturated fatty acids in Eskimos. The Lancet 314 (8140):433– GPR120. Nature Medicine 11 (1):90–4. doi: 10.1038/nm1168.
5. doi: 10.1016/S0140-6736(79)91490-9. Holy, E. W., M. Forestier, E. K. Richter, A. Akhmedov, F. Leiber, G.
Dyerberg, J., H. O. Bang, E. Stoffersen, S. Moncada, and J. R. Vane. G. Camici, P. Mocharla, T. F. Lüscher, J. H. Beer, and F. C. Tanner.
1978. Eicosapentaenoic acid and prevention of thrombosis and ath- 2011. Dietary α-linolenic acid inhibits arterial thrombus formation,
erosclerosis? Lancet (London, England) 2 (8081):117–9. doi: 10.1016/ tissue factor expression, and platelet activation. Arteriosclerosis,
s0140-6736(78)91505-2. Thrombosis, and Vascular Biology 31 (8):1772–80. doi: 10.1161/
Emre, C., L. E. Arroyo-García, K. V. Do, B. Jun, M. Ohshima, S. G. ATVBAHA.111.226118.
Alcalde, M. L. Cothern, S. Maioli, P. Nilsson, E. Hjorth, et al. 2022. Huang, S., Rutkowsky, J. M. Snodgrass, R. G. Ono-Moore, K. D.
Intranasal delivery of pro-resolving lipid mediators rescues memo- Schneider, D. A. Newman, J. W. Adams, S. H. Hwang, and D. H.
ry and gamma oscillation impairment in App NL-G-F/NL-G-F mice. S. 2012. Saturated fatty acids activate TLR-mediated proinflamma-
Communications Biology 5 (1):245. doi: 10.1038/s42003-022-03169-3. tory signaling pathways. Journal of Lipid Research 53 (9):2002–13.
Esser-von Bieren, J. 2019. Eicosanoids in tissue repair. Immunology doi: 10.1194/jlr.D029546.
and Cell Biology 97 (3):279–88. doi: 10.1111/imcb.12226. Hwang, S.-M., G. Chung, Y. H. Kim, and C.-K. Park. 2019. The role
Fahrmann, J. F., O. F. Ballester, G. Ballester, T. R. Witte, A. J. Salazar, of maresins in inflammatory pain: Function of macrophages in
B. Kordusky, K. G. Cowen, G. Ion, D. A. Primerano, G. Boskovic, wound regeneration. International Journal of Molecular Sciences 20
et al. 2013. Inhibition of nuclear factor kappa B activation in (23):5849. doi: 10.3390/ijms20235849.
early-stage chronic lymphocytic leukemia by omega-3 fatty acids. Ishitani, T., G. Takaesu, J. Ninomiya-Tsuji, H. Shibuya, R. B. Gaynor,
Cancer Investigation 31 (1):24–38. doi: 10.3109/07357907.2012.743553. and K. Matsumoto. 2003. Role of the TAB2-related protein TAB3
Feng, C., L. Li, Q. Li, K. Switzer, M. Liu, S. Han, and B. Zheng. 2021. in IL-1 and TNF signaling. The EMBO Journal 22 (23):6277–88.
Docosahexaenoic acid ameliorates autoimmune inflammation by doi: 10.1093/emboj/cdg605.
activating GPR120 signaling pathway in dendritic cells. International Itoh, Y., Y. Kawamata, M. Harada, M. Kobayashi, R. Fujii, S. Fukusumi,
Immunopharmacology 97:107698. doi: 10.1016/j.intimp.2021.107698. K. Ogi, M. Hosoya, Y. Tanaka, H. Uejima, et al. 2003. Free fatty
Francque, S. M., P. Bedossa, V. Ratziu, Q. M. Anstee, E. Bugianesi, A. acids regulate insulin secretion from pancreatic β cells through
J. Sanyal, R. Loomba, S. A. Harrison, R. Balabanska, L. Mateva, GPR40. Nature 422 (6928):173–6. doi: 10.1038/nature01478.
et al. 2021. A Randomized, Controlled Trial of the Pan-PPAR Jeansen, S., Witkamp, R. F. Garthoff, J. A. Helvoort, A. Van, and
Agonist Lanifibranor in NASH. The New England Journal of Medicine Calder, P. C. 2018. Fish oil LC-PUFAs do not affect blood coagu-
385 (17):1547–58. doi: 10.1056/NEJMoa2036205. lation parameters and bleeding manifestations: Analysis of 8 clini-
Frigerio, F., G. Pasqualini, I. Craparotta, S. Marchini, E. A. van Vliet, cal studies with selected patient groups on omega-3-enriched med-
P. Foerch, C. Vandenplas, K. Leclercq, E. Aronica, L. Porcu, et al. ical nutrition. Clinical Nutrition (Edinburgh, Scotland) 37 (3):948–57.
2018. n-3 Docosapentaenoic acid-derived protectin D1 promotes doi: 10.1016/j.clnu.2017.03.027.
resolution of neuroinflammation and arrests epileptogenesis. Brain: Kain, V., K. A. Ingle, M. Kachman, H. Baum, G. Shanmugam, N. S.
A Journal of Neurology 141 (11):3130–43. doi: 10.1093/brain/awy247. Rajasekaran, M. E. Young, and G. V. Halade. 2018. Excess ω-6
Gabbs, M., P. Zahradka, C. G. Taylor, and H. M. Aukema. 2021. Time fatty acids influx in aging drives metabolic dysregulation, electro-
course and sex effects of α-linolenic acid-rich and DHA-rich cardiographic alterations, and low-grade chronic inflammation.
14 E. DA SILVA BATISTA ET AL.

American Journal of Physiology. Heart and Circulatory Physiology Liu, T., L. Zhang, D. Joo, and S.-C. Sun. 2017. NF-κB signaling in
314 (2):H160–H169. doi: 10.1152/ajpheart.00297.2017. inflammation. Signal Transduction and Targeted Therapy 2 (1):17023.
Kang, J. Y, and J. O. Lee. 2011. Structural biology of the Toll-like doi: 10.1038/sigtrans.2017.23.
receptor family. Annual Review of Biochemistry 80:917–41. doi: Magee, P., S. Pearson, J. Whittingham-Dowd, and J. Allen. 2012. PPARγ
10.1146/annurev-biochem-052909-141507. as a molecular target of EPA anti-inflammatory activity during
Kim, S. F., D. A. Huri, and S. H. Snyder. 2005. Medicine: Inducible TNF-α-impaired skeletal muscle cell differentiation. The Journal of
nitric oxide synthase binds, S-nitrosylates, and activates Nutritional Biochemistry 23 (11):1440–8. doi: 10.1016/j.jnut-
cyclooxygenase-2. Science (New York, N.Y.) 310 (5756):1966–70. doi: bio.2011.09.005.
10.1126/science.1119407. Marcinak, J., C. Cao, D. Lee, and Z. Ye. 2017. Fasiglifam for glycemic
Kliewer, S. A., S. S. Sundseth, S. A. Jones, P. J. Brown, G. B. Wisely, control in patients with type 2 diabetes: A phase 3, placebo-controlled
C. S. Koble, P. Devchand, W. Wahli, T. M. Willson, J. M. Lenhard, study. Diabetes, Obesity & Metabolism 19 (12):1714–21. doi: 10.1111/
et al. 1997. Fatty acids and eicosanoids regulate gene expression dom.13004.
through direct interactions with peroxisome proliferator-activated Marks, K. A., P. M. Marvyn, J. J. Henao, R. M. Bradley, K. D. Stark,
receptors α and γ. Proceedings of the National Academy of Sciences and R. E. Duncan. 2015. Fasting enriches liver triacylglycerol with
of the United States of America 94 (9):4318–23. doi: 10.1073/ n-3 polyunsaturated fatty acids: Implications for understanding the
pnas.94.9.4318. adipose-liver axis in serum docosahexaenoic acid regulation. Genes
Knez Hrnčič, M., M. Ivanovski, D. Cör, and Ž. Knez. Ž 2019. Chia & Nutrition 10 (6):39. doi: 10.1007/s12263-015-0490-2.
seeds (Salvia hispanica L.): An overview-phytochemical profile, iso- Martínez-Micaelo, N., N. González-Abuín, M. Pinent, A. Ardévol, and
lation methods, and application. Molecules 25 (1):11. doi: 10.3390/ M. Blay. 2016. Dietary fatty acid composition is sensed by the
molecules25010011. NLRP3 inflammasome: Omega-3 fatty acid (DHA) prevents NLRP3
Komatsu, W., K. Ishihara, M. Murata, H. Saito, and K. Shinohara. activation in human macrophages. Food & Function 7 (8):3480–7.
2003. Docosahexaenoic acid suppresses nitric oxide production and doi: 10.1039/c6fo00477f.
inducible nitric oxide synthase expression in interferon-γ plus Mas, E., A. Barden, V. Burke, L. J. Beilin, G. F. Watts, R. C. Huang,
lipopolysaccharide-stimulated murine macrophages by inhibiting the I. B. Puddey, A. B. Irish, and T. A. Mori. 2016. A randomized
oxidative stress. Free Radical Biology & Medicine 34 (8):1006–16. controlled trial of the effects of n-3 fatty acids on resolvins in
doi: 10.1016/s0891-5849(03)00027-3. chronic kidney disease. Clinical Nutrition (Edinburgh, Scotland) 35
Krishnamoorthy, S., A. Recchiuti, N. Chiang, S. Yacoubian, C. H. Lee, (2):331–6. doi: 10.1016/j.clnu.2015.04.004.
R. Yang, N. A. Petasis, and C. N. Serhan. 2010. Resolvin D1 binds Mastalerz, L., K. E. Tyrak, M. Ignacak, E. Konduracka, F. Mejza, A.
human phagocytes with evidence for proresolving receptors. Ćmiel, M. Buczek, A. Kot, K. Oleś, and M. Sanak. 2019. Prostaglandin
Proceedings of the National Academy of Sciences of the United States E2 decrease in induced sputum of hypersensitive asthmatics during
of America 107 (4):1660–5. doi: 10.1073/pnas.0907342107. oral challenge with aspirin. Allergy 74 (5):922–32. doi: 10.1111/
Kumar, N., G. Gupta, K. Anilkumar, N. Fatima, R. Karnati, G. V. all.13671.
Reddy, P. V. Giri, and P. Reddanna. 2016. 15-Lipoxygenase metab- Mauricio, D., L. Meneghini, J. Seufert, L. Liao, H. Wang, L. Tong, A.
olites of α-linolenic acid, [13-(S)-HPOTrE and 13-(S)-HOTrE], me- Cali, P. Stella, P. Carita, and K. Khunti. 2017. Glycaemic control
diate anti-inflammatory effects by inactivating NLRP3 inflammasome. and hypoglycaemia burden in patients with type 2 diabetes initiat-
Scientific Report 18 (6):31649. ing basal insulin in Europe and the USA. Diabetes, Obesity &
Lancaster, G. I., K. G. Langley, N. A. Berglund, H. L. Kammoun, S. Metabolism 19 (8):1155–64. doi: 10.1111/dom.12927.
Reibe, E. Estevez, J. Weir, N. A. Mellett, G. Pernes, J. R. W. Conway, Mihaly, S. R., J. Ninomiya-Tsuji, and S. Morioka. 2014. TAK1 control
et al. 2018. Evidence that TLR4 is not a receptor for saturated of cell death. Cell Death and Differentiation 21 (11):1667–76. doi:
fatty acids but mediates lipid-induced inflammation by reprogram- 10.1038/cdd.2014.123.
ming macrophage metabolism. Cell Metabolism 27 (5):1096–110.e5. Mikroulis, A., M. Ledri, G. Ruffolo, E. Palma, G. Sperk, J. Dalli, A.
doi: 10.1016/j.cmet.2018.03.014. Vezzani, and M. Kokaia. 2022. Lipid mediator n-3 docosapentae-
Lee, K. R., Y. Midgette, and R. Shah. 2019. Fish oil derived omega 3 noic acid-derived protectin D1 enhances synaptic inhibition of hip-
fatty acids suppress adipose NLRP3 inflammasome signaling in pocampal principal neurons by interaction with a G-protein-coupled
human obesity. Journal of the Endocrine Society 3 (3):504–15. doi: receptor. FASEB Journal: Official Publication of the Federation of
10.1210/js.2018-00220. American Societies for Experimental Biology 36 (3):e22203. doi:
Lefere, S., T. Puengel, J. Hundertmark, C. Penners, A. K. Frank, A. 10.1096/fj.202101815R.
Guillot, K. de Muynck, F. Heymann, V. Adarbes, E. Defrêne, et al. Miles, E. A., T. Banerjee, M. M. B. W. Dooper, L. M. Rabet, Y. M. F.
2020. Differential effects of selective- and pan-PPAR agonists on Graus, and P. C. Calder. 2004. The influence of different combina-
experimental steatohepatitis and hepatic macrophages. Journal of tions of g -linolenic acid, stearidonic acid and EPA on immune
Hepatology 73 (4):757–70. doi: 10.1016/j.jhep.2020.04.025. function in healthy young male subjects. The British Journal of
Leslie, C. A., W. A. Gonnerman, M. D. Ullman, K. C. Hayes, C. Nutrition 91 (6):893–903. doi: 10.1079/BJN20041131.
Franzblau, and E. S. Cathcart. 1985. Dietary fish oil modulates Mo, Z., C. Tang, H. Li, J. Lei, L. Zhu, L. Kou, H. Li, S. Luo, C. Li,
macrophage fatty acids and decreases arthritis susceptibility in mice. W. Chen, et al. 2020. Eicosapentaenoic acid prevents inflammation
The Journal of Experimental Medicine 162 (4):1336–49. doi: 10.1084/ induced by acute cerebral infarction through inhibition of NLRP3
jem.162.4.1336. inflammasome activation. Life Sciences 242:117133. doi: 10.1016/j.
Li, X.-J., Y.-M. Mu, Q.-F. Qin, Z.-X. Zeng, Y.-S. Li, W. K. Zhang, H.- lfs.2019.117133.
B. Tang, G.-H. Tian, and H.-C. Shang. 2017. Chronic high-dosage Mortimer, L., F. Moreau, J. A. MacDonald, and K. Chadee. 2016.
fish oil exacerbates gut–liver axis injury in alcoholic steatohepatitis NLRP3 inflammasome inhibition is disrupted in a group of
in mice: The roles of endotoxin and IL-4 in Kupffer cell polarization auto-inflammatory disease CAPS mutations. Nature Immunology 17
imbalance. Toxicology Research 6 (5):611–20. doi: 10.1039/c7tx00037e. (10):1176–86. doi: 10.1038/ni.3538.
Liu, G. J., T. Tao, H. Wang, Y. Zhou, X. Gao, Y. Y. Gao, C. H. Hang, Moura-Assis, A., M. S. Afonso, V. de Oliveira, J. Morari, G. A. Dos
and W. Li. 2020. Functions of resolvin D1-ALX/FPR2 receptor in- Santos, M. Koike, A. M. Lottenberg, R. Ramos Catharino, L. A.
teraction in the hemoglobin-induced microglial inflammatory re- Velloso, A. Sanchez Ramos da Silva, et al. 2018. Flaxseed oil rich
sponse and neuronal injury. Journal of Neuroinflammation 17 (1):239. in omega-3 protects aorta against inflammation and endoplasmic
doi: 10.1186/s12974-020-01918-x. reticulum stress partially mediated by GPR120 receptor in obese,
Liu, R. Z., R. Mita, M. Beaulieu, Z. Gao, and R. Godbout. 2010. Fatty diabetic and dyslipidemic mice models. The Journal of Nutritional
acid binding proteins in brain development and disease. The Biochemistry 53:9–19. doi: 10.1016/j.jnutbio.2017.09.015.
International Journal of Developmental Biology 54 (8-9):1229–39. Mukherjee, P. K., V. L. Marcheselli, C. N. Serhan, and N. G. Bazan. 2004.
doi: 10.1387/ijdb.092976rl. Neuroprotectin D1: A docosahexaenoic acid-derived docosatriene
Critical Reviews in Food Science and Nutrition 15

­rotects human retinal pigment epithelial cells from oxidative stress.


p Raclot, T. 2003. Selective mobilization of fatty acids from adipose
Proceedings of the National Academy of Sciences of the United States of tissue triacylglycerols. Progress in Lipid Research 42 (4):257–88. doi:
America 101 (22):8491–6. doi: 10.1073/pnas.0402531101. 10.1016/s0163-7827(02)00066-8.
Naderali, E., M.-M. Naderali, and A.-R. Abubakari. 2014. Omega-3 Raptis, D. A., P. Limani, J. H. Jang, U. Ungethüm, C. Tschuor, R. Graf,
fatty acid supplementation and cognitive function: Are smaller dos- B. Humar, and P. A. Clavien. 2014. GPR120 on Kupffer cells me-
ages more beneficial? International Journal of General Medicine 7 diates hepatoprotective effects of ω3-fatty acids. Journal of Hepatology
(7):463–73. doi: 10.2147/IJGM.S67065. 60 (3):625–32. doi: 10.1016/j.jhep.2013.11.006.
Neuhofer, A., M. Zeyda, D. Mascher, B. K. Itariu, I. Murano, L. Leitner, Risérus, U., D. Sprecher, T. Johnson, E. Olson, S. Hirschberg, A. Liu,
E. E. Hochbrugger, P. Fraisl, S. Cinti, C. N. Serhan, et al. 2013. Z. Fang, P. Hegde, D. Richards, L. Sarov-Blat, et al. 2008. Activation
Impaired local production of proresolving lipid mediators in obe- of peroxisome proliferator – activated receptor (PPAR)δ promotes
sity and 17-HDHA as a potential treatment for obesity-associated reversal of multiple metabolic abnormalities, reduces oxidative stress,
inflammation. Diabetes 62 (6):1945–56. doi: 10.2337/db12-0828. and increases fatty acid oxidation in moderately obese men. Diabetes
Nicholls, S. J., A. M. Lincoff, M. Garcia, D. Bash, C. M. Ballantyne, 57 (2):332–9. doi: 10.2337/db07-1318.
P. J. Barter, M. H. Davidson, J. J. P. Kastelein, W. Koenig, D. K. Satoh, M., T. Tabuchi, T. Itoh, and M. Nakamura. 2014. NLRP3 in-
McGuire, et al. 2020. Effect of high-dose omega-3 fatty acids vs flammasome activation in coronary artery disease: Results from
corn oil on major adverse cardiovascular events in patients at high prospective and randomized study of treatment with atorvastatin
cardiovascular risk: The STRENGTH randomized clinical trial. JAMA or rosuvastatin. Clinical Science 126 (3):233–41. doi: 10.1042/
324 (22):2268–80. doi: 10.1001/jama.2020.22258. CS20130043.
Ochiai, Y., Y. Uchida, S. Ohtsuki, M. Tachikawa, S. Aizawa, and T. Schulze, M. B., A. M. Minihane, R. N. M. Saleh, and U. Risérus. 2020.
Terasaki. 2017. The blood-brain barrier fatty acid transport protein Intake and metabolism of omega-3 and omega-6 polyunsaturated
1 (FATP1/SLC27A1) supplies docosahexaenoic acid to the brain, fatty acids: Nutritional implications for cardiometabolic diseases.
and insulin facilitates transport. Journal of Neurochemistry 141 The Lancet. Diabetes & Endocrinology 8 (11):915–30. doi: 10.1016/
(3):400–12. doi: 10.1111/jnc.13943. S2213-8587(20)30148-0.
Oh, D. Y., S. Talukdar, E. J. Bae, T. Imamura, H. Morinaga, W. Fan, Serhan, C. N. 2017. Treating inflammation and infection in the 21st
P. Li, W. J. Lu, S. M. Watkins, and J. M. Olefsky. 2010. GPR120 Is century: New hints from decoding resolution mediators and mech-
an omega-3 fatty acid receptor mediating potent anti-inflammatory anisms. FASEB Journal: Official Publication of the Federation of
and insulin-sensitizing effects. Cell 142 (5):687–98. doi: 10.1016/j. American Societies for Experimental Biology 31 (4):1273–88. doi:
cell.2010.07.041. 10.1096/fj.201601222R.
Oliveira, V., R. Marinho, D. Vitorino, G. A. Santos, J. C. Moraes, N. Serhan, C. N., S. Hong, K. Gronert, S. P. Colgan, P. R. Devchand, G.
Dragano, A. Sartori-Cintra, L. Pereira, R. R. Catharino, A. S. R. da Mirick, and R. L. Moussignac. 2002. Resolvins: A family of bioac-
Silva, et al. 2015. Diets containing α-linolenic (ω3) or oleic (ω9) tive products of omega-3 fatty acid transformation circuits initiated
fatty acids rescues obese mice from insulin resistance. Endocrinology by aspirin treatment that counter proinflammation signals. The
156 (11):4033–46. doi: 10.1210/en.2014-1880. Journal of Experimental Medicine 196 (8):1025–37. doi: 10.1084/
Paschoal, V. A., E. Walenta, S. Talukdar, A. R. Pessentheiner, O. jem.20020760.
Osborn, N. Hah, T. J. Chi, G. L. Tye, A. M. Armando, R. M. Evans, Serhan, C. N., R. Yang, K. Martinod, K. Kasuga, P. S. Pillai, T. F.
et al. 2020. Positive reinforcing mechanisms between GPR120 and Porter, S. F. Oh, and M. Spite. 2009. Maresins: Novel macrophage
PPARγ modulate insulin sensitivity. Cell Metabolism 31 (6):1173–88. mediators with potent antiinflammatory and proresolving actions.
e5. doi: 10.1016/j.cmet.2020.04.020. The Journal of Experimental Medicine 206 (1):15–23. doi: 10.1084/
Payan, D. G., M. Y. Wong, T. Chernov-Rogan, F. H. Valone, W. C. jem.20081880.
Pickett, V. A. Blake, W. M. Gold, and E. J. Goetzl. 1986. Alterations Shen, L., Y. Yang, T. Ou, C. C. Key, S. H. Tong, R. C. Sequeira, J. M.
in human leukocyte function induced by ingestion of eicosapentae- Nelson, Y. Nie, Z. Wang, E. Boudyguina, et al. 2017. Dietary PUFAs
noic acid. Journal of Clinical Immunology 6 (5):402–10. doi: 10.1007/ attenuate NLRP3 inflammasome activation via enhancing macro-
BF00915380. phage autophagy. Journal of Lipid Research 58 (9):1808–21. doi:
Picklo, M. J, and E. J. Murphy. 2016. A high-fat, high-oleic diet, but 10.1194/jlr.M075879.
not a high-fat, saturated diet, reduces hepatic α-linolenic acid and Shi, C. S, and J. H. Kehrl. 2010. TRAF6 and A20 regulate lysine
eicosapentaenoic acid content in mice. Lipids 51 (5):537–47. doi: 63-linked ubiquitination of Beclin-1 to control TLR4-induced au-
10.1007/s11745-015-4106-9. tophagy. Science Signaling 3 (123):ra42. doi: 10.1126/scisig-
Pishva, H., M. Amini, M. R. Eshraghian, S. Hosseini, and S. A. nal.2000751.
Mahboob. 2012. Effects of EPA supplementation on plasma fatty Simopoulos, A. P. 2016. An increase in the omega-6/omega-3 fatty
acids composition in hypertriglyceridemic subjects with FABP2 and acid ratio increases the risk for obesity. Nutrients 8 (3):128. doi:
PPARα genotypes. Journal of Diabetes and Metabolic Disorders 11 10.3390/nu8030128.
(1):25. doi: 10.1186/2251-6581-11-25. Sinclair, A. J., X. F. Guo, and L. Abedin. 2022. Dietary Alpha-Linolenic
Polus, A., B. Zapala, U. Razny, A. Gielicz, B. Kiec-Wilk, M. Acid Supports High Retinal DHA Levels. Nutrients 14 (2):301. doi:
Malczewska-Malec, M. Sanak, C. E. Childs, P. C. Calder, and A. 10.3390/nu14020301.
Dembinska-Kiec. 2016. Omega-3 fatty acid supplementation influ- Singer, P., W. Jaeger, I. Berger, H. Barleben, M. Wirth, E.
ences the whole blood transcriptome in women with obesity, asso- Richter-Heinrich, S. Voigt, and W. Gödicke. 1990. Effects of dietary
ciated with pro-resolving lipid mediator production. Biochimica et oleic, linoleic, and α-linolenic acids on blood pressure, serum lipids,
Biophysica Acta 1861 (11):1746–55. doi: 10.1016/j.bbalip.2016.08.005. lipoproteins and the formation of eicosanoid precursors in patients
Poreba, M., M. Mostowik, A. Siniarski, R. Golebiowska-Wiatrak, K. P. with mild essential hypertension. Journal of Human Hypertension 4
Malinowski, M. Haberka, E. Konduracka, J. Nessler, A. Undas, and (3):227–33.
G. Gajos. 2017. Treatment with high-dose n-3 PUFAs has no effect Sinha, S., G. Perdomo, N. F. Brown, and R. M. O’Doherty. 2004. Fatty
on platelet function, coagulation, metabolic status or inflammation acid-induced insulin resistance in L6 myotubes is prevented by
in patients with atherosclerosis and type 2 diabetes. Cardiovascular inhibition of activation and nuclear localization of nuclear factor
Diabetology 16 (1):50. doi: 10.1186/s12933-017-0523-9. κB. The Journal of Biological Chemistry 279 (40):41294–301. doi:
Quezada, N., I. Valencia, J. Torres, G. Maturana, J. Cerda, J. P. Arab, 10.1074/jbc.M406514200.
J. J. Fuentes, C. Pinto, D. Turiel, and V. Cortés. 2021. Insulin re- Spencer, M., B. S. Finlin, R. Unal, B. Zhu, A. J. Morris, L. R. Shipp,
sistance and liver histopathology in metabolically unhealthy subjects J. Lee, R. G. Walton, A. Adu, R. Erfani, et al. 2013. Omega-3 fatty
do not correlate with the hepatic abundance of NLRP3 inflam- acids reduce adipose tissue macrophages in human subjects with
masome nor circulating IL-1β levels. BMJ Open Diabetes Research insulin resistance. Diabetes 62 (5):1709–17. doi: 10.2337/db12-1042.
& Care 9 (1):e001975. doi: 10.1136/bmjdrc-2020-001975.
16 E. DA SILVA BATISTA ET AL.

Stivala, S., S. Gobbato, N. Bonetti, G. G. Camici, T. F. Lüscher, and macrophage inflammasome activation by inhibiting NF-κB activation
J. H. Beer. 2022. Dietary alpha‐linolenic acid reduces platelet acti- and enhancing autophagy. PloS One 9 (6):e97957. doi: 10.1371/
vation and collagen‐mediated cell adhesion in sickle cell disease journal.pone.0097957.
mice. Journal of Thrombosis and Haemostasis: JTH 20 (2):375–86. Wree, A., M. D. McGeough, C. A. Peña, M. Schlattjan, H. Li, M. E.
doi: 10.1111/jth.15581. Inzaugarat, K. Messer, A. Canbay, H. M. Hoffman, and A. E.
Su, H. M., L. Bernardo, M. Mirmiran, X. H. Ma, T. N. Corso, P. W. Feldstein. 2014. NLRP3 inflammasome activation is required for
Nathanielsz, and J. T. Brenna. 1999. Bioequivalence of dietary fibrosis development in NAFLD. Journal of Molecular Medicine
α-linolenic and docosahexaenoic acids as sources of docosahexaenoate (Berlin, Germany) 92 (10):1069–82. doi: 10.1007/s00109-014-1170-1.
accretion in brain and associated organs of neonatal baboons. Pediatric Wu, C. T., K. I. Hilgendorf, R. J. Bevacqua, Y. Hang, J. Demeter, S.
Research 45 (1):87–93. doi: 10.1203/00006450-199901000-00015. K. Kim, and P. K. Jackson. 2021. Discovery of ciliary G
Sutter, A. G., A. P. Palanisamy, J. H. Lench, S. Esckilsen, T. Geng, D. protein-coupled receptors regulating pancreatic islet insulin and
N. Lewin, L. A. Cowart, and K. D. Chavin. 2016. Dietary saturat- glucagon secretion. Genes & Development 35 (17–18):1243–55. doi:
ed fat promotes development of hepatic inflammation through 10.1101/gad.348261.121.
toll-like receptor 4 in mice. Journal of Cellular Biochemistry 117 Yamada, H., E. Oshiro, S. Kikuchi, M. Hakozaki, H. Takahashi, and
(7):1613–21. doi: 10.1002/jcb.25453. K. I. Kimura. 2014. Hydroxyeicosapentaenoic acids from the Pacific
Park, S. W., M. W. Sung, D. S. Heo, H. Inoue, S. H. Shim, and K. H. krill show high ligand activities for PPARs. Journal of Lipid Research
Kim. 2005. Nitric oxide upregulates the cyclooxygenase-2 expression 55 (5):895–904. doi: 10.1194/jlr.M047514.
through the cAMP-response element in its promoter in several Yan, Y., W. Jiang, T. Spinetti, A. Tardivel, R. Castillo, C. Bourquin, G.
cancer cell lines. Oncogene 24 (44):6689–98. doi: 10.1038/sj. Guarda, Z. Tian, J. Tschopp, and R. Zhou. 2013. Omega-3 fatty
onc.1208816. acids prevent inflammation and metabolic disorder through inhibi-
Tanaka, H., F. Soga, K. Tatsumi, Y. Mochizuki, H. Sano, H. Toki, K. tion of NLRP3 inflammasome activation. Immunity 38 (6):1154–63.
Matsumoto, J. Shite, H. Takaoka, T. Doi, et al. 2020. Positive effect doi: 10.1016/j.immuni.2013.05.015.
of dapagliflozin on left ventricular longitudinal function for type 2 Yang, R. H., J. Lin, X. H. Hou, R. Cao, F. Yu, H. Q. Liu, A. L. Ji, X.
diabetic mellitus patients with chronic heart failure. Cardiovascular N. Xu, L. Zhang, and F. Wang. 2014. Effect of docosahexaenoic acid
Diabetology 19 (1):6. doi: 10.1186/s12933-019-0985-z. on hippocampal neurons in high-glucose condition: Involvement of
Tashjian, A. H., E. F. Voelkel, D. R. Robinson, and L. Levine. 1984. PI3K/AKT/nuclear factor-κB-mediated inflammatory pathways.
Dietary menhaden oil lowers plasma prostaglandins and calcium in Neuroscience 274:218–28. doi: 10.1016/j.neuroscience.2014.05.042.
mice bearing the prostaglandin-producing HSDM1 fibrosarcoma. Yip, P. K., C. Pizzasegola, S. Gladman, M. L. Biggio, M. Marino, M.
The Journal of Clinical Investigation 74 (6):2042–8. doi: 10.1172/ Jayasinghe, F. Ullah, S. C. Dyall, A. Malaspina, C. Bendotti, et al.
JCI111627. 2013. The omega-3 fatty acid eicosapentaenoic acid accelerates dis-
Udipi, S., S. Karandikar, R. Mukherjee, S. Agarwal, and P. Ghugre. 2006. ease progression in a model of amyotrophic lateral sclerosis. PLoS
Variations in fat and fatty acid intakes of adult males from three One 8 (4):e61626. doi: 10.1371/journal.pone.0061626.
regions of India. Indian Journal of Public Health 50 (3):179–86. Yokoyama, M., H. Origasa, M. Matsuzaki, Y. Matsuzawa, Y. Saito, Y.
Unamuno, X., J. Gómez-Ambrosi, B. Ramírez, A. Rodríguez, S. Becerril, Ishikawa, S. Oikawa, J. Sasaki, H. Hishida, H. Itakura, et al. 2007.
V. Valentí, R. Moncada, C. Silva, J. Salvador, G. Frühbeck, et al. 2021. Effects of eicosapentaenoic acid on major coronary events in hy-
NLRP3 inflammasome blockade reduces adipose tissue inflammation percholesterolaemic patients (JELIS): A randomised open-label,
and extracellular matrix remodeling. Cellular & Molecular Immunology blinded endpoint analysis. Lancet (London, England) 369 (9567):1090–
18 (4):1045–57. doi: 10.1038/s41423-019-0296-z. 8. doi: 10.1016/S0140-6736(07)60527-3.
Vandanmagsar, B., Y. H. Youm, A. Ravussin, J. E. Galgani, K. Stadler, Yu, D., M. Zou, Q. Pan, Y. Song, M. Li, X. Zhang, Y. Zhou, X. Wang,
R. L. Mynatt, E. Ravussin, J. M. Stephens, and V. D. Dixit. 2011. and L. Guo. 2022. Effects of liraglutide or lifestyle interventions
The NLRP3 inflammasome instigates obesity-induced inflammation combined with other antidiabetic drugs on abdominal fat distribu-
and insulin resistance. Nature Medicine 17 (2):179–88. doi: 10.1038/ tion in people with obesity and type 2 diabetes mellitus evaluated
nm.2279. by the energy spectrum ct: A prospective randomized controlled
Vegiopoulos, A., M. Rohm, and S. Herzig. 2017. Adipose tissue: study. Frontier Endocrinology 13:951570.
Between the extremes. The EMBO Journal 36 (14):1999–2017. doi: Zhang, J. Y., K. S. D. Kothapalli, and J. T. Brenna. 2016. Desaturase
10.15252/embj.201696206. and elongase-limiting endogenous long-chain polyunsaturated fatty
Wang, B., X. Gong, J. Y. Wan, L. Zhang, Z. Zhang, H. Z. Li, and S. acid biosynthesis. Current Opinion Clinical. Nutrition Metabolism
Min. 2011. Resolvin D1 protects mice from LPS-induced acute lung Care 19 (2):103–10.
injury. Pulmonary Pharmacology & Therapeutics 24 (4):434–41. doi: Zhao, N., L. Wang, and N. Guo. 2016. α-Linolenic acid increases the
10.1016/j.pupt.2011.04.001. G0/G1 switch gene 2 mRNA expression in peripheral blood mono-
Williams-Bey, Y., C. Boularan, A. Vural, N. N. Huang, I. Y. Hwang, nuclear cells from obese patients: A pilot study. Lipids in Health
C. Shan-Shi, and J. H. Kehrl. 2014. Omega-3 free fatty acids suppress and Disease 15:36. doi: 10.1186/s12944-016-0207-6.

You might also like