You are on page 1of 11

Pharmacology & Therapeutics 141 (2014) 272282

Contents lists available at ScienceDirect

Pharmacology & Therapeutics


journal homepage: www.elsevier.com/locate/pharmthera

Associate Editor: M. Madhani

Pharmacology and therapeutics of omega-3 polyunsaturated fatty acids


in chronic inammatory disease
Clara M. Yates a, Philip C. Calder b, G. Ed Rainger a,
a
Centre for Cardiovascular Sciences, School of Clinical and Experimental Medicine, The University of Birmingham, B15 2TT, UK
b
Human Development & Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK

a r t i c l e i n f o a b s t r a c t

Keywords: Omega-3 (n3) polyunsaturated fatty acids (n3 PUFAs) have well documented anti-inammatory properties,
Omega-3 polyunsaturated fatty acid and consequently therapeutic potential in chronic inammatory diseases. Here we discuss the effects of n3
Eicosapentaenoic acid PUFAs on various inammatory pathways and how this leads to alterations in the function of inammatory
Docosahexaenoic acid cells, most importantly endothelial cells and leukocytes. Strong evidence indicates n3 PUFAs are benecial
Inammation as a dietary supplement in certain diseases such as rheumatoid arthritis; however for other conditions such as
Eicosanoid
asthma, the data are less robust. A clearer understanding of the pharmacology of n3 PUFAs will help to establish
Resolvin
targets to modulate chronic inammatory diseases.
2013 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 272
2. n3 polyunsaturated fatty acids and cardiovascular disease . . . . . . . . . . . . . . . . . . . . . . . . 276
3. The use of n3 polyunsaturated fatty acids as a dietary intervention in asthma . . . . . . . . . . . . . . . 277
4. The use of n3 polyunsaturated fatty acids as a dietary intervention in rheumatoid arthritis . . . . . . . . . 278
5. Inammatory bowel diseases and n3 polyunsaturated fatty acids . . . . . . . . . . . . . . . . . . . . . . 278
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 279
Conict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 280
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 280

1. Introduction adjacent carbon atoms), monounsaturated (one double bond) or poly-


unsaturated (more than one double bond) forms. Fatty acids have
1.1. Omega-3 polyunsaturated fatty acids (n3 PUFAs) both systematic and common names. They are also described by a
nomenclature which includes the number of carbon atoms in the
Fatty acids are carboxylic acids with a variable number of carbons chain (including the terminal carbons), the number of double bonds
atoms forming a hydrocarbon chain terminated by carboxyl and methyl in the chain, and the location of the rst double bond in the chain
groups (Fig. 1). The chains vary in length from two, to greater than thirty from the terminal methyl group, known as the n or carbon. For ex-
carbon atoms and exist in either saturated (no double bonds between ample, arachidonic acid (AA) is shown as 20:4n6 using this nomen-
clature, thus describing a molecule with 20 carbon atoms in the chain,
four double bonds, with the rst double bond at the 6th carbon from
Abbreviations: AA, Arachidonic acid; ALA, alpha-linolenic acid; CD, Crohn's disease; the methyl terminus. This nomenclature enables discrimination of
COX, cyclooxygensae; CVD, cardiovascular disease; DHA, docosahexaenoic acid; EC, endo-
different families of polyunsaturated fatty acids (PUFAs) based on the
thelial cells; EPA, eicosapentaenoic acid; FFA, free fatty acids; IBD, inammatory bowel dis-
eases; ICAM-1, intercellular adhesion molecule-1; IL, interleukin; LA, linoleic acid; LOX, position of the rst double bond in the chain, i.e. n3 and n6 PUFAs
lipoxygenase; LT, leukotrienes; LX, lipoxin; n3 PUFAs, omega-3 polyunsaturated fatty (Ratnayake & Galli, 2009).
acids; NPD, neuroprotectin; PBMC, peripheral blood mononuclear cell; PD, protectin; PG, In this review we will focus on the biological effects of the so called
prostaglandin; PI, phosphatidylinositol; RA, rheumatoid arthritis; RCT, randomised con- marine n3 PUFAs, eicosapentaenoic acid (EPA; 20:5n3) (Fig. 1A)
trolled trial; Rv, resolvin; SPM, specialised pro-resolving mediators; TNF, tumour necrosis
factor; UC, ulcerative colitis; VCAM-1, vascular cell adhesion molecule-1.
and docosahexaenoic acid (DHA; 22:6n3) (Fig. 1B). In mammals,
Corresponding author. Tel.: +44 121 414 4040; fax: +44 121 414 6919. EPA and DHA can be synthesised from the dietary precursor and essen-
E-mail address: g.e.rainger@bham.ac.uk (G. Ed Rainger). tial fatty acid, -linolenic acid (ALA; 18:3n3). Synthesis requires a

0163-7258/$ see front matter 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.pharmthera.2013.10.010
C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282 273

A n-3 histamine, leukocytes are captured from owing blood by specialised


carbon receptors of the selectin and immunoglobulin super families (IgSF).
These molecules also permit a form of dynamic adhesion termed
H3C COOH rolling, during which the velocity of leukocytes is dramatically reduced
in comparison to those being transported in the bulk ow of the blood.
EPA 20:5 n-3 Rolling cells are able to assimilate EC borne signals from agents such as
/n-
carbon
chemokines, which stabilise adhesion by activating leukocyte 1- and
2-integrins. Integrin-mediated adhesion to specic counter ligands
on the EC surface, in the basement membrane and in stromal tissue sup-
B n-3
port migration of leukocytes across the vascular barrier and towards the
carbon
inammatory locus. Once in tissue, leukocytes have powerful cytotoxic
COOH
and tissue remodelling capabilities which must be tightly regulated, not
H3C least because uncontrolled or non-resolving leukocyte recruitment may
be pathogenic. Indeed, it has been postulated that chronic inammation
DHA 22:6 n-3 represents a situation where normal programmes of resolution fail,
-/n- resulting in continual inux of leukocytes into tissue, where their exces-
carbon sive activity results in inappropriate tissue remodelling and ultimately
Fig. 1. Schematic structure of the n3 PUFAs EPA and DHA. A. EPA has 20 carbons and 5
to loss of tissue function (reviewed in McGettrick et al., 2012).
double bonds; B. DHA has 22 carbons and has 6 double bonds. In both fatty acids the A hallmark of chronic inammatory disease is the continual inltra-
rst double bond occurs at the third carbon from the /n-end of the chain. tion of leukocytes from blood, across activated EC and into the affected
tissue. Although the agents that activate EC may show variation in a dis-
number of elongation and desaturation steps and is inefcient in ease specic manner, in general many of the receptors and mediators
humans. This makes dietary intake of pre-formed EPA and DHA a more induced by these agents are common to the leukocyte recruitment
effective route of assimilation. EPA and DHA in the human diet are de- cascade (Fig. 2). It is thus probable that n3 PUFAs can regulate the
rived indirectly from marine algae (higher plants lack the enzymes for magnitude, and possibly the identity, of the leukocytic inltrate, in a
their biosynthesis). Their availability is dramatically increased as they range of inammatory conditions by regulating aspects of EC activation
pass up the food chain, becoming concentrated in the esh of marine generic to the inammatory process. It is important to appreciate that
sh. Both EPA and DHA are present in high amounts in the esh of oily studying the effects of n3 PUFAs on EC function in the context of
sh e.g. herring, mackerel and salmon, and they are the main PUFAs in leukocyte recruitment is difcult in vivo. Indeed most of the data avail-
sh oil supplements. Indications of the efcacy of these fatty acids as di- able to date have been generated in vitro using cultured cells with the
etary interventions for chronic inammatory disease originated from addition of free fatty acids (FFA) to culture medium as a source of sup-
epidemiological studies conducted on populations of Greenland Inuits, plementary lipids. However, the signicance of some of these studies
native Alaskans and the residents of Okinawa, Japan. These studies uni- must be questioned due the concentrations of FFA used. Physiologically,
formly showed a relationship between the burden of chronic disease n3 PUFAs are found in the blood plasma as FFA at concentrations less
and life style, with the consumption of high levels of n3 PUFAs being than 1 M (Cawood et al., 2010) (but at higher concentrations in ester-
common to all of the study populations (Dyerberg et al., 1978; ied forms such as triglycerides and phospholipids) and many supple-
Kromann & Green, 1980; Kagawa et al., 1982; Newman et al., 1993). mentation regimens do not greatly increase the levels of the FFA. Most
Thus, the postulate that dietary marine n3 PUFAs may be protective in vitro studies which investigate mechanisms of n3 PUFA action uti-
against chronic inammatory diseases, in particular cardiovascular dis- lise free n3 PUFAs at concentrations 10100 fold higher than this
ease (CVD), was engendered. Since these initial studies, much research which may equate to total plasma concentrations of n3 PUFAs achiev-
has been carried out to conrm this relationship and to investigate the able with supplementation. However, this does not account for the fact
efcacy of n3 PUFAs, in other disease states with a chronic inamma- that the majority of circulating n3 PUFAs are transported in esteried
tory component such as rheumatoid arthritis (RA), inammatory bowel form within lipoprotein particles which do not have the same prole
diseases (IBD) and asthma. Here we describe inammation and the of bioavailability as FFA. This is an important point, as in our own
mechanisms by which EPA and DHA can modify inammatory re- experiments, n3 PUFAs as FFA at concentrations of 10 M or above
sponses, and the relevance of this to chronic inammatory diseases. induce non-specic calcium transients in cells such as neutrophils
(unpublished observation; Yates & Rainger). Thus, we routinely utilise
1.2. Inammation free fatty acid at sub-M concentrations and do not normally exceed
5 M, as we feel that this represents the maximal ceiling of free n3
Inammation is a physiological response to infection or injury PUFA levels achievable in vivo with oral supplementation.
characterised by ve classical signs described by Celsus and Galen in
antiquity, i.e. pain, heat, redness, swelling and loss of function. The 1.3. Polyunsaturated fatty acid-derived molecules as mediators of the in-
overriding purpose of an acute, resolving inammatory response is to ammatory response
protect the body from invasion and damage and to re-establish physio-
logical homeostasis. Following infection, for example, inammatory Dietary fatty acids are intimately linked with the inammatory
mediators with vasodilatory capacity increase blood ow, thereby caus- response, For example the n6 PUFA, AA, is the main precursor of sever-
ing redness and heat, and vessels also become more permeable leading al important lipid mediators. AA can be derived directly from the diet
to oedema (swelling). Sensitivity to pain is increased in response to (Jonnalagadda et al., 1995) but like the n3 PUFAs, it can also be synthe-
agents such as bradykinin (Kenji, 2007). A major function of the inam- sised through a series of desaturation and elongation reactions from the
matory response is to deliver the molecular and cellular mediators of essential fatty acid linoleic acid (LA; 18:2-n6). AA from the diet or after
immunity to affected tissues. Thus, the concentration of plasma borne synthesis is stored in membrane phospholipids, e.g. phosphatidylcholine
agents such as complement increases due to changes in vascular perme- (PC), phosphatidylethanolamine (PE) and phosphatidylinositol (PI),
ability. Leukocytes are recruited by appropriately activated vascular mainly at the sn2 position and is liberated under appropriate stimulatory
endothelial cells (EC) which support a tightly regulated series of events conditions by the enzyme phospholipase A2 (PLA2). Phospholipase C
termed the leukocyte adhesion cascade (Ley et al., 2007) (Fig. 2). In re- (PLC) is also able to release AA specically from PI via a series of reactions
sponse to stimulation by inammatory cytokines, or agents such as involving the formation of diacylglycerol (DAG) by the enzyme DAG
274 C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282

Capture Stabilisation

Rolling Arrest Transendothelial


migration

Capture receptors e.g. selectins Chemokines IgSF e.g. ICAM-1 or VCAM-1 integrins

Lipids e.g. PGD2 Junctional molecules e.g. CD31 or JAMs

Fig. 2. The leukocyte adhesion cascade. Leukocytes are captured from owing blood and roll on selectin molecules presented by activated EC. EC derived chemokine and lipid signals are
assimilated by rolling leukocytes, stabilising adhesion and resulting in activation of leukocyte integrins. Integrins bind cognate receptors e.g. ICAM-1 or VCAM-1 on EC and migrate on
the EC surface to intercellular junctions. By engaging junctional adhesion molecules, leukocytes undergo transendothelial migration and enter the underlying tissue. (Figure adapted
from (McGettrick et al., 2012).

lipase (Whatley et al., 1993). Free AA is metabolised by three main Alpha-linolenic acid (ALA; 18:3 n3) is the parent n3 PUFA, and
classes of enzymes; (i) the cyclooxygenases (COX), producing prosta- like LA, must be wholly derived from the diet. Although humans have
glandins and thromboxanes, (ii) the lipoxygenases (LOX), generating the enzymes to synthesise EPA and DHA from ALA, the conversion
leukotrienes (LTs), hydroxyeicosatetraenoic acids (HETEs) and lipoxins rate is low, in particular for DHA. Indeed, a diet rich in ALA results in
(LX) and (iii) p450 epoxygenases which synthesise epoxyeicosatrienoic increased levels of EPA but not DHA in platelet, leukocyte and plasma
acids (EETs) and HETEs (Fig. 3). The production of these eicosanoids phospholipids (Li et al., 1999). In the absence of dietary supplementa-
in vivo can be inuenced by neighbouring cells leading to the process tion, EPA is present at low levels in the majority of tissues. DHA, however,
of transcelluar biosynthesis, where an intermediate generated from comprises up to 50% of all fatty acids in PE and phosphatidylserine
AA in one cell is fully processed into the nal product in a second cell (PS) in the outer cone and rod cells of the retina and is also found at
(Folco & Murphy, 2006). high levels in the brain (Fliesler & Anderson, 1983) (and reviewed in

Membrane
phospholipids

Arachidonic Acid
(AA)

5-LOX
COX-1,2 P450 15-LOX (leukocytes)
epoxyganase

PGG2/PGH2
Epoxyeicosatrienoic acids 5-HPETE
5,6-EET 5-HPETE
8,9-EET
11,12-EET
14,15-EET LTA4

TXA2 PGE2 PGI2 PGD2 PGF2


5-LOX 12-LOX

LTB4 LTC4
Lipoxin LTD4
15deoxy 12-14PGJ 2 hydrolayses LTE4

Lipoxin A4
Lipoxin B4

Fig. 3. Eicosanoid biosynthesis from AA. Cyclooxygenase (COX)-1,2 metabolise AA to PGH2 by sequential cyclooxygenation and hydroperoxidation reactions. PGH2 is converted via specic
synthases generating thromboxane A2 (TXA2) and the prostaglandins including PGE2, PGI2, PGD2 and PGF2. These products can be non-enzymatically converted into further metabolites,
including 15-deoxy-12,14-PGJ2. 5-lipoxygenase (LOX) binds 5-lipoxygenase activating protein (FLAP) upon cell activation converting AA to 5-HPETE, which is further metabolized to LTA4.
LTA4 can also be converted into LTB4 by LTA4 hydrolase, or the cysteinyl leukotrienes (Cys-LTs) LTC4, LTD4 or LTE4. LTA4 can be metabolised by platelet 12-LOX to lipoxin A4 and lipoxin B4.
15-LOX also metabolises AA to generate 5-HEPTE, which is further converted by 5-LOX and lipoxin hydrolases to lipoxin A4 and lipoxin B4.
C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282 275

(Lauritzen et al., 2001)). Following dietary supplementation, the levels of of eicosanoid production to a less inammatory prole and could be ben-
EPA and DHA in cellular phospholipids increases, sometimes at the ex- ecial in the context of chronic inammatory disease. Interestingly some
pense of AA (Healy et al., 2000). This is important in the context of an in- AA derived mediators do anti-inammatory properties. For example,
ammatory response, as EPA can be metabolised by the same enzymes PGE2 was equally able to inhibit endotoxin-induced TNF- secretion by
as AA to generate alternative series eicosanoids. Although the biological human mononuclear cells as PGE3 (Miles et al., 2002). Additionally, AA
functions of alternative series eicosanoids are poorly described, some derived PGI2 is a potent inhibitor of platelet aggregation and EPA
have been shown to have less pro-inammatory actions than the AA de- derived PGI3 has been show to be as effective in human platelets
rived analogues and some may even have anti-inammatory actions (Kobzar et al., 2001).
(Fig. 4). For example, EPA derived LTB5 is 1030 times less potent as a Competition between the metabolites of EPA with those derived
neutrophil chemoattractant than AA derived LTB4 (Strasser et al., from AA does explain some of the anti-inammatory effects of n3
1985). We have demonstrated that treating EC with EPA results in syn- PUFAs. In addition free DHA and EPA can modulate inammation by
thesis of PGD3 via COX enzymes, which antagonised the neutrophil binding to and activating plasma membrane-bound and cytosolic
PGD2 receptor, inhibiting neutrophil migration (Tull et al., 2009). In- receptors such as GPR120 and the members of the PPAR family of
creasing the ratio of n3:n6 PUFAs may therefore shift the balance transcription factors (Im, 2012). GRP120 is a G-protein coupled receptor
(GPCR), expressed at high levels in macrophages and adipocytes
(Miyauchi et al., 2009), which binds unsaturated fatty acids with a
Membrane
A phospholipids chain length of 1622 carbons (Hirasawa et al., 2005), including EPA
and DHA. In the Raw 264.7 macrophage cell line, DHA inhibited TNF-
and LPS induced cytokine production, with an EC50 of 110 M, an effect
lost in the GPR120 knockdown (although most experiments were per-
Eicosapentaenoic
acid formed with 100 M DHA) (Oh et al., 2010). A similar dependence on
(EPA) GRP120 to mediate anti-inammatory effects of DHA was observed in
Acetylated
COX-1,2 5-LOX COX-2 or the GRP120/ mouse (Oh et al., 2010). These recent ndings suggest
15-LOX an important role for GPR120 in mediating the anti-inammatory
effects of n3 PUFAs. The peroxisome proliferator-activated receptor
(PPAR) family regulate transcription of genes involved in lipid metabo-
PGG3/PGH3
5-HPETE lism and homeostasis by forming heterodimers with retinoid X
18R HPETE receptors (RXR) (Moraes et al., 2006). In addition, PPAR activation
may inhibit pro-inammatory gene transcription, in part by inhibiting
NFB activation (reviewed in Moraes et al., 2006). Of interest both
LTA5 DHA and EPA bind PPARs and in vitro EPA binds PPAR-, and with
TXA3 PGE3 PGI3 PGD3 PGF3 5-LOX IC50 of 14 M (Xu et al., 1999). Binding of free n3 PUFAs to these
receptor classes may represent an alternative mechanism of action.
The concept that there are specic and inducible pathways which
LTB5 LTC5
LTD5 RvE 1,2 support the resolution phase of inammation is attractive, and one
LTE5 that is receiving much attention. Within this eld, specialised pro-
resolving mediators (SPM) derived from EPA and DHA, the resolvins
B Membrane and protectins, have generated signicant interest (Serhan, 2010)
phospholipids
(Fig. 4A and B), and synthetic versions of these molecules have anti-
inammatory and inammation resolving efcacy in vitro, and when
provided exogenously in vivo. The rst pro-resolving mediator to be de-
Docosahexaenoic
acid scribed was resolvin E1 (RvE1), synthesised via the metabolism of EPA
(DHA) by aspirin acetylated COX-2 and transcellular processing by neutrophil
Acetylated
COX-2
15-LOX 5-LOX (Serhan et al., 2000). The precursor of RvE1, 18R-HEPE, can be
synthesised from EPA by microbial cytochrome P450 and is subsequent-
ly metabolised by host 5-LOX to RvE1 in the absence of aspirin (Arita,
Bianchini, et al., 2005, Arita, Clish, et al., 2005). DHA derived resolvins
17R HDHA 17S HDHA were rst described in peritoneal exudates and whole mouse brain
in the presence of aspirin (Serhan et al., 2002), although aspirin is
Epoxygenation not required for the synthesis of D-series resolvins by neutrophil
Enzyme hydrolysis
5-LOX (Hong et al., 2003). Following on from these studies, a series of
5-LOX DHA derived molecules with pro-resolution properties, the protectins,
NPD1 neuroprotectins and maresins, have been identied (Hong et al., 2003;
PD1 Serhan et al., 2009). Recent studies have described the detection of en-
dogenous SPM in murine models of infection (Chiang et al., 2012) and
RvD 1-4
inammation (Oh et al., 2012; Dalli et al., 2013). In a self-resolving
Fig. 4. n3 PUFA derived lipid mediators. A. EPA liberated from membrane phospholipids model of E. coli infection the precursor 17-HDHA and its downstream
is metabolised by COX-1 or 2 to the common precursor PGH3. Subsequent conversion by products, protectin D1 and RvD5, were detected in peritoneal exudates
synthases results in TXA3, PGE3, PGI3, PGD3 and PGF3. 5-LOX also metabolises EPA to
in the ng range (Chiang et al., 2012). In addition, administration of phys-
the 5-series leukotrienes via the formation of 5-HPETE. B. DHA liberated from membrane
phospholipids is metabolised by COX-1 or 2 to the common precursor PGH3. Subsequent iological concentrations of RvD1 or RvD5, in a model of lethal E. coli
conversion by synthases results in TXA3, PGE3, PGI3, PGD3 and PGF3. 5-LOX also infection, led to an increase in macrophage bacterial load and increased
metabolises EPA to the 5-series leukotrienes via the formation of 5-HPETE. EPA is also survival (Chiang et al., 2012). Both RvE2 and the intermediate 18-HEPE
converted by acetylated COX-2 or 15-LOX to 18RHPETE, which is converted via 5-LOX to have been detected in peritoneal exudates from mice with self limiting,
the RvE1 or RvE2. B. DHA is converted by acetylated COX-2 or 15-LOX to 17R HDHA or
17S HDHA respectively. 5-LOX converts both intermediates to the RvD series 14.
zymosan-induced, peritonitis (Oh et al., 2012). Similarly DHA derived
Additionally 17S HDHA can undergo epoxygenation and enzymatic hydrolysis to produce RvD1, 3, 5 and aspirin triggered RvD3 are present in exudates from
PD1, or NPD1 in neural tissue. the same mouse peritonitis model (Dalli et al., 2013).
276 C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282

The human serum metabolome has been published which, using per week of oily sh or with sh oil (Burr et al., 1989). It was the rst
multiple methodologies, has produced a comprehensive data set of me- controlled trial which examined the effect of dietary sh (approx.
tabolites in human serum (Psychogios et al., 2011). This includes levels 300 g/week) or sh oils (500900 mg EPA + DHA/d) on the secondary
of DHA (4.7 3.3 M) and EPA (1.1 0.7 M), and the SPMs RvD1 prevention of myocardial infarction. Overall there was a 29% reduction
(0.0454 5 0.027 nM) and RvE1 (0.52 0.98 nM) in serum samples in all-cause mortality risk during the rst two years, compared to the
from healthy humans by LC-MS/MS (Psychogios et al., 2011). Addition- control group (Burr et al., 1989). The GISSI-Prevenzione trial examined
ally, SPM have been detected in plasma and serum of 20 healthy 11,324 patients who had recently survived a myocardial infarction
human subjects following 3 weeks supplementation with 1 g/d DHA (GISSI-Prevenzione Investigators, 1999). Subjects were randomised to
and 1.4 g/d EPA; RvD1 at 24.4 2.5 pg/ml, RvD2 at 26.6 4.7 pg/ml n3 PUFA (885 mg EPA + DHA/d) and/or vitamin E, or no supplemen-
and 17R-RvD1 at 53.3 6.0 pg/ml (serum measurements) (Mas tation, with up to 3.5 year follow up. The primary combined endpoint
et al., 2012). Interestingly, the concentrations reported are within the was death, non-fatal myocardial infarction and stroke. n3 PUFAs sig-
range at which SPM have anti-inammatory and pro-resolving proper- nicantly reduced this endpoint (by 15%), almost entirely attributed
ties in in vivo and in vitro models (Mas et al., 2012). The receptors re- to the reduction in cardiovascular death (GISSI-Prevenzione Investiga-
sponsible for mediating the effects of SPM are starting to be identied. tors, 1999). The Japan EPA Lipid Intervention Study, (JELIS), set out to
RVE1 binds and activates the LTB4 receptor, BLT1 on neutrophils and test the hypothesis that EPA would be effective as a preventative mea-
ChemR23 (Arita, Bianchini, et al., 2005, Arita, Clish, et al., 2005; Arita sure against a major coronary event in 18,645 hypercholersterolaemic
et al., 2007), expressed in macrophages and neutrophils. Both cell Japanese patients taking statins (Yokoyama et al., 2007). There was a
types also express the ALX receptor and GPR32 which recognise RvD1 19% reduction in the risk in subjects who were randomised to 1.8 g/d
(Krishnamoorthy et al., 2010). EPA plus 5 mg/d simvastatin or 10 mg/d pravastatin compared to
These molecules switch the inammatory environment to one statins alone (Yokoyama et al., 2007). Effects of n3 PUFAs on mortality
which promotes tissue homeostasis by inhibiting further neutrophil from heart failure was addressed in the GISSI-HF trial (GISSI-HF Investi-
recruitment, blocking the production of pro-inammatory PGs, LTs gators, 2004). The trial followed 7000 patients randomised to 850 mg
and cytokines, and promoting the restoration of tissue homeostasis by EPA + DHA/d, or a placebo control, for a median of 3.9 years. All
inducing macrophage phagocytosis of apoptotic cells (Serhan, 2010). cause mortality was reduced in the n3 PUFA group by 9% indicating
These molecules represent an opportunity to pharmacologically address that n3 PUFAs can confer benet in addition to conventional
non-resolving chronic diseases by inducing a programme of resolution treatment.
by which diseased tissues are returned to a normal. Future studies Although these four studies reported positive outcomes, three re-
are required to assay the levels of SPM in plasma in a larger cohort of cent trials report no effect of n3 PUFA on the same CVD outcomes.
healthy subjects and those with inammatory disease. In addition the Subjects in the SU.FOL.OM3 trial had a history of myocardial infarction,
effect of dietary supplementation with n3 PUFAs on SPM levels in ischaemic stroke, or unstable angina (Galan et al., 2010). The study
healthy and diseased subjects is of interest and may offer insight into aimed to test the impact of supplementing 2501 subjects with folate,
the roles of SPM under physiological and pathophysiological conditions. B vitamins and/or 600 mg/d EPA + DHA on fatal and non-fatal CVD
events with a median follow up of 4.7 years. No signicant effect of
2. n3 polyunsaturated fatty acids and cardiovascular disease n3 PUFAs was reported (Galan et al., 2010). The ALPHA-OMEGA
trial gave 4837 myocardial infarction survivors one of four trial marga-
Following the observations of lower risk of CVD observed in those rines, two of which provided 400 mg/d EPA + DHA, for 40 months
consuming traditional diets in Greenland, Alaska and Japan (Dyerberg (Kromhout et al., 2010). Again, EPA + DHA had no effect on the
et al., 1978; Kromann & Green, 1980; Bjerregaard & Dyerberg, 1988; rate of major CVD events (Kromhout et al., 2010). Finally, the OMEGA
Newman et al., 1993), studies in populations consuming a more trial supplemented 3851 survivors of myocardial infarction with
western diet were initiated to determine if n3 PUFA or oily sh ~900 mg/d EPA + DHA and followed them for one year (Rauch et al.,
consumption affected cardiovascular risk factors and reduced CVD 2010). n3 PUFAs had no affect on sudden cardiac death, total mortal-
morbidity or mortality. Varying outcomes were documented in the ity or major CVD events. These studies have limitations that include a
studies, which have been discussed in detail elsewhere (Kris-Etherton low dose of EPA + DHA, insufcient sample size, and recruitment of pa-
et al., 2002; Calder, 2004; von Schacky, 2004; Wang et al., 2006; tients long after the occurrence of a cardiovascular event. Alternatively
London et al., 2007); outcomes studied included coronary heart disease the background medication may inuence the efcacy of n3 PUFAs
rate and mortality, cardiovascular mortality, stroke incidence and myo- in secondary prevention of CVD events and mortality.
cardial infarction. Despite the conicting results reported in the intervention studies
Crucially, studies involving sh have been the subject of meta- discussed above, the potential mechanisms of action of n3 PUFAs in
analyses investigating either coronary heart disease mortality or stroke CVD continue to be extensively investigated. CVD encompasses many
incidence. (He et al., 2004a, 2004b) The rst of these (He et al., 2004a) conditions, all commonly underlined by the formation of atheromatous
analysed data from 13 cohorts and 11 independent studies, totalling plaques. These plaques have both a lipid and inammatory component
222,364 participants where both frequency of sh consumption and rel- (Libby et al., 2002) and develop as a consequence of EC activation lead-
ative risk of coronary heart disease mortality were reported. If the risk of ing to the inux of leukocytes into the intimal layer of the artery wall.
coronary heart disease in those eating sh less than once a month was n3 PUFAs are capable of modulating both EC and leukocyte function
set to 1, those consuming more portions, up to 5 per week, had signi- and it has been hypothesised that this may be responsible for the re-
cantly reduced (by 38%) risk (He et al., 2004a). In the study investigating duced CVD risk in populations consuming large amounts of oily sh.
the effect of sh consumption on stroke incidence, 9 cohorts and 8 To this end a large number of studies have investigated the effects of
independent studies comprising 200,575 participants were included EPA and DHA, either alone or in combination, on endothelial and leuko-
(He et al., 2004b). Similar to the coronary heart disease meta-analysis, cyte function. These will be discussed in detail below.
the risk of stroke decreased with greater sh intake. Indeed, in those One measure of the anti-inammatory properties of n3 PUFAs is
who ate 5 portions of sh per week the risk was reduced by 31% their ability to reduce cytokine induced expression of key EC adhesion
compared to those who never consumed sh (He et al., 2004b). molecules involved in leukocyte recruitment. Several studies have in-
Large intervention studies have investigated the effect of n3 vestigated this possibility using EC of varying origins, as well as different
PUFA supplementation in individuals with existing CVD. The Diet and stimuli, and different concentrations and durations of exposure to n3
Reinfarction Trial (DART), published in 1989, supplemented the diet of PUFAs. Human saphenous vein endothelial cells (HSVEC) treated with
1015 men who had survived a myocardial infarction with two portions 10 M DHA for 24 h, prior to 6 h TNF- stimulation, exhibited reduced
C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282 277

E-selectin protein and reduced VCAM-1 (vascular cell adhesion 26 weeks in healthy elderly subjects decreased PBMC expression of
molecule-1) protein and mRNA levels compared to TNF- stimulation genes associated with inammation and atherogenesis including NFB
alone (De Caterina et al., 1994). 65 M DHA and EPA attenuated IL-1 and eicosanoid synthesis (Bouwens et al., 2010).
induced ICAM-1 (intercellular adhesion molecule-1), VCAM-1 and E- In a study designed to compare the effects of DHA vs. EPA on a vari-
selectin mRNA in HUVEC (Collie-Duguid & Wahle, 1996). Conrming ety of leukocyte functions, participants were supplemented with oil rich
this data, 20160 M DHA inhibited TNF- induced VCAM-1 and in either DHA (4.9 g/d) or EPA (4.7 g/d) for 4 weeks. Neither DHA nor
ICAM-1 protein expression in human aortic endothelial cells (HAEC) EPA had any effect on the ability of isolated PBMCs to secrete cytokines,
(Wang et al., 2011). Our own studies indicate that 5 M DHA does not including TNF- and IL-10, in response to ex vivo stimulation with ConA
alter mRNA or total cellular levels of E-selectin protein, however the or LPS (Kew et al., 2004). In addition there was no effect of supplemen-
level of E-selectin expressed on the EC surface was signicantly down- tation on the ability of monocytes or neutrophils to phagocytose uo-
regulated due to disturbance of intracellular trafcking mechanisms rescently labelled E. coli or the expression of a number of monocyte
(Yates et al., 2011). The current evidence suggests a role for DHA in adhesion molecules, including CD18 and CD11b (Kew et al., 2004).
inhibiting cytokine-induced adhesion molecule expression. Mechanisti- DHA supplementation did reduce T lymphocyte expression of CD69,
cally this may be explained by inhibition of NF-B translocation to the an early marker of activation (Kew et al., 2004). Gorjao et al. investigat-
nucleus, for example, resulting in reduced transcription of VCAM-1 ed the effect of supplementation with 1.62 g/d DHA in 10 male subjects
(Wang et al., 2011). In addition EPA at a concentration of 25 M has for 8 weeks (Gorjo et al., 2006). They reported that DHA increased
been reported to inhibit EC surface expression of ICAM-1 (Goua et al., lymphocyte proliferation in response to ConA, increased monocyte
2008). and neutrophil phagocytosis of opsonised zymosan and increased neu-
The functional signicance of the modulation of EC expressed adhe- trophil chemotaxis to complement C5a. In vitro, 100 M DHA inhibited
sion molecules has been investigated in leukocyte adhesion assays. In LPS induced TNF- and IL-1 secretion by the THP-1 monocytic cell
vitro adhesion assays carried out in the absence of the hemodynamic line (Zhao et al., 2005), an observation conrmed in subsequent studies
factors imposed by ow demonstrate that DHA inhibits the adhesion (Weldon et al., 2007). There is also data indicating that EPA can modu-
of both THP-1 monocytes (De Caterina et al., 1994; Wang et al., 2011) late NFB activation in primary human monocytes (Zhao et al., 2004). In
and neutrophils (Wang et al., 2003) to EC. Our own ow based adhesion addition to modulating cytokine secretion, DHA (6 g/d) reduced LPS
studies indicate that DHA and EPA modulate different stages of leuko- stimulated PGE2 and LTB4 release by mononuclear cells (Kelley et al.,
cyte recruitment. Thus, DHA inhibited surface expression of E-selectin 1999). There is also data indicating that EPA can modulate up-stream
in response to TNF-, resulting in reduced recruitment of neutrophils signalling events which control pro-inammatory transcription factors.
from ow (Yates et al., 2011). EPA, in contrast, did not affect the number EPA inhibited NFB activation in the THP-1 cell line (Mullen et al., 2010)
of neutrophils recruited, but adherent neutrophils were unable to as well as in human monocytes by preventing phosphorylation and
migrate across the EC efciently. This was achieved by metabolism of subsequent degradation of the inhibitory subunit IkappaB-alpha (Zhao
EPA into the anti-inammatory eicosanoid PGD3, using the metabolic et al., 2004). A daily dose of 4 g n3 PUFA (EPA + DHA) for 9 months
pathways normally utilised to convert the n6 PUFA AA into the pro- signicantly reduced monocyte chemotaxis towards the bacterial peptide
inammatory agent PGD2. PGD3 effectively antagonised the functions formyl-methionyl-leucyl-phenylalanine (fMLP) (Schmidt et al., 1991).
of PGD2 which were necessary to support neutrophil transit across the
EC monolayer. 3. The use of n3 polyunsaturated fatty acids as a dietary interven-
The incorporation of DHA into membrane phospholipids may alter tion in asthma
the nature of cholesterol rich microdomains which play key roles in
anchoring receptors and signalling molecules (Insel & Patel, 2009). For Asthma is a chronic inammatory disease of the airways; sufferers
example, in EC supplemented with 100 M DHA, cholesterol was are hypersenstitve to specic allergens and exaggerated Th2-like cellu-
displaced from lipid rafts, resulting in reduced association of src family lar responses are considered to be crucial for the initiation and progres-
kinases with the raft (Chen et al., 2007), indicating that DHA may also sion of the disease (Georas et al., 2005). AA derived eicosanoids
modulate EC signalling. In addition 50 M DHA has been shown to including LTB4, LTC4, and LTD4 are thought to be important mediators
displace caveolin-1 and endothelial nitric oxide synthase (eNOS) from of airway inammation and airway obstruction in asthma (Hallstrand
caveolae in EC (Li et al., 2007). & Henderson, 2010). Indeed, LTB4 is increased in bronchoalveolar lavage
The ability of n3 PUFAs to modulate leukocyte function has been (BAL) uid of asthmatics compared with healthy controls (Wardlaw
investigated both in vitro using FFA as the supplemental lipids, but et al., 1989) and may be responsible for the inux of neutrophils, eosin-
more importantly, ex vivo after supplementing trial participants with ophils and mast cells into the lung (Ohnishi et al., 2008). Owing to the
DHA, EPA or a mixture of these, usually in the form of sh oils. Studies important role of n6 PUFA derived mediators in asthma, the hypoth-
using sh oil are too numerous to examine in detail and only a few esis that a diet high in these lipids might contribute to the onset of
will be discussed here. PBMCs (peripheral blood mononuclear cell) iso- asthma was postulated (Black & Sharpe, 1997). Moreover, rebalancing
lated from 9 subjects supplemented with 18 g/d of sh oil concentrate the dietary prole of n3 PUFAs and n6 PUFAs might prevent, or at
secreted reduced levels of IL-1 and TNF- in response to stimulation the least reduce, morbidity associated with the disease.
with LPS (lipopolysaccaride; endotoxin) (Endres et al., 1989). 9.4 g/d Many dietary intervention studies have been conducted, mostly
EPA + 5 g/d DHA for 10 weeks resulted in decreased neutrophil che- with sh oils rather than individual n3 PUFAs. Measured endpoints
motaxis to LTB4 (Sperling et al., 1993). Supplementation of 28 healthy for efcacy have been retarding disease onset and/or reducing patho-
male volunteers with 1.62 g/d EPA + 1.08 g/d DHA for 4 weeks logical changes occurring in the lung. Epidemiological data indicated
reduced secretion of TNF- and IL-1 by PBMCs, in response to LPS com- that children who did not eat sh were three times more likely to devel-
pared to secretion prior to dietary supplementation; the reduction in cy- op asthma than those who ate oily sh more than once a week (Hodge
tokine secretion inversely correlated with the EPA content of et al., 1996). Other studies suggest a positive association of oily sh
mononuclear cells (Caughey et al., 1996). 2.3 g/d n3 PUFAs for 3 - intake with lung function and fewer asthma symptoms (Smit et al.,
months reduced ex vivo IL-1 and TNF- secretion by PBMCs in response 1999). The data from clinical trials is more varied. Randomised con-
to concanavalin A (ConA) or phytohaemagglutinin (PHA) (Meydani trolled trials (RCTs) have investigated if n3 PUFA supplementation
et al., 1991). Trebble et al. demonstrated that a regime supplementing has any effect on established disease in adults. Twelve asthmatic
16 healthy men with increasing concentrations of EPA + DHA over patients were randomised to either low dose (0.1 g/d) or high dose
12 weeks (0.32.0 g/d) inhibited TNF- and IL-6 production in re- (4.0 g/d) EPA for 8 weeks, and lung function tests carried out before
sponse to LPS (Trebble et al., 2003). 1.8 g/d DHA and EPA for and during supplementation (Kirsch et al., 1988). The authors reported
278 C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282

no effect of EPA on clinical status of disease or lung function. Arm et al. inammation, PGE2 and LTB4 have been detected in synovial uid
(Arm et al., 1988) investigated the effect of n3 PUFA supplementation (Sperling, 1995) and are thought to contribute to the inltration of leu-
on mild asthma. 12 asthmatics were randomised to receive 3.2 g kocytes into the diseased tissue, both being powerful chemo-attractants
EPA + 2.2 g DHA/d and 8 to receive placebo (olive oil) for 10 weeks. for cells such as neutrophils.
A number of clinical parameters relating to lung health were measured In view of the role for AA derived lipid mediators in RA, the utility of
at baseline and 10 weeks including airway responsiveness to histamine supplementing the diet with n3 PUFAs to reduce the level of AA de-
and exercise, diurnal peak expiratory ow, symptom scores and bron- rived eicosanoids and to promote formation of n3 PUFA derived
chodilator use. In addition neutrophil fatty acid composition was deter- agents with putative anti-inammatory activity, has been tested . One
mined and calcium ionophore induced LTB4 and LTB5 synthesis, as well of the rst studies to address this hypothesis supplemented the
as neutrophil chemotaxis towards fMLP, was measured. There was no diet of patients with 1.8 g EPA for 12 weeks and reported both a re-
effect of n3 PUFAs on any of the clinical measurements of pulmonary duction in joint stiffness in the morning and in the number of tender
function, despite the fact that there was a signicant increase in neutro- joints (Kremer et al., 1985). Since then numerous trials have been
phil EPA levels, a 50% inhibition of LTB4 + LTB5 synthesis and a signi- reported with the supplemental levels of DHA and EPA varying
cant reduction in neutrophil chemotaxis. A study comparing 15 to from 1.6 g/d to 7.1 g/d (Kremer et al., 1995, Remans et al., 2004,
20 ml/d of evening primrose oil or sh oil with an olive oil placebo for Calder, 2006). An overview of placebo controlled studies using long
10 weeks as a supplementary treatment of asthmatic patients reported chain n3 fatty acids and published prior to 2006, concluded that
no effect of either oil on any clinical readout (Stenius-Aarniala et al., there is sound evidence suggesting that long-chain n3 PUFAs
1989). 46 patients with mild to moderate asthma were randomised to have some clinical benets in rheumatoid arthritis (Calder, 2006).
receive 50 mg/d of DHA + EPA plus 100 mg/d olive oil or 150 mg/d Similarly a meta-analysis published in 2007 set out to provide infor-
olive oil (as a placebo) for 2 months (Emelyanov et al., 2002). Compared mation concerning the efcacy of n3 PUFA supplementation on pain
to the placebo group, daytime wheezing was signicantly reduced in management, primarily in RA, but also in inammatory bowel disease
the n3 PUFA group. However there was no difference in the forced (IBD) and dysmenorrhea (Goldberg & Katz, 2007). 17 studies were
expiratory volume in one second (FEV1), a measure of airway obstruc- included which had supplemented over 800 patients. The dosage of n
tion, between groups. A smaller study supplemented 7 individuals 3 PUFA ranged from 1.7 to 9.6 g/d in the following combinations:
with 3 g/d DHA + EPA and reported a signicant decrease in airway re- EPA or ALA alone; a combination of DHA and EPA; a combination of
sponsiveness following challenge with an allergen. In agreement with DHA, EPA and ALA; krill oil (n3 content not reported); DHA and
Emelyanov et al. (2002), there was no change in FEV1 following n3 docosapentaenoic acid (DPA; 22:5n3) and EPA. Pain was quantied
PUFA supplementation. Conversely a study has reported a positive effect using six different measures: patient assessed, physician assessed, dura-
on FEV1 following 9 months of supplementation with 1 g/d DHA + EPA tion of morning joint stiffness, number of painful joints, joint tenderness
compared to a control group (Dry & Vincent, 1991). A recent meta- (Ritchie articular index (Ritchie et al., 1968)) and NSAID consumption.
analysis and systematic review of the use of n3 PUFA containing sup- The meta-analysis concluded that n3 PUFAs reduce the intensity of pa-
plements in asthma, suggested that supplementation with sh oil was tient assessed joint pain, joint stiffness in the morning, number of painful
unlikely to be useful in the primary prevention of asthma (or other aller- and/or tender joints and NSAID consumption. The authors suggest that in
gic diseases including eczema, allergic rhinitis and food allergies) order to maximise therapeutic effects future trials should supplement
(Anandan et al., 2009). The conclusions drawn about efcacy in asthma with N2.7 g/d EPA + DHA and should last for a minimum of 3 months.
were based on data from four studies which assessed disease incidence Overall n3 PUFAs appear to reduce pain and therefore have an
in high and low risk cohorts (Marks et al., 2006, Dunstan et al., 2003; NSAID sparing effect in patients with RA. This is potentially important
Lauritzen et al., 2005, Olsen et al., 2008). A comprehensive report from as prolonged NSAID use is associated with an increased risk of develop-
a United States governmental agency also concluded there was not ing gastrointestinal ulcers and gastrointestinal bleeding (Traversa et al.,
enough consistent evidence from such studies to state denitively that 1995), therefore any intervention which may reduce NSAID use could
n3 PUFAs were or were not effective in improving respiratory out- have signicant benet to patient welfare.
comes in adults or children (Schachter et al., 2004).
5. Inammatory bowel diseases and n3 polyunsaturated fatty acids
4. The use of n3 polyunsaturated fatty acids as a
dietary intervention in rheumatoid arthritis Inammatory bowel diseases (IBD) encompass two main subtypes
of disease, ulcerative colitis (UC) and Crohn's disease (CD) (Abraham
Rheumatoid arthritis (RA) is a disease characterised by chronic in- & Cho, 2009). Both are chronic inammatory diseases; UC mainly affects
ammation of the joints. It can be an extremely painful and debilitating the colon, whereas CD can occur in any part of the gastrointestinal (GI)
condition which affects any peripheral joint, but most commonly the tract. The precise causative agent of IBD is unknown, however mounting
feet, knees and hands (Smolen et al., 1995). The aetiological agent re- evidence indicates that inappropriate activation of the immune system
sponsible for triggering the onset of RA (if such an agent exists) is un- by intestinal bacteria is key (Cho, 2008). Interestingly, in the absence of
known. However, it is hypothesised that infection or traumatic insult enteric bacteria in a rat model of colitis, there was no evidence of
may induce the initial inammatory response in the synovial lining of inammation (Sellon et al., 1998). Familial and twin studies have dem-
the affected joint(s) (Firestein et al., 1987). Healthy joints do not contain onstrated the role of genetics in disease, indeed polymorphisms in
leukocytes; however in RA, neutrophils, B lymphocytes, T lymphocytes genes associated with microbial defence and intestinal immunity are as-
and monocyte-derived macrophages are abundant (Firestein et al., sociated with increased disease susceptibility (Cho, 2008). Environmen-
1987). As the disease develops, the cells of the synovial lining prolifer- tal factors, including smoking, the composition of the diet and hygiene,
ate, forming an invasive pannus which ultimately results in loss of have been linked with the development of IBD (Koutroubakis et al.,
cartilage, bone destruction and dramatic loss of function (Wong & 1996). The gut mucosa of healthy individuals ordinarily contains macro-
Lord, 2004). Cytokines and chemokines, including IL-1, IL-6 and TNF- phages, T cells and dendritic cells which secrete both pro- and anti-
, have been detected in the synovial uid of patients with RA and inammatory cytokines. There is a balance maintained which works
have been implicated in the development of the disease (Brennan & to both restrict excessive levels of enteric microbiota and protect against
McInnes, 2008). Indeed, all of these mediators are therapeutic targets infections. In IBD this balance between the immune system and gut ora
in RA, with anti-TNF- treatment proving highly effective at controlling is disrupted due to greater intestinal permeability and the breakdown of
disease progression in a signicant proportion of patients (Moreland epithelial tight junctions (Abraham & Cho, 2009). This leads to increased
et al., 1997). Importantly, the AA derived lipid mediators of inltration of neutrophils and T cells and enhanced levels of pro-
C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282 279

inammatory cytokines including TNF- (Sands & Kaplan, 2007). Here respectively. Despite the lack of effect on relapse, there are reports of
we discuss whether n3 PUFA as a dietary supplementation may be a decreased gut mucosal disease activity and improved histological scores
useful tool in the primary prevention or management of the disease. following supplementation (Stenson et al., 1992, Almallah et al., 1998,
The European prospective cohort study investigated the risk of de- Almallah et al., 2000).
veloping UC with dietary fatty acid intake in 139 patients compared Interestingly although there appears to be some benet of n3
with age and sex matched controls (Hart et al., 2008). There was no PUFA in IBD, meta-analyses have failed to conrm this. MacLean et al.
correlation between saturated or monounsaturated intake and UC, (2004) published a meta-analysis of 13 studies of sh oil supplementa-
however there was a slight increase in disease risk with total PUFA in- tion in IBD and concluded there was sufcient data to perform meta-
take (Hart et al., 2008). Further analysis of the study revealed that the analysis for relapse in UC only. The analysis concluded that n3 fatty
high intake of the n6 PUFA LA was responsible for the increased UC acids have no effect on relative risk of relapse in ulcerative colitis and
risk, and there was no association with DHA or EPA intake (The IBD in there was a statistically non-signicant reduction in requirement for
EPIC Study Investigators, 2009). A prospective cohort study of 25,639 corticosteroids for n3 fatty acids relative to placebo in two studies
patients between the ages of 4574 years used 7 d food diaries to inves- (MacLean et al., 2004). The most recent meta-analysis set out to review
tigate the hypothesis that n3 PUFA intake is protective in UC (John the efcacy and safety of n3 PUFA in the maintenance of remission in
et al., 2010). Total dietary n3 PUFAs, in particular DHA, were associat- IBD in three trials of UC and six of CD (Turner et al., 2011). Having exam-
ed with protection from UC in this cohort (John et al., 2010). Overall ined the pooled data the authors found a higher rate of diarrhoea and
these data suggest that n3 PUFA intake is associated with a decreased symptoms of the GI tract in the n3 PUFA treatment group. Overall
risk, and higher n6 intake with an increased risk of developing UC. the conclusion was there are insufcient data to recommend the use
This protective effect of n3 PUFAs may be due to modulation of of omega 3 fatty acids for maintenance of remission in CD and UC. It
inammatory events which take place within the GI environment therefore appears that although there is some benet reported in ani-
(Vilaseca et al., 1990). mal models of IBD, in particular UC, there is limited data to suggest
Rat colitis models demonstrate that inammation in the colonic that this translates into benet for human patients.
mucosa results in an increase in the levels of AA derived LTB4 and
PGE2 (Hirata et al., 2001, Nieto et al., 2002) and mucosal LTB4 is also el- 6. Conclusion
evated in humans with IBD (Sharon & Stenson, 1984). LTB4 is a potent
leukocyte chemoattractant and plays a role in mediating the inamma- The marine n3 PUFAs EPA and DHA exert a range of anti-
tion associated with IBD. Inhibition of the LT biosynthetic pathway inammatory effects including reduced eicosanoid, cytokine and adhe-
in colitic rats reduced mucosal injury and decreased levels of LTB4 sion molecule production, enhanced SPM production and decreased
(Empey et al., 1992). Interestingly, this reduction in LTB4 was accompa- leukocyte-EC adhesive interactions. These effects suggest that n3
nied by an increase in PGE2, suggesting a shift towards PG synthesis PUFAs would be useful as therapies in patients with inammatory con-
(Empey et al., 1992). PGE2 has reported anti-inammatory actions, in- ditions. However, data from patient or laboratory based studies can be
cluding inhibition of 5-LO, resulting in reduced 4-series LT synthesis difcult to interpret due to variations in dosage, treatment length,
(Levy et al., 2001). confounding medication, or translating evidence from the laboratory
IBD clearly has an inammatory component, consequently a number to the clinic. Nevertheless, supplementation with n3 PUFAs may be
of animal studies have taken place to assess the efcacy of n3 PUFAs in benecial in some settings such as RA, although high doses are required.
IBD. Many of these studies used chemically induced models of colitis. The lack of consistent data in other diseases such as asthma and IBD sug-
In a UC model, induced using trinitrobenzene sulphonic acid (TNBS), gests that n3 PUFAs may not be benecial in all settings, despite the
rats supplemented with cod liver oil had reduced colonic damage and presence of common cells and mediators in these different conditions.
inammation compared to the sunower oil group (Vilaseca et al., One explanation for this could be that n3 PUFAs may not have the
1990). Using the same TNBS model, sh oil supplementation resulted same effects on inammation in healthy, compared with diseased indi-
in lower colonic inammation and a decrease in LTB4 in colonic tissue viduals. There is the possibility that long term supplementation of
(Yuceyar et al., 1999). Recently in two mouse models of colitis, systemic healthy individuals may reset the inammatory threshold of the indi-
treatment with aspirin triggered resolvin D1 (RvD1), the precursor vidual, providing a degree of protection from cumulative inammatory
(17(R)-hydroxy docosahexaenoic acid [17R-HDHA]) or resolvin D2 injury. However, at least in the context of advanced arterial disease,
(RvD2) resulted in an improved disease activity score, reduced colonic short term supplements may not be effective at reversing systemic in-
damage and a decrease in neutrophil inltration (Bento et al., 2011). ammation associated with the disease process. This has been reviewed
Many human supplementation studies in IBD do not assess the (Sijben & Calder, 2007), and current data suggests that supplementation
ability of n3 PUFAs to modulate cellular readouts of inammation of healthy subjects with n3 PUFAs may be effective in preventing the
but report the effect on clinical outcomes. However, it has been reported onset of chronic inammatory diseases such as CVD, whereas those
that n3 PUFAs are incorporated into the colonic mucosa of patients with established disease may receive less benet from dietary supple-
with IBD and that sh oil reduces LTB4 secretion from patient's neutro- mentation with n3 PUFAs. CVD and a number of other chronic inam-
phils (McCall et al., 1989, Hawthorne et al., 1992; Shimizu et al., 2003). matory diseases are associated with ageing, and it is possible that ageing
Randomised, placebo-controlled, double-blind studies of sh oil in IBD itself may inuence the effectiveness of supplementation. Although
have been published. One of the rst studies supplemented both UC there are data on the effect of n3 PUFA in the elderly in regards to
and CD patients with 1.8 g/d EPA + 1.3 g/d DHA for 7 months cognitive function (Denis et al., 2013), this idea has not been thoroughly
(Lorenz et al., 1989). Clinical activity was unchanged in CD patients investigated in chronic inammatory disease.
with supplementation but fell in UC patients, although this did not Underlying genetic factors controlling the processing of dietary
reach signicance. In a 12 month RCT, 87 patients received 4.5 g/d lipids may offer a further explanation as to why supplementation has
EPA and 1.1 g/d DHA (Hawthorne et al., 1992) Supplementation in- varying effects in individuals (Madden et al., 2011). Recent genome
creased the rectal mucosa EPA to 3.2% of total fatty acids compared to wide association study (GWAS) studies have identied new loci in-
0.63% in the placebo olive oil group. There was limited clinical benet; volved in lipid metabolism (Kim et al., 2011; Teslovich et al., 2010). Of
patients who started the trial in relapse reported signicantly reduced interest is sirtuin 1 (SIRT1) a member of NAD+ dependant histone
corticosteroid use after one and two months of n3 PUFA, but there deacetylase family, which can target nuclear receptors/transcription
was no reduction in the rate of relapse (Hawthorne et al., 1992). Simi- factors related to lipid metabolism (Finkel et al., 2009). A recent
larly both Lorenz-Meyer et al. (1996) and Feagan et al. (2008) reported Japanese study of N1400 individuals linked SITR1 haplotype 2 with
no effect on relapse with n3 PUFA supplementation in UC and CD low circulating levels of LDL and high circulating HDL in those who
280 C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282

consumed a diet with a low n6/n3 ratio(Inamori et al., 2013). In Bouwens, M., Grootte, Bromhaar M., Jansen, J., Mller, M., & Afman, L. A. (2010). Postpran-
dial dietary lipid-specic effects on human peripheral blood mononuclear cell gene
addition a variant of the n3 lipid sensing GPR120 gene, R270H, has expression proles. Am J Clin Nutr. 91, 208217.
been identied (Ichimura et al., 2012). Expression of this polymorphism Brennan, F. M., & McInnes, I. B. (2008). Evidence that cytokines play a role in rheumatoid
in a European population is strongly associated with obesity in humans arthritis. J. Clin. Invest. 118, 35373545.
Burr, M. L., Fehily, A.M., Gilbert, J. F., Rogers, S., Holliday, R. M., Sweetnam, P.M., et al.
and inhibits signalling via GPR120 (Ichimura et al., 2012). Whether this (1989). Effects of changes in fat, sh, and bre intakes on death and myocardial
gene mutation affects the ability of people to respond to dietary n3 reinfarction: diet and reinfarction trial (DART). Lancet 2, 757761.
PUFA supplementation has not been addressed, however these data Calder, P. C. (2004). n3 Fatty acids and cardiovascular disease: evidence explained and
mechanisms explored. Clin. Sci. (Lond.) 107, 111.
raise interesting questions about the role of genetics in lipid metabolism. Calder, P. C. (2006). n-3 polyunsaturated fatty acids, inammation, and inammatory dis-
Genetic factors may direct the way individuals respond to dietary lipid in- eases. Am J Clin Nutr 83, 1505S1519S.
take and could indicate why there is inter-individual heterogeneity in re- Caughey, G. E., Mantzioris, E., Gibson, R. A., Cleland, L. G., & James, M. J. (1996). The ef-
fect on human tumor necrosis factor alpha and interleukin 1 beta production of
sponse to n3 PUFA supplementation. Further studies are required to
diets enriched in n3 fatty acids from vegetable oil or sh oil. Am. J. Clin. Nutr.
determine if interactions between genetic and modiable environmental 63, 116122.
factors occur and, if so, whether they could exert protective functions. Cawood, A. L., Ding, R., Napper, F. L., Young, R. H., Williams, J. A., Ward, M. J. A., et al.
The current literature indicates that dietary supplementation may (2010). Eicosapentaenoic acid (EPA) from highly concentrated n3 fatty acid ethyl
esters is incorporated into advanced atherosclerotic plaques and higher plaque
be benecial in some chronic inammatory diseases. However, there EPA is associated with decreased plaque inammation and increased stability.
is now an opportunity to identify novel approaches to preventing and Atherosclerosis 212, 252259.
reversing/resolving chronic inammatory disease based upon the Chen, W., Jump, D. B., Esselman, W. J., & Busik, J. V. (2007). Inhibition of cytokine signaling
in human retinal endothelial cells through modication of caveolae/lipid rafts by
downstream metabolic products of EPA and DHA and their mechanisms docosahexaenoic acid. Invest. Ophthalmol. Vis. Sci. 48, 1826.
of action or the interaction of these n3 PUFAs with free fatty acid Chiang, N., Fredman, G., Backhed, F., Oh, S. F., Vickery, T., Schmidt, B.A., et al. (2012).
receptors. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature
484, 524-285.
Cho, J. H. (2008). The genetics and immunopathogenesis of inammatory bowel disease.
Nat. Rev. Immunol. 8, 458466.
Conict of interest statement Collie-Duguid, E. S., & Wahle, K. W. (1996). Inhibitory effect of sh oil N3 polyunsatu-
rated fatty acids on the expression of endothelial cell adhesion molecules. Biochem.
CMY and GER have no conict of interest to report. PCC serves on Biophys. Res. Commun. 220, 969974.
Dalli, J., Winkler, J., Colas, R., Arnardottir, H., Cheng, C. Y., Chiang, N., et al. (2013). Resolvin
Scientic Advisory Boards of the Danone Research Centre in Specialised D3 and aspirin-triggered resolvin D3 are potent immunoresolvents. Chem. Biol. 20,
Nutrition, Aker Biomarine, Pronova Biopharma and Smartsh. He acts as 188201.
a consultant to Mead Johnson Nutritionals, Vifor Pharma and Amarin De Caterina, R., Cybulsky, M., Clinton, S., Gimbrone, M., & Libby, P. (1994). The omega-3
fatty acid docosahexaenoate reduces cytokine-induced expression of proatherogenic
Corporation. He has received speaking honoraria from Solvay Healthcare, and proinammatory proteins in human endothelial cells. Arterioscler. Thromb. 14,
Solvay Pharmaceuticals, Pronova Biopharma, Fresenius Kabi, B. Braun, 18291836.
Abbott Nutrition, Baxter Healthcare, Nestle, Unilever and DSM. He cur- Denis, I., Potier, B., Vancassel, S., Heberden, C., & Lavialle, M. (2013). Omega-3 fatty acids
and brain resistance to ageing and stress: body of evidence and possible mechanisms.
rently receives research funding from Vifor Pharma. He is immediate
Ageing Res. Rev. 12, 579594.
Past-President of the International Society for the Study of Fatty Acids Dry, J., & Vincent, D. (1991). Effect of a sh oil diet on asthma: results of a 1-year
and Lipids, an organisation that is partly supported by corporate double-blind study. Int. Arch. Allergy Appl. Immunol. 95, 156157.
membership fees, mainly the food and supplements industries. He is a Dunstan, J. A., Mori, T. A., Barden, A., Beilin, L. J., Taylor, A. L., Holt, P. G., et al.
(2003). Maternal sh oil supplementation in pregnancy reduces interleu-
member of the Board of Directors of ILSI Europe, the Board of Directors kin-13 levels in cord blood of infants at high risk of atopy. Clin Exper Allergy
of the European Nutraceutical Association, and the Council of the British 33, 442448.
Nutrition Foundation; these organisations are each supported in part by Dyerberg, J., Bang, H. O., Stofferson, E., Moncada, S., & Vane, J. R. (1978). Eicosapentaenoic
acid and prevention of thrombosis and atherosclerosis. Lancet 2, 117119.
the food and supplements industries. Emelyanov, A., Fedoseev, G., Krasnoschekova, O., Abulimity, A., Trendeleva, T., & Barnes, P.
J. (2002). Treatment of asthma with lipid extract of New Zealand green-lipped mus-
sel: a randomised clinical trial. Eur. Respir. J. 20, 596600.
References Empey, L. R., Walker, K., & Fedorak, R. N. (1992). Indomethacin worsens and a leukotriene
biosynthesis inhibitor accelerates mucosal healing in rat colitis. Can. J. Physiol.
Abraham, C., & Cho, J. H. (2009). Inammatory bowel disease. N. Engl. J. Med. 361, Pharmacol. 70, 660668.
20662078. Endres, S., Ghorbani, R., Kelley, V. E., Georgilis, K., Lonnemann, G., van der Meer, J. W., et al.
Almallah, Y. Z., Richardson, S., O'Hanrahan, T., Mowat, N. A., Sinclair, T. S., Ewen, S., et al. (1989). The effect of dietary supplementation with n3 polyunsaturated fatty acids
(1998). Distal procto-colitis, natural cytotoxicity, and essential fatty acids. Am J on the synthesis of interleukin-1 and tumor necrosis factor by mononuclear cells.
Gastroentero 93, 804809. N. Engl. J. Med. 320, 265271.
Almallah, Y. Z., Ewen, S. W., El-Tahir, A., Mowat, N. A., Brunt, P. W., Sinclair, T. S., et al. Feagan, B. G., Sandborn, W. J., Mittmann, U., Bar-Meir, S., D'Haens, G., Bradette, M., et al.
(2000). Distal Proctocolitis and n-3 Polyunsaturated Fatty Acids (n-3 PUFAs): The (2008). Omega-3 Free Fatty Acids for the Maintenance of Remission in Crohn Disease.
Mucosal Effect In Situ. J Clin Immumol 20, 6876. JAMA 299, 16901697.
Anandan, C., Nurmatov, U., & Sheikh, A. (2009). Omega 3 and 6 oils for primary prevention Finkel, T., Deng, C. X., & Mostoslavsky, R. (2009). Recent progress in the biology and
of allergic disease: systematic review and meta-analysis. Allergy 64, 840848. physiology of sirtuins. Nature 460, 587591.
Arita, M., Bianchini, F., Aliberti, J., Sher, A., Chiang, N., Hong, S., et al. (2005). Stereochemical Firestein, G. S., Tsai, V., & Zvaier, N. J. (1987). Cellular immunity in the joints of patients
assignment, antiinammatory properties, and receptor for the omega-3 lipid mediator with rheumatoid arthritis and other forms of chronic synovitis. Rheum. Dis. Clin. N.
resolvin E1. J. Exp. Med. 201, 713722. Am. 13, 191213.
Arita, M., Clish, C. B., & Serhan, C. N. (2005). The contributions of aspirin and microbial Fliesler, S. J., & Anderson, R. E. (1983). Chemistry and metabolism of lipids in the verte-
oxygenase to the biosynthesis of anti-inammatory resolvins: novel oxygenase prod- brate retina. Prog. Lipid Res. 22, 79131.
ucts from [omega]-3 polyunsaturated fatty acids. Biochem. Biophys. Res. Commun. 338, Folco, G., & Murphy, R. C. (2006). Eicosanoid transcellular biosynthesis: from cellcell
149157. interactions to in vivo tissue responses. Pharmacol. Rev. 58, 375388.
Arita, M., Ohira, T., Sun, Y. P., Elangovan, S., Chiang, N., & Serhan, C. N. (2007). Resolvin E1 Galan, P., Kesse-Guyot, E., Czernichow, S., Briancon, S., Blacher, J., Hercberg, S., et al.
selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate (2010). Effects of B vitamins and omega 3 fatty acids on cardiovascular diseases: a
inammation. J. Immunol. 178, 39123917. randomised placebo controlled trial. Br. Med. J. 341, c6273.
Arm, J. P., Horton, C. E., Mencia-Hueta, J. M., Eiser, N. M., Clark, T. J., Spur, T. J., et al. (1988). Georas, S. N., Guo, J., De Fanis, U., & Casolaro, V. (2005). T-helper cell type-2 regulation in
Effect of dietary supplementation with sh oil lipids on mild asthma. Thorax 43, allergic disease. Euro. Respir. J26, 11191137.
8492. GISSI-HF Investigators (2004). Effect of n3 polyunsaturated fatty acids in patients
Bento, A. F., Claudino, R. F., Dutra, R. C., Marcon, R., & Calixto, J. B. (2011). Omega-3 fatty with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-
acid-derived mediators 17(R)-hydroxy docosahexaenoic acid, aspirin-triggered controlled trial. Lancet 372, 12231230.
resolvin D1 and resolvin D2 prevent experimental colitis in mice. J Immunol 187, GISSI-Prevenzione Investigators (1999). Dietary supplementation with n3 polyunsatu-
1957-69. rated fatty acids and vitamin E after myocardial infarction: results of the GISSI-
Bjerregaard, P., & Dyerberg, J. (1988). Mortality from ischaemic heart disease and cerebro- Prevenzione trial. Lancet 354, 447455.
vascular disease in Greenland. Int. J. Epidemiol. 17, 514519. Goldberg, R. J., & Katz, J. (2007). A meta-analysis of the analgesic effects of omega-3
Black, P. N., & Sharpe, S. (1997). Dietary fat and asthma: is there a connection? Eur. Respir. polyunsaturated fatty acid supplementation for inammatory joint pain. Pain 129,
J. 10, 612. 210223.
C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282 281

Gorjo, R., Verlengia, R., Lima, T. M., Soriano, F. G., Boaventura, M. F., Kanunfre, C. C., et al. Lauritzen, L., Hansen, H. S., Jorgensen, M. H., & Michaelsen, K. F. (2001). The essentiality of
(2006). Effect of docosahexaenoic acid-rich sh oil supplementation on human leu- long chain n3 fatty acids in relation to development and function of the brain and
kocyte function. Clin Nutr. 25, 923938. retina. Prog. Lipid Res. 40, 194.
Goua, M., Mulgrew, S., Frank, J., Rees, D., Sneddon, A. A., & Wahle, K. W. J. (2008). Lauritzen, L., Kjaer, T. M., Fruekilde, M. B., Michaelsen, K. F., & Frokiaer, H. (2005). Fish oil
Regulation of adhesion molecule expression in human endothelial and smooth supplementation of lactating mothers affects cytokine production in 2 1/2-year-old
muscle cells by omega-3 fatty acids and conjugated linoleic acids: Involvement children. Lipids 40, 669678.
of the transcription factor NF-kappa b? Prostaglandins Leukot. Essent. Fatty Acids Levy, B.D., Clish, C. B., Schmidt, B., Gronert, K., & Serhan, C. N. (2001). Lipid mediator class
78, 3343. switching during acute inammation: signals in resolution. Nat. Immunol. 2, 612619.
Hallstrand, T. S., & Henderson, W. R., Jr. (2010). An update on the role of leukotrienes in Ley, K., Laudanna, C., Cybulsky, M. I., & Nourshargh, S. (2007). Getting to the site of inam-
asthma. Curr. Opin. Allergy Clin. Immunol. 10, 6066. mation: the leukocyte adhesion cascade updated. Nat. Rev. Immunol. 7, 678689.
Hart, A. R., Luben, R., Olsen, A., Tjonneland, A., Linseisen, J., Nagel, G., et al. (2008). Diet in Li, D., Sinclair, A., Wilson, A., Nakkote, S., Kelly, F., Abedin, L., et al. (1999). Effect of dietary
the aetiology of ulcerative colitis: a European prospective cohort study. Digestion 77, alpha-linolenic acid on thrombotic risk factors in vegetarian men. Am. J. Clin. Nutr. 69,
5764, http://dx.doi.org/10.1159/000121412. 872882.
Hawthorne, A.B., Daneshmend, T. K., Hawkey, C. J., Belluzzi, A., Everitt, S. J., Holmes, G. K., Li, Q., Zhang, Q., Wang, M., Liu, F., Zhao, S., Ma, J., et al. (2007). Docosahexaenoic acid
et al. (1992). Treatment of ulcerative colitis with sh oil supplementation: a prospec- affects endothelial nitric oxide synthase in caveolae. Arch. Biochem. Biophys. 466,
tive 12 month randomised controlled trial. Gut 33, 922928. 250259.
He, K., Song, Y., Daviglus, M. L., Liu, K., Van Horn, L., Dyer, A.R., et al. (2004a). Accumulated Libby, P., Ridker, P.M., & Maseri, A. (2002). Inammation and atherosclerosis. Circulation
evidence on sh consumption and coronary heart disease mortality: a meta-analysis 105, 11351143.
of cohort studies. Circulation 109, 27052711. London, B., Albert, C., Anderson, M. E., Giles, W. R., Van Wagoner, D. R., Balk, E., et al.
He, K., Song, Y., Daviglus, M. L., Liu, K., Van Horn, L., Dyer, A.R., et al. (2004b). Fish (2007). Omega-3 fatty acids and cardiac arrhythmias: prior studies and recommen-
consumption and incidence of stroke. Stroke 35, 15381542. dations for future research. Circulation 116, e320e335.
Healy, D. A., Wallace, F. A., Miles, E. A., Calder, P. C., & Newsholm, P. (2000). Effect of Lorenz, R., Weber, P. C., Szimnau, P., Heldwein, W., Strasser, T., & Loeschke, K. (1989).
low-to-moderate amounts of dietary sh oil on neutrophil lipid composition ad Supplementation with n3 fatty acids from sh oil in chronic inammatory bowel
function. Lipids 35, 763768. diseasea randomized, placebo-controlled, double-blind cross-over trial. J. Int. Med.
Hirata, I., Murano, M., Nitta, M., Sasaki, S., Toshina, K., Maemura, K., et al. (2001). Estima- Suppl. 731, 225232.
tion of mucosal inammatory mediators in rat DSS-induced colitis. Possible role of Lorenz-Meyer, H., Bauer, P., Nicolay, C., Schulz, B., Purrmann, J., Fleig, W. E., et al. (1996).
PGE(2) in protection against mucosal damage. Digestion 63 Suppl 1:73-80. Omega-3 fatty acids and low carbohydrate diet for maintenance of remission in
Hirasawa, A., Tsumaya, K., Awaji, T., Katsuma, S., Adachi, T., Yamada, M., et al. (2005). Free Crohn's disease. A randomized controlled multicenter trial. Study Group Members
fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. (German Crohn's Disease Study Group). Scand. J. Gastroenterol. 31, 778785.
Nat. Med. 11, 9094. Madden, J., Williams, C. M., Calder, P. C., Leitz, G., Miles, E. A., Cordell, H., et al. (2011). The
Hodge, L., Salome, C. M., Peat, J. K., Haby, M. M., Xuan, W., & Woolcock, A. J. (1996). impact of common gene variants on the response of biomarkers of cardiovascular dis-
Comsumption of oily sh and childhood asthma risk. Med. J. Aust. 164, 137140. ease (CVD) risk to increased sh oil fatty acids intakes. Annu. Rev. Nutr. 31, 203234.
Hong, S., Gronert, K., Devchand, P. R., Moussignac, R. L., & Serhan, C. N. (2003). Novel Marks, G. B., Mullen, S. M., Kemp, A. S., Tovey, E. R., Webb, K., Almqvist, C., et al. (2006).
docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine Prevention of asthma during the rst 5 years of life: A randomized controlled trial.
brain, human blood, and glial cells. J. Biol. Chem. 278, 1467714687. J. Allergy. Clin. Immunol. 118, 5361.
Im, D. S. (2012). Omega-3 fatty acids in anti-inammation (pro-resolution) and GPCRs. Mas, E., Croft, K. D., Zahra, P., Barden, A., & Mori, T. A. (2012). Resolvins D1, D2, and other
Prog. Lipid Res. 51, 232237. mediators of self-limited resolution of inammation in human blood following n3
Ichimura, A., Hirasawa, A., Poulain-Godefroy, O., Bonnefond, A., Hara, T., Yengo, L., et al. fatty acid supplementation. Clin. Chem. 58, 14761484.
(2012). Dysfunction of lipid sensor GPR120 leads to obesity in both mouse and McCall, T. B., O'Leary, D., Bloomeld, J., & O'Morin, C. A. (1989). Therapeutic potential of
human. Nature 483, 350354. sh oil in the treatment of ulcerative colitis. Aliment Pharmacol Ther 3, 415424.
Inamori, T., Goda, T., Kasezawa, N., & Yamakawa-Kobayashi, K. (2013). The combined McGettrick, H. M., Butler, L. M., Buckley, C. D., Ed Rainger, G., & Nash, G. B. (2012). Tissue
effects of genetic variation in the SIRT1 gene and dietary intake of n3 and n6 stroma as a regulator of leukocyte recruitment in inammation. J. Leukoc. Biol. 91,
polyunsaturated fatty acids on serum LDL-C and HDL-C levels: a population based 385400.
study. Lipids Health Dis. 12, 4. MacLean, C. H., Mojica, W. A., Morton, S. C., Pencharz, J., Hasenfeld, Garland R., et al.
Insel, P. A., & Patel, H. H. (2009). Membrane rafts and cardiovascular signaling. Curr. Opin. (2004). Effects of omega-3 fatty acids on lipids and glycemic control in type II diabe-
Nephrol. Hypertens. 18, 5056. tes and the metabolic syndrome and on inammatory bowel disease, rheumatoid ar-
John, S., Luben, R., Shrestha, S. S., Welch, A., Khaw, K. T., & Hart, A.R. (2010). Dietary n3 thritis, renal disease, systemic lupus erythematosus, and osteoporosis. Evid Rep
polyunsaturated fatty acids and the aetiology of ulcerative colitis: a UK prospective Technol Assess (Summ) 89, 14.
cohort study. Eur. J. Gastroenterol. Hepatol. 22. Meydani, S. N., Endres, S., Woods, M. M., Goldin, B. R., Soo, C., Morrill-Labrode, A., et al.
Jonnalagadda, S. S., Egan, S. K., Heimbach, J. T., Harris, S. S., & Kris-Etherton, P.M. (1995). (1991). Oral (n3) fatty acid supplementation suppresses cytokine production and
Fatty acid consumption pattern of Americans: 19871988 USDA Nationwide Food lymphocyte proliferation: comparison between young and older women. J. Nutr.
Consumption Survey. Nutr. Res. 15, 17671781. 121, 547555.
Kagawa, Y., Nishizawa, M., Suzuki, M., Miyatake, T., Hamamoto, T., Goto, K., et al. (1982). Miles, E. A., Allen, E., & Calder, P. C. (2002). In vitro effects of eicosanoids derived from dif-
Eicosapolyenoic acids of serum lipids of Japanese islanders with low incidence of ferent 20-carbon fatty acids on production of monocyte-derived cytokines in human
cardiovascular diseases. J. Nutr. Sci. Vitaminol. 28, 441453. whole blood cultures. Cytokine 20, 215223.
Kelley, D. S., Taylor, P. C., Nelson, G. J., Schmidt, P. C., Ferretti, A., Erickson, K. L., et al. Miyauchi, S., Hirasawa, A., Iga, T., Lui, N., Chisato, I., Sadakane, K., et al. (2009). Distribution
(1999). Docosahexaenoic acid ingestion inhibits natural killer cell activity and pro- and regulation of protein expression of the free fatty acid receptor GPR120. Naunyn
duction of inammatory mediators in young healthy men. Lipids 34, 317324. Schmiedebergs Arch. Pharmacol. 379, 427434.
Kenji, O. (2007). Pain signalling pathways: From cytokines to ion channels. Int. J. Biochem. Moraes, L. A., Piqueras, L., & Bishop-Bailey, D. (2006). Peroxisome proliferator-activated
Cell Biol. 39, 490496. receptors and inammation. Pharmacol. Ther. 110, 371385.
Kew, S., Mesa, M. D., Tricon, S., Buckley, R., Minihane, A. M., & Yaqoob, P. (2004). Effects of Moreland, L. W., Baumgartner, S. W., Schiff, M. H., Tindall, E. A., Fleischmann, R. M.,
oils rich in eicosapentaenoic and docosahexaenoic acids on immune cell composition Weaver, A. L., et al. (1997). Treatment of rheumatoid arthritis with a recombinant
and function in healthy humans. Am J Clin Nutr. 79, 674681. human tumor necrosis factor receptor (p75)-Fc fusion protein. N. Engl. J. Med. 337,
Kim, Y. J., Go, M. J., Hu, C., Hong, C. B., Kim, Y. K., Lee, J. Y., et al. (2011). Large-scale 141147.
genome-wide association studies in east Asians identify new genetic loci inuencing Mullen, A., Loscher, C. E., & Roche, H. M. (2010). Anti-inammatory effects of EPA and
metabolic traits. Nat Genet 43, 990995. DHA are dependent upon time and doseresponse elements associated with LPS
Kirsch, C. M., Payan, D.G., Wong, M. Y., Dohlman, J. G., Blake, V. A., Petri, M.A., et al. (1988). stimulation in THP-1-derived macrophages. J. Nutr. Biochem. 21, 444450.
Effect of eicosapentaenoic acid in asthma. Clin. Allergy 18, 177187. Newman, W. P., Middaugh, J. P., Propst, M. T., & Rogers, D. R. (1993). Atherosclerosis in
Kobzar, G., Mardla, V., Jarving, I., & Samel, N. (2001). Comparison of anti-aggregatory Alaska Natives and non-natives. Lancet 341, 10561057.
effects of PGI2, PGI3 and iloprost on human and rabbit platelets. Cell. Physiol. Nieto, N., Torres, M. I., Ros, A., & Gil, A. (2002). Dietary polyunsaturated fatty acids im-
Biochem. 11, 279284. prove histological and biochemical alterations in rats with experimental ulcerative
Koutroubakis, I., Manousos, O. N., Meuwissen, S. G., & Pena, A. S. (1996). Environmental colitis. J Nutr 132, 1119.
risk factors in inammatory bowel disease. Hepatogastroenterology 43, 381393. Oh, D. Y., Talukdar, S., Bae, E. J., Imamura, T., Morinaga, H., Fan, W., et al. (2010). GPR120
Kremer, J. M., Bigauoette, J., Michalek, A. V., Timchalk, M.A., Lininger, L., Rynes, R. I., et al. is an Omega-3 Fatty acid receptor mediating potent anti-inammatory and
(1985). Effects of manipulation of dietary fatty acids on clinical manifestations of insulin-sensitizing effects. Cell 142, 687698.
rheumatoid arthritis. Lancet 1, 184187. Oh, S. F., Dona, M., Fredman, G., Krishnamoorthy, S., Irimia, D., & Serhan, C. N. (2012).
Kris-Etherton, P.M., Harris, W. S., Appel, L. J., & for the Nutrition Committee (2002). Fish Resolvin E2 formation and impact in inammation resolution. J. Immunol. 188,
consumption, sh oil, omega-3 fatty acids, and cardiovascular disease. Circulation 45274534.
106, 27472757. Ohnishi, H., Miyahara, N., & Gelfand, E. W. (2008). The role of leukotriene B4 in allergic
Krishnamoorthy, S., Recchiuti, A., Chiang, N., Yacoubian, S., Lee, C. H., Yang, R., et al. diseases. Allergol. Int. 57, 291298.
(2010). Resolvin D1 binds human phagocytes with evidence for proresolving recep- Olsen, S. F., sterdal, M. L., Salvig, J. D., Mortensen, L. M., Rytter, D., Secher, N. J., et al.
tors. Proc. Natl. Acad. Sci. U. S. A. 107, 16601665. (2008). Fish oil intake compared with olive oil intake in late pregnancy and asthma
Kromann, N., & Green, A. (1980). Epidemiological studies in the Upernavik district, Green- in the offspring: 16 y of registry-based follow-up from a randomized controlled
land. Incidence of some chronic diseases 19501974. Acta Med. Scand. 208, 401406. trial. Am J Clin Nutr 88, 167-75.
Kromhout, D., Giltay, E. J., & Geleijnse, J. M. (2010). n3 Fatty acids and cardiovascular Psychogios, N., Hau, D.D., Peng, J., Guo, A.C., Mandal, R., Bouatra, S., et al. (2011). The
events after myocardial infarction. New Engl. J. Med. 363, 20152026. human serum metabolome. PLoS ONE 6, e16957.
282 C.M. Yates et al. / Pharmacology & Therapeutics 141 (2014) 272282

Ratnayake, W. M. N., & Galli, C. (2009). Fat and fatty acid terminology, methods of analysis and The IBD in EPIC Study Investigators (2009). Linoleic acid, a dietary n6 polyunsaturated
fat digestion and metabolism: a background review paper. Ann. Nutr. Metab. 55, 843. fatty acid, and the aetiology of ulcerative colitis: a nested case control study within a
Rauch, B., Schiele, R., Schneider, S., Diller, F., Victor, N., Gohlke, H., et al. (2010). OMEGA, a European prospective cohort study. Gut 58, 16061611.
randomized, placebo-controlled trial to test the effect of highly puried omega-3 fatty Traversa, G., Walker, A.M., Ippolito, F. M., Caffari, B., Capurso, L., Dezi, A., et al. (1995). Gas-
acids on top of modern guideline-adjusted therapy after myocardial infarction/clinical troduodenal toxicity of different nonsteroidal antiinammatory drugs. Epidemiology
perspective. Circulation 122, 21522159. 6, 4954.
Remans, P. H. J., Sont, J. K., Wagenaar, L. W., Wouters-Wesseling, W., Zuijderduin, W. M., Trebble, T., Arden, N. K., Stroud, M.A., Wootton, S. A., Burdge, G. C., Miles, E. A.,
Jongma, A., et al. (2004). Nutrient supplementation with polyunsaturated fatty acids et al. (2003). Inhibition of tumour necrosis factor-alpha and interleukin 6 pro-
and micronutrients in rheumatoid arthritis: clinical and biochemical effects. Eur J Clin duction by mononuclear cells following dietary sh-oil supplementation in
Nutr 58, 839845. healthy men and response to antioxidant co-supplementation. Br. J. Nutr. 90,
Ritchie, D.M., Boyle, J. A., McInnes, J. M., Jasani, M. K., Dalkos, T. G., Grievson, P., et al. 405412.
(1968). Clinical studies with an articular index for the assessment of joint tenderness Tull, S. P., Yates, C. M., Maskrey, B. H., O'Donnell, V., Madden, J., Grimble, R. F., et al. (2009).
in patients with rheumatoid arthritis. Q. J. Med. 37, 393406. Omega-3 fatty acids and inammation: novel interactions reveal a new step in
Sands, B. E., & Kaplan, G. G. (2007). The role of TNF-a in ulcerative colitis. J. Clin. Pharmacol. neutrophil recruitment. PLOS Biol. 7, e1000177.
47, 930941. Turner, D., Shah, P.S., Steinhart, A. H., Zlotkin, S., & Grifths, A.M. (2011). Mainte-
Schachter, H., Reisman, J., Tran, K., et al. (2004). Health effects of omega-3 fatty acids on nance of remission in inammatory bowel disease using omega-3 fatty acids
asthma. Evidence report/technical assessment No. 91. AHRQ publication No.04-E013-2 (sh oil): a systematic review and meta-analyses. Inamm. Bowel Dis. 17,
Agency for Healthcare Research and Quality. 336345.
Schmidt, E. B., Pedersen, J. O., Varming, K., Ernst, E., Jersild, C., Grunnet, N., et al. Vilaseca, J., Salas, A., Guarner, F., Rodriguez, R., Martinez, M., & Malagelada, J. R. (1990).
(1991). n3 fatty acids and leukocyte chemotaxis. Effects in hyperlipidemia and Dietary sh oil reduces progression of chronic inammatory lesions in a rat model
doseresponse studies in healthy men. Arterioscler. Thromb. Vasc. Biol. 11, 429435. of granulomatous colitis. Gut 31, 539544.
Sellon, R. K., Tonkonogy, S., Schultz, M., Dieleman, L. A., Grenther, W., Balish, E., et al. (1998). von Schacky, C. (2004). Omega-3 fatty acids and cardiovascular disease. Curr. Opin. Clin.
Resident enteric bacteria are necessary for development of spontaneous colitis and im- Nutr. Metab. Care 7, 131136.
mune system activation in interleukin-10-decient mice. Infect. Immun. 66, 52245231. Wang, C., Harris, W. S., Chung, M., Lichtenstein, A. H., Balk, E. M., Kupelnick, B., et al.
Serhan, C. N. (2010). Novel lipid mediators and resolution mechanisms in acute inam- (2006). n3 Fatty acids from sh or sh-oil supplements, but not alpha-linolenic acid,
mation: to resolve or not? Am J Patholo 177, 15761591. benet cardiovascular disease outcomes in primary- and secondary-prevention studies:
Serhan, C. N., Clish, C. B., Brannon, J., Colgan, S. P., Chiang, N., & Gronert, K. (2000). Novel a systematic review. Am. J. Clin. Nutr. 84, 517.
functional sets of lipid-derived mediators with antiinammatory actions generated Wang, T. M., Chen, C. J., Lee, T. S., Chao, H. Y., Wu, W. H., Hsieh, S.C., et al. (2011).
from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal antiinammatory Docosahexaenoic acid attenuates VCAM-1 expression and NF-[kappa]B activa-
drugs and transcellular processing. J. Exp. Med. 192, 11971204. tion in TNF-[alpha]-treated human aortic endothelial cells. J. Nutr. Biochem. 22,
Serhan, C. N., Hong, S., Gronert, K., Colgan, S. P., Devchand, P. R., Mirick, G., et al. (2002). 187194.
Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits Wang, Y., Liu, Q., & Thorlacius, H. (2003). Docosahexaenoic acid inhibits cytokine-induced
initiated by aspirin treatment that counter proinammation signals. JExpMed 196, expression of P-selectin and neutrophil adhesion to endothelial cells. Eur. J. Pharmacol.
10251037. 459, 269273.
Serhan, C. N., Yang, R., Martinod, K., Kasuga, K., Pillai, P.S., Porter, T. F., et al. (2009). Maresins: Wardlaw, A. J., Hay, H., Cromwell, O., Collins, J. V., & Kay, A.B. (1989). Leukotrienes, LTC4
novel macrophage mediators with potent antiinammatory and proresolving actions. and LTB4, in bronchoalveolar lavage in bronchial asthma and other respiratory
J ExpMed 206, 1523. diseases. J. Allergy Clin. Immunol. 84, 1926.
Sharon, P., & Stenson, W. F. (1984). Enhanced synthesis of leukotriene B4 by colonic Weldon, S. M., Mullen, A. C., Loscher, C. E., Hurley, L. A., & Roche, H. M. (2007).
mucosa in inammatory bowel disease. Gastroenterol 86, 453460. Docosahexaenoic acid induces an anti-inammatory prole in lipopolysaccharide-
Sijben, J. W., & Calder, P. C. (2007). Differential immunomodulation with long-chain n3 stimulated human THP-1 macrophages more effectively than eicosapentaenoic acid.
PUFA in health and chronic disease. Proc. Nutr. Soc. 66, 237259. J Nutr Biochem. 18, 250258.
Smit, H. A., Grievink, L., & Tabak, C. (1999). Dietary inuences on chronic obstructive lung Whatley, R. E., Stroud, E. D., Bunting, M., Zimmerman, G. A., McIntyre, T. M., & Prescott, S.
disease and asthma: a review of the epidemiological evidence. Proceedings of the M. (1993). Growth-dependent changes in arachidonic acid release from endothelial
Nutrition Society 58, 309319. cells are mediated by protein kinase C and changes in diacylglycerol. J. Biol. Chem.
Shimizu, T., Suzuki, M., Lee, T., Igarashi, J., Kaneko, K., & Yamashiro, Y. (2003). Effects of 268, 1613016138.
n-3 polyunsaturated fatty acids on indomethacin-induced changes in eicosanoid pro- Wong, S. H., & Lord, J. M. (2004). Factors underlying chronic inammation in rheumatoid
duction and blood ow in the gastric mucosa of rats. Prostaglandins, Leukotrienes and arthritis. Arch. Immunol. Ther. Exp. 52, 379388.
Essential Fatty Acids 69, 3337. Xu, H. E., Lambert, M. H., Montana, V. G., Parks, D. J., Blanchard, S. G., Brown, P. J., et al.
Smolen, J. S., Breedveld, F. C., Eberi, G., Jones, I., Leeming, M., Wylie, G. L., et al. (1995). Va- (1999). Molecular recognition of fatty acids by peroxisome proliferator-activated re-
lidity and reliability of the twenty-eight-joint count for the assessment of rheumatoid ceptors. Mol. Cell 3, 397403.
arthritis activity. Arthritis Rheum. 38, 3843. Yates, C. M., Tull, S. P., Madden, J., Calder, P. C., Grimble, R. F., Nash, G. B., et al. (2011).
Sperling, R. I., Benincaso, A. J., Knoell, C. T., Larkin, J. K., Austen, K. F., & Robinson, D. R. Docosahexaenoic acid inhibits the adhesion of owing neutrophils to cytokine stim-
(1993). Dietary omega-3 polyunsaturated fatty acids inhibit phosphoinositide ulated human umbilical vein endothelial cells. J. Nutr. 141, 13311334.
formation and chemotaxis in neutrophils. J. Clin. Invest. 91, 651660. Yokoyama, M., Origasa, H., Matsuzaki, M., Matsuzawa, Y., Saito, Y., Ishikawa, Y., et al.
Sperling, R. I. (1995). Eicosanoids in rheumatoid arthritis. Rheum. Dis. Clin. North Am. 21, (2007). Effects of eicosapentaenoic acid on major coronary events in hypercholester-
741758. olaemic patients (JELIS): a randomised open-label, blinded endpoint analysis. Lancet
Stenius-Aarniala, B., Aro, A., Hakulinen, A., Ahola, I., Seppala, E., & Vapaatalo, H. (1989). 369, 10901098.
Evening primrose oil and sh oil are ineffective as supplementary treatment of Yuceyar, H., Ozutemiz, O., Huseyinov, A., Saruc, M., Alkanat, M., Bor, S., et al. (1999).
bronchial asthme. Ann. Allergy 62, 534537. Is administration of n3 fatty acids by enema mucosal protective against
Stenson, W. F., Cort, D., Rodgers, J., Burakoff, R., DeSchryver-Kecskemeti, K., et al. (1992). Di- trinitrobenzene-induced colitis in rats? Prostaglandins Leukot. Essent. Fatty
etary supplementation with sh oil in ulcerative colitis. Ann Intern Med 116, 609614. Acids 61, 339345.
Strasser, T., Fischer, S., & Weber, P. C. (1985). Leukotriene B5 is formed in human Zhao, G., Etherton, T. D., Martin, K. R., Vanden Heuvel, J. P., Gillies, P. J., West, S. G., et al.
neutrophils after dietary supplementation with icosapentaenoic acid. Proc. Natl. (2005). Anti-inammatory effects of polyunsaturated fatty acids in THP-1 cells.
Acad. Sci. U. S. A. 82, 15401543. Biochem. Biophys. Res. Commun. 336, 909917.
Teslovich, T. M., Musunuru, K., Smith, A. V., Edmondson, A. C., Stylianou, I. M., Koseki, M., Zhao, Y., Joshi-Barve, S., Barve, S., & Chen, L. H. (2004). Eicosapentaenoic acid prevents
et al. (2010). Biological, clinical and population relevance of 95 loci for blood lipids. LPS-induced TNF-alpha expression by preventing NF-kappaB activation. J. Am. Coll.
Nature 466, 707713. Nutr. 23, 7178.

You might also like