You are on page 1of 17

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/6630679

Essential Fatty Acids - A Review

Article in Current pharmaceutical biotechnology · January 2007


DOI: 10.2174/138920106779116856 · Source: PubMed

CITATIONS READS

226 897

1 author:

Undurti Das
UND Life Sciences
544 PUBLICATIONS 14,969 CITATIONS

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Diabetes mellitus View project

All content following this page was uploaded by Undurti Das on 23 March 2014.

The user has requested enhancement of the downloaded file.


Current Pharmaceutical Biotechnology, 2006, 7, 000-000 1

Essential Fatty Acids - A Review

Undurti N. Das*

UND Life Sciences, 13800 Fairhill Road, Shaker Heights, OH 44120, USA

Abstract: Essential fatty acids (EFAs): cis-linoleic acid (LA) and α-linolenic acid (ALA) are essential for humans and
their deficiency is rare in humans due to their easy availability in diet. EFAs are metabolized to their respective long-chain
metabolites: dihomo-gamma-linolenic acid (DGLA), and arachidonic acid (AA) from LA; and eicosapentaenoic acid
(EPA) and docosahexaenoic acid (DHA) from ALA. Some of these long-chain metabolites form precursors to respective
prostaglandins (PGs), thromboxanes (TXs), and leukotrienes (LTs), lipoxins (LXs) and resolvins. EFAs and their metabo-
lites may function as endogenous angiotensin converting enzyme and HMG-CoA reductase inhibitors, nitric oxide en-
hancers, anti-hypertensives, and anti-atherosclerotic molecules. EFAs react with nitric oxide (NO) to yield respective ni-
troalkene derivatives that have cell-signaling actions via ligation and activation of peroxisome proliferator-activated re-
ceptors (PPARs). In several diseases such as obesity, hypertension, diabetes mellitus, coronary heart disease, alcoholism,
schizophrenia, Alzheimer’s disease, atherosclerosis, and cancer the metabolism of EFAs is altered. Thus, EFAs and their
derivatives have significant clinical implications.
Key Words: Essential fatty acids, linoleic acid, alpha-linolenic acid, gamma-linolenic acid, arachidonic acid, eicosapentaenoic
acid, docosahexaenoic acid, prostaglandins, lipoxins, nitric oxide, atherosclerosis, Alzheimer’s disease.

INTRODUCTION The “ω -7” series derived from palmitoleic acid (PA, 16:1, ω
-7).
Essential fatty acids (EFAs) are important constituents of
all cell membranes and alter membrane fluidity and thus, LA is converted to γ-linolenic acid (GLA, 18:3, n-6) by
determine and influence the behaviour of membrane-bound the enzyme Δ6 desaturase (d-6-d) and GLA is elongated to
enzymes and receptors. EFAs are essential for survival of form dihomo-GLA (DGLA, 20:3, ω-6), the precursor of the
humans and as are not synthesized in the body; have to be 1 series of prostaglandins (PGs). DGLA can also be con-
obtained in our diet [1]. EFAs are of two types as they occur verted to arachidonic acid (AA, 20:4, ω-6) by the enzyme Δ5
in the body, the ω-6 series derived from cis-linoleic acid desaturase (d-5-d). AA forms the precursor of 2 series of
(LA, 18:2) and the ω-3 series derived from α-linolenic acid PGs, thromboxanes (TXs) and the 4 series of leukotrienes
(ALA, 18:3). There is another sequence of fatty acids de- (LTs). ALA is converted to eicosapentaenoic acid (EPA,
rived from oleic acid (OA, 18:1 ω-9), and OA is not an EFA. 20:5, ω-3) by d-6-d and d-5-d. EPA forms the precursor of
The ω-9, ω-6, and ω-3 series of fatty acids are metabolized the 3 series of PGs, TXs and the 5 series of LTs. LA, GLA,
by the same set of enzymes to their respective long-chain DGLA, AA, ALA, EPA and docosahexaenoic acid (DHA,
metabolites. Further discussion is centered on ω-6 LA and ω- 22:6, ω-3) are all PUFAs, but only LA and ALA are EFAs
3 A fatty acids and their metabolites, since they are the (see Fig. 1 for metabolism of EFAs). AA and EPA also are
EFAs. It is important to note that while some of the actions converted to their respective LTs. PGs, TXs, and LTs are
and functions of EFAs require their conversion to eicosa- biologically active and involved in diseases such as athero-
noids and other products, in majority of the instances the sclerosis, bronchial asthma, inflammatory bowel disease, and
fatty acids themselves are active. several other inflammatory conditions. In the present discus-
sion, the term “PUFAs” is used to refer to all unsaturated
METABOLISM OF EFAs fatty acids: LA, GLA, DGLA, AA, ALA, EPA, and DHA;
and the term EFAs refers to LA and ALA. Although the
EFAs are polyunsaturated fatty acids (PUFAs) since they
terms EFAs and PUFAs are used interchangeably for the
contain two or more double bonds. There are at least 4 inde-
sake of convenience it should be understood that all EFAs
pendent families of PUFAs. They include:
are PUFAs but all PUFAs are not EFAs. Many actions of
The “ω -3” series derived from α-linolenic acid (ALA, 18:3, EFAs are also brought about by PUFAs and so EFA-
ω -3). deficiency can be corrected to a large extent by PUFAs, and
hence, PUFAs are termed as “functional EFAs”. In view of
The “ω -6” series derived from cis-linoleic acid (LA, 18:2, ω
this, the terms EFAs and PUFAs are used interchangeably.
-6).
EFAs/PUFAs play a significant role in collagen vascular
The “ω -9” series derived from oleic acid (OA, 18:1, ω -9).
diseases, hypertension, diabetes mellitus, metabolic syn-
drome X, psoriasis, eczema, atopic dermatitis, coronary heart
*Address correspondence to this author at the UND Life Sciences, 13800 disease, atherosclerosis, and cancer [2-5]. This is in addition
Fairhill Road, #321, Shaker Heights, OH 44120, USA; Tel: 216-231-5548; to the role of PGs and LTs in these conditions. The molecu-
Fax: 928-833-0316; E-mail: undurti@hotmail.com
lar mechanism(s) by which various stimuli preferentially

1389-2010/06 $50.00+.00 © 2006 Bentham Science Publishers Ltd.


2 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

Fig. (1). Scheme showing the metabolism of essential fatty acids and co-factors that enhance the activity of Δ6 and Δ5 desaturases and elon-
gases and formation of PGs. Ethanol blocks both Δ6 and Δ5 desaturases.
(+) Indicates enhancement of the activity of the enzyme or increase in the formation of the product.
(-) Indicates either in the inhibition of the activity of the enzyme or decrease in the formation of the product.

induce the release of AA, EPA and/or DHA and convert LA: cereals, eggs, poultry, most vegetable oils, whole-grain
them to their respective products is not clear. AA, EPA and breads, baked goods, and margarine. Sunflower, saffola, and
DHA give rise to anti-inflammatory molecules lipoxins corn oils are rich in LA [1].
(LXs) and resolvins that suppress inflammation. Thus, PU- ALA: Canola oil, flaxseed oil, linseed and rapeseed oils,
FAs form precursors to both pro- and anti-inflammatory
walnuts, and leafy green vegetables. Human milk is rich in
molecules and the balance between these mutually antago-
EFAs and GLA, DGLA, AA, EPA, and DHA. Olive oil is
nistic compounds could determine the final outcome of the
rich in OA, whereas palm and coconut oils contain virtually
disease process. Biologically active compounds formed due
none. The average daily intake of EFAs, in general, is
to the nitration of unsaturated fatty acids called as nitrolipids
around 7-15 g/day in Europe and USA.
have also been identified. Nitration of linoleate by nitric ox-
ide-derived reactive species forms novel derivatives includ- GLA: Human milk contains 0.3-1.0% of its fat as GLA.
ing nitrolinoleate that stimulates smooth muscle relaxation, Thus, breast fed babies get significant amounts of GLA.
blocks platelet activation, inhibits human neutrophil func- Evening primrose oil (EPO), borage oil, black currant oil,
tions and suppresses inflammation. These studies suggest and hemp seed oil contain substantial amounts of GLA. GLA
that PUFAs have important actions not only by themselves is present in EPO at concentrations of 7-14% of total fatty
but also by giving raise to various biologically active com- acids; in borage seed oil it is 20-27%; and in black currant
pounds. seed oil at 15-20%. GLA is also found in some fungal
sources.
DIETARY SOURCES OF EFAs
DGLA: Moderate amounts are found in human milk, liver,
The main dietary sources of EFAs are: testes, adrenals, and kidneys.
Essential Fatty Acids Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 3

AA: Human milk contains modest amounts and cow’s milk EFAs and promote inflammation, atherosclerosis and coro-
small amounts. Meat, egg yolks, some seaweeds, and some nary heart disease [1, 11]. The pro-inflammatory action of
shrimps contain substantial amounts. Average daily intake of trans fats is due to their ability to interfere with EFA metabo-
AA is estimated to be in the region of 100-200 mg/day, more lism. Several PUFAs, especially EPA and DHA inhibit the
than enough to account for the total daily production of vari- production of pro-inflammatory cytokines: interleukin-6 (IL-
ous PGs, which is estimated to be about 1 mg/day. 6), tumor necrosis factor-α (TNF-α), IL-1, and IL-2 [1].
Saturated fatty acids and cholesterol interfere with the me-
Adrenic acid (22:44 ω-6): The main sources of adrenic acid
tabolism of EFAs and thus, promote the production of pro-
are adrenals, kidneys, testes, and brain.
inflammatory cytokines, which explains their ability to cause
EPA and DHA: The major source of these two fatty acids is atherosclerosis and coronary heart disease (CHD). Thus,
marine fish. These fatty acids may be denatured and con- trans fats, saturated fats, and cholesterol have pro-inflamma-
verted into trans fats that are harmful to the body during tory actions whereas PUFAs possess anti-inflammatory
processing [6, 7]. Substantial fall in the intake of ω-3 fatty properties. Interference with the metabolism of EFAs by
acids: EPA and DHA could be one of the major changes in saturated fats, cholesterol and trans fats reduces the forma-
Western nutrition in the last 50 years that contributed to the tion of GLA, DGLA, AA, EPA, and DHA that are essential
increasing incidence of atherosclerosis, CHD, hypertension, for the formation of biologically beneficial prostacyclin
metabolic syndrome X, obesity, collagen vascular diseases (PGI2), PGI3, lipoxins, and resolvins. Deficiency and/or ab-
and cancer. sence of PGI2, PGI3, lipoxins and resolvins initiate and ac-
celerate the progression of atherosclerosis, persistence of
MODULATORS OF METABOLISM OF EFAs inflammation, CHD and failure of the healing process.
Dietary LA and ALA are metabolized by the enzymes Δ6
and Δ5 desaturases to their respective metabolites as shown ACTIONS OF EFAS AND THEIR METABOLITES
in Fig. (1). LA, ALA, and OA are metabolized by the same Cell Membrane Fluidity
set of Δ6 and Δ5 desaturases and elongases. As a result, these
3 series compete with one another for the same set of en- Cell membrane fluidity is determined by its lipid compo-
zymes, though the enzymes seem to prefer ω-3 to ω-6 and n- sition: increasing its content of saturated fatty acids and cho-
6 over ω-9. Hence, under normal physiological conditions lesterol renders the membrane more rigid, whereas increas-
the metabolites of ω-9 are formed only in trivial amounts in ing unsaturated fatty acids makes it more fluid. This is an
the cells. Thus, presence of significant amounts of 20:3 ω-9 important function of lipids since the number of receptors
suggests that there is deficiency of ω-3 and ω-6 and is used and their affinity to their respective hormones/growth fac-
as an indicator of EFA deficiency. The activities of Δ6 and tors/ proteins depends on the fluidity of the cell membrane.
Δ5 desaturases are slow in humans (Δ5 > Δ6). As a result, the For instance, a rigid cell membrane shows reduced number
conversion of LA and ALA to their respective metabolites of insulin receptors and their affinity to insulin that, in turn,
may be inadequate under certain conditions. In such in- causes insulin resistance. In contrast, increase in cell mem-
stance, it is necessary to supplement GLA and DGLA (to brane fluidity due to its high content of unsaturated fatty
bypass Δ6 desaturase) and AA and EPA and DHA (to bypass acids and reduced cholesterol, increases the number of insu-
Δ6 and Δ5 desaturases). Generally, supplementation of AA is lin receptors on the membrane and their affinity to insulin
not necessary since; it can be obtained from the diet. Western and thus, decreases insulin resistance [1, 12, 13].
diet is rich in ω-6 fatty acids compared to ω-3 fatty acids (ω- Availability of appropriate amounts of ω-3 and ω-6 fatty
6 to ω-3 ratio is 10:1), whereas the recommended ratio is acids and various growth factors is essential for the growth
~1:1 [1]. of brain during the perinatal period and adolescence [1, 14,
Saturated fats, cholesterol, trans-fatty acids, alcohol, 15]. Deficiency of ω-3 EPA and DHA and ω-6 AA during
adrenaline, and glucocorticoids inhibit Δ6 and Δ5 desaturases the critical period impairs brain growth and the development
[1, 8, 9]. Pyridoxine, zinc, nicotinic acid, and magnesium are of appropriate synaptic connections that, in turn, could lead
co-factors for normal Δ6 desaturase activity. Insulin activates to developmental disorders of the brain and neuropsy-
Δ6 desaturase whereas diabetics have reduced Δ6 desaturase chological conditions: dementia, depression, schizophrenia,
activity. The activity of Δ6 desaturase falls with age. Onco- Alzheimer’s disease, and neurodegenerative diseases: Hunt-
genic viruses and radiation inhibit Δ6 desaturase activity. ington’s disease, Parkinson’s disease, spinocerebellar degen-
Total fasting, protein deficiency, and a glucose-rich diet re- eration, etc, and may impair memory formation and consoli-
duce, whereas fat- free diet and partial caloric restriction dation.
enhance Δ6 desaturase activity. Activity of Δ6 and Δ5 desatu-
rases are regulated by sterol regulatory element binding pro- Second Messenger Action
tein-1 (SREBP-1) and peroxisome proliferator-activated re- Growth factors and hormones activate phospholipase A2
ceptor-α (PPAR-α), two reciprocal transcription factors for (PLA2) leading to the release of DGLA, AA, EPA, and DHA
fatty acid metabolism, and some of their (SREBP-1 and from the cell membrane lipid pool. Fatty acids thus released
PPAR- α) lipogenic functions are brought about by their ac- are utilized for the formation of eicosanoids to bring about
tion on PUFAs [10]. some of their actions. For example, the tumoricidal action of
Activities of Δ6 and Δ5 desaturases are decreased in dia- TNF-α is dependent on its ability to induce PLA2, and inhibi-
betes mellitus, hypertension, hyperlipidemia, and metabolic tors of PLA2 completely inhibited this action. I observed that
syndrome X. Trans fats interfere with the metabolism of TNF-α-resistant tumor cells can be rendered sensitive to
4 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

TNF-α by the addition of various PUFAs especially, GLA. studies showed that AA, EPA, and DHA could give rise to
PUFAs enhance the activity of protein kinase C (PKC), a anti-inflammatory compounds such as lipoxins (LXs) and
well-known second messenger; activate macrophages, poly- resolvins that are essential to limit and resolve inflammation
morphonuclear leukocytes (PMNs), modulate TH1 and TH2 [1, 35]. These studies imply that a deficiency of LXs and
balance, and increase free radical generation by these cells resolvins could lead to the perpetuation of inflammation and
[1, 16, 17]. tissue damage. In the light of these facts, it will be interest-
ing to study whether a sub-clinical deficiency of LCPUFAs,
PUFAs HAVE ANTI-BACTERIAL, ANTI-VIRAL, decreased formation of LXs and resolvins occurs in subjects
ANTI-FUNGAL, AND ANTI-PARASITIC ACTIONS who develop various types of infections and their complica-
tions such as sepsis. Since, LCPUFAs can inactivate envel-
Linolenic acid rapidly killed cultures of Staphylococcus
aureus, and hydrolyzed linseed oil (which contains both LA oped viruses including influenza [22, 33], it is probably
worthwhile to study the effect of various fatty acids on bird
and ALA) inactivated methicillin-resistant S. aureus. ALA
flu virus and if so, whether increased intake of these fatty
promotes adhesion of Lactobacillus casei to mucosal sur-
acids could reduce the risk of flu.
faces and thus, augments their growth. Lactobacilli, in turn,
suppress the growth of pathogenic bacteria like Helicobacter
ACE ACTIVITY, ENDOTHELIAL NITRIC OXIDE
pylori, Shigella flexneri, Salmonella typhimurium, Pseudo- GENERATION, AND PUFAs
monas aeroginosa, Clostridium difficile, and Escherichia
coli. Kodicek [18] showed that both LA and ALA have bac- PUFAs inhibited leukocyte ACE activity [36, 37] sug-
teriostatic effect on both gram-positive and gram-negative gesting that they could function as endogenous regulators of
bacteria. Lacey and Lord [19] observed that cultures of ACE activity, and thus regulate the formation of (angio-
Staphylococcus aureus seeded on to human skin were rap- tensin-II) Ang-II. PUFAs enhance nitric oxide generation
idly killed after the skin has been covered with ALA and [38]. Hence, when tissue/cell concentrations of PUFAs are
suggested that it has all the attributes of an ideal anti- low the formation of Ang-II will be high whereas that of
bacterial agent. A variety of bacteria were found to be sensi- endothelial nitric oxide (eNO) will be low. Plasma concen-
tive to the growth inhibitory actions of LA and ALA in vitro trations of PUFA and eNO are low in hypertension, diabetes
[20]. Hydrolyzed linseed oil, which contains 52% ALA and mellitus, renal diseases, rheumatoid arthritis, lupus; psoria-
pure ALA were found to be capable of killing methicillin- sis, eczema, atopic and non-atopic dermatitis; atherosclero-
resistant Staphylococcus aureus [21]. Both LA and AA were sis, insulin resistance, obesity; dementia, schizophrenia, bi-
reported to inactivate animal herpes, influenza, Sendai, and polar disorders, Huntington’s disease, Alzheimer’s disease;
Sindbis virus within minutes of contact [22]. LA adminis- peptic ulcer disease; and cancer [1, 5, 39-41]. Furthermore, a
tered orally as safflower oil (which contains 76% LA) pro- 25-nucelotide ACE deletion polymorphism increases ACE
duced remission of mycosis fungoides, a rare skin disease of activity and such individuals showed a higher risk of devel-
viral etiology, in dogs that correlated with an increase in the oping stroke, obesity, emphysema, bipolar affective disor-
plasma levels of LA and AA [23]. AA, EPA, and DHA have ders, and cancers [42, 43]. This suggests that an altered ACE
been shown to induce death of Plasmodium both in vitro and activity and EFA metabolism plays a role in many diseases.
in vivo [24]. It is interesting to note that an analog of myris-
Transgenic rats overexpressing both human renin and
tic acid (14:0) showed selective toxicity to African Trypano-
angiotensinogen genes (dTGR) develop hypertension, in-
somes [25]. But no studies have been peformed with various
flammation, and renal failure and showed decreased forma-
EFAs and their metabolites for such properties. It is possible
tion epoxy-eicosatrienoic acids (5,6-, 8,9-, 11,12- and 14,15-
that several PUFAs alos may possess such anti-Trypano- EETs) and hydroxyeicosa-tetraenoic acids (19- and 20-
somal action. Furthermore, both prostaglandin E1 (PGE1) and
HETEs) from AA. These EETs and HETEs inhibited IL-6
PGA, derived from DGLA, AA, and EPA, are known to in-
and TNF-α-induced activation of NF-κB and prevented vas-
hibit viral replication and behave as anti-viral compounds
cular inflammation [44] suggesting that AA and other PU-
[26, 27]. These resuls suggest that EFAs/PUFAs may func-
FAs not only regulate ACE activity and Ang-II levels but
tion as endogenous anti-viral compounds and could be of
also possess anti-inflammatory properties.
some benefit in AIDS (acquired immunodeficiency syn-
drome) [28-30]. Thus, long-chain polyunsaturated fatty acids EPA and AA stimulate eNO synthesis [1, 38]. NO has
(LCPUFAs) and their products have anti-bacterial, anti- potent anti-atherosclerotic and anti-inflammatory actions.
fungal, anti-viral, and anti-parasitic actions. Lymphocytes Aspirin enhances the formation of eNO through the genera-
and macrophages contain significant amounts of LCPUFAs tion of epi-lipoxins that may explain its anti-inflammatory
and release them on appropriate stimulation. In addition, action [45]. Epi-lipoxins that have potent anti-inflammatory
LCPUFAs stimulate NADPH-dependent superoxide produc- actions enhance the generation of NO that, in turn, prevents
tion by macrophages, neutrophils and lymphocytes that are the interaction between leukocytes and the vascular endothe-
capable of killing the invading microorganisms [31]. lium. NO stimulates the formation of PGI2 from AA [46] and
lipoxins are derived from AA, EPA, and DHA. Aspirin in-
It will be interesting to study whether local application or
hibits TXA2 formation, a potent platelet aggregator and
intravenous infusions of PUFAs are of help in the treatment
vasoconstrictor, and enhances PGI2 formation, a platelet anti-
of various bacterial, viral and fungal infections. Since, neu- aggregator and vasodilator, and thus brings about its anti-
trophils, T cells and macrophages release PUFAs on stimula-
atherosclerotic actions. These results emphasize the close
tion; it is possible that this could be one of the defense
interaction between PUFAs, NO synthase, and COX en-
mechanisms of the body to fight infections [32-34]. Recent
zymes [47].
Essential Fatty Acids Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 5

PUFAs SUPPRESS PRODUCTION OF PRO-INFLAM- altering expression of its target genes, fatty acid synthase,
MATORY CYTOKINES acetyl-CoA carboxylase, or ATP citrate lyase and decreased
intestinal fatty acid synthesis by a posttranscriptional
ALA, DGLA, EPA, and DHA; LXs and resolvins sup-
mechanism independent of the SREBP pathway [64]. Feed-
press pro-inflammatory IL-1, IL-2, IL-6, macrophage migra-
ing mice on fish oil diet for 2 weeks decreased serum choles-
tion inhibitory factor (MIF), HMGB1 (high mobility group terol and triacylglycerol levels, by 50% and 60% respec-
box 1) and TNF-α production by T cells and other cells [1,
tively, hepatic FPP (farnesyl diphosphate synthase, a SREBP
48-50], and thus could function as endogenous anti-
target enzyme that is subject to negative-feedback regulation
inflammatory molecules. PGE2, PGF2α, TXA2 and LTs de-
by sterols in co-ordination with HMG-CoA reductase) syn-
rived from AA also modulate IL-6 and TNF-α production.
thase and HMG-CoA reductase mRNAs were decreased by
These results imply levels of IL-6 and TNF-α at the sites of
70% and 40% respectively. PUFAs down regulate hepatic
inflammation and injury may depend on the local levels of cholesterol synthesis by impairing the SREBP pathway [65].
various PUFAs and eicosanoids formed from them. In par-
PUFAs reduce SREBP-mediated gene transcription by in-
ticular, the suppressive action of DHA on IL-1β and TNF-α
creasing intracellular cholesterol content through the hy-
production by stimulated human retinal vascular endothelial
drolysis of cellular sphingomyelin, and the lipid second mes-
cells [51] is interesting since this suggests that it (DHA) and
senger ceramide, a product of sphingomyelin hydrolysis,
possibly other PUFAs may be important to prevent athero-
decreased SRE-mediated gene transcription of SREBP-1 and
sclerosis, macular degeneration, and diabetic retinopathy. SREBP-2 [66].
The ability of EPA and DHA to suppress the production of
pro-inflammatory cytokines and induce their anti-inflam- HMG-CoA reductase catalyzes the synthesis of me-
matory actions are mediated by their ability to increase valonate, which is the rate-limiting step in the mevalonate
PPAR-γ mRNA and protein activity [52]. pathway. Mevalonate is the precursor of cholesterol and a
variety of isoprenoid containing compounds. These isopre-
IL-1, IL-6, MIF (macrophage migration inhibitory factor) noid precursors are necessary for the posttranslational lipid
and TNF-α induce insulin resistance, have cytotoxic actions,
modification (prenylation) and hence, the function of Ras
are neurotoxic, and produce cachexia seen in tuberculosis,
and other small GTPases. Hence, inhibition of mevalonate
cancer, and AIDS. EPA, and other PUFAs ameliorate
pathway has the potential to disrupt the function of onco-
cachexia induced by TNF-α in animal tumor models [53].
genic forms of Ras. This explains the ability of both statins
Lipodystrophy and insulin resistance seen with the use of
and PUFAs to suppress Ras activity, anti-proliferative action
retroviral agents is due to increased levels of TNF-α and and induce apoptosis of tumor cells. In addition, small
decreased concentrations of adiponectin [54]. PUFAs de-
GTPases, which are prenylated products of the mevalonate
crease TNF-α and enhance adiponectin levels and thus, could
pathway, have negative control on the expression of BMPs
be of benefit to prevent/reverse insulin resistance [1, 55], and
(bone morphogenetic proteins). In view of this, inhibition of
side effects of retroviral drugs.
the mevalonate pathway by PUFAs will prevent the function
of small GTPases and enhance the expression of various
PUFAs DECREASE HMG-CoA REDUCTASE ACTIV-
BMPs. Various BMPs are known to be essential for neuronal
ITY
growth, proliferation, and differentiation. Thus, PUFAs
The two sterol regulatory element-binding proteins modulate brain growth and development, and neuronal dif-
(SREBPs): SREBP-1 and SREBP-2, each ~1150 amino acids ferentiation. This action is in addition to their (PUFAs) abil-
in length, control the transcription of the genes for the low- ity to form an important constituent of neuronal cell mem-
density lipoprotein (LDL) receptor and 3-hydroxy-3-methyl branes and involvement in memory formation and consolida-
glutaryl coenzyme A (HMG-CoA) synthase. The proteolytic tion [14, 15], explaining the beneficial action of PUFAs in
processing of both SREBPs is blocked by sterol overloading the prevention and treatment of dementia and Alzheimer’s
and enhanced when sterols are depleted by statins, the HMG- disease [67-73]. The beneficial actions of PUFAs in Alz-
CoA synthesis inhibitors. [56]. Cholesterol depletion that heimer’s disease, schizophrenia and dementia have been
occurs due to the use of statins leads to proteolytic activation attributed to the formation of anti-inflammatory compounds
of transcription factors of the SREBPs and also induces such as lipoxins and resolvins, whose formation is discussed
PPAR-γ expression [57], suggesting that PPAR-γ expression below.
is controlled by SREBPs. Similar to statins, AA, EPA, and
DHA are useful in the treatment of hyperlipidemias, have LIPOXINS, RESOLVINS, AND NEUROPROTECTIN
anti-proliferative action on tumor cells both in vitro and in D1
vivo, bind to DNA and regulate the expression of genes and
PUFAs also give rise to LXs and resolvins. Aspirin con-
oncogenes. More importantly, PUFAs are also potent inhibi- verts AA, EPA and DHA to form aspirin-triggered 15 epimer
tors of the HMG-CoA reductase enzyme [58, 59]. Statins
LXs (ATLs) that are potent inhibitors of acute inflammation
have the ability to enhance plasma AA concentrations and
[1, 47, 74, 75]. Acetylation of COX-2 by aspirin prevents the
decrease the ratio of EPA to AA significantly [60].
formation of prostanoids, but the acetylated enzyme remains
PUFAs have inhibitory effects on SREBP-1a and active in situ to generate 15R-hydroxy-eicosatetraenoic acid
SREBP-1c [61]. In CaCo-2 cells, PUFAs decreased gene and (15R-HETE) from AA that is released and converted by ac-
protein expression of SREBP-1 and FAS mRNA by interfer- tivated PMNs to the 15-epimeric LXs [74, 75]. This interac-
ing with LXR activity [62], and in rats PUFAs enhanced tion between endothelial cells and PMNs leading to the for-
cholesterol losses via bile acid synthesis [63]. In the intes- mation of 15R-HETE and its subsequent conversion to 15-
tine, dietary PUFAs suppress SREBP-1c mRNA without epimeric LXs by aspirin-acetylated COX-2 is a protective
6 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

mechanism to prevent local inflammation on the vessel wall Murine brain cells transformed enzymatically DHA to
by regulating the motility of PMNs, eosinophils, and mono- 17R series of hydroxy DHAs (HDHAs) that, in turn, is con-
cytes [1, 74, 75]. Endothelial cells oxidize AA (and possibly verted enzymatically by PMNs to di- and tri-hydroxy con-
EPA and DHA) via P450 enzyme system to form 11,12- taining docosanoids [77]. Similar small molecular weight
epoxy-eicosatetraenoic acid(s) that blocks endothelial cell compounds (similar to HDHAs) are generated from AA and
activation, suggesting that COX-2 enzyme is essential for the EPA. Thus, 15R-hydroxy containing compounds are formed
formation of LXs. Deficiency or absence of LXs leads to from AA, 18R series from EPA, and 17R-hydroxy series
interaction between PMN and endothelial cells as a result of from DHA that have potent anti-inflammatory actions and
which endothelial damage occurs that results in the initiation induce resolution of the inflammatory process and hence are
and progression of atherosclerosis, thrombus formation and called “resolvins” (see Fig. 2). Resolvins inhibited cytokine
coronary artery disease, and persistence of inflammation. generation, leukocyte recruitment, leukocyte diapedesis, and
exudate formation. AA, EPA, and DHA-derived resolvins
Compounds similar to 15R-HETE and 15-epimeric LXs
from acetylated COX-2 are formed due to communication
are also formed from EPA and DHA. These include conver-
between endothelial cells and PMNs. Resolvins inhibit brain
sion of EPA to 18R-HEPE (18R- hydroxy-eicosapentaenoic
ischemia-reperfusion injury [78]. Thus, lipoxins and resolv-
acid), 18-HEPE, and 15R-HEPE. Activated human PMNs, in
ins formed from AA, EPA, and DHA have cardio-protective,
turn, converted 18R-HEPE to 5,12,18R-triHEPE and 15R-
HEPE to 15-epi-LXA5 by 5-lipoxygenase. Both 18R-HEPE neuroprotective, and other cytoprotective actions.
and 5,12,18R-triHEPE inhibited LTB4-stimulated PMN tran- Of the several 17-hydroxy-containing bioactive media-
sendothelial migration similar to 15-epiLXA4. 5,12,18R- tors derived from DHA that were termed docosatrienes and
triHEPE competed with LTB4 for its receptors and inhibited 17S series resolvins, 10,17S-dihydroxydocosatriene termed
PMN infiltration, and thus, 5,12,18R-triHEPE suppresses as neuroprotectin D1 (NPD1) that reduced infiltration of
LT-mediated responses when present at the sites of inflam- PMNs, showed anti-inflammatory and neuroprotective prop-
mation [76]. erties [78, 79]. NPD1 inhibited oxidative stress-induced

COOH

COOH
Aspirin/COX2 O(O)H

eicosapentaenoic acid 18R-hydro(per)oxy-EPE

COOH
COOH

OOH
OH

RvE2 5S-hydropeoxy, 18R-hydroxy-EPE


OH
OH

OH
COOH COOH
O
OH

RvE1 HO 5,6-epoxy, 18R-hydroxy-EPE


OH

Fig. (2a). Scheme showing the formation of resolvin E (RvE) derived from EPA.
In the endothelial cells, the COX-2 enzyme that has been acetylated introduces an 18R hydroperoxy-group into the EPA molecule (c.f. the
role of aspirin in the biosynthesis of the epi-lipoxins). This is reduced to the corresponding hydroxy compound before a 5S-hydroperoxy
group is introduced into the molecule by the action of 5-lipoxygenase as in the biosynthesis of leukotrienes. A further reduction step pro-
duces 15S,18R-dihydroxy-EPE or resolvin E2. Alternatively, the 5S-hydrpperoxy, 18R-hydroxy-EPE intermediates is converted to a 5,6-
epoxy fatty acids in polymorphonuclear leukocytes I humans and eventually to 5S,12R,18R-trihydroxy-6Z,8E,10E,14Z,16E-eiocsapentaenoic
acid or resolvin E1 by process similar to the formation of leukotrienes in leukocytes.
Essential Fatty Acids Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 7

COOH COOH
HO OH

OH
OH

HO

OH
Resolvin D 1 Resolvin D2

Fig. (2b). Structures of Resolvin D1 and D2. DHA is converted to 17R-resolvins by a similar aspirin-triggered mechanism similar to the
scheme shown in Figure 2a. In the absence of aspirin, COX-2 of endothelial cells converts DHA to 13S-hydroxy-DHA. In the presence of
aspirin, the initial product is 17R-hydroxy-DHA, which is converted to 7S-hydroperoxy, 17R-hydroxy-DHA by the action of a lipoxygenase,
and thence via an epoxy intermediate to epimeric resolvins D1 and D2. An alternative lipoxygenase-generated intermediate, 4S-hydroperoxy,
17R-hydroxy-DHA, is transformed via an epoxide to epimeric resolvins D3 and D4. 17S Resolvins of the D series are produced in cells in
the absence of aspirin by a reaction catalyzed in the first step by a lipoxygenase.

COOH
DHA

COOH
OOH 17S-hydroperoxy-DHA

COOH
O
16, 17-epoxy-docosatriene

OH
OH
17

10 COOH
neuroprotectin D1

Fig. (2c). Scheme showing the synthesis of neuroprotectin D1. Resolvins are generated in brain tissue in response to aspirin treatment, and in
addition docosatrienes termed neuroprotectins are also produced. The lipoxygenase product 17S-hydroperoxy-DHA is converted first to a
16(17)-epoxide and then to the 10, 17-dihydroxy docosatriene denoted as 10, 17 S-DT or NPD1. As with the leukotrienes, there are three
double bonds in conjugation, hence the term ‘triene’, though there are six double bonds in total.

apoptosis of human retinal pigment epithelial cells [80]. for the structure of some nitrolipids and their method of de-
Both LXs and NPD1 enhanced wound healing [81], and tection by MS). These nitroalkene derivatives, termed as
promoted brain cell survival via the induction of antiapop- nitrolipids, produce vascular relaxation, inhibit neutrophil
totic and neuroprotective gene-expression programs [67, 68]. degranulation and superoxide formation, and inhibit platelet
Based on these findings, it is logical to propose that un- activation [82-84]. Nitrolipids have endogenous PPAR-γ
ligand activity and release NO [85]. These actions of nitrol-
der physiological conditions COX-1 and COX-2 enzymes
ipids prevent platelet aggregation, thrombus formation and
induce the formation of beneficial eicosanoids PGE1, PGI2,
atherosclerosis, and it is possible that they also possess anti-
and LXs, resolvins, and NPD1 to prevent inflammation.
inflammatory actions. This implies that nitrolipids may have
Failure to produce adequate amounts of LXs, resolvins, and
a significant role in low-grade systemic inflammatory condi-
NPD1 or interference with their action and/or a simultaneous
increase in the production of pro-inflammatory eicosanoids tions such as type 2 diabetes, hypertension, hyperlipidemias,
insulin resistance, and metabolic syndrome X. Since, nitrol-
and cytokines could lead to initiation and persistence of in-
ipids are present both in the plasma and urine in substantial
flammation and tissue damage.
amounts, it will be interesting to measure their levels in these
NITROLIPIDS conditions. These evidences suggest that PUFAs not only
form precursors to various eicosanoids, resolvins, LXs, and
NO can react with PUFAs to yield their respective nitro- NPD1 but also react with various other molecules and form
alkene derivatives that can be detected in plasma (see Fig. 3
8 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

O O2N

HO

O NO2

HO

Two regioisomers of OA-NO2: 9- and 10-nitro-9-cis-octadecenoic acids

A.
O O2N OH O OH NO2

-O -O
202 171

B.
O OH NO2 O OH NO2

-O -O
171 211

O O2N OH O O2N OH

-O -O
202 242

C.
O OH NO2 O O2N OH

-O -O
171 242

O O2N OH O HO NO2

-O -O
202 251
O HO NO2 O O2N OH

-O -O
211 282

A= Oleic acid-NO [18:1 (OH)-NO2 ]


B= Linoleic acid-NO [18:2 (OH)-NO2 ]
C= Linolenic acid-NO [18:3 (OH)-NO2]
Some regions of the MS/MS fragmentation patterns are shown above. The 10-nitro regioisomers of 18:1(OH)-NO2 is evident by the intense peak correspond-
ing to m/z 171; Also present are fragments consistent with the 9-nitroregioisomer (m/z 202), loss of a nitro group (m/z 297) and water (m/z 326),
18:2(OH)-NO2 shows a predominant m/z 171 fragment again consistent with an oxidation product of LNO2 nitrated at 10-carbon.
Multiple regioisomers of 18:3(OH)-NO2 are present (see C above).

Fig. (3). Nitrolipids and their detection.

novel compounds that have biological activity. It is not yet pholipid fatty acid content. Since LA and ALA can be de-
clear whether nitrolipids have any interaction with eicosa- saturated and elongated to form their long-chain metabolites,
noids, lipoxins, and resolvins. A study in this direction may it is reasonable to assume that dietary content EFAs is one
prove to be interesting. factor that influences the degree of inflammation and its sub-
sequent resolution. PUFAs alter the cell membrane fatty acid
ROLE OF EFAs/PUFAs IN SOME DISEASE PROC- composition that, in turn, modulates cell/tissue response to
ESSES infection, injury and inflammatory events. Increased dietary
intake of GLA, DGLA, and EPA/DHA substantially de-
Inflammation creases inflammatory response. GLA is elongated to form
DGLA that, in turn, is converted to PGE1 and its (DGLA)
The amount and type of PUFA(s) released in response to
15-lipoxygenase product 15-hydroxy-8, 11, 13-eicosatrienoic
inflammatory stimuli depends on the cell membrane phos-
Essential Fatty Acids Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 9

acid (15-OH-20: 3, ω6), which have anti-inflammatory ac- molecules such as lipoxins and resolvins and their temporal
tions [86-88]. GLA, DGLA, AA, and EPA inhibited IL-1 relationship is interesting. 5S,6R,15S-trihydroxy-7,9,13-
and IL-2 production without conversion into their cyclo- trans-11-cis-eicosatetraenoic acid (LXA4) generated from
oxygenase pathway products [89, 90]. Cellular phospholipid AA via the lipoxygenase pathway during cell-cell interaction
concentrations of AA, formation of LTB4, LTC4 /LTD4, 6- and aspirin-triggered 15-epi- LXA4 (ATL), an LXA4 epimer,
keto-PGF1α (a metabolite of PGI2), and TXB2 levels, mye- are endogenous anti-inflammatory lipid mediators that in-
loperoxidase (MPO) activity and neutrophil accumulation duce resolution of inflammation acting via G-protein-
were lower, whereas the concentrations of EPA and DHA coupled receptor, LXA4 receptor [109]. These lipid media-
and DGLA were higher with reduced plasma concentrations tors down-regulate PMN infiltration and thus, function as
of soluble vascular cell adhesion molecule-1 and soluble E- “breaking signals” to stop inflammation. Both EPA and
selectin in experimental animals and patients that received a DHA also form precursors to di- and trihydroxy-containing
combination of GLA and EPA (in the form of a mixture of lipid autacoids resolvins, docosatrienes, and neuroprotectins
evening primrose oil and fish oil) and fish oil rich in (see Figs. 1 and 2 for the metabolism of EFAs and formation
EPA/DHA compared to corn oil control, though no change of these molecules) that possess potent anti-inflammatory
in the TNF-α levels between the control endotoxin-treated actions and may serve not only to activate resolution of the
rats and the fish oil and fish oil plus borage oil treated groups inflammatory process but may also activate the resolution
was noted [91-94]. It is likely that when the cell membrane process and promote return to normalcy. Simultaneous side-
lipid pool is rich in GLA/DGLA/EPA/DHA and contains by-side lipidomic and proteomic-based approach [110] using
appropriate amounts of AA, specific activation of sPLA2 and the standard murine model of self-resolving inflammation
cPLA2 (soluble and cytosolic phospholipase A2 respectively) revealed that the initial response (0-12 hours) of inflamma-
in response to an inflammatory stimuli would occur that tion is composed of PMN influx followed monocyte infiltra-
leads to the formation of increased amounts of LXs, PGD 2 tion and a transient exudation of serum proteins. During this
and 15deoxyΔ12-14PGJ2, eNO, GSNO, PGE1, PGI2, PGI3, and early phase also displayed formation (0-4 hours) of both
HPETEs that dampen inflammatory process and enhance LTB4 and LXA4 from AA that showed identical kinetics. At
resolution of inflammation [95, 96]. the same time (2 hours), unesterified AA, EPA, and DHA
was found in the inflammatory exudate presumably released
DGLA, EPA, and DHA suppress NF-κB signaling [97,
due to the involvement of calcium-insensitive iPLA2. During
98], whereas AA activates NF- κB [99]. PUFAs serve as
this time (2 hours), S100A9, a cytosolic PMN protein that
endogenous ligands for PPARs and thus, suppress inflamma-
can be secreted and exhibits potent actions on inflammatory
tory events [100, 101]. Oxidized PUFAs prevented LPS- or cell recruitment and a known AA-binding protein, was pre-
PMA-induced activation of transcription factor NF-κB and
sent in the inflammatory exudate from the initiation of in-
consequent induction of adhesion molecules, and prevented
flammation till the onset of resolution paralleling the PMN
the binding of monocytes to cultured endothelium due to the
infiltration time course and decreased by 24 hours. Serum
upregulation of adhesion molecules by exposure to LPS, IL-
proteins plasminogen, fibrinogen, serum albumin, and α1 -
1α, TNF-α, or phorbol myristate acetate (PMA). The active
macroglobulin were abundant in inflammatory exudates 4
principle of oxidized PUFAs was found to be hydroperoxy hours after the initiation of inflammation. α1-macroglobulin
fatty acids [102, 103]. This ability of oxidized lipids to in-
binds to tissue-plasminogen activator and may also bind to
hibit the expression of endothelial cell adhesion molecules
pro-inflammatory cytokines [111] present in the inflamma-
could be a natural mechanism whereby inflammation-
tory exudate and thus, may help to lower their effective local
mediated oxidation of endothelial PUFAs retard unrestrained
concentrations and initiate the onset of resolution. The dis-
inflammatory responses. Oxidized ω-3 fatty acids inhibit
tinct events observed during the phase of resolution of in-
NF-κB activation via a PPAR-α-dependent pathway. flammation include: (a) an increase in the levels of hapto-
It is generally believed that increased intake of PUFAs globin whose purpose seems to be to prevent oxidative dam-
may enhance lipid peroxidation, and that these oxidized age, (b) activation of CD163, a scavenger receptor that has
products are harmful to tissues. But, it was reported that in- the ability to trigger anti-inflammatory pathways [112], (c)
creased intake of EPA/DHA, in fact, reduces in vivo lipid an increase in inflammatory exudate 10,17S-docosatriene,
peroxidation and oxidative stress in humans [104-106]. (d) a remarkable rise in the concentrations of TGF-β due to
its release from macrophages, and (d) an increase in PGE2
It is now believed that the central causative event for levels at the beginning of resolution and a decline during the
several of the most common diseases such as atherosclerosis, active phase of resolution with an increase in the levels of
cancer, asthma, collagen vascular diseases such as lupus, PGD2 coinciding with the decrease in PGE2 (at 24 hours).
rheumatoid arthritis, and scleroderma; CHD, hypertension, Based on these findings it is apparent that both PGE2 and
type 2 diabetes mellitus, obesity, and neuropathological dis- PGD2 switch eicosanoid biosynthesis from predominantly
eases such as schizophrenia, stroke, Alzheimer’s, and Park- “pro-inflammatory” LTB4 to “anti-inflammatory” LXA4 pro-
inson’s diseases is low grade systemic inflammation [16, duction that suppresses PMN infiltration at the site of in-
107]. Hence, understanding factors that initiate, modulate, flammation. It is noted that some of these temporal events
perpetuate, and inhibit inflammation are of great signifi- could be different during TNF-α-induced inflammatory
cance. Recent studies suggest that inflammation to the “nor- process suggesting that the different kinetics in eicosanoid
mal” non-inflamed state is not merely the passive termina- generation may reflect the nature of the experimental models
tion of inflammation but rather an actively regulated pro- used and/or stimulus-specific signaling pathways. Neverthe-
gram of resolution [108]. In this context, the interaction be- less, it is clear that ATLa inhibits PMN infiltration and pro-
tween pro-inflammatory cytokines and anti-inflammatory inflammatory cytokine and chemokine release, and enhances
10 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

TGF-β release, whereas resolvin E1 and 10,17S-docosatriene free radicals that results in the development of insulin resis-
activated and shortened and thus, accelerated resolution and tance; decrease in plasma and cellular HDL concentrations,
the healing process. and decrease the formation of eNO, PGE1, PGI2, PGI3, LXs,
resolvins, and NPD1 that may ultimately initiate and per-
In this context, it should be noted that (a) LXA4 potently
inhibited TNF-α-induced IL-8 release [113], whose receptor petuate atherosclerosis.
(LTA4 receptor) is induced by IL-13 and IFN-γ; (b) LXA4 Normally a balance is maintained between pro- and anti-
and ATLa inhibited TNF-α secretion from activated T cells inflammatory cytokines and eicosanoids, and pro- and anti-
[114], and LXA4 suppressed IL-12-induced responsiveness platelet aggregation-inducing molecules such that atheroscle-
of murine splenic dendritic cells after microbial stimulation rosis is prevented. For instance, TXA2 is a platelet aggrega-
[115]. IL-12 production by antigen presenting cells (APCs) tor and vasoconstrictor; whereas PGI2 has opposite actions.
is essential for the induction of IFN-γ that mediates host re- PGI2 prevents whereas TXA2 promotes atherosclerosis [118].
sistance to bacterial and protozoan intracellular pathogens. Both PGE2 and PGI2, formed from AA, participate in the
Both LXA4 and LXB4 inhibit neutrophil chemotaxis, NK pathogenesis of RA (rheumatoid arthritis). PGI receptor de-
(natural killer) cell cytotoxicity, monokine and metalloprote- ficient (IP-/-) mice exhibited significant reduction in arthritic
inase production, TNF-α-induced superoxide anion, IL-1β, scores, reduction in IL-1β and IL-6 levels in the arthritic
and macrophage inflammatory protein-2 production by neu- paws when were subjected to collagen-induced arthritis. Ad-
trophils [115]. These evidences suggest that lipoxins and dition of IP agonist to cultured synovial fibroblasts enhanced
resolvins could be potent immunosuppressors. IL-6 production. In contrast, inhibition of PGE receptor sub-
type (EP2 or EP4) alone did not affect inflammatory events
The various anti-inflammatory products such as lipoxins
in collagen-induced arthritis, whereas suppression of arthritis
and resolvins formed from AA/EPA/DHA may explain the
occurred when both EP2 and EP4 receptors were inhibited,
beneficial actions of EPA/DHA noted in several clinical
conditions and the anti-inflammatory actions of aspirin. The suggesting that both PGE2 and PGI2 participate in RA [119].
These results emphasize the fact that the type of products
ability of EPA/DHA supplementation and aspirin use to pre-
formed from various PUFAs determines the final outcome of
vent CHD could be attributed to the formation of various
any physiological and pathological process.
lipoxins and resolvins. It is possible that there may be indi-
vidual differences in their ability to form various lipoxins In this context, the interaction between various PUFAs is
and resolvins that may explain the differences in the benefits particularly important. In perfused vascular tissue, DGLA
noted with the use of EPA/DHA and aspirin that could be and AA increased the conversion of EPA to PGI3, a vasodila-
attributed to the genetic differences in the expression and tor and platelet anti-aggregator [120-122], whereas orally
activation of enzymes concerned with their synthesis and administered EPA enhanced AA conversion to PGI2 [123]
degradation. Decreased production of lipoxins and resolvins and inhibited the activity of Δ6 and Δ5 desaturases (Δ6 > Δ5 )
could be responsible for the low-grade inflammation seen in which resulted in enhanced tissue levels of DGLA as a result
obesity, hypertension, type 2 diabetes, metabolic syndrome of decreased conversion of DGLA to AA. The enhanced
X, CHD, atherosclerosis, and persistent inflammation ob- levels of DGLA could lead to an increase in the formation of
served in collagen vascular diseases such as lupus, rheuma- PGE1, a vasodilator and platelet anti-aggregator. This close
toid arthritis and scleroderma. In contrast, immunosuppres- interaction between DGLA, AA, and EPA [124] implies that
sion seen in leprosy, miliary tuberculosis, some bacterial and optimal levels of DGLA, AA, EPA, and DHA need to be
protozoal infections might be due to inappropriate produc- present in the tissues to optimize the formation of various
tion of lipoxins and resolvins. beneficial eicosanoids and lipoxins and resolvins to prevent
atherosclerosis (Fig. 4).
Atherosclerosis
Fetal-Alcohol Syndrome
Atherosclerosis is a disease of low-grade systemic in-
flammation [116] and is closely linked to the integrity of Ethanol exposure during brain development induces neu-
endothelial cells. Healthy endothelial cells synthesize and rodevelopmental defects referred to as fetal-alcohol syn-
release adequate amounts of GLA/DGLA/EPA/DHA, NO, drome (FAS) that is characterized by hyperactivity, learning
PGI2, PGI3, PGE1, and possibly, lipoxins and resolvins to and memory deficits, mental retardation, psychosis, depres-
prevent platelet aggregation on their surface, prevent inap- sion, and schizophrenia. Ethanol-induced neurotoxicity, oxi-
propriate expression of adhesion molecules, and production dative stress, induction of apoptosis, excitotoxicity, interfer-
of pro-inflammatory cytokines. Pro-inflammatory cytokines ence with the action of growth factors, and EFA metabolism
IL-1, IL-2, IL-6, and TNF-α induce oxidant stress Shear may all could be responsible for the development of FAS.
stress, hyperglycemia, clinical or sub-clinical infections, and Neurons are susceptible to ethanol-induced apoptotic cell
low-grade systemic inflammation as seen in type 2 diabetes death during synaptogenesis during brain growth spurt,
mellitus, hypertension, hyperlipidemia, and metabolic syn- which occurs during the third trimester of pregnancy and
drome X could induce the production of pro-inflammatory perinatal period.
cytokines. EPA/ DHA/GLA and HDL suppress IL-6 and
Recent studies showed that nicotinamide enhanced neu-
TNF-α synthesis and secretion, inhibit free radical generation ronal survival following free radical exposure and oxidative
and adhesion molecule expression, enhance eNO generation
stress [125]. The mechanism of nicotinamide-mediated neu-
and thus, prevent oxidant stress [117]. This suggests that
roprotection has been attributed to inhibition of caspase-3
endothelial cell deficiency of GLA/ DGLA/EPA/DHA might
and release of cytochrome-c from mitochondria during oxy-
increase the production of pro-inflammatory cytokines and
gen-glucose deprivation [126]. Subsequent studies showed
Essential Fatty Acids Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 11

Fig. (4). Scheme showing interaction(s) between ω-3 and ω-6 fatty acids and their effect on the formation of PGI2, PGI3, PGE1, and LXs,
resolvins and NPD1.
(-) Indicates inhibition or block in the synthesis, formation or release.
(+) Indicates enhancement in the formation or release.
It is proposed that LXs, resolvins, and NPD1 enhance the formation and/or action of PGI2, PGI3 and suppress that of TXA2 and TXA 3 thus
may prevent atherosclerosis and show their cytoprotective actions. LXs, resolvins and NPD1 suppress formation of LTs.

that nicotinamide can prevent deleterious effects of ethanol sumption is substantial, tissue concentrations of PUFAs de-
on the developing mouse brain when given shortly after cline leading to fall in the synthesis of various PGs, LXs,
ethanol exposure which led to the suggestion that nicotina- resolvins, and NPD1. This is supported by our recent obser-
mide may hold promise as preventive therapy of FAS [127]. vation that in rats fed alcohol, the plasma levels of lipoxins
Nicotinamide is a co-factor in the metabolism of EFAs, and resolvins is decreased whereas those of TXs and LTs are
which could explain the beneficial action of nicotinamide in increased (unpublished data).
FAS.
DHA and other PUFAs released from the membrane in
EFAs are not only structural components of the brain but response to neurotransmitters [131] activate RXR as PUFAs
also influence nerve conduction, transmitter release and ac- form ligands for RXR receptor [132]. DHA deficiency re-
tion [128]. Prostaglandins (PGs) derived from EFAs modify sults in impaired spatial learning and other abnormalities
nerve conduction and transmitter function. Ethanol (a) re- [133]. RXR heterodimerization partners PPARs, liver X re-
duces blood linoleic acid (LA) levels; (b) blocks conversion ceptors, and farnesoid X receptors that are essential for regu-
of LA and ALA to AA, and EPA and DHA respectively by lating energy and nutritional homeostasis. PUFAs being
inhibiting Δ6 and Δ5 desaturases; and (c) enhances the con- ligands for RXR and PPARs modulate these and other regu-
version of DGLA to PGE1 [128]. Ethanol enhanced IL-6 and latory events by binding to these nuclear receptor heterodi-
TNF-α production suggesting that immunological mecha- mers.
nisms play a role in ethanol-induced diseases. Cerebral cor-
tex from chronic ethanol-fed rats showed up-regulation of AA, DHA, EPA, and LXs, resolvins, and NPD1 have
inducible nitric oxide (iNOS), COX-2, IL-1β, and activation neuroprotective actions [47, 68, 78, 80], inhibit IL-6 and
of transcription factors NF-κB and AP-1, effects that in- TNF-α production that are neurotoxic, and increase synthesis
creased both caspase-3 and apoptosis [129]. Brains of etha- of endothelial nitric oxide (eNO), a neurotransmitter. When
nol treated mice show raised phospholipase A2 and phos- neurons are depleted of their PUFA content, as happens after
pholipase C activity [130] that can cause the release of AA, high alcohol intake, formation of LXs, resolvins, and NPD 1
EPA, and DHA from membrane phospholipids. LXs, resolv- will be suboptimal and hence, neuronal death induced by
ins, and NPD1 formed from AA, EPA, and suppress IL-6 and TNF-α cannot be antagonized. This results in FAS. Thus,
TNF-α production [47, 110]. PGs, LXs, resolvins, and NPD 1 nicotinamide by augmenting the action of Δ6 desaturase and
precursor supplies are limited; therefore when ethanol con- enhancing the formation of various PUFAs and their prod-
12 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

ucts [134] may be able to bring about its neuroprotective with EPA- and DHA-rich oil; (ii) highly purified EPA re-
action against ethanol induced FAS. duced insulin resistance and decreased the incidence of type
2 diabetes in experimental animals; and (iii) decreased con-
In addition, EPA and AA protected heart against ische-
centrations of EPA, DHA and AA in skeletal muscle phos-
mia/reperfusion-induced injury by inhibiting caspase-3 acti-
pholipids was found to be associated with decreased insulin
vation and PARP-cleavage and reducing the apoptotic index sensitivity in humans [1, 152-155]. I observed that plasma
during reperfusion by increasing ERK phosphorylation and
phospholipid concentrations of EPA, DHA and AA were low
decreasing p38 phosphorylation during reperfusion [135].
in subjects with hypertension, diabetes mellitus and CHD
DHA promoted neuronal survival by facilitating membrane
[156]. Metabolic syndrome X is uncommon in Greenland
translocation/activation of Akt. In vivo reduction of DHA by
Eskimos whose traditional diet is rich in EPA and DHA,
dietary depletion increased hippocampal neuronal suscepti-
whereas South Asian Indians, who are at high risk of devel-
bility to apoptosis [70]. Retinal pigment epithelium and syn- oping metabolic syndrome X, have significantly lower con-
aptic membranes contain the highest content of DHA of all
centrations of AA, EPA, and DHA compared to healthy Ca-
cell membranes, and DHA is required for retinal pigment
nadians and Americans [157]. These observations suggest
epithelium functional integrity. NPD1 that is formed from
that PUFAs, cytokines, and NO interact with each other and
DHA upon its release from the cell membrane by the action
play an important role in the pathobiology of metabolic syn-
of phospholipase A2 counteracted H2O2/TNF-α-induced
drome X [1-3]. There is evidences to suggest that PUFAs by
apoptosis by upregulating anti-apoptotic proteins Bcl-2 and their ability to influence brain growth and development, hy-
Bcl-XL and decreasing pro-apoptotic Bax and Bad proteins.
pothalamic neurons, and the secretion and actions of various
In addition, NPD1 inhibited oxidative stress-induced caspase-
neuropeptides and their receptors and insulin receptors in
3 activation, IL-1β-stimulated expression of cyclo-oxygen-
various regions of the brain during the perinatal period may
ase-2 and thus, prevented oxidative stress-induced apoptosis
have significant impact on the development of obesity, hy-
and inflammation [80]. EFA deficiency promotes respiratory
pertension, type 2 diabetes, metabolic syndrome X, CHD,
uncoupling [136, 137]. Mild respiratory uncoupling-a form and atherosclerosis in adult life [1-5, 73, 107].
of mitochondrial dysfunction-increases oxidative stress and
decreases nitric oxide availability or biological action [138]. SCHIZOPHRENIA, HUNTINGTON’S DISEASE, AND
Furthermore, PUFAs modulate the activities of uncoupling ALZHEIMER’S DISEASE
proteins [5], and under certain specific conditions PUFAs
function as anti-oxidants and prevent free radical-induced Low-grade systemic inflammation seems to play a sig-
damage to cells [139, 140]. PUFAs have the ability to modu- nificant role in the pathobiology of schizophrenia,
late the expression, properties, and action dopamine, sero- Huntington’s disease and Alzheimer’s disease. In patients
tonin, and acetylcholine [141-147], especially during the with schizophrenia, both circulating and cerebrospinal fluid
perinatal period during which the growth and development (CSF) concentrations of pro-inflammatory cytokines are in-
of brain is critical [14, 15]. This evidence suggests that PU- creased and the plasma phospholipid concentrations of EPA
FAs, especially EPA/DHA could be of benefit in the preven- and DHA are decreased. Supplementation of EPA (espe-
tion and treatment of FAS. cially ethyl EPA) was reported to be of some benefit to these
patients [reviewed in 158, 159].
Metabolic Syndrome X
It was reported that diet high in DHA slowed the pro-
Metabolic syndrome X is characterized by abdominal gression of Alzheimer's disease in mice. Specifically, DHA
obesity, atheroslcerosis, insulin resistance and hyperinsu- cut the harmful brain plaques that mark the disease. Mice
linemia, hyperlipidemias, essential hypertension, type 2 dia- genetically altered to develop Alzheimer's disease, when
betes mellitus, and coronary heart disease (CHD). Plasma were fed with DHA-fortified chow had 70-percent less
levels of C-reactive protein (CRP), TNF-α, and IL-6, mark- buildup of amyloid protein in the brain compared with con-
ers of inflammation, are elevated in subjects with obesity, trol or DHA-deficient mice [67-69]. DHA protected against
insulin resistance, essential hypertension, type 2 diabetes, damage to the "synaptic" areas and enabled mice to perform
and CHD, suggesting that metabolic syndrome X is a low- better on memory tests. These studies indicate that increased
grade systemic inflammatory condition [148]. intake of DHA could be of benefit in people who are geneti-
cally or otherwise predisposed to develop the disease.
EPA, DHA, and AA, inhibit TNF-α and IL-6 production;
enhance eNO generation, inhibit 3-hydroxy-3-methylglutaryl Huntington’s disease is an inherited neurodegenerative
coenzyme A (HMG-CoA) reductase and angiotensin con- disorder due to a mutation in exon 1 of the Huntingtin gene
verting enzyme activities, function as endogenous ligands for that encodes a stretch of polyglutamine (poly Q) residues
PPARs, and modulate leptin gene expression [1, 5, 37, 58- close to the N-terminus of the Huntingtin protein. Aggre-
60, 149, 150]. Thus, PUFAs suppress inflammation, regulate gated poly Q residues are toxic to the neuronal cells. Trans-
cholesterol metabolism, enhance the production of adi- genic R6/1 mice that develop late-onset neurologic deficits
ponectin, and decrease insulin résistance. This may explain similar to the motor abnormalities of Huntington’s disease
why and how PUFAs are useful in protection against CHD, seen in humans showed increased survival rates and de-
atheroslcerosis, and decrease blood pressure. PUFAs bind to creased neurologic deficits when were supplemented with
PPARs (similar to thiazolidinediones) and thus, enhance PUFAs, especially ethyl EPA [41], suggesting that unsatu-
adiponectin levels [151] that, in turn, improve insulin resis- rated fatty acids may prevent or arrest poly Q aggregation.
tance. Furthermore, (i) hypertension and insulin resistance These results suggest that PUFAs, in general, are useful in
was ameliorated in experimental animals by feeding them the treatment of various neurological diseases. Some of the
Essential Fatty Acids Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 13

beneficial actions of these PUFAs in neurological diseases Recently it was reported that PUFAs participate in athe-
could be due to the increased formation of lipoxins and re- rosclerosis by modulating the expression of uncoupling pro-
solvins that have neuroprotective actions. But, it is not clear tein-1 (UCP-1) in the vascular tissue. Oxidative stress is
why and how a particular fatty acid is useful only in a spe- known to play a major role in atherosclerosis. Mitochondrial
cific neurological condition. For instance, DHA is useful in electron transport accounts for most of the reactive oxygen
Alzheimer’s disease whereas ethyl EPA is of benefit in species (ROS) generated. Uncoupling proteins (UCPs) (inner
Huntington’s disease and schizophrenia. More research is mitochondrial membrane anion transporters) allow protons
needed to understand the molecular mechanisms of action of to leak back into the mitochondrial matrix, thereby decreas-
EPA/DHA in these neurological conditions. ing energy available for ROS generation. Superoxide anion
activates UCPs [160], which limits further superoxide anion
CONCLUSIONS generation by dissipating protonmotive force. Uncoupling
decreases glucose-induced ROS formation, and thus prevents
It is evident from the preceding discussion that EFAs and
vascular damage in cultured endothelial cells [161]. Respira-
their metabolites including eicosanoids, LXs, resolvins, and
tory uncoupling is increased in the aortae of pigeons suscep-
NPD1 have many biological actions and participate in sev-
tible to atherosclerosis [162]. Smooth muscle cells are a ma-
eral diseases processes (Fig. 5).
jor source of ROS in the vasculature [163]. It was reported

Fig. (5). Scheme showing the relationship between various mediators of tissue damage/resolution and clinical conditions and the role of PU-
FAs and their metabolites in these processes.
14 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

that UCP-1 expression in aortic smooth muscle cells pro- [5] Das, U. N. A Perinatal Strategy for Preventing Adult Diseases: The
duced hypertension, increased high fat diet-induced athero- Role of Long-Chain Polyunsaturated Fatty Acids. Kluwer Aca-
demic Publishers, Boston, 2002.
sclerosis without affecting cholesterol levels, enhanced su- [6] Cantwell, M. M.; Flynn, M. A.; Cronin, D.; O’Neill, J. P.; Gibney,
peroxide anion production, and decreased the availability of M. J. (2005) J. Hum. Nutr. Diet. 18, 377-385.
nitric oxide, suggesting that mild respiratory uncoupling due [7] Lopez-Garcia, E.; Schultze, M. B.; Meigs, J. B.; Manson, J. E.;
to mitochondrial dysfunction causes atherosclerosis [164]. Rifai, N.; Stampfer, M. J.; Willett, W. C.; Hu, F. B. (2005) J. Nutr.
135, 562-566.
EFA deficiency promotes respiratory uncoupling [165-167] [8] Mozaffarian, D.; Pischon, T.; Hankinson, S. E.; Rifai, N.;
and atherosclerosis [117, 168]. Atherosclerosis-free aortae Joshipura, K.; Willett, W. C.; Rimm, E. B. (2004) Am. J. Clin.
have abundant amounts of EFA linoleate, whereas fatty Nutr. 79, 606-612.
streaks of early atherosclerosis are deficient in EFAs [169, [9] Brenner, R. R. (1982) Prog. Lipid Res. 20, 41-48.
170]. Pro-inflammatory eicosanoids stimulate the formation [10] Matsuzaka, T.; Shimano, H.; Yahagi, N.; Amemiya-Kudo, M.;
Yoshikawa, T.; Hasty, A. H.; Tamura, Y.; Osuga, J.; Okazaki, H.;
and the activity of adhesion molecules (integrins), and thus, Iizuka, Y.; Takahashi, A.; Sone, H.; Gotoda, T.; Ishibashi, S.; Ya-
promote atherosclerosis and inflammation, whereas GLA, mada, N. (2002) J. Lipid Res. 43, 107-114.
DGLA, EPA, and DHA, and their products such as LXs, [11] Cook, H. W. (1981) Lipids 16, 920-926.
resolvins, and NPD1 suppress the expression of adhesion [12] Das, U. N. (2005) Prostaglandins Leukot. Essen. Fatty Acids 72,
343-350.
molecules and thus, inhibit atherosclerosis and inflammation. [13] Coetzer, H.; Claassen, N.; van Papendorp, D. H.; Kruger, M. C.
LTB4, a 5-lipoxygenase (5-LO) product derived from (1994) Prostaglandins Leukot. Essen. Fatty Acids 50, 257-266.
[14] Das, U. N. (2003) Nutrition 19, 62-65.
AA, is a potent leukocyte chemoattractant, a pro- [15] Calderon, F.; Kim. H. Y. (2004) J. Neurochem. 90, 979-988.
inflammatory molecule, and participates in atherosclerosis [16] Das, U. N. (2006) Med. Sci. Res. 12, RA99-RA111.
through its receptors BLT-1 and BLT-2. LTB4 promotes [17] Das, U. N. (2006) Adv. Clin. Chem. 41, 189-229.
atherosclerosis by chemo-attracting monocytes, and enhanc- [18] Kodicek, E. (1949) Symposia Soc. Exp. Biol. 3, 217-232.
ing the formation of foam cells [171]. It was reported that the [19] Lacey, R.W.; Lord, V.L. (1981) J. Med. Microbiol. 14, 41-49.
[20] Galbraith, H.; Miller, T.B.; Paton, A.M.; Thompson, J.K. (1971) J.
expression of 5-LO is elevated in symptomatic compared Appl. Bact. 34, 803-813.
with asymptomatic plaques [172], suggesting that LTB4 has [21] McDonald, M.I.; Graham, I.; Harvey, K.J.; Sinclair, A. (1981)
an important role in atherosclerosis. Lancet 2, 1056.
[22] Kohn, A.; Gitelman, J.; Inbar, M. (1980) Arch. Virol. 66, 301-307.
DHA constitutes about 30 to 50% of total fatty acids in [23] Iwamoto, K.S.; Bennett, L.R.; Norman, A.; Villalobos, A.E.; Hut-
the mammalian brain, where it is predominantly associated son, C.A. (1992) Cancer Lett. 64, 17-22.
with membrane phospholipids [1, 14, 173, 174]. PUFAs [24] Krugloak, M.; Deharo, E.; Shalmiev, G.; Sauvain, M.; Moretti, C.;
Ginsburg, H. (1995) Exp. Parasitol. 81, 97-105.
serve as ligands for RXR receptor. DHA is essential for [25] Doering, T.L.; Raper, J.; Buxbaum, L.U.; Adams, S.P.; Gordon,
brain growth, development, and maturation, and DHA defi- J.I.; Hart, G.W.; Englund, P.T. (1991) Science 252, 1851-1854.
ciency results in impaired spatial learning as well as other [26] Giron, D.J. (1982) Proc. Soc. Exp. Biol. Med. 170, 25-28.
abnormalities [14, 175-177], defects that are also seen in [27] Santoro, M.G.; Benedetto, A.; Carruba, G.; Garaci, E.; Jaffe, B.M.
RXRγ-deficient mice [178]. DHA reduces blood cholesterol (1980) Science 209, 1032-1034.
[28] Das, U.N. (1985) Canadian Med. Assoc. J. 132, 900.
and enhances insulin sensitivity, actions that are also seen [29] Das, U.N. (1985) Canadian Med. Assoc. J. 132, 1350.
with synthetic RXR ligands [179, 180], implying that DHA [30] Das, U.N. (2005) Med. Sci. Monit. 11, RA 206-RA211.
brings about its actions by RXR activation. Several RXR [31] Bromberg, Y.; Pick, E. (1984) Cell Immunol. 88, 213-221.
heterodimerization partners such as PPARs, the liver X re- [32] Sun, C. Q.; O’Connor, C. J.; Robertson, A. (2003) FEMS Immunol.
Med. Microbiol. 36, 9-17.
ceptors, and farnesoid X receptors are essential for regulating [33] Das, U. N. (2006) Am. J. Clin. Nutr. 83, 390-391.
energy and nutritional homeostasis. Since DHA serves as a [34] Giamarellos-Bourboulis, E. J.; Mouktaroudi, M.; Adamis, T.;
ligand for RXR and PPARs, it is likely that it (DHA) modu- Koussoulas, V.; Baziaka, F.; Perrea, D.; Karavannacos, P. E.; Gia-
lates these and several other regulatory events by binding to marellou, H. (2004) Antimicrob. Agents Chemother. 48, 4713-
these nuclear receptor heterodimers. 4717.
[35] Serhan, C.N.; Hong, S.; Gronert, K.; Colgan, S.P.; Devchand, P.R.;
Since several biologically active molecules are formed Mirick, G.; Moussignac, R-L. (2002) J. Exp. Med. 196, 1025-1037.
from PUFAs, it is important to know the molecular triggers [36] Das, U.N. (2005) Med. Sci. Monit. 11, RA155-RA162.
[37] Kumar, K.V.; Das, U. N. (1997) Proc. Soc. Exp. Biol. Med. 214,
that facilitate their formation and understand their actions in 374-379.
various diseases. If methods to selectively enhance the syn- [38] Okuda, Y.; Kawashima, K.; Sawada, T.; Tsurumaru, K.; Asano,
thesis of desirable molecules such as LXs, resolvins, NPD1, M.; Suzuki, S.; Soma, M.; Nakajima, T.; Yamashita, K. (1997)
nitroalkenes are developed, then it may be possible to sup- Biochem. Biophys. Res. Commun. 232, 487-491.
[39] Das, U. N.; Mohan, I. K.; Raju, T. R. (2001) Prostaglandins Leu-
press inappropriate inflammation. Possible use of LXs and kot. Essen. Fatty Acids 65, 197-203.
resolvins and/or their structural analogues in the manage- [40] Das, U. N. (1995) Prostaglandins Leukot. Essen. Fatty Acids 52,
ment of asthma, metabolic syndrome X, neurodegenerative 387-391.
conditions, stroke, CHD, psoriasis, collagen vascular dis- [41] Das, U. N.; Vaddadi, K. S. (2004) Nutrition 20, 942-947.
eases, and cancer need to studied. [42] Bunk, S. (2002) Scientist 16(24), 22-24.
[43] Moskowitz, D. W. (2002) Diabetes Tech. Therapeutics 4, 683-711.
[44] Kaergel, E.; Muller, D. N.; Honeck, H.; Theuer, J.; Shagdarsuren,
REFERENCES E.; Mullally, A.; Luft, F. C.; Schunck, W-H. (2002) Hypertension
[1] Das, U. N. (2006) Biotech. J. 1, 420-439. 40, 273-279.
[45] Gilroy, D. W. (2005) Mem. Inst. Oswaldo Cruz. 100(Suppl. 1), 49-
[2] Das, U. N.; Mohan, I. K.; Raju, T. R. (2001) Prostaglandins Leu-
kot. Essen. Fatty Acids 65, 197-203. 54.
[46] Wang, W.; Diamond, S. L. (1997) Biochem. Biophys. Res. Com-
[3] Das, U. N. (2004) Eur. J. Clin. Nutr. 58, 195-203.
[4] Das, U. N. (2003) Eur. J. Clin. Nutr. 57, 218-226. mun. 233, 748-751.
[47] Das, U. N. (2005) Med. Sci. Monit. 11, R233-R237.
Essential Fatty Acids Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 15

[48] Kumar, G. S.; Das, U. N. (1994) Prostaglandins Leukot. Essen. L. M. S.; Branchaud, B. P.; Chen, Y.; Freeman, B. A. (2005) J.
Fatty Acids 50, 331-334. Biol. Chem. 280, 42464-42475.
[49] Arita, M.; Bianchini, F.; Aliberti, J.; Sher, A.; Chiang, N.; Hong, [83] Coles, B.; Bloodsworth, A.; Clark, S.R.; Lewis, M. J.; Cross, A. R.;
S.; Yang, R.; Petasis, N. A.; Serhan, C. N. (2005) J. Exp. Med. 201, Freeman, B. A.; O’Donnell, V. B. (2002) Circ. Res. 91, 375-381.
713-722. [84] Lima, E. S.; Bonim, M. G.; Augusto, O.; Barbeiro, H. V.; Souza,
[50] Dooper, M. M.; van Riel, B.; Graus, Y. M.; M’Rabet, L. (2003) H. P.; Abdalla, D. S. P. (2005) Free Rad. Biol. Med. 39, 532-539.
Immunology 110, 348-57. [85] Wright, M.M.; Schopfer, F.J.; Baker, P.R.S.; Vidyasagar, V.; Pow-
[51] Chen, W.; Esselman, W. J.; Jump, D. B.; Busik, J. V. (2005) Invest. ell, P.; Chumley, P.; Iles, K.E.; Freeman, B.A.; Agarwal, A. (2006)
Ophthalmol. Vis. Sci. 46, 4342-4347. Proc. Natl. Acad. Sci. USA 103, 4299-4304.
[52] Li, H.; Ruan, X. Z.; Powis, S. H.; Fernando, R.; Mon, W. Y.; [86] Miller, C. C.; McCreedy, C. A.; Jones, A. D.; Ziboh, V. A. (1988)
Wheeler, D. C.; Moohead, J. F.; Varghese, Z. (2005) Kidney Int. Prostaglandins 35, 917-938.
67, 867-874. [87] Tate, G.; Mandell, B. F.; Laposata, M.; Ohliger, D.; Baker, D. G.;
[53] Beck, S. A.; Smith, K. L.; Tisdale, M. J. (1991) Cancer Res. 51, Schumacher, H. R.; Zurier, R. B. (1989) J. Rheumatol. 16, 729-
6089-6093. 734.
[54] Lihn, A. S.; Richelsen, B.; Pedersen, S. B.; Hangaard, S. B.; [88] Heitmann, J.; Iversen, L.; Kragballe, K.; Ziboh, V. A. (1995) Exp.
Rathje, G. S.; Madsbad, S.; Andersen, O. (2003) Am. J. Physiol. Dermatol. 4, 74-78.
Endocrinol. Metab. 285, E1072-E1080. [89] Santoli, D.; Zurier, R. B. (1989) J. Immunol. 143, 1303-1309.
[55] Miller, J.; Carr, A.; Emery, S.; Law, M.; Mallal, S.; Baker, D.; [90] DeLuca, P.; Rossetti, R. G.; Alavian, C.; Karim, P.; Zurier, R. B.
Smith, D.; Kaldor, J.; Cooper, D. A. (2003) HIV Med. 4, 293-301. (1999) J. Invest. Med. 47, 246-250.
[56] Sheng, Z.; Otani, H.; Brown, M. S.; Goldstein, J. L. (1995) Proc. [91] Mancuso, P.; Whelan, J.; DeMichele, S. J.; Snider, C. C.; Guszcza,
Natl. Acad. Sci. USA 92, 935–938. J. A.; Karlstad, M. D. (1997) Crit. Care Med. 25, 1198-1206.
[57] Fajas, L.; Schoonjans, K.; Gelman, L.; Kim, J. B.; Najib, J.; Martin, [92] Palombo, J. D.; DeMichele, S. J.; Boyce, P. J.; Lydon, E. E.; Liu, J.
G.; Fruchart, J. C.; Briggs, M.; Spiegelman, B. M.; Auwerx, J. W.; Huang, Y. S.; Forse, R. A.; Mizgerd, J. P.; Bistrian, B. R.
(1999) Mol. Cell Biol. 19, 5495-5503. (1999) Crit. Care Med. 27, 1908-1915.
[58] El-Sohemy, A.; Archer, M. C. (1999) Lipids 34, 1037-1043. [93] Barham, J. B.; Edens, M. B.; Fonteh, A. N.; Johnson, M. M.;
[59] Das, U. N. (2000) Nutrition 16, 286-290. Easter, L.; Chilton, F. H. (2000) J. Nutr. 130, 1925-1931.
[60] Nakamura, N.; Hamazaki, T.; Jokaji, H.; Minami, S.; Kobayashi, [94] Thies, F.; Miles, E. A.; Nebe-von-Caron, G.; Powell, J. R.; Hurst,
M. (1998) Int. J. Clin. Lab. Res. 28, 192-195. T. L.; Newsholme, E. A.; Calder, P. C. (2001) Lipids 36, 1183-
[61] Hannah, V. C.; Ou, J.; Luong, A.; Goldstein, J. L.; Brown, M. S. 1193.
(2001) J. Biol. Chem. 276, 4365-4372. [95] Kambe, T.; Murakami, M.; Kudo, I. (1999) FEBS Lett. 453, 81-84.
[62] Field, F. J.; Born, E.; Murthy, S.; Mathur, S. N. (2002) Biochem. J. [96] Gilroy, D. W.; Newson, J.; Sawmynaden, P.; Willoughby, D. A.;
368(Pt 3), 855-864. Croxtall, J. D. (2004) FASEB J. 18, 489-498.
[63] Xu, J.; Cho, H.; O’Malley, S.; Park, J. H.; Clarke, S. D. (2002) J. [97] Lo, C. J.; Chiu, K. C.; Fu, M.; Lo, R.; Helton, S. (1999) J. Surg. 82,
Nutr. 132, 3333-3339. 216-221.
[64] Field, F. J.; Born, E.; Mathur, S. N. (2003) J. Lipid Res. 44, 1199- [98] Borghaei, H.; Borghaei, R. C.; Ni, X.; Pease, E.; Thornton, R.;
1208. Mochan, E. (1997) Inflamm. Res. 46(Suppl. 2), S177-178.
[65] Le Jossic-Corcos, C.; Gonthier, C.; Zaghini, I.; Logette, E.; Shech- [99] Camandola, S.; Leonarduzzi, G.; Musso, T.; Varesio, L.; Carini, R.;
ter, I.; Bournot, P. (2005) Biochem. J. 385(Pt 3), 787-794. Scavazza, A.; Chiarpotto, E.; Baeuerle, P. A.; Poli, G. (1996) Bio-
[66] Worgall, T. S.; Johnson, R. A.; Seo, T.; Gierens, H.; Deckelbaum, chem. Biophys. Res. Commun. 229, 643-647.
R. J. (2002) J. Biol. Chem. 277, 3878-3885. [100] Li, H.; Ruan, X. Z.; Powis, S. H.; Fernando, R.; Mon, W. Y.;
[67] Calon, F.; Lim, G. P.; Yang, F.; Morihara, T.; Teter, B.; Ubeda, O.; Wheeler, D. C.; Moorhead, J. F.; Varghese, Z. (2005) Kidney Int.
Rostaing, P.; Triller, A.; Salem, Jr. N.; Ashe, K. H.; Frautschy, S. 67, 867-874.
A.; Cole, G. M. (2004) Neuron 43, 633-645. [101] Kliewer, S. A.; Sundseth, S. S.; Jones, S. A.; Brown, P. J.; Wisely,
[68] Lukiw, W. J.; Cui, J-G.; Marcheselli, V. L.; Bodker, M.; Botkjaer, G. B.; Koble, C. S.; Devchand, P.; Wahli, W.; Willson, T. M.; Len-
A.; Gotlinger, K.; Serhan, C. N.; Bazan, N. G. J. (2005) Clin. In- hard, J. M.; Lehmann, J. M. (1997) Proc. Natl. Acad. Sci. USA 94,
vest. 115, 2774-2783. 4318-4323.
[69] Calon, F.; Lim, G. P.; Morihara, T.; Yang, F.; Ubeda, O.; Salem, N. [102] Sethi, S.; Eastman, A. Y.; Eaton, J. W. (1996) J. Lab. Clin. Med.
Jr.; Frautschy, S. A.; Cole, G. M. (2005) Eur. J. Neurosci. 22, 617- 128, 27-38.
626. [103] Mishra, A.; Chaudhary, A.; Sethi, S. (2004) Arterioscler. Thromb.
[70] Akbar, M.; Calderon, F.; Wen, Z.; Kim, H. Y. (2005) Proc. Natl. Vasc. Biol. 24, 1621-1627.
Acad. Sci. USA 102, 10858-10863. [104] Mori, T. A.; Woodman, R. J.; Burke, V.; Puddey, I. B.; Croft, K.
[71] Hashimoto, M.; Tanabe, Y.; Fujii, Y.; Kikuta, T.; Shibata, H.; D.; Beilin, L. J. (2003) Free Radic. Biol. Med. 35, 772-781.
Shido, O. (2005) J. Nutr. 135, 549-555. [105] Mori, T. A.; Puddey, I. B.; Burke, V.; Croft, K. D.; Dunstan, D.
[72] Aravindakshan, M.; Ghate, M.; Ranjekar, P. K.; Evans, D. R.; W.; Rivera, J. H.; Beilin, L. (2000) J. Redox. Rep. 5, 45-46.
Mahadik, S. P. (2003) Schizophr. Res. 62, 195-204. [106] Barden, A. E.; Mori, T. A.; Dunstan, J. A.; Taylor, A. L.; Thornton,
[73] Das, U. N. (2004) Med. Sci. Monit. 10, HY33-HY37. C. A.; Croft, K. D.; Beilin, L. J.; Prescott, S. L. (2004) Free Radic.
[74] Claria, J.; Serhan, C. N. (1995) Proc. Natl. Acad. Sci. USA 92, Res. 38, 233-239.
9475-9479. [107] Das, U.N. (2005) Nutrition 21, 762-773.
[75] Chiang, N.; Gronert, K.; Clish, C. B.; O’Brien, J. A.; Freeman, M. [108] Levy, B.D.; Clish, C.B.; Schmidt, B.; Gronert, K.; Serhan, C.N.
W.; Serhan, C. N. (1999) J. Clin. Invest. 104, 309-316. (2001) Nature Immunol. 2, 612-
[76] Serhan, C. N.; Clish, C. B.; Brannon, J.; Colgan, S. P.; Chiang, N.; [109] Devchand, P.R.; Arita, M.; Hong, S.; Bannenberg, G.L.; Mous-
Gronert, K. (2000) J. Exp. Med. 192, 1197-1204. signac, R-L.; Gronert, K.; Serhan, C.N. (2003) FASEB J. 17, 652-
[77] Serhan, C. N.; Hong, S.; Gronert, K.; Colgan, S. P.; Devchand, P. [110] Bannenberg, G.L.; Chiang, N.; Ariel, A.; Arita, M.; Tjonahen, E.;
R.; Mirick, G.; Moussignac, R-L. (2002) J. Exp. Med. 196, 1025- Gotlinger, K.H.; Hong, S.; Serhan, C.N. (2005) J. Immunol. 174,
1037. 4345-4355.
[78] Marcheselli, V. L.; Hong, S.; Lukiw, W. J.; Tian, X. H.; Gronet, [111] LaMarre, J.; Wollenberg, G.K.; Gonias, S.L.; Hayes, M.A. (1991)
K.; Musto, A.; Hardy, M.; Gimenez, J. M.; Chiang, N.; Serhan, C. Lab. Invest. 65, 3-
N, Bazan, N. G. (2003) J. Biol. Chem. 278, 43807-43817. [112] Philippidis, P.; Mason, J.C.; Evans, B.J.; Nadra, I.; Taylor, K.M.;
[79] Hong, S.; Gronert, K.; Devchand, P. R.; Moussignac, R. L.; Serhan, Haskard, D.O.; Landis, R.C. (2004) Circ. Res. 94, 119-
C. N. (2003) J. Biol. Chem. 278, 14677-14687. [113] Gronert, K.; Gewirtz, A.; Madara, J.L.; Serhan, C.N. (1998) J. Exp.
[80] Mukherjee, P. K.; Marcheselli, V. L.; Serhan, C. N.; Bazan, N. G. Med. 187, 1285-1294.
(2004) Proc. Natl. Acad. Sci. USA 101, 8491-8496. [114] Ariel, A.; Chiang, N.; Arita, M.; Petasis, N.A.; Serhan, C.N. (2003)
[81] Gronert, K.; Maheshwari, N.; Khan, N.; Hasan, I. R.; Dunn, M.; J. Immunol. 170, 6266-6272.
Laniado Schwartzman, M. (2005) Biol. Chem. 280, 15267-15278. [115] Aliberti, J.; Hieny, S.; Reis, E.; Sousa, C.; Serhan, N.C.; Sher, A.
[82] Baker, P. R. S.; Lin, Y.; Schopfer, F. J.; Woodcock, S. R.; Groeger, (2002) Nature Immunol. 3, 76-82.
A. L.; Batthyany, C.; Swooney, S.; Long, M. H.; Iles, K. E.; Baker,
16 Current Pharmaceutical Biotechnology, 2006, Vol. 7, No. 6 Undurti N. Das

[116] Mullenix, P. S.; Andersen, C. A.; Starnes, B. W. (2005) Ann. Vasc. Polyunsaturated fatty acids of marine origin induce adiponectin in
Surg. 19, 130-138. mice fed a high-fat diet. Diabetologia 2006 Jan 6, 1-4.
[117] Das, U. N. (2005) Prostaglandins Leukot. Essen. Fatty Acids 72, [152] Huang, Y-J.; Fang, V. S.; Chou, Y-C.; Kwok, C-F.; Ho, L-T.
173-179. (1997) Metabolism 46, 1252-1258.
[118] Kobayashi, T.; Tahara, Y.; Matsumoto, M Iguchi, M.; Sano, H.; [153] Mori, Y.; Murakawa, Y.; Katoh, S.; Hata, S.; Yokoyama, J.; Ta-
Murayama, T.; Arai, H.; Oida, H.; Kobayashi, T. Y.; Yamashita, J. jima, N.; Ikeda, Y.; Nobukata, H.; Ishikawa, T.; Shibutani, Y.
K.; Katagiri, H.; Majima, M.; Yokode, M.; Kita, T.; Narumiya, S. (1997) Metabolism 46, 1458-1464.
(2004) J. Clin. Invest. 114, 784-794. [154] Nobukata, H.; Ishikawa, T.; Obata, M.; Shibutani, Y. (2000) Me-
[119] Honda, T.; Segi-Nishida, E.; Miyachi, Y.; Narumiya, S. (2006) J. tabolism 49, 912-919.
Exp. Med. 203, 325-335. [155] Borkman, M.; Storlien, L. H.; Pan, D. A Jenkins, A. B.; Chisholm,
[120] Juan, H.; Sametz, W. (1985) Prostaglandins Leukot. Med. 19, 79- D. J.; Campbell, L. V. (1993) N. Engl. J. Med. 328, 238-244.
86. [156] Das, U. N. (1995) Prostaglandins Leukot. Essen. Fatty Acids 52,
[121] Bordet, J. C.; Guichardant, M.; Lagarde, M. (1988) Biochim. Bi- 387-391.
ophys. Acta 958, 460-468. [157] Das, U. N.; Kumar, K. V.; Ramesh, G. (1994) Prostaglandins
[122] Bordet, J. C.; Guichardant, M.; Lagarde, M. (1986) Biochem. Bi- Leukot. Essen. Fatty Acids 50, 253-255.
ophys. Res. Commun. 135, 403-410. [158] Aravindakshan, M.; Ghate, M.; Ranjekar, P. K.; Evans, D. R.;
[123] Hamazaki, T.; Hirai, A.; Terano, T.; Sajiki, J.; Kondo, S.; Fujita, Mahadik, S. P. (2003) Schizophr. Res. 62, 195-204.
T.; Tamura, Y.; Kumagai, A. (1982) Prostaglandins 23, 557-567. [159] Das, U. N. (2004) Med. Sci. Monit. 10, HY33-HY37.
[124] Das, U. N. (2000) Prostaglandins Leukot. Essen. Fatty Acids 63, [160] Echtay, K. S.; Roussel, D.; St-Pierre, J.; Jekabsons, M. B.; Cade-
351-362. nas, S.; Stuart, J. A.; Harper, J. A.; Roebuck, S. J.; Morrison, A.;
[125] Mukherjee, S.K.; Klaidman, L.K.; Yasharel, R.; Adams, J.D. Jr. Pickering, S.; Clapham, J. C.; Brand, M. D. (2002) Nature 415, 96-
(1997) Eur. J. Pharmacol. 330, 27-34. 99.
[126] Chong, Z.Z.; Lin, S.H.; Maise, K. (2004) J. Cereb. Blood Flow [161] Nishikawa, T.; Edelstein, D.; Du, X. L.; Tamagishi, S.; Matsumura,
Metab. 24, 728-743. T.; Kaneda, Y.; Yorek, M. A.; Beebe, D.; Oates, P. J.; Hammes, H.
[127] Ieraci, A.; Herrera, D.G. (2006) PloS Med. 3, e101. P.; Giardino, I.; Brownlee, M. (2000) Nature 404, 787-790.
[128] Horrobin, D.F. (1987) Alcohol Clin. Exp. Res. 11, 2-9. [162] Santerre, R. F.; Nicolosi, R. J.; Smith, S. C. (1974) Exp. Mol.
[129] Valles, S.L.; Blanco, A.M.; Pascual, M.; Guerri, C. (2004) Brain Pathol. 20, 397-406.
Pathol. 14, 365-371. [163] Droge, W. (2002) Physiol. Rev. 82, 47-95.
[130] Das, U.N. (2002) Lancet 360, 490. [164] Bernal-Mizrachi, C.; Gates, A. C.; Weng, S.; Imamura, T.; Knut-
[131] Garcia, M.C.; Kim, H.Y. (1997) Brain Res. 768, 43-48. sen, R. H.; DeSantis, P.; Coleman, T.; Townsend, R. R.; Muglia, L.
[132] Mata de Urquiza, A.; Liu, S.; Sjoberg, M.; Zetterstrom, R.H.; J.; Semenkovich, C. F. (2005) Nature 435, 502-506.
Griffiths, W.; Sjovall, J.; Perlmann, T. (2000) Science 290, 2140- [165] Klein, P. D.; Johnson, R. M. (1954) J. Biol. Chem. 211, 103-110.
2144. [166] Hayashida, T.; Portman, O. W. (1960) Proc. Soc. Exp. Biol. Med.
[133] Gamoh, S.; Hashimoto, M.; Hossain, S.; Masumura, S. (2001) Clin. 103, 656-659.
Exp. Pharmacol. Physiol. 28, 266-270. [167] Cha, S. H.; Fukushima, A.; Sakuma, K.; Kagawa, Y. (2001) J.
[134] Saareks, V.; Mucha, I.; Sievi, E.; Riutta, A. (1999) Pharmacol. Nutr. 131, 2636-2642.
Toxicol. 84, 274-280. [168] Cornwell, D. G.; Panganamala, R. V. (1981) Prog. Lipid Res. 20,
[135] Engelbrecht, A.M.; Engelbrecht, P.; Genade, S.; Niesler, C.; Page, 365-376.
C.; Smuts, M.; Lochner, A. (2005) J. Mol. Cell Cardiol. 39, 940- [169] Smith, E. B. (1962) Biochem. J. 84, 49.
954. [170] Smith, E. B. (1962) Lancet 2, 530-534.
[136] Klein, P.D.; Johnson, R.M. (1954) J. Biol. Chem. 211, 103-110. [171] Subbarao, K.; Jala, V. R.; Mathias, S.; Suttles, J.; Zacharias, W.;
[137] Hayashida, T.; Portman, O.W. (1960) Proc. Soc. Exp. Biol. Med. Ahamed, J.; Ali, H.; Tseng, M. T.; Haribabu, B. (2004) Arterio-
103, 656-659. scler. Thromb. Vasc. Biol. 24, 369-3675.
[138] Bernal-Mizrachi, C.; Gates, A.C.; Weng, S.; Imamura, T.; Knutsen, [172] Cipollone, F.; Mezzetti, A.; Fazia, M. L.; Cuccurullo, C.; Iezzi, A.;
R.H.; DeSantis, P.; Coleman, T.; Townsend, R.R.; Muglia, L.J.; Ucchino, S.; Spigonardo, F.; Bucci, M.; Cuccurullo, F.; Prescott, S.
Semenkovich, C.F. (2005) Nature 435, 502-506. M.; Stafforini, D. M. (2005) Arterioscler. Thromb. Vasc. Biol. 25,
[139] Suresh, Y.; Das, U.N. (2003) Nutrition 19, 93-114. 1665-1670.
[140] Suresh, Y.; Das, U.N. (2003) Nutrition 19, 213-228. [173] Connor, W. E.; Neuringer, M.; Reisbick, S. (1992) Nutr. Rev. 50(Pt
[141] Piomelli, D.; Pilon, C.; Giros, B.; Sokoloff, P.; Martres, M.P.; 2), 21-29.
Schwartz, J.C. (1991) Nature 353, 164-167. [174] Connor, W. E.; Neuringer, M.; Reisbick, S. (1991) World Rev.
[142] Zhang, L.; Reith, M.E. (1996) Eur. J. Pharmacol. 315, 345-354. Nutr. Diet. 66, 118-132.
[143] Blanchet, F.; Gauchy, C.; Perez, S.; Glowinski, J.; Kemel, M.L. [175] Lim, S. Y.; Hoshiba, J.; Moriguchi, T.; Salem, N. Jr. (2005) Pedi-
(1999) Synapse 31, 140-150. atr. Res. 58, 741-748.
[144] de La Presa Owens, S.; Innis, S.M. (1999) J. Nutr. 129, 2088-2093. [176] Lim, S. Y.; Hoshiba, J.; Salem, N. Jr. (2005) J. Neurochem. 95,
[145] de La Presa Owens, S.; Innis, S.M. (2000) Pediatr. Res. 48, 125- 848-857.
130. [177] Garcia-Calatayud, S.; Redondo, C.; Martin, E.; Ruiz, J. I.; Garcia-
[146] Innis, S.M.; de La Presa Owens, S. (2001) J. Nutr. 131, 118-122. Fuentes, M.; Sanjurjo, P. (2005) Pediatr. Res. 57(5 Pt 1), 719-723.
[147] Kuperstein, F.; Yakubov, E.; Dinerman, P. (2005) J. Neurochem. [178] Chiang, M. Y.; Misner, D.; Kempermann, G.; Schikorski, T.;
95, 1550-1562. Giguere, V.; Sucov, H. M.; Gage, F. H.; Stevens, C. F.; Evans, R.
[148] Das, U.N. (2002) Exp. Biol. Med. 227, 989-997. M. (1998) Neuron 21, 1353-1361.
[149] Peyron-Caso, E.; Taverna, M.; Guerre-Millo, M.; Veronese, A.; [179] Argmann, C. A.; Sawyez, C. G.; McNeil, C. J.; Hegele, R. A.;
Pacher, N.; Slama, G.; Rizkalla, S. W. (2002) J. Nutr. 132, 2235- Huff, M. W. (2003) Arterioscler. Thromb. Vasc. Biol. 23, 475-482.
2240. [180] Kim, H. I.; Cha, J. Y.; Kim, S. Y.; Kim, J. W.; Roh, K. J.; Seong, J.
[150] Reseland, J. E.; Haugen, F.; Hollung, K.; Solvoll, K.; Halvorsen, K.; Lee, N. T.; Choi, K. Y.; Kim, K. S.; Ahn, Y. H. (2002) Diabe-
B.; Brude, I. R.; Nenseter, M. S.; Christiansen, E. N.; Drevon, C. tes 51, 676-685.
A. (2001) J. Lipid Res. 42, 743-750.
[151] Flachs, P.; Mohamed-Ali, V.; Horakova, O.; Rossmeisl, M.;
Hosseinzadeh-Attar, M. J.; Hensler, M.; Ruzickova, J.; Kopecky, J.

View publication stats

You might also like