Vous êtes sur la page 1sur 14

REVIEW ARTICLE

published: 09 September 2013


doi: 10.3389/fncel.2013.00144

Fatting the brain: a brief of recent research


Ghulam Hussain1,2 , Florent Schmitt1,2 , Jean-Philippe Loefer1,2 * and Jose-Luis Gonzalez de Aguilar1,2 *
1
UMR_S 1118, Universit de Strasbourg, Strasbourg, France
2
Mcanismes Centraux et Priphriques de la Neurodgnrescence, U1118, Institut National de la Sant et de la Recherche Mdicale, Facult de Mdecine,
Universit de Strasbourg, Strasbourg, France

Edited by: Fatty acids are of paramount importance to all cells, since they provide energy, function as
Johannes Le Coutre, Nestle Research signaling molecules, and sustain structural integrity of cellular membranes. In the nervous
Center, Switzerland
system, where fatty acids are found in huge amounts, they participate in its development
Reviewed by:
Jean-Pierre Montmayeur, The
and maintenance throughout life. Growing evidence strongly indicates that fatty acids in
Coca-Cola Company, USA their own right are also implicated in pathological conditions, including neurodegenerative
Takumi Takizawa, Gunma University, diseases, mental disorders, stroke, and trauma. In this review, we focus on recent studies
Japan that demonstrate the relationships between fatty acids and function and dysfunction of
*Correspondence: the nervous system. Fatty acids stimulate gene expression and neuronal activity, boost
Jean-Philippe Loefer and Jose-Luis
Gonzalez de Aguilar, Mcanismes
synaptogenesis and neurogenesis, and prevent neuroinammation and apoptosis. By
Centraux et Priphriques de la doing so, they promote brain development, ameliorate cognitive functions, serve as
Neurodgnrescence, U1118, anti-depressants and anti-convulsants, bestow protection against traumatic insults, and
Institut National de la Sant et de la enhance repairing processes. On the other hand, unbalance between different fatty acid
Recherche Mdicale, Facult de
Mdecine, Universit de Strasbourg,
families or excess of some of them generate deleterious side effects, which limit the
Btiment 3, Etage 8, 11 rue Humann, translatability of successful results in experimental settings into effective therapeutic
F-67085 Strasbourg, France strategies for humans. Despite these constraints, there exists realistic evidence to consider
e-mail: loefer@unistra.fr;
that nutritional therapies based on fatty acids can be of benet to several currently incurable
gonzalez@unistra.fr
nervous system diseases.
Keywords: monounsaturated fatty acid, neurodegenerative disease, neurological disease, peripheral nerve,
saturated fatty acid, polyunsaturated fatty acid

INTRODUCTION SOME ASPECTS OF THE BIOCHEMISTRY OF FATTY ACIDS


Fatty acids represent a class of lipids that are crucial components of According to the IUPAC denition, fatty acids arealiphatic mono-
all mammalian cells. They display a variety of biological functions carboxylic acids derived from or contained in esteried form in
to maintain vital cellular processes at various levels. Thus, fatty an animal or vegetable fat, oil or wax (IUPAC, 1997). Naturally
acids provide energy, function as signaling molecules, and sustain occurring fatty acids mostly consist of an unbranched 428 car-
structural integrity of cellular membranes. They are of particular bon chain that is usually composed of an even number of carbon
importance for the nervous system for two major reasons. First, atoms. On the basis of the carbon chain length, fatty acids are
the nervous system possesses a very high concentration of fatty classied into short- (less than six carbon atoms), medium- (612
acids, second only to adipose tissue (Etschmaier et al., 2011). Sec- carbon atoms), long- (1422 carbon atoms), and very long chain
ond, these fatty acids participate actively both in the development fatty acids (more than 22 carbon atoms). Fatty acids in which the
of the nervous system during embryonic and early postnatal life, aliphatic chain is fully composed of single bonds between carbon
and in its maintenance throughout adulthood and natural aging atoms are named as saturated fatty acids (SFAs), whereas fatty
(Uauy and Dangour, 2006; Rombaldi Bernardi et al., 2012). Along acids with one or more than one carboncarbon double bond are
with these actions, currently incurable pathological conditions of called unsaturated fatty acids. Based on the number of double
the nervous system, including neurodegenerative diseases, mental bonds, unsaturated fatty acids are further divided into mono-
disorders, stroke, and trauma, involve deregulated contents of fatty unsaturated fatty acids (MUFAs) and polyunsaturated fatty acids
acids. It is therefore believed that these changes contribute in their (PUFAs; Table 1). Long chain SFAs have relatively high melt-
own right by as yet incompletely understood mechanisms to those ing points that make them to appear solid at room temperature.
pathological processes. In consequence, the roles of fatty acids in Therefore, the body possesses a mechanism to introduce double
health and disease of the nervous system have been intensively bonds in the carbon chain, which lowers the melting point and
investigated in the last few decades. In this piece of work, we focus permits functioning in a physiological environment. There are
mainly on studies published during the last ve years to show the four fatty acid desaturases documented in humans that selectively
diversity in the fatty acids implicated in function and dysfunction catalyze the introduction of a double bond in different positions
of the nervous system. The detailed mechanisms of action of fatty of the carbon chain. -9 desaturase, also known as stearoyl-CoA
acids at the molecular level are not treated in this article, since they desaturase (SCD), is charged with synthesizing MUFAs, mainly
are the subject of other recently published reviews (Georgiadi and palmitoleic acid (16:1) and oleic acid (18:1), by introducing a dou-
Kersten, 2012; Yamashima, 2012). ble bond between carbon atoms nine and 10 from the carboxylic

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 1

fncel-07-00144 2013/9/6 13:17 page 1 #1


Hussain et al. Fatty acids and nervous system

Table 1 | Most typical fatty acids.

Systematic name Common name Abbreviation C:D n-X1

Saturated fatty acids (SFAs)


Butanoic Butyric 4:0
Hexanoic Caproic 6:0
Octanoic Caprylic 8:0
Decanoic Capric 10:0
Dodecanoic Lauric 12:0
Tetradecanoic Myristic 14:0
Hexadecanoic Palmitic 16:0
Octadecanoic Stearic 18:0
Eicosanoic Arachidic 20:0
Docosanoic Behenic 22:0
Tetracosanoic Lignoceric 24:0
Mono-unsaturated fatty acids (MUFAs)
cis-9-Hexadecenoic Palmitoleic 16:1
cis-9-Octadecenoic Oleic 18:1
cis-13-Docosenoic Erucic 22:1
cis-15-Tetracosenoic Nervonic 24:1
Polyunsaturated fatty acids (PUFAs)
cis-9,cis-12-cis-15-Octadecatrienoic -Linolenic ALA 18:3 n-3
cis-6,cis-9,cis-12,cis-15-Octadecatetraenoic Stearidonic SDA 18:4 n-3
cis-8,cis-11,cis-14,cis-17-Eicosatetraenoic Eicosatetraenoic ETE 20:4 n-3
cis-5, cis-8,cis-11,cis-14,cis-17-Eicosapentaenoic Eicosapentaenoic EPA 20:5 n-3
cis-7,cis-10,cis-13,cis-16, cis-19-Docosapentaenoic Docosapentaenoic DPA 22:5 n-3
cis-4,cis-7,cis-10,cis-13,cis-16,cis-19-Docosahexaenoic Docosahexaenoic DHA 22:6 n-3
cis-9,cis-12-Octadecadienoic Linoleic LA 18:2 n-6
cis-6,cis-9,cis-12-Octadecatrienoic -Linolenic GLA 18:3 n-6
cis-8,cis-11,cis-14-Eicosatrienoic Dihomo--linolenic DHGLA 20:3 n-6
cis-5,cis-8,cis-11,cis-14-Eicosatetraenoic Arachidonic AA 20:4 n-6
cis-7,cis-10,cis-13,cis-16-Docosatetrtaenoic Docosatetraenoic 22:4 n-6
cis-4,cis-7,cis-10,cis-13,cis-16-Docosapentaenoic Docosapentaenoic 22:5 n-6

1This nomenclature designates the number of carbon atoms in the fatty acid (C), the number of double bonds (D), and the position of the rst double bond counting
from the terminal methyl carbon (n-X).

acid end (Figure 1; Enoch et al., 1976). -4, -5, and -6 desat- 22:6 n-3; Figure 1). Both LA (18:2 n-6) and ALA (18:3 n-3) cannot
urases introduce a double bond at carbon positions 4, 5, and 6, be synthesized indigenously by the human body, so that they must
respectively, and work cooperatively with elongases, which are be supplied with food, and such fatty acids are termed as essential
responsible for the extension of the aliphatic chain. The combined fatty acids (Ruzickova et al., 2004; Lands, 2012). In spite of the fact
actions of desaturases and elongases are implicated in the synthesis that the body is able to metabolize these essential fatty acids, the
of PUFAs (Nakamura and Nara, 2004). efciency of conversion is low. Hence, the availability not only of
According to the position of the rst double bond from the essential precursors but also of some of their metabolites, such as
methyl end of the fatty acid chain, the most important PUFAs for EPA (20:5 n-3) and DHA (22:6 n-3), greatly depends on dietary
humans can be divided into two families: n-6 and n-3 PUFAs. support (Brenna et al., 2009). Alternatively, PUFAs can also be
Linoleic acid (LA, 18:2 n-6) is the parent fatty acid of n-6 made available by enzymatic processing of membrane phospho-
PUFAs, which produces principally arachidonic acid (AA, 20:4 n- lipids by phospholipases (Lee et al., 2011). Whatever pathway is
6), whereas -linolenic acid (ALA, 18:3 n-3) is the parent fatty acid involved, several PUFAs can be metabolized by cyclo-oxygenases,
of n-3 PUFAs, which gives rise mainly to eicosapentaenoic acid lipo-oxygenases, or cytochrome P450 mono-oxygenases to pro-
(EPA, 20:5 n-3) and subsequently docosahexaenoic acid (DHA, duce other compounds with important biological functions. AA

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 2

fncel-07-00144 2013/9/6 13:17 page 2 #2


Hussain et al. Fatty acids and nervous system

FIGURE 1 | Biosynthesis of fatty acids. Medium- to long chain SFAs to AA (20:4 n-6) in the n-6 series. Afterward, these end products are
are successively transformed by the action of elongases (E) into palmitic further elongated, desaturated, and submitted to peroxisomal -
acid (16:0), which is then either elongated to stearic acid (18:0), and other oxidation (all three steps indicated by OX) to yield DHA (22:6 n-3) and
long chain SFAs, or desaturated, together with stearic acid (18:0), by 9 docosapentaenoic acid (22:5 n-6), respectively. Finally, AA (20:4 n-6)
desaturase to produce de novo MUFAs of the n-7 and n-9 series, such as is the precursor of potent pro-inammatory eicosanoids. EPA (20:5 n-3)
palmitoleic acid (16:1) and oleic acid (18:1). In the case of PUFAs, 6 and produces less potent (dashed arrow) eicosanoids and, together with DHA
5 desaturases work cooperatively with elongases to introduce double (22:6 n-3), gives rise to docosanoids with anti-inammatory properties (i.e.,
bonds and extend the aliphatic chain in a successive manner, from resolvins and protectins). GLA, -linolenic acid; DHGLA, dihomo--linolenic
ALA (18:3 n-3) to EPA (20:5 n-3) in the n-3 series, and from LA (18:2 n-6) acid.

(20:4 n-6) and, to a lesser extent, EPA (20:5 n-3) are trans- specic enolase, glial brillary acidic protein, and myelin basic pro-
formed into potent pro-inammatory eicosanoids. Additionally, tein was observed in pups from mice fed on n-3 PUFA enriched
EPA (20:5 n-3) and DHA (22:6 n-3) generate opposing anti- diet, administered from two months prior to mouse conception
inammatory docosanoids, including resolvins and protectins to end of lactation period (Tian et al., 2011). Similarly, postna-
such as neuroprotectin-D1 (NPD1; Bazan, 2009; Figure 1). tal supplementation of ALA (18:3 n-3), the parent precursor of
n-3 PUFAs, enhanced cell proliferation and early neuronal differ-
EVIDENCE OF THE IMPORTANCE OF FATTY ACIDS FOR entiation, while its deprivation resulted in increased proportion
HEALTH AND DISEASE OF THE NERVOUS SYSTEM of apoptosis in the dentate gyrus of unweaned pups. This ame-
FATTY ACIDS AND BRAIN DEVELOPMENT liorating effect was offset by maternal ALA (18:3 n-3) deciency
Mothers own resources, via placenta and milk, provide most of during gestation period, suggesting that ALA (18:3 n-3) is not
the n-3 PUFAs necessary for brain development during fetal and only required at postnatal stages but is also essential for fetal brain
early postnatal life. Due to this high demand of the developing ner- development (Niculescu et al., 2011). Importantly, such diets given
vous system in the progeny, maternal brain levels of DHA (22:6 at perinatal stages may have long lasting consequences in the adult-
n-3) exhaust during pregnancy and lactation period (Chen and hood. Thus, the abundance of n-3 PUFAs in the diet of pregnant
Su, 2012). Thus, enhanced provision or adequate supply of n- females revealed essential for the development of the glutamatergic
3 PUFAs at these stages can yield positive effects on offspring system and normal behavior performance in the adult offspring
brain development. For instance, increased expression of neuron (Moreira et al., 2010a). Also, motor coordination was ameliorated

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 3

fncel-07-00144 2013/9/6 13:17 page 3 #3


Hussain et al. Fatty acids and nervous system

in adulthood when rats were fed on DHA (22:6 n-3) and EPA either SFAs or MUFAs, MUFAs promoted insulin sensitivity and
(20:5 n-3) supplementation starting from gestation stage to post- cortical activity while SFAs did not (Sartorius et al., 2012). Lastly,
natal age of 90 days (Coluccia et al., 2009). Finally, n-3 PUFA it is noteworthy that the intake of sufcient quantity of MUFAs
enriched diets also improved reference and working memory in prevented the age related deletion of mitochondrial DNA in the
offspring rats when supplied to mother at gestation stage (Chung brain of aged animals (Ochoa et al., 2011).
et al., 2008).
Frequently, the impact of dietary fatty acids depends on a bal- FATTY ACIDS AND NEURODEGENERATIVE DISORDERS
ance between different types. In a study to assess the effects of The altered amounts of different classes of fatty acids in the ner-
quality and quantity of several high fat diets, mice were nourished vous system may contribute positively or negatively to any given
with various concentrations and types of fats mingled with nor- neuropathological process (Table 2). Using APP-C99-transfected
mal chow. It was noticed that these diets not only modied the COS-7 cells, a cellular model of Alzheimers disease-like degen-
lipid prole in brain but also altered spatial memory and learn- eration, a study was carried out to investigate the class of fatty
ing ability of the pups in a different manner (Yu et al., 2010). In acids that was thought to inuence the production of A peptide,
another study, when mice were fed on diets supplemented with which is a major neuropathological hallmark of disease. It was

Table 2 | Changes in brain fatty acid composition in pathological conditions.

Fatty acid Disease Tendency Reference1

Saturated fatty acids (SFAs)


Myristic (14:0) Depression Down Conklin et al. (2010)
Palmitic (16:0) Alzheimer Up Fraser et al. (2010)
Palmitic (16:0) Parkinson Up Fabelo et al. (2011)
Palmitic (16:0) Depression Down Conklin et al. (2010)
Palmitic (16:0) Depression Up Hamazaki et al. (2012)
Stearic (18:0) Alzheimer Down Fraser et al. (2010)
Stearic (18:0) Parkinson Up Fabelo et al. (2011)
Stearic (18:0) Depression Down Conklin et al. (2010)
Mono-unsaturated fatty acids (MUFAs)
Palmitoleic (16:1) Alzheimer Up Astarita et al. (2011)
Palmitoleic (16:1) Depression Down Conklin et al. (2010)
Oleic (18:1) Alzheimer Down Martn et al. (2010)
Oleic (18:1) Alzheimer Up Fraser et al. (2010)
Oleic (18:1) Alzheimer Up Astarita et al. (2011)
Oleic (18:1) Depression Up Hamazaki et al. (2012)
Erucic (22:1) Alzheimer Up Astarita et al. (2011)
Nervonic (24:1) Alzheimer Up Astarita et al. (2011)
Polyunsaturated fatty acids (PUFAs)
EPA (20:5 n-3) Depression Down Lin et al. (2010)
DPA (22:5 n-3) Depression Down Conklin et al. (2010)
DHA (22:6 n-3) Alzheimer Down Martn et al. (2010)
DHA (22:6 n-3) Parkinson Down Fabelo et al. (2011)
DHA (22:6 n-3) Depression Down Conklin et al. (2010)
DHA (22:6 n-3) Depression Down Lin et al. (2010)
LA (18:2 n-6) Depression Down Conklin et al. (2010)
AA (20:4 n-6) Parkinson Down Fabelo et al. (2011)
AA (20:4 n-6) Depression Down Conklin et al. (2010)
Docosatetraenoic (22:4 n-6) Depression Down Conklin et al. (2010)
Docosatetraenoic (22:4 n-6) Schizophrenia Down Hamazaki et al. (2012)

1This table summarizes recent studies cited in the text. It is concluded that, whatever disease condition is considered, PUFA levels were systematically decreased,
whereas MUFA amounts often appeared increased. In contrast, there was no clear-cut tendency in the changes of the proportions of SFAs.

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 4

fncel-07-00144 2013/9/6 13:17 page 4 #4


Hussain et al. Fatty acids and nervous system

shown that palmitic acid (16:0), stearic acid (18:0), upstream n-3 behavioral decits induced by cuprizone, whereas a diet contain-
PUFAs, and AA (20:4 n-6) triggered higher secretion of A pep- ing n-3 PUFAs from cod affected similarly as n-6 PUFA enriched
tide compared to long chain downstream n-3 PUFAs and MUFAs or control diet did, suggesting that not only the type of PUFA
(Amtul et al., 2011a). These ndings were corroborated in vivo by but its origin is also to consider when prescribing a diet based
using a transgenic mouse model of early-onset Alzheimers disease remedy (Torkildsen et al., 2009). Contrasting these ndings, other
that expresses the double-mutant form of human APP, which is studies did not corroborate the protective effects of n-3 PUFAs
the precursor protein responsible for the synthesis of A peptide. against multiple sclerosis and concluded that neither n-3 nor n-6
Decreased levels of A peptide and less accumulation in the form of PUFAs had any effect on disease progression or remedial inuence
amyloid plaques were observed in the brain of mice nourished with (Wergeland et al., 2012). Moreover, dietary administration of EPA
a diet enriched in n-3 PUFAs, mainly DHA (22:6 n-3; Amtul et al., (20:5 n-3) even accelerated disease progression in mice expressing
2011a). Not only extraneously supplied but endogenously syn- a mutated form of Cu/Zn-superoxide dismutase (SOD1), which is
thesized n-3 PUFAs can suppress the synthesis of A peptide and a model of neuromuscular degeneration as caused by amyotrophic
the formation of amyloid plaques. Lebbadi et al. (2011) crossed lateral sclerosis (Yip et al., 2013).
3xTg-AD mice, a model of Alzheimers disease, with transgenic
mice expressing -3 desaturase (Fat-1) from Caenorhabditis ele- FATTY ACIDS AND TRAUMATIC INJURY TO THE NERVOUS SYSTEM
gans, which endogenously converts n-6 PUFAs into n-3 PUFAs. Several recent studies have provided evidence that n-3 PUFAs can
It was observed that the double transgenic 3xTg-AD/Fat-1 mice exert protection against neuronal injury triggered by hypoxia or
had increased brain levels of DHA (22:6 n-3) and lower levels of ischemia. In neonates, these fatty acids protected neurons follow-
A peptide. Similarly, MUFAs, mainly oleic acid (18:1 n-9), were ing hypoxia/ischemia by modulating the microglial inammatory
also shown to inhibit the production of A peptide and amyloid response through inhibition of the nuclear factor-B (NF-B)
plaques both in vitro and in vivo (Amtul et al., 2011b). In contrast, dependent pathway (Zhang et al., 2010). However, it is impor-
n-6 PUFAs, such as AA (20:4 n-6), aggravated Alzheimers disease tant to mention that consistent increased intake of n-3 PUFAs can
neuropathology, by increasing the synthesis of A peptide (Amtul also affect adversely in some cases. In this respect, a diet rich in
et al., 2012). EPA (20:5 n-3) and DHA (22:6 n-3) enhanced the risk for intrac-
The results obtained in experimental models of Alzheimers erebral hemorrhagic stroke in rats, and caused oxidative damage
disease have been conrmed, at a certain extent, by studies per- to the brain, probably due to the fact that a high PUFA con-
formed on human brain. Thus, decreased levels of PUFAs and tent increased the danger of lipid peroxidation. Alternatively, n-3
MUFAs, particularly DHA (22:6 n-3) and oleic acid (18:1 n-9), PUFA intake was reported to affect blood viscosity, vasoconstric-
respectively, were observed in the brain of Alzheimers disease tion, platelet aggregation, and blood clotting ultimately leading to
patients (Martn et al., 2010). However, other studies reported that, hemorrhaging (Park et al., 2009).
although the abundance of DHA (22:6 n-3) varied highly among There is also evidence that certain fatty acids have the poten-
patients, the mean quantity of this PUFA did not differ signi- tial to improve the recovery of the injured spinal cord. Hirakawa
cantly when compared to healthy brains (Fraser et al., 2010). This et al. (2010) reported that trans-2-decenoic acid ethyl ester, a
study also showed that levels of stearic acid (18:0) were reduced medium-chain fatty acid derivative, increased the expression of
remarkably in frontal and temporal cortex, while those of oleic extracellular signal-regulated protein kinases 1 and 2 (ERK1/2)
acid (18:1 n-9) were increased in both parts; also, levels of palmitic in cultured cortical neurons and at the site of injury in a rat
acid (16:0) appeared increased in the parietal cortex (Fraser et al., spinal cord injury model. Indeed, the administration of trans-
2010). These a priori puzzling abnormalities in MUFAs could be a 2-decenoic acid ethyl ester ameliorated functional recovery and
result of alterations in the expression of MUFA synthesizing genes. reduced lesion size in response to injury, by increasing the expres-
Thus, levels of MUFAs in hippocampus, frontal cortex and tem- sion of ERK1/2, brain-derived neurotrophic factor (BDNF), and
poral cortex were elevated in Alzheimers disease patients, as was anti-apoptotic Bcl-2. Similarly, DHA (22:6 n-3) pre-treatment in
the expression of the SCD isomers SCD1, SCD5a, and SCD5b. an acute spinal cord injury model diminished the extent of func-
In addition, the ratio of MUFAs to SFAs, an index of desaturase tional decits as compared to that observed in the control group,
activity, was reported to be negatively correlated with the degree and this protective effect was associated with increased survival of
of cognitive performance (Astarita et al., 2011). precursor cells, sparing of white matter and axonal preservation
Less is known about the changes of fatty acids in other neurode- (Figueroa et al., 2012; Lim et al., 2013b). In the same way, mice
generative conditions. Fabelo et al. (2011) reported that lipid rafts carrying the Fat-1 transgene for boosting endogenous synthesis of
from brain cortices of patients with Parkinson disease displayed n-3 PUFAs showed better outcome after spinal cord injury (Lim
signicantly decreased levels of n-3 and n-6 PUFAs, particularly et al., 2013a). Finally, in relation to diabetes, it was shown that
DHA (22:6 n-3) and AA (20:4 n-6), respectively, while SFAs, the augmentation of epoxy-fatty acid resources, as obtained by
mainly palmitic acid (16:0) and stearic acid (18:0), were noted inhibiting soluble epoxide hydrolase, resulted in a dose dependent
augmented, as compared to control subjects. In another study, anti-allodynic effect on neuropathic pain due to glucose toxicity
the effects of diets rich in n-3 or n-6 PUFAs were assessed on (Inceoglu et al., 2012).
cuprizone-induced experimental demyelination, an animal model
for multiple sclerosis. It was observed that n-3 PUFAs from various FATTY ACIDS AND NEUROLOGICAL DISORDERS
sources affected the pathological phenotype differently; for exam- Particular changes in brain fatty acid composition appear to be
ple, a diet containing n-3 PUFAs from salmon ameliorated the intimately connected to a series of neurological diseases, as recently

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 5

fncel-07-00144 2013/9/6 13:17 page 5 #5


Hussain et al. Fatty acids and nervous system

reported in several studies. Thus, Conklin et al. (2010) observed the amelioration of memory decits in aged mice (Labrousse et al.,
a reduction in the quantity of both saturated and unsaturated 2012). Dietary DHA (22:6 n-3) also enhanced the expression of F-
fatty acids of various types, including n-3 and n-6 PUFAs, in ATPase involved in mitochondrial ATP synthesis in the CA1 region
the cingulate cortex of depressive patients. Similar alternations of the hippocampus, whereas its deciency led to decreased glu-
in n-3 PUFAs, including EPA (20:5 n-3) and DHA (22:6 n-3), cose transporter expression and defective glucose transport in the
were also shown by others (Lin et al., 2010). In another study, cerebral cortex (Harbeby et al., 2012). The stimulatory action of
it was noticed that the altered concentrations of MUFAs and n-3 PUFAs on gene expression also appears to affect neurotrans-
PUFAs were region-specic. In fact, no changes in n-3 and n- mission. In fact, recent proteomics studies performed on mouse
6 PUFAs were found in hippocampus and orbitofrontal cortex brain decient in DHA (22:6 n-3) revealed a loss of synaptic pro-
of patients with depression but concentrations of MUFAs, such teins associated with altered synaptic transmission (Sidhu et al.,
as oleic acid (18:1 n-9), and SFAs, such as palmitic acid (16:0), 2011). In contrast, expression of vesicular glutamate transporters
appeared augmented (Hamazaki et al., 2012). A partial conr- 1 and 2, which are implicated in glutamatergic neurotransmission,
mation of these ndings emerged from another study showing was increased in response to ALA (18:3 n-3) exposure (Blondeau
lowered expression of genes involved in PUFA and MUFA synthe- et al., 2009). Similarly, DHA (22:6 n-3) provision to rats with trau-
sis in the frontal cortex of depressed patients (McNamara and matic brain injury enhanced learning ability, by modulating the
Liu, 2011). It is also noteworthy that lifelong n-3 PUFA de- expression levels of synapsin-1, cAMP response element-binding
ciency perturbed normal endocannabinoid function in prelimbic protein-1 and calcium/calmodulin-dependent protein kinase-2 in
prefrontal cortex and accumbens, and this effect was related to the hippocampus of treated animals (Wu et al., 2008, 2011). DHA
impaired emotional behavior (Lafourcade et al., 2011). Although (22:6 n-3) also ameliorated spatial memory in rats by increas-
less investigated, several studies also detected changes in fatty acids ing the expression of subtypes of endocannabinoid/endovanilloid
in patients with schizophrenia. A decrease in docosatetraenoic receptors (Pan et al., 2011). Last, n-3 PUFAs augmented the
acid (22:4 n-6) was observed in the nuclei of the amygdala of these expression of a series of transcription factors involved in learning
patients but other PUFAs, including DHA (22:6 n-3) and AA (20:4 and memory, including retinoic acid receptor, retinoic X recep-
n-6), remained unchanged (Hamazaki et al., 2010, 2012). Interest- tor and peroxisome proliferator-activated receptor (Dyall et al.,
ingly, the decrease in total membrane PUFAs found in erythrocytes 2010).
of young patients with schizophrenia correlated with the degree Many positive actions of DHA (22:6 n-3), and likely other n-3
of demyelination in brain white matter (Peters et al., 2009). PUFAs, may therefore converge to enhance synaptic transmission,
Lastly, several lines of evidence support the anticonvulsant and ameliorate spatial learning and memory (Connor et al., 2012).
effects of certain fatty acids in animal models of epileptogenesis, In a mouse model of systemic lupus erythematosus and Sjgrens
and the administration of PUFA enriched diets has been envisaged syndrome, which is characterized by behavioral abnormalities,
to treat epileptogenic convulsions. Using the pentylenetetrazol- reduced aged hippocampal neurogenesis and loss of long-term
induced epilepsy rat model, Porta et al. (2009) showed that a PUFA potentiation (LTP), the dietary supplementation with n-3 PUFAs
containing diet increased the threshold level for pentylenetetra- corrected LTP at synapses in the medial perforant pathway/dentate
zol to induce convulsions. A contemporary study conrmed that gyrus and enhanced the amount of adult-born neurons in the hip-
rats nourished with n-3 PUFAs exhibited greater resistance to pocampus (Crupi et al., 2012). Similarly, docosapentaenoic acid
pentylenetetrazol-induced seizures (Taha et al., 2009). In the kin- (DPA, 22:5 n-3) also ameliorated hippocampal function by attenu-
dling model of epilepsy, intracerebroventricular injection of DHA ating the reduction in LTP in aged brain (Kelly et al., 2011). Finally,
(22:6 n-3), or its derivative NPD1, limited the progression in the in vitro studies showed that treatment of differentiated PC12 cells
hippocampus of the electrically induced neuronal hyperexcitabil- with EPA (20:5 n-3) resulted in activation of the neuroprotec-
ity characteristic of seizures (Musto et al., 2011). In contrast, other tive PI3-kinase/Akt signaling pathway, a mechanism that might
studies did not corroborate these ndings, since DHA (22:6 n- account for the increase in LTP observed in vivo following EPA
3) or EPA (20:5 n-3) showed neither anticonvulsant activity nor (20:5 n-3) treatment (Wu et al., 2008; Kawashima et al., 2010).
protection against pentylenetetrazol-induced seizures (Willis et al., In Alzheimers disease, A peptide induces neuronal apopto-
2009). sis through degradation of the adaptor protein insulin receptor
substrate-1 in a c-Jun N-terminal kinase (JNK)-dependent man-
CELLULAR ROLES OF FATTY ACIDS IN THE NERVOUS SYSTEM ner. An n-3 PUFA enriched diet prevented the phosphorylation
ACTIONS OF FATTY ACIDS IN THE HIPPOCAMPUS of JNK, and ultimately protected neurons from death in vitro and
Many recent studies have investigated the implication of fatty acids improved cognitive decit in vivo (Ma et al., 2009). Also, lower
in learning and memory processes occurring in the hippocampus levels of phosphorylated tau protein and improved brain function
(Figure 2). In general, n-3 PUFAs were shown to foster neuronal were observed by crossing 3xTg-AD mice with Fat-1 express-
activity and hence counteract memory decits. It is well known ing mice to enhance the endogenous production of n-3 PUFAs
that increased c-Fos expression is an indicator of neuronal activ- (Lebbadi et al., 2011). Nevertheless, it is noteworthy that 12/15-
ity in response to extracellular signals like growth factors, and it lipo-oxygenase adversely affected Alzheimers disease pathology
is initiated when neurons re action potentials. Commonly, the by synthesizing pro-inammatory and pro-oxidant hydroperox-
activity of c-Fos decreases as age extends and spatial memory goes yacids resulting from the oxidation of PUFAs, so that genetic
off. Provision of n-3 PUFAs restored c-Fos expression in the hip- ablation of this enzyme ameliorated cognitive function (Yang et al.,
pocampus, and enhanced neuronal activity ultimately leading to 2010).

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 6

fncel-07-00144 2013/9/6 13:17 page 6 #6


Hussain et al. Fatty acids and nervous system

FIGURE 2 | Multiple effects of fatty acids in the hippocampus. n-3 and n-6 PUFAs exert a variety of positive actions that promote formation, storage and
processing of learning and memory in the hippocampus. In contrast, SFAs display rather negative actions. Green arrows indicate stimulatory effects while
orange arrows represent inhibitory effects.

Neuroinammation is one of the distinctive features of aged or apoptotic demise. Interestingly, these pro-inammatory actions of
diseased brain, as demonstrated by the activation of glial cells and SFAs could be reverted by n-3 PUFAs like DHA (22:6 n-3; Gupta
the increase in the expression of a variety of pro-inammatory et al., 2012; Wang et al., 2012).
factors. In this respect, it was reported that n-3 PUFA provi- Another way by which n-3 PUFAs can afford neuroprotection
sion restored spatial memory loss in aged animals by suppressing is by preventing apoptosis. The mouse model of infantile neu-
pro-inammatory interleukin-1 and reverting to normal the ronal ceroid lipofuscinosis, a neurodegenerative disease caused
morphology of microglia and astrocytes in the hippocampus by palmitoyl-protein thioesterase-1 (PPT1) deciency, manifests
(Labrousse et al., 2012; Park et al., 2012). n-3 PUFAs also yielded enhanced endoplasmic reticulum- and oxidative stress that lead
protecting effects to neurons by blocking microglia activation in to apoptotic cell demise. In PPT1-decient cells from such mice,
a transgenic mouse model of systemic lupus erythematosus and intervention of n-3 PUFAs attenuated stress and repressed apop-
Sjgrens syndrome (Crupi et al., 2012). In the same way, DPA totic death casting a protection to neuronal cells (Kim et al., 2010;
(22:5 n-3) inactivated microglia attenuating neuroinammation Wu et al., 2011). Similarly, differentiated PC12 cells treated with
and counteracting spatial learning decit in aged brain (Kelly et al., EPA (20:5 n-3) showed lower rates of apoptosis and suppressed
2011). Contrary to the protective effects of PUFAs, SFAs stimulated activity of the apoptotic effector caspase-3 (Boudrault et al., 2009;
the secretion of pro-inammatory cytokines and induced apop- Kawashima et al., 2010). Conjugated LA (18:2 n-6) also protected
tosis in astrocytes. Particularly, palmitic acid (16:0), lauric acid neurons from mitochondrial dysfunction and demise. Treatment
(12:0), and stearic acid (18:0) triggered the secretion of tumor of cortical neurons with this fatty acid following excitotoxic glu-
necrosis factor- (TNF-) and interleukin-6 by engaging toll-like tamate exposure resulted in decreased glutamate-induced loss
receptor-4 (TLR-4). Moreover, palmitic acid (16:0) also activated of mitochondrial function, increased Bcl-2 expression and pro-
caspase-3 and modied the Bax/Bcl-2 ratio in these glial cells for longed neuronal survival (Hunt et al., 2010). In the same manner,

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 7

fncel-07-00144 2013/9/6 13:17 page 7 #7


Hussain et al. Fatty acids and nervous system

administration of sh oil, that is a rich source of n-3 PUFAs, pro- neurotrophic N-docosahexaenoylethanolamine, stimulated neu-
tected hippocampal neurons from diabetic insult by precluding rogenic differentiation of neural stem cells (Katakura et al., 2013;
the expression of apoptosis inducing genes in both CA1 region and Rashid et al., 2013). The importance of the stimulatory role of
cultured cells, and by increasing the expression of anti-apoptotic PUFAs for neurogenesis is also illustrated by experiments report-
genes, such as Bcl-2 and Bcl-xL (Zhang and Bazan, 2010; Zhao ing increased expression of fatty acid binding proteins (FABPs) in
et al., 2012). Together with caspase-3, ceramides, resulting from the ischemic hippocampus. FABPs are carriers of PUFAs in the
the hydrolysis of sphingomyelin by sphingomyelinase, are well- cytoplasm, and their expression declines with age in association
known apoptosis inducing factors. Treatment with DPA (22:5 n-3) with reduced synaptic activity and other cellular functions. CA1
inactivated sphingomyelinase and caspase-3 in the hippocampus and dentate gyrus regions in the hippocampus showed augmented
of elderly rats (Kelly et al., 2011). On the other hand, n-3 PUFA levels of FABP-5 and FABP-7 after ischemia, suggesting elevated
withdrawal modulated the phosphorylation of glycogen-synthase transportation of PUFAs in these regions to restore cellular neu-
kinase-3 and ERK1/2, predisposing more hippocampal neurons rophysiology (Liu et al., 2010; Ma et al., 2010). More importantly,
to damage in an in vitro oxygen and glucose deprivation model of at post-ischemic stages, the subgranular zone in the dentate gyrus
ischemia (Moreira et al., 2010b). Along with this, a decrease in the of the hippocampus, a niche of adult neurogenesis, displayed a
release of PUFAs from cell membranes in the rat hippocampus, as concomitant increase in the neuronal expression of FABPs and
a result of reduced phospholipase-A2 activity, caused alterations in the fatty acid receptor GPR40, representing compensatory pro-
membrane uidity that could account for loss of spatial memory cesses of newborn cells (Boneva et al., 2011a,b; Yamashima, 2012).
and cognitive impairment in Alzheimers disease (Schaeffer et al., Finally, it is noteworthy that many of the benecial actions of
2011). However, the protective effects of n-3 PUFAs under certain PUFAs on hippocampal function were associated with an increase
conditions seemed to be limited to some of the members of this in the production of BDNF, which is a member of the neurotrophin
class of fatty acids. Thus, only DHA (22:6 n-3) offset the expression family of growth factors involved in supporting growth, differen-
of AMPA receptors in the membrane of hippocampal neurons and tiation and survival of neurons (Wu et al., 2008, 2011; Blondeau
attenuated neurotoxicity leading to improved cognitive function. et al., 2009; Venna et al., 2009; Avraham et al., 2011; Vines et al.,
Other members of the n-3 PUFA family, especially EPA (20:5 n- 2012).
3), lacked such a protective effect against AMPA-mediated toxicity
(Mnard et al., 2009). ACTIONS OF FATTY ACIDS IN THE HYPOTHALAMUS
Synaptogenesis is one of the mechanisms by which memory The central regulation of energy balance involves a number of
process takes place. Hence, the loss of synapses is characteristic neuronal circuits in the hypothalamus that either exert anorexic
of neurodegenerative conditions and aging. For instance, cortical actions or stimulate food intake. In this respect, it was recently
or hippocampal neurons incubated with the neurotoxic prion- shown that certain fatty acids could inuence the control of energy
derived peptide PrP82146, and pre-treated with DHA (22:6 homeostasis by the hypothalamus. In general, dietary supple-
n-3) or EPA (20:5 n-3), showed less loss of synaptophysin-1 and mentation with sh oil, rich in n-3 PUFAs, normalized several
reduced accumulation of prion peptide (Bate et al., 2010). ALA hypothalamic neurochemical systems in food restricted animals
(18:3 n-3) also stimulated the expression of genes involved in (Avraham et al., 2011). However, supplementation of diet with
synaptic function, like VAMP-2, SNAP-25 and synaptophysin-1, SFAs induced endoplasmic reticulum stress and expression of
that led to improved stability and physiology of synapses (Blon- cytokines via TLR-4 signaling in the hypothalamus, and this effect
deau et al., 2009). Similarly, the chronic supplementation of n-3 resulted in resistance to anorexigenic signals (Milanski et al., 2009).
PUFAs yielded anti-depressant effects by increasing the expres- At the cellular level, treating hypothalamic mHy-poE-44 cells with
sion of synaptophysin-1 in the hippocampus (Venna et al., 2009). palmitic acid (16:0) increased the expression of the orexigenic
However, another study performed on SH-SY5Y cells reported that neuropeptide-Y, suggesting that this fatty acid could enhance food
DHA (22:6 n-3) did not affect the neurotransmission machinery, intake (Fick et al., 2011). Moreover, palmitic acid (16:0) faded
as evaluated by the expression of synaptotagmin-1, syntaxin-1A, insulin signaling and enhanced endoplasmic reticulum stress and
and synaptobrevin-1, although the release of noradrenaline by caspase-3 cleavage in the same cell line, which resulted in apop-
these cells was enhanced (Mathieu et al., 2010). tosis in a JNK-dependent manner (Mayer and Belsham, 2010). In
Hippocampal neurogenesis also contributes to learning and another study, exposure to palmitic acid (16:0) displayed no effects
memory processes. The mouse model of systemic lupus erythe- on insulin resistance and inammatory process activation but cor-
matosus and Sjgrens syndrome typically exhibits age-dependent roborated the stimulation of endoplasmic reticulum stress and
reduced hippocampal neurogenesis. Supplementation of diet with apoptosis, along with the activation of mitogen-activated protein
n-3 PUFAs to these mice enhanced the density of bromodeoxyuri- kinase (Choi et al., 2010).
dine (BrdU)- and doublecortin positive cells in the hippocampus,
suggesting an ongoing neurogenesis (Crupi et al., 2012). Similar ACTIONS OF FATTY ACIDS IN THE NIGROSTRIATAL PATHWAY
neurogenesis enhancement was also reported in response to ALA Growing evidence supports a link between the dietary intake of
(18:3 n-3) treatment (Blondeau et al., 2009). In addition, AA (20:4 n-3 PUFAs and the function (or dysfunction) of the nigrostri-
n-6) even increased neurogenesis at postnatal stages when admin- atal pathway involved in the control of movement (Figure 3).
istered at gestation period (Maekawa et al., 2009). Several in vitro This relationship was particularly investigated in a number of ani-
studies revealed that not only n-3 PUFA precursors, such as EPA mal models of Parkinson disease, which is a neurodegenerative
(20:5 n-3), but also naturally derived metabolites, including the condition primarily characterized by the loss of dopaminergic

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 8

fncel-07-00144 2013/9/6 13:17 page 8 #8


Hussain et al. Fatty acids and nervous system

FIGURE 3 | Conflicting effects of n-3 PUFAs in the nigrostriatal pathway. pro-inammatory cues. However, extreme caution should be exercised since
n-3 PUFAs are commonly endowed with a wide range of helpful effects, as these same PUFAs may not provide complete safety to halt degeneration
illustrated by the protective benet that these fatty acids offer to induced by parkinsonian toxins or even trigger adverse effects, which
dopaminergic neurons in the nigrostriatal tract against apoptotic and eventually aggravates the extent of the pathological process.

neurons connecting the substantia nigra to the striatum. In pathogenesis. Indeed, the presence of DHA (22:6 n-3) augmented
several recent studies, n-3 PUFAs were shown to be benecial neuritic injury and astrocytosis in mice transgenic for a Parkin-
by reverting disease phenotype. In the MPTP model of Parkin- son disease causing mutation in human -synuclein. In addition,
son disease, pre-treatment of mice with n-3 PUFAs bestowed DHA (22:6 n-3) triggered oligomerization of -synuclein, through
protection by increasing the expression of BDNF and involv- the activation of retinoic X receptor and peroxisome proliferator-
ing its TrkB receptor (Bousquet et al., 2009; Balanz-Martnez activated receptor-2. Interestingly, its withdrawal from diet was
et al., 2011). In other studies, it was found that exposure to the found to be benecial against the deleterious effects caused by
n-3 PUFA ethyl-eicosapentaenoate derivative lowered the expres- it provision (Yakunin et al., 2012). Finally, structural and con-
sion of Bax and caspase-3, and enhanced cortical dopamine formational modications in -synuclein leading to pathological
levels (Bousquet et al., 2008; Meng et al., 2010). Furthermore, aggregation were brought by DHA (22:6 n-3; De Franceschi et al.,
n-3 PUFAs also yielded protective inuence indirectly, by atten- 2009, 2011; Bousquet et al., 2011).
uating inammation-causing factors. These fatty acids targeted
the NFB signaling pathway in microglia to suppress their over- ACTIONS OF FATTY ACIDS IN THE PERIPHERAL NERVES
activated response and hence protect dopaminergic neurons A subset of peripheral sensory neurons expresses transient recep-
(Boudrault et al., 2009; Zhang et al., 2010; Ji et al., 2012; Zhou tor potential cation channel-A1 (TRPA1), which is involved in
et al., 2012). pain and neurogenic inammation. TRPA1 is a target for a vari-
Other ndings, however, did not support the benecial ety of noxious and inammatory irritant substances. In addition,
effects of n-3 PUFAs on Parkinson disease. It was reported it was found that n-3 PUFAs could act as a ligand for TRPA1 to
that treatment with ethyl-eicosapentaenoate, although minimized excite sensory neurons and hence regulate their responses in vivo
pro-inammatory cytokines and yielded positive effects on pro- (Motter and Ahern, 2012). Transient receptor potential vanilloid
cedural memory decit, it was unable to preclude the loss of cation channel-1 (TRPV1), which is another member of the fam-
nigrostriatal dopamine in MPTP mice (Shchepinov et al., 2011; ily, is also found mainly in nociceptive neurons of the peripheral
Luchtman et al., 2012). Similarly, the parkinsonian neurotoxin 6- nervous system, where they are involved in transmission and mod-
hydroxydopamine caused lesions in the medial forebrain bundle ulation of pain. In this respect, it was shown that NPD1, which has
of rats and motor decits that remained unaffected by sh oil anti-inammatory properties, inhibited TRPV1 currents induced
derived n-3 PUFAs (Delattre et al., 2010). A chronic interven- by capsaicin in dorsal root ganglion neurons, and modulated
tion of a DHA (22:6 n-3) containing diet modied neither the TRPV1/TNF--mediated synaptic plasticity in the spinal cord,
number of cortical glial cells nor the expression of -synuclein, suggesting a novel analgesic role (Park et al., 2011). The effects of
which is typically involved in disease pathogenesis (Muntan et al., fatty acids on sensory neurons go beyond receptor signaling. Both
2010). The use of different animal models of Parkinson disease n-6 and n-3 PUFAs promoted neurite outgrowth in sensory neu-
and the different ways of treating these mice to counteract the rons from dorsal root ganglia of embryos but also adult and aged
pathological process may explain the observed discrepancies. In animals (Robson et al., 2010). Enhanced levels of endogenously
this respect, it is important to mention that some studies indi- synthesized n-3 PUFAs also bestowed benecial effects in various
cated even adverse effects of n-3 PUFAs on Parkinson disease aspects. Thus, dorsal root ganglion neurons from Fat-1 expressing

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 9

fncel-07-00144 2013/9/6 13:17 page 9 #9


Hussain et al. Fatty acids and nervous system

mice exhibited more resistance to hypoxia and mechanical injury During the last century, however, the n-6:n-3 ratio has dramat-
as compared to neurons from wild-type littermates. Furthermore, ically increased up to 2025:1, particularly in Western societies,
Fat-1 expressing mice showed better functional recovery after sci- because of a high consumption of n-6 PUFAs to the detriment
atic nerve crush. The increased endogenous levels of n-3 PUFAs of n-3 PUFA intake (Simopoulos, 2011). Once the equilibrium
reduced the expression of the stress sensor activating transcription is broken, an excessively high n-6:n-3 ratio would impair normal
factor-3 in dorsal root ganglion neurons, and diminished muscle brain function and, importantly, predispose to disease (Palacios-
atrophy (Gladman et al., 2012). Similarly, our own studies also Pelaez et al., 2010). According to what we have exposed herein, a
reported that the down-regulation of SCD1, which is in charge of huge amount of studies have shown the good and the bad side
the production of MUFAs such as oleic acid (18:1), triggered accel- of different fatty acids in many experimental models of trauma
erated motor function recovery after sciatic nerve crush, providing and disease. Nevertheless, the diversity in modeling any given
evidence for a new role of this fatty acid desaturase in modulating physiopathological condition, together with differences in time,
the restorative potential of the neuromuscular axis (Hussain et al., dose and type of fatty acid used to counteract the insult, cer-
2013). tainly account for a number of conicting results concerning the
The retina possesses a high concentration of n-3 PUFAs, par- nature of the observed effects. In addition, it must be taken into
ticularly DHA (22:6 n-3). Many studies have shown that this fatty consideration that particular fatty acids are assumed to foster neu-
acid not only has a structural function but also protects visual roprotection but engender indeed a series of collateral deleterious
neurons from trauma and disease. Recently, it was noticed that actions, such as increasing oxidative stress susceptibility or favor-
the retinal dysfunction induced by diabetes could be recovered to ing neurodegenerative protein aggregation, which may preclude
some extent by supplementing DHA (22:6 n-3) extraneously. In the use of these fatty acids under certain (pathological) condi-
fact, diabetes resulted in reduced levels of n-3 PUFAs, by affecting tions (Figure 3). Finally, it is also noteworthy that, frequently,
n-3 fatty acid desaturase enzymatic activity, so that the provision studies used nutritional approaches consisting in giving a specic
of a DHA (22:6 n-3) enriched diet prevented dysfunction of rods fatty acid or its precursor mixed with others and forming part
and ameliorated vision (Yee et al., 2010). Also, n-3 PUFA derived of foods relatively more complex than desired, since they also
NPD1, together with pigment epithelial-derived growth factor, contain other substances with potential, uncontrolled positive or
promoted corneal nerve regeneration in a rat model of surgi- negative effects. Taken together, these drawbacks limit the trans-
cal injury (Cortina et al., 2010, 2012; Kenchegowda et al., 2013). latability of successful results in terms of neuroprotection obtained
However, other studies rather obtained contradictory results. in animal experiments into effective therapeutic interventions in
Therefore, augmented levels of DHA (22:6 n-3) bestowed no humans. Numerous epidemiological studies have put fatty acids
protection against retinal degeneration in mice carrying a disease- forward as key factors contributing to neuropathology but, in some
causing VPP rhodopsin mutation and expressing Fat-1 (Li et al., cases, discrepant concentrations of fatty acids were reported in
2009, 2010). In the same way, it was also reported that high levels the corresponding diseased brain regions (Table 2). Despite these
of DHA (22:6 n-3) in the retina could generate oxidative stress, constraints, on the basis of these epidemiological studies and sup-
instead of protection, and hence enhance the susceptibility to ported by experimental research, there is quite realistic evidence
degeneration (Tanito et al., 2009). to envisage that nutritional therapies based on fatty acids can
be of benet to several neurodegenerative and neurological dis-
CONCLUSION eases, such as age-related macular degeneration, cognitive decline,
The biological functions of fatty acids have been investigated depression, and some related behavioral disorders (Prior and Gal-
intensively during these last years, due to their active involvement durz, 2012; Schleicher et al., 2013). More research is needed now
in the physiology of both central and peripheral nervous system. for arriving at the nal and conclusive result concerning the type
They promote brain development, ameliorate cognitive functions of fatty acid, number of double bonds, origin, particular stage and
in normal and diseased conditions, serve as anti-depressants and proper concentration to achieve benecial therapeutic potential
anti-convulsants, bestow protection against traumatic insults, and against otherwise incurable diseases.
elevate repairing processes. At the cellular level, fatty acids stimu-
late gene expression and neuronal activity, and boost synaptogen- ACKNOWLEDGMENTS
esis and neurogenesis while preventing from neuroinammatory This work was supported by funds from European Communitys
toxicity and apoptosis (Figure 2). Although the demand for fatty Health Seventh Framework Programme under grant agreement
acids in a healthy body applies to any of them, it can be said that, No. 259867, and Thierry Latran Foundation to Jean-Philippe
in general, excess of SFAs and, to some extent, n-6 PUFAs brings Loefer; and Association pour la Recherche sur la Sclrose
about negative consequences, whereas MUFAs and n-3 PUFAs Latrale Amyotrophique et autres Maladies du Motoneurone
are endowed with rather benecial properties. In this respect, to Jose-Luis Gonzalez de Aguilar. Ghulam Hussain is supported
the ratio of n-6 to n-3 PUFAs is of special interest. It has been by the Higher Education Commission of the Pakistani govern-
postulated that a relatively constant n-6:n-3 ratio of about 1:1 ment and Association pour la Recherche et le Dveloppement de
constituted a major breakthrough in the expansion of gray mat- Moyens de Lutte contre les Maladies Neurodgnratives (ARE-
ter in the cerebral cortex of modern human beings (Bradbury, MANE). Florent Schmitt is granted by Association Franaise
2011). In the brain, the preservation of an optimal n-6:n-3 ratio contre les Myopathies and AREMANE. Jose-Luis Gonzalez de
is crucial to the maintenance of the variety of the cellular pro- Aguilar is recipient of a Chaire dExellence INSERM/Universit
cesses in which PUFAs participate (Luchtman and Song, 2013). de Strasbourg.

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 10

fncel-07-00144 2013/9/6 13:17 page 10 #10


Hussain et al. Fatty acids and nervous system

REFERENCES stroke. Neuropsychopharmacology Chen, H.-F., and Su, H.-M. (2012). surgery. Arch. Ophthalmol. 130,
Amtul, Z., Uhrig, M., Rozma- 34, 25482559. doi: 10.1038/npp. Fish oil supplementation of mater- 7683. doi: 10.1001/archophthalmol.
hel, R. F., and Beyreuther, K. 2009.84 nal rats on an n-3 fatty acid-decient 2011.287
(2011a). Structural insight into the Boneva, N. B., Kaplamadzhiev, D. diet prevents depletion of maternal Crupi, R., Cambiaghi, M., Deckel-
differential effects of omega-3 and B., Sahara, S., Kikuchi, H., Pyko, brain regional docosahexaenoic acid baum, R., Hansen, I., Mindes, J.,
omega-6 fatty acids on the produc- I. V, Kikuchi, M., et al. (2011a). levels and has a postpartum anxi- Spina, E., et al. (2012). n-3 fatty acids
tion of Abeta peptides and amy- Expression of fatty acid-binding pro- olytic effect. J. Nutr. Biochem. 23, prevent impairment of neurogene-
loid plaques. J. Biol. Chem. 286, teins in adult hippocampal neuro- 299305. doi: 10.1016/j.jnutbio.2010. sis and synaptic plasticity in B-cell
61006107. doi: 10.1074/jbc.M110. genic niche of postischemic monkeys. 12.010 activating factor (BAFF) transgenic
183608 Hippocampus 21, 162171. doi: Choi, S. J., Kim, F., Schwartz, M. mice. Prev. Med. 54(Suppl.), S103
Amtul, Z., Westaway, D., Cechetto, D. 10.1002/hipo.20732 W., and Wisse, B. E. (2010). Cul- S108. doi: 10.1016/j.ypmed.2011.
F., and Rozmahel, R. F. (2011b). Boneva, N. B., Kikuchi, M., Minabe, tured hypothalamic neurons are 12.019
Oleic acid ameliorates amyloidosis Y., and Yamashima, T. (2011b). resistant to inammation and De Franceschi, G., Frare, E., Bubacco,
in cellular and mouse models of Neuroprotective and ameliorative insulin resistance induced by sat- L., Mammi, S., Fontana, A., and
Alzheimers disease. Brain Pathol. actions of polyunsaturated fatty acids urated fatty acids. Am. J. Physiol. De Laureto, P. P. (2009). Molec-
21, 321329. doi: 10.1111/j.1750- against neuronal diseases: impli- Endocrinol. Metab. 298, E1122 ular insights into the interaction
3639.2010.00449.x cation of fatty acid-binding pro- E1130. doi: 10.1152/ajpendo.00 between alpha-synuclein and docosa-
Amtul, Z., Uhrig, M., Wang, L., Rozma- teins (FABP) and G protein-coupled 006.2010 hexaenoic acid. J. Mol. Biol. 394,
hel, R. F., and Beyreuther, K. (2012). receptor 40 (GPR40) in adult Chung, W.-L., Chen, J.-J., and Su, 94107. doi: 10.1016/j.jmb.2009.
Detrimental effects of arachidonic neurogenesis. J. Pharmacol. Sci. H.-M. (2008). Fish oil supplementa- 09.008
acid and its metabolites in cellular 116, 163172. doi: 10.1254/jphs. tion of control and (n-3) fatty acid- De Franceschi, G., Frare, E., Pivato,
and mouse models of Alzheimers 10R34FM decient male rats enhances reference M., Relini, A., Penco, A., Greggio,
disease: structural insight. Neu- Boudrault, C., Bazinet, R. P., and and working memory performance E., et al. (2011). Structural and
robiol. Aging 33, 831.e2131. doi: Ma, D. W. L. (2009). Experimental and increases brain regional docosa- morphological characterization of
10.1016/j.neurobiolaging.2011.07.014 models and mechanisms underlying hexaenoic acid levels. J. Nutr. 138, aggregated species of -synuclein
Astarita, G., Jung, K.-M., Vasilevko, the protective effects of n-3 polyun- 11651171. induced by docosahexaenoic
V., Dipatrizio, N. V., Martin, S. saturated fatty acids in Alzheimers Coluccia, A., Borracci, P., Renna, G., acid. J. Biol. Chem. 286, 22262
K., Cribbs, D. H., et al. (2011). disease. J. Nutr. Biochem. 20, Giustino, A., Latronico, T., Ric- 22274. doi: 10.1074/jbc.M110.
Elevated stearoyl-CoA desaturase in 110. doi: 10.1016/j.jnutbio.2008. cio, P., et al. (2009). Developmental 202937
brains of patients with Alzheimers 05.016 omega-3 supplementation improves Delattre, A. M., Kiss, A., Szawka, R.
disease. PLoS ONE 6:e24777. doi: Bousquet, M., Calon, F., and Cic- motor skills in juvenile-adult rats. E., Anselmo-Franci, J. A., Bagatini,
10.1371/journal.pone.0024777 chetti, F. (2011). Impact of -3 fatty Int. J. Dev. Neurosci. 27, 599 P. B., Xavier, L. L., et al. (2010).
Avraham, Y., Saidian, M., Burston, J. J., acids in Parkinsons disease. Age- 605. doi: 10.1016/j.ijdevneu.2009. Evaluation of chronic omega-3 fatty
Mevorach, R., Vorobiev, L., Magen, I., ing Res. Rev. 10, 453463. doi: 05.011 acids supplementation on behavioral
et al. (2011). Fish oil promotes sur- 10.1016/j.arr.2011.03.001 Conklin, S. M., Runyan, C. A., Leonard, and neurochemical alterations in 6-
vival and protects against cognitive Bousquet, M., Gibrat, C., Saint- S., Reddy, R. D., Muldoon, M. F., hydroxydopamine-lesion model of
decline in severely undernourished Pierre, M., Julien, C., Calon, F., and Yao, J. K. (2010). Age-related Parkinsons disease. Neurosci. Res. 66,
mice by normalizing satiety signals. and Cicchetti, F. (2009). Modula- changes of n-3 and n-6 polyunsatu- 256264. doi: 10.1016/j.neures.2009.
J. Nutr. Biochem. 22, 766776. doi: tion of brain-derived neurotrophic rated fatty acids in the anterior cingu- 11.006
10.1016/j.jnutbio.2010.07.001 factor as a potential neuroprotec- late cortex of individuals with major Dyall, S. C., Michael, G. J., and Michael-
Balanz-Martnez, V., Fries, G. R., tive mechanism of action of omega-3 depressive disorder. Prostaglandins Titus, A. T. (2010). Omega-3 fatty
Colpo, G. D., Silveira, P. P., Portella, fatty acids in a parkinsonian animal Leukot. Essent. Fatty Acids 82, acids reverse age-related decreases in
A. K., Tabars-Seisdedos, R., et al. model. Prog. Neuropsychopharma- 111119. doi: 10.1016/j.plefa.2009. nuclear receptors and increase neuro-
(2011). Therapeutic use of omega-3 col. Biol. Psychiatry 33, 1401 12.002 genesis in old rats. J. Neurosci. Res. 88,
fatty acids in bipolar disorder. Expert 1408. doi: 10.1016/j.pnpbp.2009. Connor, S., Tenorio, G., Clandinin, 20912102. doi: 10.1002/jnr.22390
Rev. Neurother. 11, 10291047. doi: 07.018 M. T., and Sauv, Y. (2012). DHA Enoch, H. G., Catala, A., and Strittmat-
10.1586/ern.11.42 Bousquet, M., Saint-Pierre, M., Julien, supplementation enhances high- ter, P. (1976). Mechanism of rat liver
Bate, C., Tayebi, M., Salmona, M., C., Salem, N., Cicchetti, F., and frequency, stimulation-induced microsomal stearyl-CoA desaturase.
Diomede, L., and Williams, A. (2010). Calon, F. (2008). Benecial effects synaptic transmission in mouse Studies of the substrate specicity,
Polyunsaturated fatty acids pro- of dietary omega-3 polyunsaturated hippocampus. Appl. Physiol. Nutr. enzyme-substrate interactions, and
tect against prion-mediated synapse fatty acid on toxin-induced neuronal Metab. 37, 880887. doi: 10.1139/ the function of lipid. J. Biol. Chem.
damage in vitro. Neurotox. Res. 17, degeneration in an animal model h2012-062 251, 50955103.
203214. doi: 10.1007/s12640-009- of Parkinsons disease. FASEB J. Cortina, M. S., He, J., Li, N., Etschmaier, K., Becker, T., Eichmann,
9093-2 22, 12131225. doi: 10.1096/fj.07- Bazan, N. G., and Bazan, H. E. P. T. O., Schweinzer, C., Scholler,
Bazan, N. G. (2009). Cellular and 9677com (2010). Neuroprotectin D1 synthesis M., Tam-Amersdorfer, C., et al.
molecular events mediated by Bradbury, J. (2011). Docosahexaenoic and corneal nerve regeneration after (2011). Adipose triglyceride lipase
docosahexaenoic acid-derived acid (DHA): an ancient nutrient for experimental surgery and treatment affects triacylglycerol metabolism at
neuroprotectin D1 signaling in the modern human brain. Nutri- with PEDF plus DHA. Invest. Oph- brain barriers. J. Neurochem. 119,
photoreceptor cell survival and brain ents 3, 529554. doi: 10.3390/nu30 thalmol. Vis. Sci. 51, 804810. doi: 10161028. doi: 10.1111/j.1471-
protection. Prostaglandins Leukot. 50529 10.1167/iovs.09-3641 4159.2011.07498.x
Essent. Fatty Acids 81, 205211. doi: Brenna, J. T., Salem, N., Sinclair, Cortina, M. S., He, J., Li, N., Bazan, Fabelo, N., Martn, V., Santpere, G.,
10.1016/j.plefa.2009.05.024 A. J., and Cunnane, S. C. (2009). N. G., and Bazan, H. E. P. (2012). Marn, R., Torrent, L., Ferrer, I., et al.
Blondeau, N., Nguemeni, C., Debruyne, alpha-Linolenic acid supplementa- Recovery of corneal sensitivity, (2011). Severe alterations in lipid
D. N., Piens, M., Wu, X., Pan, H., et al. tion and conversion to n-3 long- calcitonin gene-related peptide- composition of frontal cortex lipid
(2009). Subchronic alpha-linolenic chain polyunsaturated fatty acids positive nerves, and increased rafts from Parkinsons disease and
acid treatment enhances brain plas- in humans. Prostaglandins Leukot. wound healing induced by pigment incidental Parkinsons disease. Mol.
ticity and exerts an antidepressant Essent. Fatty Acids 80, 8591. doi: epithelial-derived factor plus docosa- Med. 17, 11071118. doi: 10.2119/
effect: a versatile potential therapy for 10.1016/j.plefa.2009.01.004 hexaenoic acid after experimental molmed.2011.00119

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 11

fncel-07-00144 2013/9/6 13:17 page 11 #11


Hussain et al. Fatty acids and nervous system

Fick, L. J., Fick, G. H., and Belsham, D. Leukot. Essent. Fatty Acids 86, 211 hippocampus of the aged rat. Neu- with high endogenous omega-3 fatty
D. (2011). Palmitate alters the rhyth- 220. doi: 10.1016/j.plefa.2012.04.008 robiol. Aging 32, 2318.e115. doi: acids are protected from spinal cord
mic expression of molecular clock Hirakawa, A., Shimizu, K., Fukumitsu, 10.1016/j.neurobiolaging.2010.04.001 injury. Neurobiol. Dis. 51, 104112.
genes and orexigenic neuropeptide H., Soumiya, H., Iinuma, M., and Kenchegowda, S., He, J., and doi: 10.1016/j.nbd.2012.10.021
Y mRNA levels within immor- Furukawa, S. (2010). 2-Decenoic acid Bazan, H. E. P. (2013). Involve- Lim, S.-N., Huang, W., Hall, J. C.
talized, hypothalamic neurons. ethyl ester, a derivative of unsaturated ment of pigment epithelium-derived E., Michael-Titus, A. T., and Priest-
Biochem. Biophys. Res. Commun. 413, medium-chain fatty acids, facilitates factor, docosahexaenoic acid and ley, J. V. (2013b). Improved outcome
414419. doi: 10.1016/j.bbrc.2011. functional recovery of locomotor neuroprotectin D1 in corneal inam- after spinal cord compression injury
08.103 activity after spinal cord injury. mation and nerve integrity after in mice treated with docosahexaenoic
Figueroa, J. D., Cordero, K., Neuroscience 171, 13771385. doi: refractive surgery. Prostaglandins acid. Exp. Neurol. 239, 1327. doi:
Baldeosingh, K., Torrado, A. I., 10.1016/j.neuroscience.2010.10.004 Leukot. Essent. Fatty Acids 88, 2731. 10.1016/j.expneurol.2012.09.015
Walker, R. L., Miranda, J. D., et al. Hunt, W. T., Kamboj, A., Anderson, H. doi: 10.1016/j.plefa.2012.03.010 Lin, P.-Y., Huang, S.-Y., and Su, K.-
(2012). Docosahexaenoic acid pre- D., and Anderson, C. M. (2010). Pro- Kim, S.-J., Zhang, Z., Saha, A., Sarkar, P. (2010). A meta-analytic review of
treatment confers protection and tection of cortical neurons from exci- C., Zhao, Z., Xu, Y., et al. (2010). polyunsaturated fatty acid compo-
functional improvements after acute totoxicity by conjugated linoleic acid. Omega-3 and omega-6 fatty acids sitions in patients with depression.
spinal cord injury in adult rats. J. Neurochem. 115, 123130. doi: suppress ER- and oxidative stress in Biol. Psychiatry 68, 140147. doi:
J. Neurotrauma 29, 551566. doi: 10.1111/j.1471-4159.2010.06908.x cultured neurons and neuronal pro- 10.1016/j.biopsych.2010.03.018
10.1089/neu.2011.2141 Hussain, G., Schmitt, F., Henriques, A., genitor cells from mice lacking PPT1. Liu, R.-Z., Mita, R., Beaulieu, M.,
Fraser, T., Tayler, H., and Love, S. (2010). Lequeu, T., Rene, F., Bindler, F., et al. Neurosci. Lett. 479, 292296. doi: Gao, Z., and Godbout, R. (2010).
Fatty acid composition of frontal, (2013). Systemic down-regulation of 10.1016/j.neulet.2010.05.083 Fatty acid binding proteins in brain
temporal and parietal neocortex in delta-9 desaturase promotes muscle Labrousse, V. F., Nadjar, A., Joffre, development and disease. Int. J.
the normal human brain and in oxidative metabolism and accelerates C., Costes, L., Aubert, A., Grgoire, Dev. Biol. 54, 12291239. doi:
Alzheimers disease. Neurochem. Res. muscle function recovery following S., et al. (2012). Short-term long 10.1387/ijdb.092976rl
35, 503513. doi: 10.1007/s11064- nerve injury. PLoS ONE 8:e64525. chain omega3 diet protects from neu- Luchtman, D. W., Meng, Q., and Song,
009-0087-5 doi: 10.1371/journal.pone.0064525 roinammatory processes and mem- C. (2012). Ethyl-eicosapentaenoate
Georgiadi, A., and Kersten, S. (2012). Inceoglu, B., Wagner, K. M., Yang, ory impairment in aged mice. PLoS (E-EPA) attenuates motor impair-
Mechanisms of gene regulation by J., Bettaieb, A., Schebb, N. H., ONE 7:e36861. doi: 10.1371/jour- ments and inammation in the
fatty acids. Adv. Nutr. 3, 127134. doi: Hwang, S. H., et al. (2012). Acute nal.pone.0036861 MPTP-probenecid mouse model
10.3945/an.111.001602 augmentation of epoxygenated fatty Lafourcade, M., Larrieu, T., Mato, of Parkinsons disease. Behav.
Gladman, S. J., Huang, W., Lim, S.- acid levels rapidly reduces pain- S., Duffaud, A., Sepers, M., Brain Res. 226, 386396. doi:
N., Dyall, S. C., Boddy, S., Kang, related behavior in a rat model of Matias, I., et al. (2011). Nutri- 10.1016/j.bbr.2011.09.033
J. X., et al. (2012). Improved out- type I diabetes. Proc. Natl. Acad. tional omega-3 deciency abolishes Luchtman, D. W., and Song, C.
come after peripheral nerve injury in Sci. U.S.A. 109, 1139011395. doi: endocannabinoid-mediated neu- (2013). Cognitive enhancement by
mice with increased levels of endoge- 10.1073/pnas.1208708109 ronal functions. Nat. Neurosci. 14, omega-3 fatty acids from child-
nous -3 polyunsaturated fatty acids. IUPAC. (1997). Compendium of Chem- 345350. doi: 10.1038/nn.2736 hood to old age: ndings from
J. Neurosci. 32, 563571. doi: ical Terminology, 2nd Edn (the Lands, B. (2012). Consequences of animal and clinical studies. Neu-
10.1523/JNEUROSCI.3371-11.2012 Gold Book). Compiled by A. D. essential fatty acids. Nutrients 4, ropharmacology 64, 550565. doi:
Gupta, S., Knight, A. G., Gupta, S., McNaught and A. Wilkinson. Oxford: 13381357. doi: 10.3390/nu4091338 10.1016/j.neuropharm.2012.07.019
Keller, J. N., and Bruce-Keller, A. Blackwell Scientic Publications. Lebbadi, M., Julien, C., Phivilay, A., Ma, D., Zhang, M., Mori, Y., Yao,
J. (2012). Saturated long-chain fatty Ji, A., Diao, H., Wang, X., Yang, Tremblay, C., Emond, V., Kang, J. C., Larsen, C. P., Yamashima, T.,
acids activate inammatory signal- R., Zhang, J., Luo, W., et al. X., et al. (2011). Endogenous conver- et al. (2010). Cellular localization
ing in astrocytes. J. Neurochem. 120, (2012). N-3 Polyunsaturated fatty sion of omega-6 into omega-3 fatty of epidermal-type and brain-type
10601071. acids inhibit lipopolysaccharide- acids improves neuropathology in an fatty acid-binding proteins in adult
Hamazaki, K., Choi, K. H., and induced microglial activation animal model of Alzheimers disease. hippocampus and their response to
Kim, H.-Y. (2010). Phospholipid pro- and dopaminergic injury in rats. J. Alzheimers Dis. 27, 853869. doi: cerebral ischemia. Hippocampus 20,
le in the postmortem hippocam- Neurotoxicology 33, 780788. doi: 10.3233/JAD-2011-111010 811819. doi: 10.1002/hipo.20682
pus of patients with schizophrenia 10.1016/j.neuro.2012.02.018 Lee, J. C.-M., Simonyi, A., Sun, A. Y., Ma, Q.-L., Yang, F., Rosario, E. R.,
and bipolar disorder: no changes Katakura, M., Hashimoto, M., Okui, T., and Sun, G. Y. (2011). Phospholipases Ubeda, O. J., Beech, W., Gant,
in docosahexaenoic acid species. J. Shahdat, H. M., Matsuzaki, K., and A2 and neural membrane dynamics: D. J., et al. (2009). Beta-amyloid
Psychiatr. Res. 44, 688693. doi: Shido, O. (2013). Omega-3 polyun- implications for Alzheimers disease. oligomers induce phosphorylation of
10.1016/j.jpsychires.2009.11.017 saturated fatty acids enhance neu- J. Neurochem. 116, 813819. doi: tau and inactivation of insulin recep-
Hamazaki, K., Hamazaki, T., and ronal differentiation in cultured rat 10.1111/j.1471-4159.2010.07033.x tor substrate via c-Jun N-terminal
Inadera, H. (2012). Fatty acid com- neural stem cells. Stem Cells Int. 2013, Li, F., Marchette, L. D., Brush, R. S., kinase signaling: suppression by
position in the postmortem amygdala 490476. doi: 10.1155/2013/490476 Elliott, M. H., Davis, K. R., Ander- omega-3 fatty acids and curcumin.
of patients with schizophre- Kawashima, A., Harada, T., Kami, H., son, A. G., et al. (2010). High levels J. Neurosci. 29, 90789089. doi:
nia, bipolar disorder, and major Yano, T., Imada, K., and Mizuguchi, of retinal docosahexaenoic acid do 10.1523/JNEUROSCI.1071-09.2009
depressive disorder. J. Psychi- K. (2010). Effects of eicosapentaenoic not protect photoreceptor degenera- Maekawa, M., Takashima, N., Mat-
atr. Res. 46, 10241028. doi: acid on synaptic plasticity, fatty tion in VPP transgenic mice. Mol. Vis. sumata, M., Ikegami, S., Kontani,
10.1016/j.jpsychires.2012.04.012 acid prole and phosphoinositide 3- 16, 16691679. M., Hara, Y., et al. (2009). Arachi-
Harbeby, E., Jouin, M., Alessandri, J.- kinase signaling in rat hippocam- Li, F., Marchette, L. D., Brush, R. S., donic acid drives postnatal neuro-
M., Lallemand, M.-S., Linard, A., pus and differentiated PC12 cells. J. Elliott, M. H., Le, Y.-Z., Henry, K. genesis and elicits a benecial effect
Lavialle, M., et al. (2012). n-3 PUFA Nutr. Biochem. 21, 268277. doi: A., et al. (2009). DHA does not pro- on prepulse inhibition, a biological
status affects expression of genes 10.1016/j.jnutbio.2008.12.015 tect ELOVL4 transgenic mice from trait of psychiatric illnesses. PLoS
involved in neuroenergetics differ- Kelly, L., Grehan, B., Chiesa, A. D., retinal degeneration. Mol. Vis. 15, ONE 4:e5085. doi: 10.1371/jour-
ently in the fronto-parietal cortex OMara, S. M., Downer, E., Sahyoun, 11851193. nal.pone.0005085
compared to the CA1 area of the hip- G., et al. (2011). The polyunsat- Lim, S.-N., Gladman, S. J., Dyall, S. Martn, V., Fabelo, N., Santpere, G.,
pocampus: effect of rest and neuronal urated fatty acids, EPA and DPA C., Patel, U., Virani, N., Kang, J. Puig, B., Marn, R., Ferrer, I.,
activation in the rat. Prostaglandins exert a protective effect in the X., et al. (2013a). Transgenic mice et al. (2010). Lipid alterations in

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 12

fncel-07-00144 2013/9/6 13:17 page 12 #12


Hussain et al. Fatty acids and nervous system

lipid rafts from Alzheimers disease hippocampal ischemic insult in adult J. Neurosci. 31, 1507215085. doi: Sartorius, T., Ketterer, C., Kullmann, S.,
human brain cortex. J. Alzheimers rats. J. Nutr. Biochem. 21, 351356. 10.1523/JNEUROSCI.2443-11.2011 Balzer, M., Rotermund, C., Binder,
Dis. 19, 489502. doi: 10.3233/JAD- doi: 10.1016/j.jnutbio.2009.01.013 Park, Y., Moon, H.-J., and Kim, S.- S., et al. (2012). Monounsaturated
2010-1242 Motter, A. L., and Ahern, G. P. H. (2012). N-3 polyunsaturated fatty fatty acids prevent the aversive effects
Mathieu, G., Graldine, M., Denis, (2012). TRPA1 is a polyunsaturated acid consumption produces neu- of obesity on locomotion, brain
S., Stphanie, D., Langelier, fatty acid sensor in mammals. PLoS robiological effects associated with activity, and sleep behavior. Dia-
B., Bndicte, L., et al. (2010). ONE 7:e38439. doi: 10.1371/jour- prevention of depression in rats betes 61, 16691679. doi: 10.2337/
DHA enhances the noradrenaline nal.pone.0038439 after the forced swimming test. J. db11-1521
release by SH-SY5Y cells. Neu- Muntan, G., Janu, A., Fernandez, N., Nutr. Biochem. 23, 924928. doi: Schaeffer, E. L., Skaf, H. D., Novaes
rochem. Int. 56, 94100. doi: Odena, M. A., Oliveira, E., Boluda, S., 10.1016/j.jnutbio.2011.04.018 Bde, A., Da Silva, E. R., Martins,
10.1016/j.neuint.2009.09.006 et al. (2010). Modication of brain Park, Y., Nam, S., Yi, H.-J., B. A., Joaquim, H. D., et al. (2011).
Mayer, C. M., and Belsham, D. D. lipids but not phenotype in alpha- Hong, H.-J., and Lee, M. (2009). Inhibition of phospholipase A2 in
(2010). Palmitate attenuates insulin synucleinopathy transgenic mice by Dietary n-3 polyunsaturated fatty rat brain modies different mem-
signaling and induces endoplasmic long-term dietary n-3 fatty acids. acids increase oxidative stress in brane uidity parameters in opposite
reticulum stress and apoptosis in Neurochem. Int. 56, 318328. doi: rats with intracerebral hemorrhagic ways. Prog. Neuropsychopharmacol.
hypothalamic neurons: rescue of 10.1016/j.neuint.2009.10.015 stroke. Nutr. Res. 29, 812818. doi: Biol. Psychiatry 35, 16121617. doi:
resistance and apoptosis through Musto, A. E., Gjorstrup, P., and 10.1016/j.nutres.2009.10.019 10.1016/j.pnpbp.2011.05.001
adenosine 5 monophosphate- Bazan, N. G. (2011). The omega- Peters, B. D., Duran, M., Vlieger, E. Schleicher, M., Weikel, K., Garber,
activated protein kinase activation. 3 fatty acid-derived neuroprotectin J., Majoie, C. B., Den Heeten, G. C., and Taylor, A. (2013). Dimin-
Endocrinology 151, 576585. doi: D1 limits hippocampal hyperex- J., Linszen, D. H., et al. (2009). ishing risk for age-related macular
10.1210/en.2009-1122 citability and seizure susceptibility Polyunsaturated fatty acids and brain degeneration with nutrition: a cur-
McNamara, R. K., and Liu, Y. in kindling epileptogenesis. Epilepsia white matter anisotropy in recent- rent view. Nutrients 5, 24052456.
(2011). Reduced expression of 52, 16011608. doi: 10.1111/j.1528- onset schizophrenia: a prelimi- doi: 10.3390/nu5072405
fatty acid biosynthesis genes in 1167.2011.03081.x nary study. Prostaglandins Leukot. Shchepinov, M. S., Chou, V. P., Pol-
the prefrontal cortex of patients Nakamura, M. T., and Nara, T. Y. Essent. Fatty Acids 81, 6163. doi: lock, E., Langston, J. W., Cantor,
with major depressive disorder. J. (2004). Structure, function, and 10.1016/j.plefa.2009.04.007 C. R., Molinari, R. J., et al. (2011).
Affect. Disord. 129, 359363. doi: dietary regulation of delta6, delta5, Porta, N., Bourgois, B., Galabert, Isotopic reinforcement of essential
10.1016/j.jad.2010.08.021 and delta9 desaturases. Annu. Rev. C., Lecointe, C., Cappy, P., Bor- polyunsaturated fatty acids dimin-
Mnard, C., Patenaude, C., Gagn, Nutr. 24, 345376. doi: 10.1146/ det, R., et al. (2009). Anticonvul- ishes nigrostriatal degeneration in
A.-M., and Massicotte, G. (2009). annurev.nutr.24.121803.063211 sant effects of linolenic acid are a mouse model of Parkinsons dis-
AMPA receptor-mediated cell death Niculescu, M. D., Lupu, D. S., and unrelated to brain phospholipid cell ease. Toxicol. Lett. 207, 97103. doi:
is reduced by docosahexaenoic acid Craciunescu, C. N. (2011). Mater- membrane compositions. Epilepsia 10.1016/j.toxlet.2011.07.020
but not by eicosapentaenoic acid in nal -linolenic acid availability dur- 50, 6571. doi: 10.1111/j.1528- Sidhu, V. K., Huang, B. X., and Kim, H.
area CA1 of hippocampal slice cul- ing gestation and lactation alters 1167.2008.01723.x Y. (2011). Effects of docosahexaenoic
tures. J. Neurosci. Res. 87, 876886. the postnatal hippocampal develop- Prior, P. L., and Galdurz, J. C. (2012). acid on mouse brain synaptic plasma
doi: 10.1002/jnr.21916 ment in the mouse offspring. Int. (N-3) Fatty acids: molecular role membrane proteome analyzed by
Meng, Q., Luchtman, D. W., El Bahh, B., J. Dev. Neurosci. 29, 795802. doi: and clinical uses in psychiatric dis- mass spectrometry and (16)O/(18)O
Zidichouski, J. A., Yang, J., and Song, 10.1016/j.ijdevneu.2011.09.006 orders. Adv. Nutr. 3, 257265. doi: labeling. J. Proteome Res. 10, 5472
C. (2010). Ethyl-eicosapentaenoate Ochoa, J. J., Pamplona, R., Ramirez- 10.3945/an.111.001693 5480. doi: 10.1021/pr2007285
modulates changes in neurochem- Tortosa, M. C., Granados-Principal, Rashid, M. A., Katakura, M., Kharebava, Simopoulos, A. P. (2011). Evolutionary
istry and brain lipids induced S., Perez-Lopez, P., Naud, A., et al. G., Kevala, K., and Kim, H. Y. (2013). aspects of diet: the omega-6/omega-3
by parkinsonian neurotoxin 1- (2011). Age-related changes in N -Docosahexaenoylethanolamine is ratio and the brain. Mol. Neurobiol.
methyl-4-phenylpyridinium in brain mitochondrial DNA deletion a potent neurogenic factor for neu- 44, 203215. doi: 10.1007/s12035-
mouse brain slices. Eur. J. and oxidative stress are differen- ral stem cell differentiation. J. 010-8162-0
Pharmacol. 649, 127134. doi: tially modulated by dietary fat Neurochem. 125, 869884. doi: Taha, A. Y., Filo, E., Ma, D. W. L.,
10.1016/j.ejphar.2010.09.046 type and coenzyme Q1 . Free Radic. 10.1111/jnc.12255 and McIntyre Burnham, W. (2009).
Milanski, M., Degasperi, G., Coope, Biol. Med. 50, 10531064. doi: Robson, L. G., Dyall, S., Sidloff, D., Dose-dependent anticonvulsant
A., Morari, J., Denis, R., Cin- 10.1016/j.freeradbiomed.2011.02.004 and Michael-Titus, A. T. (2010). effects of linoleic and alpha-linolenic
tra, D. E., et al. (2009). Saturated Palacios-Pelaez, R., Lukiw, W. J., and Omega-3 polyunsaturated fatty acids polyunsaturated fatty acids on
fatty acids produce an inam- Bazan, N. G. (2010). Omega-3 essen- increase the neurite outgrowth of pentylenetetrazol induced seizures
matory response predominantly tial fatty acids modulate initiation rat sensory neurones throughout in rats. Epilepsia 50, 7282. doi:
through the activation of TLR4 and progression of neurodegenera- development and in aged animals. 10.1111/j.1528-1167.2008.01731.x
signaling in hypothalamus: impli- tive disease. Mol. Neurobiol. 41, 367 Neurobiol. Aging 31, 678687. doi: Tanito, M., Brush, R. S., Elliott, M.
cations for the pathogenesis of 374. doi: 10.1007/s12035-010-8139-z 10.1016/j.neurobiolaging.2008.05.027 H., Wicker, L. D., Henry, K. R.,
obesity. J. Neurosci. 29, 359370. doi: Pan, J.-P., Zhang, H.-Q., Wei-Wang, Rombaldi Bernardi, J., De Souza Esco- and Anderson, R. E. (2009). High
10.1523/JNEUROSCI.2760-08.2009 Guo, Y.-F., Na-Xiao, Cao, X.-H., et al. bar, R., Ferreira, C. F., and Pelufo levels of retinal membrane docosa-
Moreira, J. D., Knorr, L., Ganzella, M., (2011). Some subtypes of endo- Silveira, P. (2012). Fetal and neonatal hexaenoic acid increase susceptibil-
Thomazi, A. P., De Souza, C. G., De cannabinoid/endovanilloid receptors levels of omega-3: effects on neurode- ity to stress-induced degeneration.
Souza, D. G., et al. (2010a). Omega- mediate docosahexaenoic acid- velopment, nutrition, and growth. J. Lipid Res. 50, 807819. doi:
3 fatty acids deprivation affects induced enhanced spatial memory Sci. World J. 2012, 202473. doi: 10.1194/jlr.M800170-JLR200
ontogeny of glutamatergic synapses in rats. Brain Res. 1412, 1827. doi: 10.1100/2012/202473 Tian, C., Fan, C., Liu, X., Xu, F.,
in rats: relevance for behavior alter- 10.1016/j.brainres.2011.07.015 Ruzickova, J., Rossmeisl, M., Prazak, and Qi, K. (2011). Brain histologi-
ations. Neurochem. Int. 56, 753759. Park, C.-K., L, N., Xu, Z.-Z., T., Flachs, P., Sponarova, J., Veck, cal changes in young mice submit-
doi: 10.1016/j.neuint.2010.02.010 Liu, T., Serhan, C. N., and Ji, M., et al. (2004). Omega-3 PUFA of ted to diets with different ratios of
Moreira, J. D., Knorr, L., Thomazi, A. R.-R. (2011). Resolving TRPV1- marine origin limit diet-induced obe- n-6/n-3 polyunsaturated fatty acids
P., Simo, F., Batt, C., Oses, J. P., and TNF--mediated spinal cord sity in mice by reducing cellularity of during maternal pregnancy and lac-
et al. (2010b). Dietary omega-3 fatty synaptic plasticity and inamma- adipose tissue. Lipids 39, 11771185. tation. Clin. Nutr. 30, 659667. doi:
acids attenuate cellular damage after a tory pain with neuroprotectin D1. doi: 10.1007/s11745-004-1345-9 10.1016/j.clnu.2011.03.002

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 13

fncel-07-00144 2013/9/6 13:17 page 13 #13


Hussain et al. Fatty acids and nervous system

Torkildsen, ., Brunborg, L. A., Willis, S., Samala, R., Rosenberger, T. Yee, P., Weymouth, A. E., Fletcher, E. L., rats. Brain Res. 1457, 3343. doi:
Milde, A. M., Mrk, S. J., Myhr, A., and Borges, K. (2009). Eicosapen- and Vingrys, A. J. (2010). A role for 10.1016/j.brainres.2012.03.067
K.-M., and B, L. (2009). A taenoic and docosahexaenoic acids omega-3 polyunsaturated fatty acid Zhou, Y., Zhang, Y., Li, J., Lv, F.,
salmon based diet protects mice from are not anticonvulsant or neuro- supplements in diabetic neuropathy. Zhao, Y., Duan, D., et al. (2012). A
behavioural changes in the cuprizone protective in acute mouse seizure Invest. Ophthalmol. Vis. Sci. 51, 1755 comprehensive study on long-term
model for demyelination. Clin. Nutr. models. Epilepsia 50, 138142. doi: 1764. doi: 10.1167/iovs.09-3792 injury to nigral dopaminergic neu-
28, 8387. doi: 10.1016/j.clnu.2008. 10.1111/j.1528-1167.2008.01722.x Yip, P. K., Pizzasegola, C., Gladman, S., rons following intracerebroventricu-
10.015 Wu, A, Ying, Z., and Gomez-Pinilla, Biggio, M. L., Marino, M., Jayasinghe, lar injection of lipopolysaccharide in
Uauy, R., and Dangour, A. D. (2006). F. (2008). Docosahexaenoic acid M., et al. (2013). The omega-3 fatty rats. J. Neurochem. 123, 771780. doi:
Nutrition in brain development and dietary supplementation enhances acid eicosapentaenoic acid acceler- 10.1111/jnc.12010
aging: role of essential fatty acids. the effects of exercise on synap- ates disease progression in a model
Nutr. Rev. 64, S24S33. doi: tic plasticity and cognition. Neuro- of amyotrophic lateral sclerosis. PLoS Conflict of Interest Statement: The
10.1301/nr.2006.may.S24-S33 science 155, 751759. doi: 10.1016/j. ONE 8:e61626. doi: 10.1371/jour- authors declare that the research was
Venna, V. R., Deplanque, D., Allet, neuroscience.2008.05.061 nal.pone.0061626 conducted in the absence of any com-
C., Belarbi, K., Hamdane, M., and Wu, A., Ying, Z., and Gomez-Pinilla, F. Yu, H., Bi, Y., Ma, W., He, L., Yuan, mercial or nancial relationships that
Bordet, R. (2009). PUFA induce (2011). The salutary effects of DHA L., Feng, J., et al. (2010). Long- could be construed as a potential con-
antidepressant-like effects in parallel dietary supplementation on cogni- term effects of high lipid and high ict of interest.
to structural and molecular changes tion, neuroplasticity, and membrane energy diet on serum lipid, brain
in the hippocampus. Psychoneu- homeostasis after brain trauma. J. fatty acid composition, and mem- Received: 27 May 2013; paper pend-
roendocrinology 34, 199211. doi: Neurotrauma 28, 21132122. doi: ory and learning ability in mice. Int. ing published: 23 June 2013; accepted:
10.1016/j.psyneuen.2008.08.025 10.1089/neu.2011.1872 J. Dev. Neurosci. 28, 271276. doi: 19 August 2013; published online: 09
Vines, A., Delattre, A. M., Lima, M. Yakunin, E., Loeb, V., Kisos, H., 10.1016/j.ijdevneu.2009.12.001 September 2013.
M. S., Rodrigues, L. S., Suchecki, D., Biala, Y., Yehuda, S., Yaari, Y., et al. Zhang, C., and Bazan, N. G. (2010). Citation: Hussain G, Schmitt F, Loef-
Machado, R. B., et al. (2012). The (2012). A-synuclein neuropathology Lipid-mediated cell signaling pro- er J-P and Gonzalez de Aguilar J-L
role of 5-HT1 A receptors in sh oil- is controlled by nuclear hormone tects against injury and neurodegen- (2013) Fatting the brain: a brief of recent
mediated increased BDNF expression receptors and enhanced by docosa- eration. J. Nutr. 140, 858863. doi: research. Front. Cell. Neurosci. 7:144.
in the rat hippocampus and cortex: hexaenoic acid in a mouse model 10.3945/jn.109.114884 doi: 10.3389/fncel.2013.00144
a possible antidepressant mechanism. for Parkinsons disease. Brain Pathol. Zhang, W., Hu, X., Yang, W., Gao, This article was submitted to the journal
Neuropharmacology 62, 184191. doi: 22, 280294. doi: 10.1111/j.1750- Y., and Chen, J. (2010). Omega- Frontiers in Cellular Neuroscience.
10.1016/j.neuropharm.2011.06.017 3639.2011.00530.x 3 polyunsaturated fatty acid Copyright 2013 Hussain, Schmitt,
Wang, Z., Liu, D., Wang, J., Liu, S., Gao, Yamashima, T. (2012). PUFA-GPR40- supplementation confers long- Loefer and Gonzalez de Aguilar. This is
M., Ling, E.-A., et al. (2012). Cyto- CREB signaling hypothesis for the term neuroprotection against an open-access article distributed under
protective effects of melatonin on adult primate neurogenesis. Prog. neonatal hypoxic-ischemic brain the terms of the Creative Commons
astroglial cells subjected to palmitic Lipid Res. 51, 221231. doi: injury through anti-inammatory Attribution License (CC BY). The use,
acid treatment in vitro. J. Pineal Res. 10.1016/j.plipres.2012.02.001 actions. Stroke 41, 23412347. doi: distribution or reproduction in other
52, 253264. doi: 10.1111/j.1600- Yang, H., Zhuo, J.-M., Chu, J., 10.1161/STROKEAHA.110.586081 forums is permitted, provided the origi-
079X.2011.00952.x Chinnici, C., and Pratic, D. Zhao, C.-H., Liu, H.-Q., Cao, R., nal author(s) or licensor are credited and
Wergeland, S., Torkildsen, ., B, L., (2010). Amelioration of the Ji, A.-L., Zhang, L., Wang, F., that the original publication in this jour-
and Myhr, K.-M. (2012). Polyunsatu- Alzheimers disease phenotype by et al. (2012). Effects of dietary nal is cited, in accordance with accepted
rated fatty acids in multiple sclerosis absence of 12/15-lipoxygenase. sh oil on learning function and academic practice. No use, distribution or
therapy. Acta Neurol. Scand. Suppl. Biol. Psychiatry 68, 922929. doi: apoptosis of hippocampal pyramidal reproduction is permitted which does not
126, 7075. doi: 10.1111/ane.12034 10.1016/j.biopsych.2010.04.010 neurons in streptozotocin-diabetic comply with these terms.

Frontiers in Cellular Neuroscience www.frontiersin.org September 2013 | Volume 7 | Article 144 | 14

fncel-07-00144 2013/9/6 13:17 page 14 #14

Vous aimerez peut-être aussi