Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Feb 9.
Published in final edited form as: Brain Res. 2014 Dec 8;0:220–246. doi: 10.1016/j.brainres.2014.11.059

Pathways of Polyunsaturated Fatty Acid Utilization: Implications for Brain Function in Neuropsychiatric Health and Disease

Joanne J Liu 1,2, Pnina Green 3, J John Mann 1,4,5, Stanley I Rapoport 6, M Elizabeth Sublette 1,4,*
PMCID: PMC4339314  NIHMSID: NIHMS647443  PMID: 25498862

Abstract

Essential polyunsaturated fatty acids (PUFAs) have profound effects on brain development and function. Abnormalities of PUFA status have been implicated in neuropsychiatric diseases such as major depression, bipolar disorder, schizophrenia, Alzheimer’s disease, and attention deficit hyperactivity disorder. Pathophysiologic mechanisms could involve not only suboptimal PUFA intake, but also metabolic and genetic abnormalities, defective hepatic metabolism, and problems with diffusion and transport. This article provides an overview of physiologic factors regulating PUFA utilization, highlighting their relevance to neuropsychiatric disease.

Keywords: Polyunsaturated fatty acids, lipid metabolism, brain, depression, bipolar disorder, schizophrenia

1. INTRODUCTION

Lipids, which compose cell membranes, occur in very high concentrations throughout the central nervous system (Jumpsen, 1995), making up about half the dry weight of the human brain (Benatti et al., 2004). Of brain lipids, approximately 35% are polyunsaturated fatty acids (PUFAs), which cannot be synthesized de novo from 2-carbon fragments and are nutritionally essential (Benatti et al., 2004). PUFAs are critical for normal brain development and functioning (Luchtman and Song, 2013). They fall into three major categories, according to the number of carbon atoms separating the first double bond of the carbon chain from the terminal methyl (omega) end: n-3, n-6, and n-9. This review will focus primarily on three highly unsaturated, long-chain PUFAs (LC-PUFAs) that appear relevant to neuropsychiatry: arachidonic acid (AA, 20:4n-6) and docosahexaenoic acid (DHA, 22:6n-3), which make up 50% and 40% of brain PUFAs, respectively (Gerster, 1998; Lauritzen et al., 2001; Singh, 2005; Spector, 1999); and eicosapentaenoic acid (EPA, 20:5n-3) (Fernstrom, 1999; McNamara and Strawn, 2013; Youdim et al., 2000; Young and Conquer, 2005), which is maintained at a much lower brain concentration (Chen et al., 2009; Chen et al., 2011) through very rapid breakdown by β-oxidation (Chen and Bazinet, 2014). Also relevant are n-6 and n-3 isomers of docosapentaenoic acid (DPA, 22:5), both of which may serve as a partial substitute for DHA in states of experimental n-3 PUFA deficiency (Kaur et al., 2010; Kaur et al., 2011; Lim et al., 2005b); and linoleic acid (LA, 18:2n-6) and alpha-linolenic acid (ALA,18:3n-3), the shorter-chain nutritionally essential precursors of longer-chain n-6 and n-3 PUFAs, respectively.

Lipidomics in neuropsychiatric illness is an emerging field (Wenk, 2005). With respect to LC-PUFA, both AA and DHA are critical to brain development and maintenance of brain structure and function. DHA is important for the health of developing neurons and for neurotransmission, with clear ramifications for cognition and behavior (Innis, 2007). Neurological health also requires sufficient levels of AA for the growth (Darios and Davletov, 2006), repair (Darios and Davletov, 2006), maintenance (Fukaya et al., 2007), and protection (Wang et al., 2006) of neurons. The role of EPA in brain functioning is less clear, but it has been found to be an important component in PUFA supplements for treatment of depression (Lin et al., 2012; Martins, 2009; Martins et al., 2012; Sublette et al., 2011), and EPA status has been specifically associated with certain aspects of substance use disorders (Beier et al., 2014; Buydens-Branchey et al., 2011). Dysregulated lipid metabolism and low dietary consumption of n-3 PUFAs have been implicated in neuropsychiatric diseases, encompassing specific domains including development (Daniels et al., 2004; Gustafsson et al., 2004; Helland et al., 2003; Jorgensen et al., 2001; Larque et al., 2002; Milte et al., 2011; Veena et al., 2010), neurodegeneration (Conquer et al., 2000; Morris et al., 2003; Schaefer et al., 2006), cognition (Barberger-Gateau et al., 2007; Beydoun et al., 2007; Devore et al., 2009; Dullemeijer et al., 2007; Eskelinen et al., 2008; Heude et al., 2003; Huang et al., 2005; Kalmijn et al., 1997a; Kalmijn et al., 1997b; Kalmijn et al., 2004; Lopez et al., 2011; Morris et al., 2003; Morris et al., 2005; Schaefer et al., 2006; Tan et al., 2012; van Gelder et al., 2007), mood (Adams et al., 1996; Amin et al., 2008; Bountziouka et al., 2009; Conklin et al., 2007; Edwards et al., 1998; Frasure-Smith et al., 2004; Green et al., 2006; Hibbeln, 1998; Hibbeln, 2002a; Lin et al., 2010; Maes et al., 1999b; McNamara et al., 2010; Peet et al., 1998; Raeder et al., 2007; Riemer et al., 2010; Sanchez-Villegas et al., 2007; Schins et al., 2007; Tanskanen et al., 2001), psychosis (Amminger et al., 2010; Evans et al., 2003a; Kaddurah-Daouk et al., 2007), suicidal behaviors (Huan et al., 2004; Lewis et al., 2011; Sublette et al., 2006), and neuroinflammation (Calder, 2006b; Kiecolt-Glaser et al., 2007).

The goal of this article is to review and integrate the scientific literature concerning aspects of PUFA physiology and metabolism that may influence neuropsychiatric diseases.

2. POLYUNSATURATED FATTY ACIDS

2.1. Sources of PUFAs

The average daily intake of total lipids by American adults is about 85g, comprising 33% of total energy intake (U.S. Department of Agriculture: Agricultural Research Service, 2008). Most lipids found in foods are in the form of triacylglycerols, while the remainder include cholesterol, cholesteryl esters, phospholipids, and nonesterified (free) fatty acids (Brown, 1991). N-3 and n-6 PUFAs are essential nutrients because they cannot be synthesized by vertebrates de novo; and the ALA and LA precursors, which can be converted to LC-PUFA in the liver and to some extent in other organs, must be obtained through direct dietary consumption (Burr, 1930).

LA, the parent compound of the n-6 family, and AA, the major brain n-6 species, are plentiful in nature. LA is found in the seeds of most plants such as soybean, canola, corn, sunflower, safflower, and cottonseed (some exceptions being coconut, cocoa and palm), whereas AA is found in most meats and products from animals fed corn-based diets high in n-6 PUFAs (Meyer et al., 2003; Simopoulos, 1999). ALA, the precursor for the n-3 PUFA family, is found in green leafy vegetables, walnuts, and the seeds of flax, rape, and chia (Sinclair et al., 2002). Seafood is currently the major source of EPA and DHA in most industrialized countries (Simopoulos, 2011), although significant amounts can be obtained through ingestion of protein from animals fed diets high in n-3 PUFAs, including eggs from n-3 PUFA-fed chickens (Trebunova et al., 2007) and beef from range-fed cattle (Howe et al., 2006; Ollis et al., 1999). According to Simopoulos (Simopoulos, 2011), changes in practices by the food industry have led to dramatic dietary imbalances, shifting from an approximately equal intake of n-6 to n-3 PUFAs 150 years ago to current ratios of about 20 to 1, a change that may contribute to modern health problems. Ironically, to some extent, this shift in PUFA balance also may be due to earlier efforts to improve public health through lowering cholesterol levels by replacing saturated fat intake with polyunsaturated fats, without an adequate understanding of the relative risks and benefits of altering the n-6 to n-3 PUFA balance, including the risks to mental health. For example, meta-analyses of mortality rates in studies of dietary interventions and in medication trials to lower cholesterol found no reduction of mortality rates, but rather increased mortality due to violent death such as suicide and accidents (Muldoon et al., 1990; Muldoon et al., 2001). A recent study suggests that elevated n-6 to n-3 PUFA ratios may be remedied by altering the diet with therapeutic benefit in brain disease (Ramsden et al., 2013).

At the present time, no official Dietary Reference Intake (DRI) criteria have been determined for LC-PUFAs in the US and Canada, as there are no agreed-upon normal ranges or clinical functional endpoints for DHA or EPA (Panel on Macronutrients, 2002). However, it is clear that individuals living in the U.S. and many other countries whose diet is importantly derived from reared animals have a very low dietary n-3 PUFA intake compared with other, more seafood-oriented cultures. In a study showing that low DHA was associated with higher suicide risk among US military personnel, Lewis et al. (Lewis et al., 2011) note that almost the entire US military study population had lower DHA levels than the lowest group in a Chinese case-control study of suicide attempters (Huan et al., 2004). Cross-national comparisons find that lower intakes of seafood, by country, are associated with higher rates of bipolar disorders (Noaghiul and Hibbeln, 2003) and post-partum depression (Hibbeln, 2002b).

2.2. Long-chain PUFA structure and synthesis

The metabolic fate of an ingested fatty acid is influenced by the lipid in which it is esterified, and by its structural characteristics, which include carbon chain length, number of double bonds, placement of double bonds relative to the omega carbon, and configuration of hydrogen atoms around the double bonds (cis or trans).

The number of double bonds influences fatty acid flexibility at double bond locations (Gawrisch et al., 2003). This has implications for cell membrane lipid matrix properties, formerly referred to as ‘fluidity’ (Shinitzky and Barenholz, 1978), but more recently described in terms of ‘lipid rafts’ or ‘membrane microdomains’, dynamic membrane aggregations of specific lipid composition that promote interactions between membrane-bound proteins by bringing them into physical proximity (Pike, 2006; Yaqoob and Shaikh, 2010). The highly flexible n-3 PUFAs incorporate directly into membranes and thereby promote the partitioning of the more rigid cholesterol and sphingolipid moleclules into lipid rafts (Altenburg and Siddiqui, 2009; Duraisamy et al., 2007; Eldho et al., 2003; Grimm et al., 2011; Huster et al., 1998; Langelier et al., 2010; Rockett et al., 2012; Rogers et al., 2010; Schley et al., 2007; Shaikh et al., 2002; Shaikh et al., 2003; Shaikh et al., 2004; Soni et al., 2008; Stillwell et al., 2005a; Stillwell et al., 2005b; Wassall et al., 2004; Wong et al., 2009b; Ye et al., 2010).

The nutritionally essential shorter chain PUFAs LA and ALA undergo a series of desaturation and elongation steps (Mayes, 1996) in the endoplasmic reticulum and peroxisomes of the liver to form n-6 (AA) and n-3 (EPA and DHA) LC-PUFAs, respectively (Figure 1), catalyzed by enzymes that are upregulated in response to experimental deficiency of n-3 PUFAs (Rao et al., 2007b). The conversion process normally occurs with low efficiency (de Gomez Dumm and Brenner, 1975; Emken, 1989b) at rates of 0.2 to 21% (Burdge et al., 2002; Burdge and Wootton, 2002; Emken et al., 1994). However, in rats fed LC-PUFA-deficient diets, net conversion of LA to AA and ALA to DHA increases; even in severe deficiency, the amount of DHA produced greatly exceeds brain PUFA consumption (Domenichiello et al., 2014; Igarashi et al., 2006; Igarashi et al., 2007c; Rapoport and Igarashi, 2009). The applicability of these preclinical studies to humans is uncertain, but they suggest that in populations with low long-chain n-3 PUFA intake, such as vegans or vegetarians, the liver likely is capable of compensating and producing adequate DHA for brain function (Igarashi et al., 2007b; Igarashi et al., 2007c).

Figure 1. Long-chain polyunsaturated fatty acid (PUFA) synthesis from parent essential fatty acids.

Figure 1

Long-chain (LC)-PUFAs not acquired directly through diet can be synthesized through desaturation, elongation, and β-oxidation steps (solid arrows) from shorter-chain precursors. N-6 and n-3 PUFAs compete for Δ5 and Δ6 desaturases. Small amounts of apparent retroconversion from DHA to DPA and EPA also have been reported (Plourde et al., 2011) (dashed arrows).

Several polymorphisms of the fatty acid desaturase gene cluster (FADS1-FADS2) (Cho et al., 1999a; Cho et al., 1999b) can affect production of LC-PUFAs from ALA and LA (Al-Hilal et al., 2013; Gillingham et al., 2013; Harslof et al., 2013; Koletzko et al., 2008; Malerba et al., 2008; Nwankwo et al., 2003; Rizzi et al., 2013; Schaeffer et al., 2006b; Solakivi et al., 2013), and thereby influence plasma concentrations of not only fatty acids, but also low density lipoprotein (LDL) and cholesterol (Tanaka et al., 2009). Postmortem studies have reported altered expression of genes regulating fatty acid biosynthesis in the prefrontal cortex of patients with schizophrenia (Liu et al., 2009a), major depressive disorder (McNamara and Liu, 2011), bipolar disorder (Liu and McNamara, 2011b), and depressed suicides compared with nonpsychiatric controls (Lalovic et al., 2010). Additionally, antipsychotic medications have been shown to upregulate mRNA expression of genes coding for biosynthetic enzymes in rats (Liu et al., 2009a; McNamara et al., 2011) and red blood cell PUFA levels in patients with psychosis (Evans et al., 2003a; Kaddurah-Daouk et al., 2007). FADS2 expression is elevated in prefrontal cortex in schizophrenia, independently of sex or medication usage (Liu et al., 2009b). Decreased conversion of EPA to DHA has also been observed in schizophrenia (Mahadik et al., 1996).

2.3. The balance of n-3 and n-6 PUFAs

The n-3 and n-6 PUFA families often have opposing physiological functions, and therefore their relative proportions have implications for physiology and pathophysiology. In mammals, LA and ALA compete with each other for enzymes involved in the elongation – desaturation process (de Gomez Dumm and Brenner, 1975; Emken, 1989a; Hague, 1984a). Although the desaturation and elongation cascade is more selective for n-3 than n-6 PUFAs, due in part to higher affinities of Δ5 and Δ6 desaturases for n-3 PUFAs (de Gomez Dumm and Brenner, 1975; Hague, 1984b; Hague, 1986), high LA intake can interfere with the desaturation and elongation of ALA (Emken, 1989b; Indu, 1992) and slows hepatic synthesis of DHA (Smink et al., 2012), whereas trans fatty acids interfere with the desaturation and elongation of both LA and ALA (Simopoulos, 1994). In addition to dietary effects on the n-6 to n-3 PUFA balance, some physiologic states can influence long-chain n-3 PUFA production, including aging (de Gomez Dumm and Brenner, 1975), infant prematurity (Carlson et al., 1986), hypertension (Singer et al., 1984) and diabetes (Honigmann, 1982).

Conversely, enhanced PUFA catabolism could also contribute to PUFA imbalances. For instance, children with ADHD exhibit abnormal plasma fatty acid profiles (Colter et al., 2008) despite a dietary intake of LC-PUFAs equivalent to that of healthy children (Chen et al., 2004; Colter et al., 2008; Ng et al., 2009; Stevens et al., 1995). This difference could be explained by more rapid PUFA breakdown, because children with ADHD exhale higher levels of ethane, a non-invasive measure of oxidative damage to n-3 PUFAs (Ross et al., 2010). Increased catabolism of n-3 PUFAs has also been implicated in the pathology of schizophrenia (Evans et al., 2003b; Horrobin et al., 1994; Peet et al., 1995).

2.4. PUFA distribution

In mammals, the n-3 PUFAs are distributed selectively among lipid classes (Simopoulos, 2008). Both ALA and EPA are found in triacylglycerols and cholesteryl esters. However, in phospholipids, ALA is minimally present, whereas DHA and EPA are both prominent components. In body organs, DHA is the most abundant n-3 PUFA, especially in brain and retina, where DHA is several hundred-fold more abundant than EPA (Arterburn et al., 2006). EPA is maintained at much lower brain concentrations (Chen et al., 2009; Chen et al., 2011) through rapid β-oxidation (Chen and Bazinet, 2014).

2.5. Overview of PUFA functions

PUFAs serve multiple important physiologic roles, falling within major categories that include energy storage and production, inflammation, and cell signaling, with effects on multiple organ systems. PUFAs impact energy by regulating lipid catabolism and lipogenesis (Kopecky et al., 2009), and PUFA metabolites serve as lipid mediators in endocrine, paracrine, and autocrine signaling within adipose tissue (Masoodi et al., 2014). Another mechanism of PUFA effects on cell signaling is the modulation of ion channel functioning, including sodium conductance in human and rodent ion channels in skeletal muscle (Wieland et al., 1992; Xiao et al., 1997) and calcium conductance in cardiomyocytes (Akhtar Khan, 2010; Chapkin et al., 2009; Ferrante et al., 1994; Kim et al., 2010; Reynolds and Roche, 2010; Szentandrassy et al., 2007; Triboulot et al., 2001; Yog et al., 2010) and T-cells (Triboulot et al., 2001; Yog et al., 2010)

Many effects of PUFAs can be attributed to their ability to regulate gene expression by affecting transcription factors (Jump, 2002; Jump et al., 2008a). Hepatic gene transcription factors affected by n-3 PUFAs include peroxisome proliferatoractivated receptor alpha, PPAR-α; sterol regulatory element-binding protein 1, SREBP-1; carbohydrate-responsive element-binding protein, ChREBP; and max-like protein X, MLX. These factors contribute to control of proteins involved in lipid synthesis and oxidation, and lipoprotein secretion (Jump et al., 1994; Jump et al., 2008b; Ntambi and Bene, 2001; Sessler and Ntambi, 1998). In cultured rat cardiomyocytes, supplementation with n-3 PUFAs causes up- and downregulation of genes related to lipid metabolism, inflammation, cell survival, cell proliferation, and cardiac contractility (Bordoni et al., 2007).

A deeper explanation for n-3 PUFAs’ widespread effects is their ability to modulate membrane functioning at a subcellular level through influencing lipid rafts (Brown and London, 2000; Chapkin et al., 2008; Fan et al., 2003; Fan et al., 2004; Kim et al., 2008a; Ma et al., 2004; Pike, 2003; Smart et al., 1999). For example, DHA has robust effects on structural and dynamic characteristics of membrane domains (Huster et al., 1998; Shaikh et al., 2002; Shaikh et al., 2003; Shaikh et al., 2004; Soni et al., 2008; Stillwell et al., 2005b; Turk and Chapkin, 2012; Wassall et al., 2004; Williams et al., 2012) such as domain size and membrane order, likely due to lateral segregation of DHA-rich membrane domains from cholesterol-rich membrane regions. The physiologic implications for neuropsychiatry are profound, since such microdomains participate in regulation of serotonin (Magnani et al., 2004; Samuvel et al., 2005), dopamine (Adkins et al., 2007; Jones et al., 2012), and norepinephrine (Jayanthi et al., 2004; Matthies et al., 2009) transporters; serotonin 1A (Kalipatnapu and Chattopadhyay, 2005; Kobe et al., 2008; Nothdurfter et al., 2011; Pucadyil and Chattopadhyay, 2004; Renner et al., 2007; Sjogren et al., 2008), 2A (Dreja et al., 2002; Mialet-Perez et al., 2012; Sommer et al., 2009), 3A (Eisensamer et al., 2005; Ilegems et al., 2005; Nothdurfter et al., 2010), and 7A (Sjogren et al., 2006; Sjogren and Svenningsson, 2007a; Sjogren and Svenningsson, 2007b) receptors; dopamine uptake (Jones et al., 2012); and dopamine D2 receptor oligomerization (Celver et al., 2012; Genedani et al., 2005).

N-3 PUFAs also consistently have been implicated in lipid raft regulation of immunomodulation (Ariel et al., 2005; Basiouni et al., 2012; Baumer et al., 2010; Bonilla et al., 2010; Chentouf et al., 2010; De Smedt-Peyrusse et al., 2008; Diaz et al., 2002; Geyeregger et al., 2005; Gurzell et al., 2013; Hou et al., 2012; Kim et al., 2008b; Li et al., 2005; Li et al., 2007; Li et al., 2008; Murakami et al., 2012; Ruth et al., 2009; Schumann et al., 2011; Teague et al., 2013; Van Laethem and Leo, 2002; Webb et al., 2000; Wong et al., 2009a; Zeyda et al., 2002), including a direct influence on lipid raft modulation of Toll-like receptor 4, a factor governing induction of genes that regulate inflammatory responsiveness (Wong et al., 2009a).

3. DIGESTION, ABSORPTION, AND SECRETION OF DIETARY PUFAS

3.1. Oral detection of dietary PUFAs

The discovery of lipid receptors CD36 and GPR120 in taste bud cells supports the existence of a gustatory oral perception of dietary lipids (Degrace-Passilly and Besnard, 2012; Martin et al., 2011a; Nozaki et al., 1995). CD36 can bind PUFAs with nanomolar affinity (Baillie et al., 1996), and inactivation of the CD36 gene disturbs the consumption of PUFAs (Laugerette et al., 2005; Martin et al., 2011b). Human data also indicate that unesterified free fatty acids are taste stimuli that can be distinguished by chain length (Chale-Rush et al., 2007a; Chale-Rush et al., 2007b) and saturation (Mattes, 2009). Taste cues from the oral detection of lipids modulate subsequent lipid responsiveness (Mattes, 2005), e.g. meals with different fatty acid profiles (Fielding et al., 1996) differentially initiate mobilization of fat from jejunal enterocytes (Robertson et al., 2003).

3.2. Digestion of PUFAs

Digestion of lipids is catalyzed by lingual lipase (Hamosh et al., 1975) and gastric lipase (Armand, 2007), both of which hydrolyze the stereospecifically numbered (sn)-3 position of triacylglycerols (Staggers et al., 1981), producing free fatty acids, diacylglycerols, and a few 2-monoacylglycerols (Phan and Tso, 2001). The long-chain free fatty acids released by gastric lipolysis stimulate cholecystokinin secretion, which in turn stimulates pancreatic lipase secretion, slows gastric emptying (Hamosh, 1990), and suppresses appetite (Little et al., 2007).

Carbohydrate (amylose)-lipid complexes in the stomach may serve as a delivery system for PUFAs (Lalush, 2005). Following enzymatic hydrolysis of the amylose, these complexes facilitate optimal release of PUFAs into the small intestine (Lalush, 2005), where most lipid digestion takes place.

In the duodenum, emulsification of dietary lipids increases the surface area of hydrophobic lipid droplets for digestive enzymes (Bauer, 2005). Emulsion is accomplished through mechanical mixing by peristalsis in the presence of bile salts, amphipathic derivatives of cholesterol (Holt, 1971). Although lipid absorption is markedly more efficient in the presence of bile, PUFAs, which are unsaturated, and therefore less hydrophobic, do not rely on bile for emulsification in the intestine as much as their saturated counterparts (Bernard and Carlier, 1991; Carlier et al., 1991; Demarne et al., 1982; Jones et al., 1985; Ockner et al., 1972; Toorop et al., 1979; Tso, 1994).

Hydrolysis of triacylglycerol into monoacylglycerol and free fatty acids is accomplished predominantly by pancreatic lipase (Bauer, 2005) which cleaves triacylglycerols at the sn-1 and sn-3 positions. Pancreatic lipase acts on EPA and DHA less than on shorter-chain fatty acids in the sn-3 position (Ikeda et al., 1995).

3.3. Absorption and secretion of PUFAs

Absorption of free fatty acids and 2-monoacylglycerol, the primary products of lipid digestion in the small intestine, is facilitated by formation of amphipathic mixed micelles, which carry their hydrophobic cargo across the enterocyte brush border membrane by simple diffusion (reviewed in (Ramirez et al., 2001). Short- and mediumchain length fatty acids do not require incorporation into mixed micelles for absorption. Triacylglycerol structure can also influence intestinal absorption, and fatty acids located in the sn-2 position can dramatically enhance absorption (Ramirez et al., 2001).

The mechanisms by which PUFAs are taken up by the enterocyte across its apical membrane likely include both facilitated (Abumrad et al., 1993; Chow and Hollander, 1979a; Chow and Hollander, 1979b; Diede et al., 1992; Hollander et al., 1984; Masson et al., 2010; Stremmel, 1988b) and passive diffusion (Chow and Hollander, 1979a; Ling et al., 1989). Uptake of PUFAs by facilitated membrane translocation involves fatty acid translocases (FAT) also known as CD36 (Abumrad et al., 1993; Diede et al., 1992; Stremmel, 1988b), fatty acid transport proteins (FATP), also known as very long-chain acyl-CoA synthetase (ACSVL) (Jia et al., 2007), and membrane-bound fatty acid binding proteins (FABP) (Masson et al., 2010).

Once inside the enterocyte, cytosolic FABP (FABPc) facilitates fatty acid transport (Storch and Corsico, 2008) to the smooth endoplasmic reticulum, where complex lipids are re-synthesized. Fatty acyl-CoA synthetase converts the fatty acids into activated acyl CoA derivatives that combine with 2-monoacylglycerols into triacylglycerols via the consecutive actions of acyl CoA:monoacylglycerol acyltransferase and acyl CoA:diacylglycerol acyltransferase, within the triacylglycerol synthase enzyme complex. The very hydrophobic triacylglycerols are re-packaged as chylomicrons and transported to the rest of the body via the lymphatic system (Fielding, 2008).

4. PERIPHERAL MECHANISMS OF PUFA TRANSPORT

4.1. Circulating lipoproteins

Lipids are transported within the plasma (Kane, 1983; Shen et al., 1977; Zilversmit, 1965) via lipoprotein particles composed of a neutral lipid core (triacylglycerol and cholesteryl esters) surrounded by a shell of amphipathic apolipoproteins, phospholipid, and nonesterified (free) cholesterol (Havel, 1975). In normal human serum, n-6 PUFAs account for 60% of fatty acids in cholesteryl esters, 22% in triacylglycerols, and 38% in phospholipids (Edelstein, 1986). LA content ranges from 21–25% in phospholipids of the VLDL, LDL, and HDL fractions in subjects on an ad lib diet (Morrisett, 1976), while AA values are much lower, ranging from 4.8% in VLDL to 9.6% in HDL fractions. By contrast, n-3 PUFAs account for 1.7% in cholesteryl esters, 1.8% in triacylglycerols, and 3.5% in phospholipids, where DHA is the most abundant at 2.2%.

Chylomicrons are the largest and lowest density lipoproteins, containing the highest percentage of lipid, of which 90% is triacylglycerol (Jonas, 2008). As the chylomicron circulates, most of the triacylglycerol is hydrolyzed by lipoprotein lipase, and the particle decreases in size and increases in density until its remnant is endocytosed by the liver and degraded in lysosomes by hepatic lipase (Naik, 2011). The free fatty acids derived from the degradation of triacylglycerol may enter adjacent muscle cells or adipocytes, be β-oxidized for energy, or be transported in blood in association with albumin until cellular uptake occurs.

4.2. Hepatic processing of PUFAs

Chylomicrons and non-esterified circulating ALA and LA are taken up into hepatocytes from the circulation and serve as precursors for the synthesis of longerchain PUFAs (Demar et al., 2005; Igarashi et al., 2007a; Igarashi et al., 2008; Rapoport et al., 2010; Scott, 1989). Liver secretion of triacylglycerol-rich VLDL particles is dependent on the synthesis, activation, and esterification of acyl-CoA PUFA intermediates into phospholipids, triacylglycerols, or cholesteryl esters (Gibbons et al., 1992; Scott and Bazan, 1989; Vance and Vance, 1990). In addition, free fatty acids can be re-esterified into triacylglycerol molecules and stored in adipocytes and hepatocytes until release for body needs. This process, however, can result in pathological hypertriglyceridemia. N-3 PUFA supplementation in the form of fish oil is FDA-approved as a treatment for hypertriglyceridemia, suppressing both fasting and postprandial hypertriglyceridemia (Ikeda et al., 2001) by reducing hepatic triacylglycerol secretion (Agren et al., 1996; Hansen et al., 1998; Ikeda et al., 1998; Ikeda et al., 2001; Surette et al., 1992; Wong et al., 1984; Yoshida et al., 1999). Thus, lipid balance within the liver affects triacylglycerol concentrations and lipoprotein composition in plasma (Soutar, 1978) and, ultimately, in brain (Caspi et al., 2007).

Peroxisome proliferator-activated receptors (PPARs) in the liver play critical roles by translating nutritional stimuli (Berger and Moller, 2002), such as PUFA status, into changes in the expression and transcription rate of genes (Desvergne and Wahli, 1999; Kliewer et al., 2001; van Raalte et al., 2004), particularly those involved in lipid metabolism (Schoonjans et al., 1996).

PUFAs are the principal natural ligands for PPARα (Price et al., 2000; Sessler and Ntambi, 1998; van Raalte et al., 2004), expressed primarily in the liver, kidney, heart, muscle and adipose tissue. PPARα regulates the expression of proteins involved in transport and β-oxidation of free fatty acids (van Raalte et al., 2004). PUFAs and other PPAR α agonists also influence metabolism of lipoproteins, particularly triacylglycerol-rich lipoproteins (van Raalte et al., 2004). Genetic polymorphisms of PPARα can modulate the response of serum lipid concentrations to dietary PUFA intake (Chan et al., 2006; Tai et al., 2005; Yamakawa-Kobayashi et al., 2002).

PPARα-stimulated fatty acid β-oxidation provides hepatocytes with an effective means for catabolism of fatty acids under conditions of fatty acid overload. Liver (L)-FABP is integral to this response and may act to reduce the toxicity of long-chain nonesterified fatty acids by binding them in the cytosolic compartment, thereby facilitating their intracellular diffusion and utilization (Bass, 1993a; Bass, 1993b; Glatz and van der Vusse, 1996; Weisinger, 1996).

Pathways of potential PPAR regulation have been elucidated whereby L-FABP facilitates rapid LC-PUFA uptake into nuclei (McIntosh et al., 2009) and directly interacts with PPARα (Hostetler et al., 2009; Hostetler et al., 2010; Tan et al., 2002). The PUFAmediated increase in the expression of PPARα-regulated LC-PUFA β-oxidative enzymes, L-FABPs, and PPARα itself is L-FABP dependent (Petrescu et al., 2013). However, it is not known if L-FABP contributes to specificity of PPARα activation with respect to n-3 vs. n-6 LC-PUFAs, or to specificity within the n-3 subfamily of PUFAs (e.g., EPA vs. DHA). Moreover, the molecular mechanisms underlying interactions between L-FABP, PPARα, and PUFAs have yet to be discerned.

4.3. Adipose storage and mobilization of PUFAs

Adipose tissue represents the primary reservoir of triacylglycerols from which free fatty acids are hydrolyzed and released into plasma (Spector, 2001), where they are transported as a physical complex highly but reversibly bound with albumin (Spector, 1986). As a result, the PUFA composition of adipose free fatty acids is very similar to that of plasma (Seidelin, 1995). Plasma free fatty acids contain approximately 17% n-6 PUFAs, of which 90–93% is LA (Edelstein, 1986). Only 1.2–2.5% of plasma free fatty acids are n-3 PUFAs, of which 59–77% is ALA. Free AA and DHA are present in very small amounts, accounting for less than 1% of all fatty acids in adipose tissue (Seidelin, 1995).

Regulation of PUFA storage in adipose tissue is not well understood. With regard to n-3 PUFAs, both slower uptake into adipose tissue (Summers et al., 2000) and higher mobilization (Raclot and Groscolas, 1994) in relation to other fatty acids have been observed. Dietary fatty acid availability, enzyme selectivity, rate of mobilization, and reuptake all likely affect the PUFA composition of adipose tissue (Raclot and Oudart, 1999).

4.4. PUFA transporters

Fatty acid binding proteins (FABPs) are small, cytosolic proteins (Hohoff, 1998; Ribarik Coe, 1998; Veerkamp, 1995) that influence fatty acid metabolism through facilitating uptake, transport, sequestration, and metabolic targeting of fatty acids from the plasma membrane to specific intracellular organelles (Ek et al., 1997; Murphy et al., 1999; Wolfrum et al., 2000) for β-oxidation, lipid synthesis, or signaling (Veerkamp et al., 1991; Weisinger, 1996). Nine isoforms of FABPs are currently known to be expressed throughout the body, including nervous tissue (Veerkamp and Zimmerman, 2001), with overlapping, but somewhat different substrate specificities (reviewed in (Storch and Corsico, 2008). Liver (L)-FABPs, in particular, have a high affinity for fatty acids (Gordon et al., 1983; Thompson et al., 1999), binding DHA and AA with comparable affinities at sub-micromolar concentrations (Maatman et al., 1994; Richieri et al., 2000; Widstrom et al., 2001; Wolfrum et al., 2000). L-FABPs facilitate DHA, oleic (18:1n-9) and palmitic (16:0) acid (Reubsaet et al., 1990) transfer to the peroxisome. Consistent with the various roles of fatty acids, the functions of FABPs may be diverse and dependent on the FABP type and the cell in which they are present (Veerkamp and Zimmerman, 2001).

Alternatively, very long-chain n-3 PUFA intermediates may be transported between organelles in the unbound state or as their acyl CoA derivatives. Nonesterified fatty acids are able to desorb from lipid bilayers and diffuse through aqueous solution in the absence of a binding protein (Hamilton et al., 2001; Zucker, 2001). However, the rate of spontaneous membrane desorption slows greatly as chain length increases, and is made faster by unsaturation (Pownall, 2001). L-FABP has been shown to slow intermembrane diffusion (Zucker, 2001) but increase cytoplasmic diffusion (Luxon, 1996; Murphy, 1998) of fatty acids. A mathematical model has been formulated that predicts both processes (Weisiger and Zucker, 2002).

Within the last decade, studies have begun to assess the substrate specificity of the fatty acid transporters. Thus, CD36 selectively transports n-6 PUFAS LA and AA (Guo et al., 2013). Overexpression or deficiency of CD36 (Febbraio et al., 2002) and FABP are associated with steatosis (Miquilena-Colina et al., 2011), insulin deficiency (Luiken et al., 2002a) and resistance (Chabowski et al., 2004; Miquilena-Colina et al., 2011), hyperinsulinemia (Corpeleijn et al., 2008; Miquilena-Colina et al., 2011), and hyperlipidemia (Makinen et al., 2010; Meex et al., 2005). These data suggest that fatty acid transporters not only facilitate but also regulate cellular lipid utilization, and it has been proposed that fatty acid uptake is subject to short-term regulation by translocation of CD36 from intracellular storage sites to the plasma membrane (Glatz et al., 2002; Luiken et al., 2002b) while liver-type FABPc functions in long-term, ligand-induced regulation of gene expression by directly interacting with nuclear receptors (Glatz et al., 2002).

5. BRAIN PUFAS

5.1. Mechanisms of PUFA supply to the brain

Several mechanisms have been described for PUFA entry into the brain from the circulation (see Figure 2), derived from both rat (Lagarde, 2001; Smith and Nagura, 2001a; Thies et al., 1994) and human (Lagarde, 2001; Spector, 2001) studies. Albumin carries PUFAs in the non-esterified form, derived from adipose triacylglycerol stores or esterified in lysophosphatidylcholine, a partially hydrolyzed phosphatidylcholine retaining sn-1 or sn-2 fatty acids (Lagarde, 2001; Thies et al., 1994). Plasma lipoproteins are another transport medium for PUFAs esterified in triacylglycerols, phospholipids, and cholesteryl esters (Spector, 2001).

Figure 2. Proposed mechanisms of Polyunsaturated Fatty Acid Passage Across the Blood-Brain Barrier.

Figure 2

Polyunsaturated fatty acids (PUFA) are primarily delivered to the brain capillary bed via lipoproteins, in esterified form (E-PUFA); or via albumin, esterified to lysophosphatidylcholine (lyso-PC) or unesterified (NE-PUFA) in a rapidly dissociable lipid-protein complex. Evidence exists for several entry mechanisms into endothelial cells of blood brain barrier and into brain: LDL receptors (LDL-R) bind to the lipoprotein, and lipoprotein lipases (LPL), present on the luminal surface of cerebral endothelial cells, liberate PUFA by hydrolysis of ester bonds. Both NE- and E-PUFA can cross the blood-brain barrier by passive diffusion through a flip-flop mechanism. Fatty acid binding proteins on the external (FABPpm) and internal (FABPc) plasma leaflets assist PUFA transport, which may occur via integral membrane proteins, fatty acid transport protein (FATP) or fatty acid translocases (FAT/CD36). FATP indirectly drives fatty acid uptake by esterifing PUFA to CoA inside the cell, effectively trapping the nondiffusible acyl-CoA-PUFA compound. DHA-containing lysoPC molecules are transported via major facilitator molecule, Mfsd2a. Abbreviations: E-PUFA = esterified polyunsaturated fatty acids; FABP = fatty acid binding protein; FABPpm = peripheral membrane fatty acid binding protein; FABPc = cytoplasmic (intracellular) fatty acid binding protein FAT/CD36 = fatty acid translocase/ cluster of differentiation 36; FATP = fatty acid transport protein; LDL-R = low density lipoprotein receptor; LPL = lipoprotein lipase; lyso-PC = lysophosphatidylcholine; NE-PUFA = non-esterified fatty acids.

However, the molecular mechanisms by which fatty acids enter the brain are not agreed upon. Passage into the brain entails traversing first the luminal and abluminal membranes of the endothelial cells that comprise the blood brain barrier (Reese and Karnovsky, 1967), and then the plasma membrane of the neural cells. There is debate concerning whether the main form of PUFA entering the brain is esterified or nonesterified; and the relative importance of energy-dependent protein facilitated transport vs. energy-independent passive diffusion.

5.1.1. PUFA esterification state during brain uptake

In situ brain perfusion in rodents (Hamilton et al., 2002; Hamilton, 2007; Ouellet et al., 2009; Smith and Nagura, 2001b) and in vitro studies in cell culture (Delton-Vandenbroucke et al., 1997; Moore et al., 1990; Moore et al., 1991) with radiolabelled fatty acids have demonstrated that nonesterified PUFAs, hydrophobic in nature, readily cross the blood brain barrier in a passive diffusion process that is not saturable and is regulated by albumin concentration. Rodent studies confirm that radiolabelled PUFAs enter the brain readily when injected into the plasma in the non-esterified form. Over 90% of radiolabelled arachidonic acid is cleared from plasma within 2 min, and brain phospholipids are almost completely pulse labeled at 1 min (DeGeorge et al., 1989; Nariai et al., 1991; Rapoport et al., 2001; Washizaki et al., 1994). Additional research suggests that lipoproteins containing esterified PUFAs are hydrolyzed at the rat brain endothelial cell surface by lipoprotein lipase (Brecher and Kuan, 1979; Purdon, 1997; Shirai et al., 1986), resulting in transfer of non-esterified fatty acids across the blood brain barrier. Incorporation rates of AA and DHA from the nonesterified plasma pool into rat brain phospholipids are rapid (approximately 5pmol/g brain/s, for AA) and closely approximate independent measures of their respective brain consumption rates (Chen et al., 2008a). Thus, although nonesterified PUFAs in serum amount to approximately 17% of n-6 PUFA and 10-fold less of n-3 PUFA (Edelstein, 1986), they likely serve as a major source of PUFAs for the brain. Taken together, these findings suggest that despite the low level in plasma, the nonesterified form is a primary source of brain PUFAs, derived from both albumin-PUFA complexes and PUFAs carried in lipoproteins.

However, evidence from one research group supports uptake of esterified PUFAs as a significant mode of transfer across the blood-brain barrier. DHA esterified to lysophophatidylcholine was taken up more efficiently than the non-esterified forms in brain-capillary endothelial cell and astrocyte cell co-cultures (Bernoud et al., 1999; Lagarde, 2001) and into brains of 20 day-old rats (Lagarde, 2001), but not into liver, heart, or kidney (Thies et al., 1994). This group found that the percentage of labelled DHA in rat brain lipids after [3H]DHA-lysophophatidylcholine infusion was approximately 10-fold higher than that of unesterified DHA (Thies et al., 1994), and that lysophophatidylcholine was primarily found in association with albumin, which they propose as an effective mechanism of delivering PUFA to brain (Croset et al., 2000).

Preliminary evidence that lysophosphatidylcholine may play a significant role in transport has been strengthened with the recent discovery that Mfsd2a, of the major facilitator superfamily and previously an orphan transporter, functions as a major, sodium-dependent symporter of lysophosphatidylcholine-DHA into brain of immature mice, and Mfsd2a-knockout mice exhibit reduced brain DHA levels (Nguyen et al., 2014). Studies using the pulse-labeling method (Robinson et al., 1992) remain to be done in adult animals to clarify this mechanism. In both rodents and humans, about 55% of DHA is esterified to lysophosphatidylcholine, with 45% in the non-esterified form (Lagarde, 2001).

An additional hypothesis is that lipoproteins themselves, containing the esterified PUFAs, are taken up by endothelium, and the resultant phospholipids are hydrolyzed intracellularly. Supporting evidence includes the presence of lipoprotein lipase and acid lipase activity in cerebral microvasculature from rabbit brain (Brecher and Kuan, 1979). and LDL receptors in the bovine cerebral vascular endothelium (de Vries, 1993; Dehouck et al., 1994; Dehouck et al., 1997; Herz and Bock, 2002; Lucarelli et al., 1997); the ability of the HDL3 subfraction to bind to bovine brain capillary endothelial cells (Martin-Nizard et al., 1989); and, in dogs and humans, the presence of apolipoproteins as well as LDL and HDL particles that have crossed the blood brain barrier into cerebrospinal fluid (Hesse et al., 2000; Pitas et al., 1987), although their presence may be due to ultrafiltration through the choroid plexus (Rapoport and Pettigrew, 1979). Furthermore, LDL can undergo transcytosis through rat and bovine brain capillary endothelial cells (Candela et al., 2008; Dehouck et al., 1997). Similarly, in rats, intravenously injected triacylglyerol-rich lipoproteins and lipid emulsions can be directly taken up in vivo by tissues as intact particles with little lipolysis (Hultin et al., 1995). However, the biological relevance of these phenomena to brain PUFA homeostasis is unclear, as studies using knock-out mice have found that neither LDL (Chen et al., 2008b) nor VLDL (Rahman et al., 2010) receptors are necessary for regulating brain PUFA concentrations. One possibility is that the receptors are critical for transport and function of the capillaries themselves, but not necessarily for neurons or glia on the abluminal surface. Capillary isolation studies are in order (Williams et al., 1997).

5.1.2. Passive diffusion or protein-facilitated transfer?

Current models for brain uptake of non-esterified fatty acids postulate passive diffusion using a “flip-flop” mechanism, fatty acid transport protein-facilitated movement, or a combination of both (Chen et al., 2008a). A rapid reversible flip-flop diffusion mechanism is consistent with known biophysical properties of LC-PUFAs in biomembranes (Hamilton, 2007; Hamilton and Brunaldi, 2007). Others have challenged this model, however, asserting that in biological membranes, flip-flop is not rapid but rather is a rate-limiting step that may only be overcome with the assistance of proteinmediated PUFA transport (Cupp et al., 2004; Kampf et al., 2006).

The facilitated transfer hypothesis is given face validity by the existence of multiple putative lipid transporter proteins including isoforms of the integral membrane protein FATP (DiRusso et al., 2005; Jia et al., 2007; Melton et al., 2011), the transmembrane protein CD36 (Bastie et al., 2004; Drover et al., 2008; Ibrahimi et al., 1996; Nickerson et al., 2009), and the peripheral membrane fatty acid binding protein (FABPpm) (Luiken et al., 1999; Potter et al., 1987; Schwieterman et al., 1988; Sorrentino et al., 1988; Sorrentino et al., 1991; Stremmel et al., 1985a; Stremmel et al., 1985b; Stremmel et al., 1986; Stremmel, 1988a; Turcotte et al., 2000). In this model, fatty acid transporters are believed to attach PUFA molecules on the luminal face of cerebral endothelial cells and diffuse across both the endothelial and the neuronal cell membranes (Abumrad et al., 1998). However, FATP do not act as simple transporters; rather, through acyl-CoA synthetase activity, they indirectly drive PUFA uptake through “quenching” (Chen and Bazinet, 2014) the fatty acids by esterifying them with CoA, forming a non-diffusible fatty acid compound that is trapped within the cell (Jia et al., 2007), a possible rate-limiting step for PUFA incorporation into brain (Black and DiRusso, 2003). Similarly, CD36 proteins have been demonstrated in HEK 293 cell lines not to physically transport PUFAs but rather to promote uptake by upregulating intracellular esterification of PUFAs to triacylglycerols (Xu et al., 2013). CD36-null mice evince learning deficits but do not differ in brain AA or DHA concentrations from wildLiu p. 30 type mice (Song et al., 2010), and thus CD36 likely does not play a major role in maintaining brain PUFA stores. Once inside the cell, the LCPUFA binds to a FABP molecule on the inner leaflet of the plasma membrane for intracellular transport. In human brain, this transport process is characterized by differential permeability with respect to the apical-basolateral direction of fatty acid movement (Mitchell et al., 2011), and selectivity, as essential PUFAs enter the brain while cholesterol and nonessential fatty acids do not (Edmond, 2001) (but see (Pardridge and Mietus, 1980).

FABPs in the brain exhibit distinct, isoform-specific temporal distributions (Storch and Corsico, 2008; Veerkamp and Zimmerman, 2001). Brain (B)-FABP and epidermal (E)-FABP (also known as keratinocyte [K]-FABP) are mainly expressed in developing brain (Owada, 2008). B-FABP preferentially binds n-3 PUFAs, especially DHA (Balendiran et al., 2000; Hanhoff et al., 2002; Xu et al., 1996), and its expression is directly correlated with DHA utilization. B-FABP null mice incorporate decreased DHA and increased AA into phospholipids (Owada, 2008). E-FABP is expressed mainly in the embryo and may also play a role in regeneration of neurons after injury (De Leon et al., 1996; Owada, 2008). During adulthood, heart (H)-FABP is also expressed in brain, where its concentration is more than 10-fold that of B-FABP, suggesting that it helps to maintain the differentiation status of adult brain cells (Owada, 2008). The efficacy of LCPUFA absorption into brain may be affected by genetic polymorphisms and epigenetic changes affecting expression patterns of transporters. For example, expression of BFABP is increased in schizophrenia (Watanabe et al., 2007) and Down syndrome (Cheon et al., 2003; Sanchez-Font et al., 2003), whereas H-FABP brain protein levels are lower in both Down syndrome and Alzheimer’s disease (Cheon et al., 2003).

5.1.3. Endogenous fatty acid synthesis by the brain

While it is generally accepted that the brain obtains essential PUFAs from blood, it may also produce LC-PUFAs from precursors (Berg JM, 2002). Endothelial cells comprising the blood-brain barrier accomplish the initial elongation and desaturation steps (Moore et al., 1990), and astrocytes may also produce DHA and AA (Moore et al., 1991). Developing rat brain is capable of converting ALA to DHA (Dhopeshwarkar and Subramanian, 1976), and can also take up, convert, and selectively esterify PUFAs into phospholipids (Green and Yavin, 1993). However, radiolabelled ALA entering the brain is almost completely metabolized to its β-oxidation products, with less than 0.2% transformed to DHA (Demar et al., 2005). Even under conditions of low dietary AA, less than 1% of the LA precursor is transformed to AA in adult rat brain (DeMar et al., 2006). Thus, even when the diet is PUFA deficient, endogenous brain production is a minor contributor compared to the liver, and is not significantly upregulated in response to deficiency states.

5.2.1. Maintenance of PUFA composition in the brain

Plasma PUFAs equilibrate to about 90% of maximal brain uptake within 15 min (Rapoport, 1999), and the higher the degree of unsaturation, the more rapid the uptake (Lagarde, 2001). In contrast, brain DHA loss is very slow: in rat pups on an n-3 PUFA adequate diet, the half-life of DHA in total brain phospholipids is 33 days (Demar et al., 2004). Moreover, the brain retains DHA longer under conditions of deficiency (Gazzah et al., 1995). For example, in rat pups deprived of dietary n−3 PUFA for 15 weeks, brain DHA half-life increases twofold (Demar et al., 2004), and the brain retains its DHA stores at the expense of other organs for long periods of deprivation (Bourre et al., 1992). An exception is that in reproducing female rats, brain DHA content falls by 20% during gestation and nursing (Levant et al., 2006), an observation that may have relevance for postpartum depression in humans. In rats fed a n-3 deficient diet for multiple generations (Contreras et al., 2000), and in nursing dams (Levant et al., 2006), the lost DHA content is replaced by 22:5n-6 DPA. In contrast to DHA, recent rodent studies of the kinetics of EPA metabolism find that the brain maintains low levels of EPA through extremely rapid rates of β-oxidation (Chen and Bazinet, 2014; Igarashi et al., 2013). Dietary PUFA supplementation can reverse reductions in PUFA concentrations in brain and peripheral organs caused by dietary deprivation. Deprived rats subsequently fed an n-3 sufficient diet were able to recover DHA levels in the brain after three weeks; however, recovery half-life was considerably shorter for non-neural tissues and blood (Ikemoto et al., 2001; Moriguchi et al., 2001). The slower recovery of DHA by the brain has been attributed to the small contributions (2–4%) of plasma PUFAs to the net quantity that is re-esterified (Shafrir et al., 1965; Wosilait and Soler-Argilaga, 1975), or to slow transfer of DHA synthesized in astrocytes to neurons (Moore, 2001). However, kinetic modeling studies with radiolabelled fatty acids in awake adult rats find that the turnover of 3–5% of AA and 2–8% of non-esterified DHA entering the esterified brain pool each day (Rapoport, 2001; Rapoport, 1999; Rapoport et al., 2001) is adequate to counterbalance metabolic loss.

5.2.2. Effects of PUFA balance on brain

One useful metric of PUFA status is the relative proportion of n-3 and n-6 PUFAs, which has been reported to affect monoaminergic transmission (Chalon, 2006b) and has ramifications for major mood disorders (Adams et al., 1996; De Vriese et al., 2003; Maes et al., 1996; Maes et al., 1999a; Mamalakis et al., 2004), functional improvement in psychotic disorders (Amminger et al., 2010), and cognitive decline (Heude et al., 2003). Low n-3 relative to n-6 PUFA status also has been implicated in risk of suicide attempts (Huan et al., 2004; Sublette et al., 2006) and suicide (Lewis et al., 2011). PUFA imbalance also may underlie the observed relationship of cholesterollowering treatments with higher rates of non-cardiac illness mortality due to suicide and other types of violent death (Muldoon et al., 1990; Muldoon et al., 2001), e.g. gemfibrozil (Nyalala et al., 2008) increases the lipid proportion of AA.

The factors governing the maintenance of PUFA balance in the brain are not completely understood. Turnover (recycling) is one nexus of regulation (Chen et al., 2008a), controlled to some extent by PUFA species-specific PLA2 isoforms: for example, cytosolic calcium-independent group IVA cPLA2 for AA (Clark et al., 1991) and calcium-independent group VIA iPLA2 for DHA (Green et al., 2008; Strokin et al., 2003), as well as acyl-CoA synthetases, for example acyl-CoA synthetase-4 for AA (Shimshoni et al., 2011; Soupene and Kuypers, 2008). H-FABP expression is also necessary to maintain the n-6:n-3 PUFA balance in adult brain cells, and lower H-FABP expression reduces the incorporation of AA into brain phospholipids (Murphy et al., 2005).

Studies of major depression suggest that the balance of individual n-3 subspecies is also important. In a meta-analyses of PUFA status in depression, both lower DHA and EPA levels distinguished depressed from healthy groups (Lin et al., 2010); however, meta-analyses of clinical trials in depression find that supplements containing higher proportions of EPA are more effective than those containing higher amounts of DHA (Lin et al., 2012; Martins, 2009; Martins et al., 2012; Ross et al., 2007). Reasons for this are unknown, although there are a number of non-brain contexts in which EPA and DHA have different functionality. Well-studied examples are that EPA competes with AA, its 20-carbon n-6 congener, for binding to cPLA2α and to COX-1 (Smith, 2005); and that EPA is a much stronger activator of PPARα than DHA (Jump, 2008; Jump et al., 2008b). Conversely, DHA has more powerful effects than EPA on lipid raft structure (Williams et al., 2012). Experimental n-3 PUFA deficiency results in compensatory elevations of DPAn-6 (22:5n-6) derived from liver elongation of AA (Kim et al., 2011; Moriguchi, 2004), which displays very different lipid matrix properties from DHA, although it contains only 1 fewer double bond (Eldho et al., 2003). However, to our knowledge, significant elevations of plasma DPAn-6 have not been demonstrated in presumed severe clinical n-3 PUFA deficiency.

5.3. PUFA roles in brain function

Development of novel, lipid-based neuropsychiatric treatments will require an indepth understanding of PUFA actions in the brain. Preclinical studies in cell cultures and rodent models have elucidated many brain PUFA functions including participation in neurotransmission, neuroinflammation, neuroprotection, and energy maintenance, discussed below. Recently, the development of fatty acid radiotracers for use in positron emission tomography (PET) has made possible the in vivo study of PUFA uptake into human brain. [1-11C]AA has been used to characterize unesterified AA incorporation into brain in healthy adults (Thambisetty et al., 2012), in aging (Esposito et al., 2007b; Giovacchini et al., 2002; Giovacchini et al., 2004; Rapoport et al., 2011; Umhau et al., 2009), and in patients with Alzheimer’s disease (Esposito et al., 2008); and during functional or pharmacological dopaminergic D2 receptor activation (Esposito et al., 2007a; Thambisetty et al., 2012), while [1-11C]DHA has been used to study baseline brain DHA consumption and chronic alcoholism (Umhau et al., 2013). Another new radiotracer, [18F]-fluoroarachidonic acid (Pichika et al., 2012), is now under development.

5.3.1. PUFAs and signaling pathways in the brain

PUFAs can modulate many signal transduction mechanisms in neuronal membranes and the synapse. As serotonin (5-HT1 and 5-HT4), beta-adrenergic and dopamine (D1 and D2) receptors are all coupled to the cAMP messenger system, PUFAs can influence them by increasing adenylate cyclase (Murphy, 1985; Nicolas et al., 1991) and protein kinase A (Speizer et al., 1991) activity. Animal studies, in piglets (de la Presa Owens and Innis, 1999) and in two generations of rats (Zimmer et al., 1999; Zimmer et al., 2000b) fed n-3 PUFA deficient diets identify many effects on dopaminergic systems, including lower levels of dopamine (de la Presa Owens and Innis, 1999; Zimmer et al., 2000c), D2 receptors, D2 receptor mRNA and dopaminergic presynaptic vesicles (Zimmer et al., 2000c), and increased breakdown of dopamine (Zimmer et al., 1998), in the prefrontal cortex. Experimental n-3 PUFA deficiency in rat dams shortly after conception also results in offspring with decreased tyrosine hydroxylase (Kuperstein et al., 2008), the rate-limiting enzyme in dopamine synthesis; fewer detectable dopaminergic neurons in the substantia nigra and ventral tegmentum (Ahmad et al., 2008); and elevated postnatal expression of dopamine receptor genes in rat pups (Kuperstein et al., 2005). After two generations of n-3 PUFA deficiency, rats also exhibit higher dopamine levels, D2 receptor mRNA, D2 receptors, and less release and breakdown of dopamine in the nucleus accumbens (Zimmer et al., 2000a). Conversely, two generations of a high-n-3 PUFA diet increases dopamine levels in rat prefrontal cortex by 40% and elevates D2 receptor binding, while lowering monoamine oxidase B activity in prefrontal cortex and D2 receptor binding in striatum (Chalon et al., 1998). PUFA associations with dopamine also have been implicated in clinical depression (Sublette et al., 2014). Less has been reported concerning dietary PUFA effects on serotonin (5-HT), although, compared with normally fed animals, rats with low brain DHA had decreased 5-HT levels and turnover in frontal cortex (nulliparous females) and higher density of hippocampal 5-HT1A receptors (parous dams) (Levant et al., 2008). Also, in 2-month old male rats, stimulated 5-HT release in the hippocampus was lower in rats fed a n-3 PUFA deficient diet for two generations, and the ability of diet alteration to reverse this deficiency decreased over time after parturition (Chalon, 2006a). However, in adult male mice, n-3 PUFA supplementation was effective in reversing 5-HT levels that had declined by 40–65% as a result of unpredictable chronic mild stress (Vancassel et al., 2008). On the other hand, in one study comparing high saturated fat, high n-3 PUFA, and high n-6 PUFA diets, 5-HT2A receptor and 5-HT transporter binding were most strongly affected by the n-6 PUFA diet (Dubois et al., 2006).

PUFAs also can interact with the phosphoinositide signaling pathway by exerting effects on phospholipase C (Irvine et al., 1979) and protein kinase C (McPhail et al., 1984a; McPhail et al., 1984b), both of which are involved in 5-HT2 and alpha-1 adrenergic mediated signal transduction.

Neurotransmitters affect PUFA turnover, as PLA2 is activated by multiple receptor types--dopamine D2 (Vial and Piomelli, 1995), serotonin 5-HT2 (Berg et al., 1996), glutamate (Tence et al., 1995), and muscarinic acetylcholine (Jones et al., 1996)--to liberate fatty acids from the sn-2 position of phospholipids. In turn, PUFAs also regulate the activity, mRNA expression, and protein levels of multiple PLA2 isoforms (Downes and Currie, 1998; Lister et al., 1988; Rao et al., 2007a).

Fatty acids that are not recycled back into phospholipids can be metabolized through several pathways, e.g. cyclooxygenase (COX)-2 (O'Banion, 1999) acts on AA, EPA and DHA (Serhan et al., 2002) to produce prostanoids (prostaglandins [PGE] and thromboxanes) (Chang and Karin, 2001; Kozak et al., 2001), while lipoxygenases produce leukotrienes and lipoxins. PGEs occur in 3 families which have different effects through interactions with specific signaling systems: the PGE-2 family transduces signals via a Gs protein, elevating cAMP levels, whereas the PGE-3 family uses a Gi protein, lowering cAMP, and the PGE-1 family acts through a phosphoinositide signaling system (Smith, 1992). Eicosanoids, 20-carbon n-6 compounds, and docosanoids, 22-carbon n-3 compounds that include resolvins or neuroprotectins, can have opposing effects on signal transduction and inflammatory processes (Calder, 2006b; Piomelli, 1994).

Release of neurotransmitters from synaptic vesicles by activation of Ca2+/calmodulin dependent protein kinase is affected by PUFAs (Piomelli et al., 1989), and Ca-ATPase in neuronal membranes is inhibited by DHA and EPA (Kearns and Haag, 2002).

5.3.2. PUFAs and neuroinflammation

PUFAs regulate inflammation largely through the actions of their bioactive eicosanoid and docosanoid metabolites, which play distinct roles and have complex interactions in the development and resolution of acute inflammation (Calder and Grimble, 2002; Stables and Gilroy, 2011) (see Figure 3). In contrast to the generally pro-inflammatory n-6 derived eicosanoids, the DHA-derived docosanoids display primarily anti-inflammatory effects, including in brain (Orr et al., 2013). For example, neuroprotectin D1 (NPD1) downregulates peptide-induced proinflammatory cytokine expression in microglia (Bazan et al., 2005; Bazan, 2009; Connor et al., 2007), and resolvins block the production of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and pro-inflammatory cytokines by microglial cells (Marcheselli et al., 2003).

Figure 3. Inflammatory status of polyunsaturated fatty acid (PUFA) metabolites.

Figure 3

PUFAs mediate inflammatory responses through the actions of bioactive eicosanoid and docosanoid metabolites. Eicosanoids derived from the n-6 PUFA family are generally pro-inflammatory, whereas the eicosanoids and docosanoids derived from the n-3 PUFA family are less pro-inflammatory or have anti-inflammatory effects, as do n-3 derived lipoxins.

Abbreviations: COX = cyclooxygenase; cPLA2 = cytosolic phospholipase A2; iPLA2 = Ca2+-independent phospholipase A2; LOX = lipoxygenase; PLA2 = phospholipase A2; PUFA = polyunsaturated fatty acids.

As in peripheral systems, the balance between n-6 and n-3 PUFAs thus can have profound effects on neuroinflammation. For example, as n-3 and n-6 LC-PUFAs compete for membrane insertion, an n-3 PUFA-enriched diet increases production of anti-inflammatory docosanoids and decreases the n-6 content of glial cell membranes, resulting in less substrate available for AA-derived eicosanoid synthesis (Calder, 2006a). Some examples with clinical relevance include rodent models of metabolic and behavioral responses to traumatic brain injury, in which n-3 PUFA deficiency states worsened post-injury anxiety-like behavior (Agrawal et al., 2014) and sensorimotor impairments (Russell et al., 2013), and resulted in lower levels of anxiolytic neuropeptide Y1 receptor (Agrawal et al., 2014) and mRNA expression of tissue inhibitor of matrix metalloproteinase-1 (Timp1) (Russell et al., 2013). Supplementation with n-3 PUFAs in the diet prior to experimental injury mitigated the usual sequelae of abnormal levels of brain-derived neurotrophic factor (BDNF), synapsin I, and cAMP responsive element-binding protein (CREB) (Wu et al., 2004), as well as axonal injury, apoptosis (Mills et al., 2011), and cognitive impairments (Mills et al., 2011; Wu et al., 2004). Moreover, in a mouse model, even after experimental brain injury, an inhibitor of fatty acid amide hydrolase (FAAH) reduced breakdown of the PUFA metabolite anandamide, and thereby mitigated fine motor and working memory impairments and anxiety-like behaviors, concomitantly reducing neurodegeneration and amyloid precursor protein, and upregulating stress-responsive growth factors and heat shock proteins (Tchantchou et al., 2014).

5.3.3. PUFAs as regulators of brain energy

DHA also has been identified as an important regulator of brain energy metabolism, having effects on both glucose uptake and on the density of glucose transporter-1 (GLUT1) in endothelial cell cultures (Pifferi et al., 2007) and in cerebral cortex from rat brain (Pifferi et al., 2005). Neuroimaging in humans with [18F]-fluoro-2-deoxyglucose positron emission tomography (FDG-PET) has identified correlations between plasma PUFA levels and cerebral metabolic rates for glucose (rCMRglu) in specific brain regions (Sublette et al., 2009).

5.3.4. PUFAs and neuroprotection

The benefits of n-3 LC-PUFAs to neuropsychiatric health are likely afforded, in part, by their neuroprotective properties. DHA and its metabolite NPD1 alter the expression of pro- and anti-apoptotic genes, including Bcl-2, Akt, and Bfl-1 (Akbar and Kim, 2002; Akbar et al., 2005; Bazan, 2007), positively affecting neuronal survival. Resolvins, too, protect the brain from ischemia (Marcheselli et al., 2003). Dietary DHA also confers neuroprotection by specifically reducing β-amyloid in an mouse model of Alzheimer’s disease (Lim et al., 2005a) and in cytokine-stressed human neural cells (Lukiw et al., 2005), while in an in vivo human study, elevated AA brain uptake was seen with PET, in patients with Alzheimer’s disease compared to healthy volunteers (Esposito et al., 2007a). Conversely, in vivo DHA depletion has been shown to result in decreased brain-derived neurotrophic factor (BDNF) in rodents (Rao et al., 2007c) and increased neuronal cell death in cell cultures (Akbar et al., 2005).

5.4. Inferring PUFA mechanisms of action from medication effects

Rapoport et al. have shown that mood stabilizers and some atypical antipsychotics FDA-approved for treating bipolar disorder have direct or indirect convergent effects on the AA cascade in rat brain, such as decreasing AA turnover in phospholipids or expression of AA-selective cPLA2 IV-A or COX-2 or inhibiting AAselective acyl-CoA synthetase-4 (Shimshoni et al., 2011). These medications include lithium (Basselin et al., 2003; Basselin et al., 2005; Basselin et al., 2006; Basselin et al., 2007), carbamazepine (Bazinet et al., 2006a), valproic acid (Bazinet et al., 2006b), clozapine (Modi et al., 2013), lamotrigine (Lee et al., 2008), and olanzapine (Cheon et al., 2011). Conversely, the medication topiramate, which did not have significant benefit in clinical trials in bipolar disorder (Delbello et al., 2005), also had no effects on rat brain AA metabolism (Lee et al., 2005). Additionally, two antidepressants implicated in triggering manic episodes, fluoxetine (Lee et al., 2006) and imipramine (Lee et al., 2010), increased AA turnover (Lee et al., 2006; Lee et al., 2010) and upregulated cPLA2 (Lee et al., 2010), while the more dopaminergic antidepressant bupropion had no effects on AA turnover (Lee et al., 2010).

Taken together, these results suggest that bipolar mood instability may be related to dysregulation of the AA cascade.

5.5. Relevance of peripheral PUFA biomarker data

Interpreting the results of studies using peripheral PUFAs as biomarkers is complicated by several factors: 1) Esterification. Few human studies analyze levels of nonesterified PUFAs. However, non-esterified PUFAs readily cross the blood brain barrier and their plasma concentrations are tied closely to brain consumption rates (Chen et al., 2008a), although the relationship between plasma and brain DHA levels may be nonlinear (Rapoport, 2013). In one study in bipolar disorder (Sublette et al., 2007), manic symptoms correlated with nonesterified but not esterified plasma PUFA concentrations. 2) Tissue source (adipose, total plasma, plasma lipid subfractions, platelets, erythrocytes). It has been assumed by some that erythrocyte PUFA concentrations give the best indication of long-term PUFA status, given the longevity of red blood cells. However, the membrane PUFA composition of erythrocytes is not static throughout their lifetime, but rather experiences turnover as in other tissues. In fact, PUFA constituents of erythrocytes exhibit diet-related changes within 24 hours (Hodson et al., 2008), similar to plasma and platelet time courses (Skeaff et al., 2006). 3) Lipoprotein status. In total plasma PUFAs, percentages of n-3 PUFAs may be strongly influenced by the relative amounts of different lipoproteins (Hodson et al., 2008). Different types of lipoproteins carry characteristic proportions of triacylglycerols, cholesteryl esters, and phospholipids, e.g., HDL contains the largest percentages of LCPUFAs. These lipids in turn exhibit individually distinct PUFA profiles. Therefore, individual differences in lipidemia may confound the study of total plasma PUFAs, and the use of phospholipid fractions may mitigate this problem. 4) PUFA species. Quantification of levels of all PUFAs that are separable by chromatography provides a comprehensive picture but incurs loss of statistical power for group comparisons. In contrast, clinical approaches which focus a priori on a few well-characterized PUFAs (frequently DHA, AA, EPA) retain more statistical power but run the risk of oversimplification and overlooking the contribution of less abundant PUFA species or their metabolites (Yuan et al., 2013). Moreover, it is unclear whether absolute levels or proportions of total plasma PUFA are more physiologically relevant.

6. CONCLUSIONS

From the perspective of precision medicine (National Research Council, 2011), patients with low n-3 PUFA levels relative to n-6 PUFA may represent a biological subgroup that is at greater risk for psychiatric illness. Suboptimal n-3 PUFA intake may also predispose such patients to other medical comorbidities, e.g. depression and cardiac illness are highly comorbid (Musselman et al., 1998), and low n-3 PUFA levels have been implicated in both conditions.

The majority of studies concerning PUFAs in neuropsychiatric illness have focused on brain development and mood, psychotic and cognitive disorders. Less is known about PUFA status in anxiety disorders, substance abuse, or personality disorders. Surprisingly little is known about the effects of low essential LC-PUFA intake in context of eating disorders, which in their most severe forms carry a high rate of major depression and suicide risk.

Given that low n-3 PUFA and excess n-6 PUFA intake are widespread in many countries, it is likely that the occurrence of psychiatric illness does not depend solely on dietary imbalance. We hypothesize that low n-3 PUFA intake is likely to have the greatest impact if combined with metabolic or genetic abnormalities affecting PUFA absorption, transport, interconversion, membrane turnover, or catabolism. A wellstudied instance is the genetically controlled expression of FADS enzymes affecting the fatty acid composition of serum phospholipids in times of suboptimal LC-PUFA dietary intake (Al-Hilal et al., 2013; Glaser et al., 2010; Nwankwo et al., 2003; Schaeffer et al., 2006a) that may lead to individual differences in dietary requirements (Koletzko et al., 2008) and contribute to psychiatric illness including schizophrenia (Liu et al., 2009a) and bipolar disorder (Liu and McNamara, 2011a). Additionally, low levels of n-3 PUFA may contribute to psychiatric illness by interacting with other types of external environmental stress or genomic factors involving stress response systems. For example, in rodents, PUFA deficiency plus environmental stress is more likely to produce anxiety-like behaviors than either alone (Bandaru et al., 2010; Harauma and Moriguchi, 2011; Liu et al., 2013; Mathieu et al., 2008; Mathieu et al., 2011). Therefore, there is a need for studies to assess environmental, genetic, and epigenetic influences on PUFA metabolism and their impact on neuroinflammation in the pathogenesis of psychiatric disorders.

Review Highlights.

  • Essential polyunsaturated fatty acids (PUFAs) are integral for brain functioning

  • Absorption, delivery, transport, or metabolic problems can influence PUFA levels

  • Understanding mechanisms of PUFA supply to the brain is relevant for neuroscientists

Acknowledgments

Funding

Work by Dr. Rapoport was supported entirely by the Intramural Program of the National Institute on Aging. Work by Dr. Sublette was supported in part by NIMH MH079033.

Dr. Mann received royalties from Research Foundation for Mental Hygiene for commercial use of the C-SSRS and stock options in Qualitas Health, a startup company developing a polyunsaturated fatty acid nutritional supplement. Dr. Sublette received a grant of nutritional supplements from Unicity International for an unrelated study.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest

All other authors have no conflicts of interest to report.

References

  1. Abumrad N, Harmon C, Ibrahimi A. Membrane transport of long-chain fatty acids: evidence for a facilitated process. Journal of lipid research. 1998;39:2309–2318. [PubMed] [Google Scholar]
  2. Abumrad NA, et al. Cloning of a rat adipocyte membrane protein implicated in binding or transport of long-chain fatty acids that is induced during preadipocyte differentiation. Homology with human CD36. The Journal of biological chemistry. 1993;268:17665–17668. [PubMed] [Google Scholar]
  3. Adams PB, et al. Arachidonic acid to eicosapentaenoic acid ratio in blood correlates positively with clinical symptoms of depression. Lipids. 1996;31(Suppl):S157–S161. doi: 10.1007/BF02637069. [DOI] [PubMed] [Google Scholar]
  4. Adkins EM, et al. Membrane mobility and microdomain association of the dopamine transporter studied with fluorescence correlation spectroscopy and fluorescence recovery after photobleaching. Biochemistry. 2007;46:10484–10497. doi: 10.1021/bi700429z. [DOI] [PubMed] [Google Scholar]
  5. Agrawal R, et al. Coupling energy homeostasis with a mechanism to support plasticity in brain trauma. Biochim Biophys Acta. 2014;1842:535–546. doi: 10.1016/j.bbadis.2013.12.004. [DOI] [PubMed] [Google Scholar]
  6. Agren JJ, et al. Fish diet, fish oil and docosahexaenoic acid rich oil lower fasting and postprandial plasma lipid levels. European journal of clinical nutrition. 1996;50:765–771. [PubMed] [Google Scholar]
  7. Ahmad SO, et al. Reduced numbers of dopamine neurons in the substantia nigra pars compacta and ventral tegmental area of rats fed an n-3 polyunsaturated fatty acid-deficient diet: a stereological study. Neurosci Lett. 2008;438:303–307. doi: 10.1016/j.neulet.2008.04.073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Akbar M, Kim HY. Protective effects of docosahexaenoic acid in staurosporine-induced apoptosis: involvement of phosphatidylinositol-3 kinase pathway. J Neurochem. 2002;82:655–665. doi: 10.1046/j.1471-4159.2002.01015.x. [DOI] [PubMed] [Google Scholar]
  9. Akbar M, et al. Docosahexaenoic acid: a positive modulator of Akt signaling in neuronal survival. Proc Natl Acad Sci USA. 2005;102:10858–10863. doi: 10.1073/pnas.0502903102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Akhtar Khan N. Polyunsaturated fatty acids in the modulation of T-cell signaling. Prostaglandins Leukot Essent Fatty Acids. 2010;82:179–187. doi: 10.1016/j.plefa.2010.02.023. [DOI] [PubMed] [Google Scholar]
  11. Al-Hilal M, et al. Genetic variation at the FADS1-FADS2 gene locus influences delta-5 desaturase activity and LC-PUFA proportions after fish oil supplement. J Lipid Res. 2013;54:542–551. doi: 10.1194/jlr.P032276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Altenburg JD, Siddiqui RA. Omega-3 polyunsaturated fatty acids down-modulate CXCR4 expression and function in MDA-MB-231 breast cancer cells. Mol Cancer Res. 2009;7:1013–1020. doi: 10.1158/1541-7786.MCR-08-0385. [DOI] [PubMed] [Google Scholar]
  13. Amin AA, et al. Acute coronary syndrome patients with depression have low blood cell membrane omega-3 fatty acid levels. Psychosom Med. 2008;70:856–862. doi: 10.1097/PSY.0b013e318188a01e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Amminger GP, et al. Long-chain omega-3 fatty acids for indicated prevention of psychotic disorders: a randomized, placebo-controlled trial. Arch Gen Psychiatry. 2010;67:146–154. doi: 10.1001/archgenpsychiatry.2009.192. [DOI] [PubMed] [Google Scholar]
  15. Ariel A, et al. The docosatriene protectin D1 is produced by TH2 skewing and promotes human T cell apoptosis via lipid raft clustering. J Biol Chem. 2005;280:43079–43086. doi: 10.1074/jbc.M509796200. [DOI] [PubMed] [Google Scholar]
  16. Armand M. Lipases and lipolysis in the human digestive tract: where do we stand? Current opinion in clinical nutrition and metabolic care. 2007;10:156–164. doi: 10.1097/MCO.0b013e3280177687. [DOI] [PubMed] [Google Scholar]
  17. Arterburn LM, Hall EB, Oken H. Distribution, interconversion, and dose response of n-3 fatty acids in humans. The American journal of clinical nutrition. 2006;83:1467S–1476S. doi: 10.1093/ajcn/83.6.1467S. [DOI] [PubMed] [Google Scholar]
  18. Baillie AG, Coburn CT, Abumrad NA. Reversible binding of long-chain fatty acids to purified FAT, the adipose CD36 homolog. The Journal of membrane biology. 1996;153:75–81. doi: 10.1007/s002329900111. [DOI] [PubMed] [Google Scholar]
  19. Balendiran GK, et al. Crystal structure and thermodynamic analysis of human brain fatty acid-binding protein. The Journal of biological chemistry. 2000;275:27045–27054. doi: 10.1074/jbc.M003001200. [DOI] [PubMed] [Google Scholar]
  20. Bandaru SS, et al. Effects of PI3K inhibition and low docosahexaenoic acid on cognition and behavior. Physiol Behav. 2010;100:239–244. doi: 10.1016/j.physbeh.2009.10.019. [DOI] [PubMed] [Google Scholar]
  21. Barberger-Gateau P, et al. Dietary patterns and risk of dementia: the Three-City cohort study. Neurology. 2007;69:1921–1930. doi: 10.1212/01.wnl.0000278116.37320.52. [DOI] [PubMed] [Google Scholar]
  22. Basiouni S, et al. Polyunsaturated fatty acid supplements modulate mast cell membrane microdomain composition. Cell Immunol. 2012;275:42–46. doi: 10.1016/j.cellimm.2012.03.004. [DOI] [PubMed] [Google Scholar]
  23. Bass NM. Cellular binding proteins for fatty acids and retinoids: similar or specialized functions? Molecular and cellular biochemistry. 1993a;123:191–202. doi: 10.1007/BF01076492. [DOI] [PubMed] [Google Scholar]
  24. Bass NM, Kaikaus RM, Ockner RK. Physiology and molecular biology of hepatic cytosolic fatty acid-binding protein, Vol. New York: Raven Press; 1993b. [Google Scholar]
  25. Basselin M, et al. Chronic lithium administration potentiates brain arachidonic acid signaling at rest and during cholinergic activation in awake rats. J Neurochem. 2003;85:1553–1562. doi: 10.1046/j.1471-4159.2003.01811.x. [DOI] [PubMed] [Google Scholar]
  26. Basselin M, et al. Chronic Lithium Administration to Rats Selectively Modifies 5-HT2A/2C Receptor-Mediated Brain Signaling Via Arachidonic Acid. Neuropsychopharmacology. 2005;30:461–472. doi: 10.1038/sj.npp.1300611. [DOI] [PubMed] [Google Scholar]
  27. Basselin M, et al. Chronic lithium chloride administration attenuates brain NMDA receptor-initiated signaling via arachidonic acid in unanesthetized rats. Neuropsychopharmacology. 2006;31:1659–1674. doi: 10.1038/sj.npp.1300920. [DOI] [PubMed] [Google Scholar]
  28. Basselin M, et al. Chronic lithium administration attenuates up-regulated brain arachidonic acid metabolism in a rat model of neuroinflammation. J Neurochem. 2007;102:761–772. doi: 10.1111/j.1471-4159.2007.04593.x. [DOI] [PubMed] [Google Scholar]
  29. Bastie CC, et al. CD36 in myocytes channels fatty acids to a lipase-accessible triglyceride pool that is related to cell lipid and insulin responsiveness. Diabetes. 2004;53:2209–2216. doi: 10.2337/diabetes.53.9.2209. [DOI] [PubMed] [Google Scholar]
  30. Bauer E, Jakob S, Mosenthin R. Principles of Physiology of Lipid Digestion. Asian-Aust. J. Anim. Sci. 2005;18:282–295. [Google Scholar]
  31. Baumer W, et al. The putative lipid raft modulator miltefosine displays immunomodulatory action in T-cell dependent dermal inflammation models. Eur J Pharmacol. 2010;628:226–232. doi: 10.1016/j.ejphar.2009.11.018. [DOI] [PubMed] [Google Scholar]
  32. Bazan NG, Marcheselli VL, Cole-Edwards K. Brain response to injury and neurodegeneration: endogenous neuroprotective signaling. Annals of the New York Academy of Sciences. 2005;1053:137–147. doi: 10.1196/annals.1344.011. [DOI] [PubMed] [Google Scholar]
  33. Bazan NG. Omega-3 fatty acids, pro-inflammatory signaling and neuroprotection. Curr Opin Clin Nutr Metab Care. 2007;10:136–141. doi: 10.1097/MCO.0b013e32802b7030. [DOI] [PubMed] [Google Scholar]
  34. Bazan NG. Neuroprotectin D1-mediated anti-inflammatory and survival signaling in stroke, retinal degenerations, and Alzheimer's disease. Journal of lipid research. 2009;50(Suppl):S400–S405. doi: 10.1194/jlr.R800068-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Bazinet R, et al. Chronic Carbamazepine Decreases the Incorporation Rate and Turnover of Arachidonic Acid but Not Docosahexaenoic Acid in Brain Phospholipids of the Unanesthetized Rat: Relevance to Bipolar Disorder. Biological Psychiatry. 2006a;59:401–407. doi: 10.1016/j.biopsych.2005.07.024. [DOI] [PubMed] [Google Scholar]
  36. Bazinet RP, et al. Valproic acid selectively inhibits conversion of arachidonic acid to arachidonoyl-CoA by brain microsomal long-chain fatty acyl-CoA synthetases: relevance to bipolar disorder. Psychopharmacology (Berl) 2006b;184:122–129. doi: 10.1007/s00213-005-0272-4. [DOI] [PubMed] [Google Scholar]
  37. Beier AM, et al. Low Plasma Eicosapentaenoic Acid Levels are Associated with Elevated Trait Aggression and Impulsivity in Major Depressive Disorder with a History of Comorbid Substance Use Disorder. Journal of Psychiatric Research. 2014 doi: 10.1016/j.jpsychires.2014.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Benatti P, et al. Polyunsaturated fatty acids: biochemical, nutritional and epigenetic properties. Journal of the American College of Nutrition. 2004;23:281–302. doi: 10.1080/07315724.2004.10719371. [DOI] [PubMed] [Google Scholar]
  39. Berg JM TJ, Stryer L. Biochemistry. New York, NY: 2002. Fatty acids are synthesized and degraded by different pathways. [Google Scholar]
  40. Berg KA, Maayani S, Clarke WP. 5-hydroxytryptamine2C receptor activation inhibits 5-hydroxytryptamine1B-like receptor function via arachidonic acid metabolism. Molecular pharmacology. 1996;50:1017–1023. [PubMed] [Google Scholar]
  41. Berger J, Moller DE. The mechanisms of action of PPARs. Annual review of medicine. 2002;53:409–435. doi: 10.1146/annurev.med.53.082901.104018. [DOI] [PubMed] [Google Scholar]
  42. Bernard A, Carlier H. Absorption and intestinal catabolism of fatty acids in the rat: effect of chain length and unsaturation. Experimental physiology. 1991;76:445–455. doi: 10.1113/expphysiol.1991.sp003511. [DOI] [PubMed] [Google Scholar]
  43. Bernoud N, et al. Preferential transfer of 2-docosahexaenoyl-1-lysophosphatidylcholine through an in vitro blood-brain barrier over unesterified docosahexaenoic acid. Journal of neurochemistry. 1999;72:338–345. doi: 10.1046/j.1471-4159.1999.0720338.x. [DOI] [PubMed] [Google Scholar]
  44. Beydoun MA, et al. Plasma n-3 fatty acids and the risk of cognitive decline in older adults: the Atherosclerosis Risk in Communities Study. The American journal of clinical nutrition. 2007;85:1103–1111. doi: 10.1093/ajcn/85.4.1103. [DOI] [PubMed] [Google Scholar]
  45. Black PN, DiRusso CC. Transmembrane movement of exogenous long-chain fatty acids: proteins, enzymes, and vectorial esterification. Microbiology and molecular biology reviews : MMBR. 2003;67:454–472. doi: 10.1128/MMBR.67.3.454-472.2003. table of contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Bonilla DL, et al. Incorporation of a dietary omega 3 fatty acid impairs murine macrophage responses to Mycobacterium tuberculosis. PLoS One. 2010;5:e10878. doi: 10.1371/journal.pone.0010878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Bordoni A, et al. N-3 PUFAs modulate global gene expression profile in cultured rat cardiomyocytes. Implications in cardiac hypertrophy and heart failure. FEBS letters. 2007;581:923–929. doi: 10.1016/j.febslet.2007.01.070. [DOI] [PubMed] [Google Scholar]
  48. Bountziouka V, et al. Long-term fish intake is associated with less severe depressive symptoms among elderly men and women: the MEDIS (MEDiterranean ISlands Elderly) epidemiological study. Journal of aging and health. 2009;21:864–880. doi: 10.1177/0898264309340693. [DOI] [PubMed] [Google Scholar]
  49. Bourre JM, et al. Dietary alpha-linolenic acid deficiency in adult rats for 7 months does not alter brain docosahexaenoic acid content, in contrast to liver, heart and testes. Biochim Biophys Acta. 1992;1124:119–122. doi: 10.1016/0005-2760(92)90087-c. [DOI] [PubMed] [Google Scholar]
  50. Brecher P, Kuan HT. Lipoprotein lipase and acid lipase activity in rabbit brain microvessels. Journal of lipid research. 1979;20:464–471. [PubMed] [Google Scholar]
  51. Brown DA, London E. Structure and function of sphingolipid-and cholesterolrich membrane rafts. The Journal of biological chemistry. 2000;275:17221–17224. doi: 10.1074/jbc.R000005200. [DOI] [PubMed] [Google Scholar]
  52. Brown WH, Foote CS. Organic Chemistry, Vol. Harcourt College Publishers; 1991. [Google Scholar]
  53. Burdge GC, Jones AE, Wootton SA. Eicosapentaenoic and docosapentaenoic acids are the principal products of alpha-linolenic acid metabolism in young men*. The British journal of nutrition. 2002;88:355–363. doi: 10.1079/BJN2002662. [DOI] [PubMed] [Google Scholar]
  54. Burdge GC, Wootton SA. Conversion of alpha-linolenic acid to eicosapentaenoic, docosapentaenoic and docosahexaenoic acids in young women. The British journal of nutrition. 2002;88:411–420. doi: 10.1079/BJN2002689. [DOI] [PubMed] [Google Scholar]
  55. Burr GOBMM. The nature and role of the fatty acids essential in nutrition. J Biol Chem. 1930;86:587–621. [Google Scholar]
  56. Buydens-Branchey L, Branchey M, Hibbeln JR. Higher n-3 fatty acids are associated with more intense fenfluramine-induced ACTH and cortisol responses among cocaine-abusing men. Psychiatry Res. 2011;188:422–427. doi: 10.1016/j.psychres.2011.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Calder PC, Grimble RF. Polyunsaturated fatty acids, inflammation and immunity. European journal of clinical nutrition. 2002;56(Suppl 3):S14–S19. doi: 10.1038/sj.ejcn.1601478. [DOI] [PubMed] [Google Scholar]
  58. Calder PC. n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. The American journal of clinical nutrition. 2006a;83:1505S–1519S. doi: 10.1093/ajcn/83.6.1505S. [DOI] [PubMed] [Google Scholar]
  59. Calder PC. n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr. 2006b;83:1505S–1519S. doi: 10.1093/ajcn/83.6.1505S. [DOI] [PubMed] [Google Scholar]
  60. Candela P, et al. Physiological pathway for low-density lipoproteins across the blood-brain barrier: transcytosis through brain capillary endothelial cells in vitro. Endothelium : journal of endothelial cell research. 2008;15:254–264. doi: 10.1080/10623320802487759. [DOI] [PubMed] [Google Scholar]
  61. Carlier H, Bernard A, Caselli C. Digestion and absorption of polyunsaturated fatty acids. Reproduction, nutrition, development. 1991;31:475–500. doi: 10.1051/rnd:19910501. [DOI] [PubMed] [Google Scholar]
  62. Carlson SE, Rhodes PG, Ferguson MG. Docosahexaenoic acid status of preterm infants at birth and following feeding with human milk or formula. The American journal of clinical nutrition. 1986;44:798–804. doi: 10.1093/ajcn/44.6.798. [DOI] [PubMed] [Google Scholar]
  63. Caspi L, Wang PY, Lam TK. A balance of lipid-sensing mechanisms in the brain and liver. Cell metabolism. 2007;6:99–104. doi: 10.1016/j.cmet.2007.07.005. [DOI] [PubMed] [Google Scholar]
  64. Celver J, Sharma M, Kovoor A. D(2)-Dopamine receptors target regulator of G protein signaling 9-2 to detergent-resistant membrane fractions. J Neurochem. 2012;120:56–69. doi: 10.1111/j.1471-4159.2011.07559.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Chabowski A, et al. Insulin stimulates fatty acid transport by regulating expression of FAT/CD36 but not FABPpm. American journal of physiology. Endocrinology and metabolism. 2004;287:E781–E789. doi: 10.1152/ajpendo.00573.2003. [DOI] [PubMed] [Google Scholar]
  66. Chale-Rush A, Burgess JR, Mattes RD. Evidence for human orosensory (taste?) sensitivity to free fatty acids. Chem Senses. 2007a;32:423–431. doi: 10.1093/chemse/bjm007. [DOI] [PubMed] [Google Scholar]
  67. Chale-Rush A, Burgess JR, Mattes RD. Multiple routes of chemosensitivity to free fatty acids in humans. American journal of physiology. Gastrointestinal and liver physiology. 2007b;292:G1206–G1212. doi: 10.1152/ajpgi.00471.2006. [DOI] [PubMed] [Google Scholar]
  68. Chalon S, et al. Dietary fish oil affects monoaminergic neurotransmission and behavior in rats. J Nutr. 1998;128:2512–2519. doi: 10.1093/jn/128.12.2512. [DOI] [PubMed] [Google Scholar]
  69. Chalon S. Omega-3 fatty acids and monoamine neurotransmission. Prostaglandins Leukot Essent Fatty Acids. 2006a;75:259–269. doi: 10.1016/j.plefa.2006.07.005. [DOI] [PubMed] [Google Scholar]
  70. Chalon S. Omega-3 fatty acids and monoamine neurotransmission. Prostaglandins, leukotrienes, and essential fatty acids. 2006b;75:259–269. doi: 10.1016/j.plefa.2006.07.005. [DOI] [PubMed] [Google Scholar]
  71. Chan E, et al. The V227A polymorphism at the PPARA locus is associated with serum lipid concentrations and modulates the association between dietary polyunsaturated fatty acid intake and serum high density lipoprotein concentrations in Chinese women. Atherosclerosis. 2006;187:309–315. doi: 10.1016/j.atherosclerosis.2005.10.002. [DOI] [PubMed] [Google Scholar]
  72. Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001;410:37–40. doi: 10.1038/35065000. [DOI] [PubMed] [Google Scholar]
  73. Chapkin RS, et al. Docosahexaenoic acid alters the size and distribution of cell surface microdomains. Biochimica et biophysica acta. 2008;1778:466–471. doi: 10.1016/j.bbamem.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Chapkin RS, et al. Dietary docosahexaenoic and eicosapentaenoic acid: emerging mediators of inflammation. Prostaglandins Leukot Essent Fatty Acids. 2009;81:187–191. doi: 10.1016/j.plefa.2009.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Chen CT, et al. Regulation of brain polyunsaturated fatty acid uptake and turnover. Prostaglandins, leukotrienes, and essential fatty acids. 2008a;79:85–91. doi: 10.1016/j.plefa.2008.09.003. [DOI] [PubMed] [Google Scholar]
  76. Chen CT, et al. The low density lipoprotein receptor is not necessary for maintaining mouse brain polyunsaturated fatty acid concentrations. J Lipid Res. 2008b;49:147–152. doi: 10.1194/jlr.M700386-JLR200. [DOI] [PubMed] [Google Scholar]
  77. Chen CT, et al. Rapid beta-oxidation of eicosapentaenoic acid in mouse brain: an in situ study. Prostaglandins, leukotrienes, and essential fatty acids. 2009;80:157–163. doi: 10.1016/j.plefa.2009.01.005. [DOI] [PubMed] [Google Scholar]
  78. Chen CT, Liu Z, Bazinet RP. Rapid de-esterification and loss of eicosapentaenoic acid from rat brain phospholipids: an intracerebroventricular study. Journal of neurochemistry. 2011;116:363–373. doi: 10.1111/j.1471-4159.2010.07116.x. [DOI] [PubMed] [Google Scholar]
  79. Chen CT, Bazinet RP. beta-oxidation and rapid metabolism, but not uptake regulate brain eicosapentaenoic acid levels. Prostaglandins Leukot Essent Fatty Acids. 2014 doi: 10.1016/j.plefa.2014.05.007. [DOI] [PubMed] [Google Scholar]
  80. Chen JR, et al. Dietary patterns and blood fatty acid composition in children with attention-deficit hyperactivity disorder in Taiwan. J Nutr Biochem. 2004;15:467–472. doi: 10.1016/j.jnutbio.2004.01.008. [DOI] [PubMed] [Google Scholar]
  81. Chentouf M, et al. The lipid-modulating effects of a CD4-specific recombinant antibody correlate with ZAP-70 segregation outside membrane rafts. Immunol Lett. 2010;133:62–69. doi: 10.1016/j.imlet.2010.07.003. [DOI] [PubMed] [Google Scholar]
  82. Cheon MS, et al. Heart type fatty acid binding protein (H-FABP) is decreased in brains of patients with Down syndrome and Alzheimer's disease. Journal of neural transmission. 2003;(Supplementum):225–234. doi: 10.1007/978-3-7091-6721-2_20. [DOI] [PubMed] [Google Scholar]
  83. Cheon Y, et al. Chronic olanzapine treatment decreases arachidonic acid turnover and prostaglandin E(2) concentration in rat brain. J Neurochem. 2011;119:364–376. doi: 10.1111/j.1471-4159.2011.07410.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Cho HP, Nakamura M, Clarke SD. Cloning, expression, and fatty acid regulation of the human delta-5 desaturase. J Biol Chem. 1999a;274:37335–37339. doi: 10.1074/jbc.274.52.37335. [DOI] [PubMed] [Google Scholar]
  85. Cho HP, Nakamura MT, Clarke SD. Cloning, expression, and nutritional regulation of the mammalian Delta-6 desaturase. J Biol Chem. 1999b;274:471–477. doi: 10.1074/jbc.274.1.471. [DOI] [PubMed] [Google Scholar]
  86. Chow SL, Hollander D. A dual, concentration-dependent absorption mechanism of linoleic acid by rat jejunum in vitro. Journal of lipid research. 1979a;20:349–356. [PubMed] [Google Scholar]
  87. Chow SL, Hollander D. Linoleic acid absorption in the unanesthetized rat: mechanism of transport and influence of luminal factors on absorption. Lipids. 1979b;14:378–385. doi: 10.1007/BF02533421. [DOI] [PubMed] [Google Scholar]
  88. Clark JD, et al. A novel arachidonic acid-selective cytosolic PLA2 contains a Ca(2+)-dependent translocation domain with homology to PKC and GAP. Cell. 1991;65:1043–1051. doi: 10.1016/0092-8674(91)90556-e. [DOI] [PubMed] [Google Scholar]
  89. Colter AL, Cutler C, Meckling KA. Fatty acid status and behavioural symptoms of attention deficit hyperactivity disorder in adolescents: a case-control study. Nutr. J. 2008:7–8. doi: 10.1186/1475-2891-7-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Conklin SM, et al. High omega-6 and low omega-3 fatty acids are associated with depressive symptoms and neuroticism. Psychosomatic medicine. 2007;69:932–934. doi: 10.1097/PSY.0b013e31815aaa42. [DOI] [PubMed] [Google Scholar]
  91. Connor KM, et al. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nature medicine. 2007;13:868–873. doi: 10.1038/nm1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Conquer JA, et al. Fatty acid analysis of blood plasma of patients with Alzheimer's disease, other types of dementia, and cognitive impairment. Lipids. 2000;35:1305–1312. doi: 10.1007/s11745-000-0646-3. [DOI] [PubMed] [Google Scholar]
  93. Contreras MA, et al. Nutritional deprivation of alpha-linolenic acid decreases but does not abolish turnover and availability of unacylated docosahexaenoic acid and docosahexaenoyl-CoA in rat brain. J Neurochem. 2000;75:2392–2400. doi: 10.1046/j.1471-4159.2000.0752392.x. [DOI] [PubMed] [Google Scholar]
  94. Corpeleijn E, et al. Insulin acutely upregulates protein expression of the fatty acid transporter CD36 in human skeletal muscle in vivo. Journal of physiology and pharmacology : an official journal of the Polish Physiological Society. 2008;59:77–83. [PubMed] [Google Scholar]
  95. Croset M, et al. Characterization of plasma unsaturated lysophosphatidylcholines in human and rat. The Biochemical journal. 2000;345(Pt 1):61–67. [PMC free article] [PubMed] [Google Scholar]
  96. Cupp D, Kampf JP, Kleinfeld AM. Fatty acid-albumin complexes and the determination of the transport of long chain free fatty acids across membranes. Biochemistry. 2004;43:4473–4481. doi: 10.1021/bi036335l. [DOI] [PubMed] [Google Scholar]
  97. Daniels JL, et al. Fish intake during pregnancy and early cognitive development of offspring. Epidemiology. 2004;15:394–402. doi: 10.1097/01.ede.0000129514.46451.ce. [DOI] [PubMed] [Google Scholar]
  98. Darios F, Davletov B. Omega-3 and omega-6 fatty acids stimulate cell membrane expansion by acting on syntaxin 3. Nature. 2006;440:813–817. doi: 10.1038/nature04598. [DOI] [PubMed] [Google Scholar]
  99. de Gomez Dumm IN, Brenner RR. Oxidative desaturation of alpha-linoleic, linoleic, and stearic acids by human liver microsomes. Lipids. 1975;10:315–317. doi: 10.1007/BF02532451. [DOI] [PubMed] [Google Scholar]
  100. de la Presa Owens S, Innis SM. Docosahexaenoic and arachidonic acid prevent a decrease in dopaminergic and serotoninergic neurotransmitters in frontal cortex caused by a linoleic and alpha-linolenic acid deficient diet in formula-fed piglets. J Nutr. 1999;129:2088–2093. doi: 10.1093/jn/129.11.2088. [DOI] [PubMed] [Google Scholar]
  101. De Leon M, et al. Fatty acid binding protein is induced in neurons of the dorsal root ganglia after peripheral nerve injury. Journal of neuroscience research. 1996;44:283–292. doi: 10.1002/(SICI)1097-4547(19960501)44:3<283::AID-JNR9>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  102. De Smedt-Peyrusse V, et al. Docosahexaenoic acid prevents lipopolysaccharide-induced cytokine production in microglial cells by inhibiting lipopolysaccharide receptor presentation but not its membrane subdomain localization. J Neurochem. 2008;105:296–307. doi: 10.1111/j.1471-4159.2007.05129.x. [DOI] [PubMed] [Google Scholar]
  103. de Vries HF, Kuiper J, de Boer AG, van Verkel TJ, Breimer DD. Characterization of the scavenger receptor on bovine cerebral endothelial cells in vitro. J. Neurochem. 1993;61:1813–1821. doi: 10.1111/j.1471-4159.1993.tb09821.x. [DOI] [PubMed] [Google Scholar]
  104. De Vriese SR, Christophe AB, Maes M. Lowered serum n-3 polyunsaturated fatty acid (PUFA) levels predict the occurrence of postpartum depression: further evidence that lowered n-PUFAs are related to major depression. Life sciences. 2003;73:3181–3187. doi: 10.1016/j.lfs.2003.02.001. [DOI] [PubMed] [Google Scholar]
  105. DeGeorge JJ, et al. Intravenous injection of [1-14C]arachidonate to examine regional brain lipid metabolism in unanesthetized rats. Journal of neuroscience research. 1989;24:413–423. doi: 10.1002/jnr.490240311. [DOI] [PubMed] [Google Scholar]
  106. Degrace-Passilly P, Besnard P. CD36 and taste of fat. Current opinion in clinical nutrition and metabolic care. 2012;15:107–111. doi: 10.1097/MCO.0b013e32834ff19c. [DOI] [PubMed] [Google Scholar]
  107. Dehouck B, et al. Upregulation of the low density lipoprotein receptor at the blood-brain barrier: intercommunications between brain capillary endothelial cells and astrocytes. The Journal of cell biology. 1994;126:465–473. doi: 10.1083/jcb.126.2.465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Dehouck B, et al. A new function for the LDL receptor: transcytosis of LDL across the blood-brain barrier. The Journal of cell biology. 1997;138:877–889. doi: 10.1083/jcb.138.4.877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Delbello MP, et al. A pilot controlled trial of topiramate for mania in children and adolescents with bipolar disorder. J Am Acad Child Adolesc Psychiatry. 2005;44:539–547. doi: 10.1097/01.chi.0000159151.75345.20. [DOI] [PubMed] [Google Scholar]
  110. Delton-Vandenbroucke I, Grammas P, Anderson RE. Polyunsaturated fatty acid metabolism in retinal and cerebral microvascular endothelial cells. Journal of lipid research. 1997;38:147–159. [PubMed] [Google Scholar]
  111. Demar J, et al. Half-lives of docosahexaenoic acid in rat brain phospholipids are prolonged by 15 weeks of nutritional deprivation of n-3 polyunsaturated fatty acids. J Neurochem. 2004;91:1125–1137. doi: 10.1111/j.1471-4159.2004.02789.x. [DOI] [PubMed] [Google Scholar]
  112. Demar JC, Jr, et al. alpha-Linolenic acid does not contribute appreciably to docosahexaenoic acid within brain phospholipids of adult rats fed a diet enriched in docosahexaenoic acid. Journal of neurochemistry. 2005;94:1063–1076. doi: 10.1111/j.1471-4159.2005.03258.x. [DOI] [PubMed] [Google Scholar]
  113. DeMar JC, Jr, et al. Brain elongation of linoleic acid is a negligible source of the arachidonate in brain phospholipids of adult rats. Biochim Biophys Acta. 2006;1761:1050–1059. doi: 10.1016/j.bbalip.2006.06.006. [DOI] [PubMed] [Google Scholar]
  114. Demarne Y, et al. Fat absorption in germ-free and conventional rats artificially deprived of bile secretion. Gut. 1982;23:49–57. doi: 10.1136/gut.23.1.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocrine reviews. 1999;20:649–688. doi: 10.1210/edrv.20.5.0380. [DOI] [PubMed] [Google Scholar]
  116. Devore EE, et al. Dietary intake of fish and omega-3 fatty acids in relation to long-term dementia risk. The American journal of clinical nutrition. 2009;90:170–176. doi: 10.3945/ajcn.2008.27037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Dhopeshwarkar GA, Subramanian C. Biosynthesis of polyunsaturated fatty acids in the developing brain: I. Metabolic transformations of intracranially administered 1-14C linolenic acid. Lipids. 1976;11:67–71. doi: 10.1007/BF02532586. [DOI] [PubMed] [Google Scholar]
  118. Diaz O, et al. The mechanism of docosahexaenoic acid-induced phospholipase D activation in human lymphocytes involves exclusion of the enzyme from lipid rafts. J Biol Chem. 2002;277:39368–39378. doi: 10.1074/jbc.M202376200. [DOI] [PubMed] [Google Scholar]
  119. Diede HE, et al. Identification and characterization of a monoclonal antibody to the membrane fatty acid binding protein. Biochimica et biophysica acta. 1992;1125:13–20. doi: 10.1016/0005-2760(92)90149-p. [DOI] [PubMed] [Google Scholar]
  120. DiRusso CC, et al. Comparative biochemical studies of the murine fatty acid transport proteins (FATP) expressed in yeast. The Journal of biological chemistry. 2005;280:16829–16837. doi: 10.1074/jbc.M409598200. [DOI] [PubMed] [Google Scholar]
  121. Domenichiello AF, et al. Whole body synthesis rates of DHA from alpha-linolenic acid are greater than brain DHA accretion and uptake rates in adult rats. J Lipid Res. 2014;55:62–74. doi: 10.1194/jlr.M042275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Downes CP, Currie RA. Lipid signalling. Current biology : CB. 1998;8:R865–R867. doi: 10.1016/s0960-9822(07)00546-5. [DOI] [PubMed] [Google Scholar]
  123. Dreja K, et al. Cholesterol depletion disrupts caveolae and differentially impairs agonist-induced arterial contraction. Arterioscler Thromb Vasc Biol. 2002;22:1267–1272. doi: 10.1161/01.atv.0000023438.32585.a1. [DOI] [PubMed] [Google Scholar]
  124. Drover VA, et al. CD36 mediates both cellular uptake of very long chain fatty acids and their intestinal absorption in mice. The Journal of biological chemistry. 2008;283:13108–13115. doi: 10.1074/jbc.M708086200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Dubois T, et al. Fatty acids differentially affect serotonin receptor and transporter binding in the rat brain. Neuroscience. 2006;139:1397–1403. doi: 10.1016/j.neuroscience.2006.02.068. [DOI] [PubMed] [Google Scholar]
  126. Dullemeijer C, et al. n 3 fatty acid proportions in plasma and cognitive performance in older adults. The American journal of clinical nutrition. 2007;86:1479–1485. doi: 10.1093/ajcn/86.5.1479. [DOI] [PubMed] [Google Scholar]
  127. Duraisamy Y, et al. Differential incorporation of docosahexaenoic acid into distinct cholesterol-rich membrane raft domains. Biochemical and biophysical research communications. 2007;360:885–890. doi: 10.1016/j.bbrc.2007.06.152. [DOI] [PubMed] [Google Scholar]
  128. Edelstein C. General properties of plasma lipoproteins and apolipoproteins, Vol. New York, NY: Marcel Dekker ; 1986. [Google Scholar]
  129. Edmond J. Essential polyunsaturated fatty acids and the barrier to the brain: the components of a model for transport. Journal of molecular neuroscience : MN. 2001;16:181–193. doi: 10.1385/JMN:16:2-3:181. discussion 215-21. [DOI] [PubMed] [Google Scholar]
  130. Edwards R, et al. Omega-3 polyunsaturated fatty acid levels in the diet and in red blood cell membranes of depressed patients. Journal of affective disorders. 1998;48:149–155. doi: 10.1016/s0165-0327(97)00166-3. [DOI] [PubMed] [Google Scholar]
  131. Eisensamer B, et al. Antidepressants and antipsychotic drugs colocalize with 5-HT3 receptors in raft-like domains. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2005;25:10198–10206. doi: 10.1523/JNEUROSCI.2460-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Ek BA, et al. Fatty acid binding proteins reduce 15-lipoxygenase-induced oxygenation of linoleic acid and arachidonic acid. Biochimica et biophysica acta. 1997;1346:75–85. doi: 10.1016/s0005-2760(97)00021-0. [DOI] [PubMed] [Google Scholar]
  133. Eldho NV, et al. Polyunsaturated docosahexaenoic vs docosapentaenoic acid-differences in lipid matrix properties from the loss of one double bond. J Am Chem Soc. 2003;125:6409–6421. doi: 10.1021/ja029029o. [DOI] [PubMed] [Google Scholar]
  134. Emken EA, Adlof RO, Gulley RM. Dietary linoleic acid influences desaturation and acylation of deuterium-labeled linoleic and linolenic acids in young adult males. Biochimica et biophysica acta. 1994;1213:277–288. doi: 10.1016/0005-2760(94)00054-9. [DOI] [PubMed] [Google Scholar]
  135. Emken EA, Adlof RO, Rakoff H, Rohwedder WK. Synthesis and Application of Isotopically Labelled COmpounds 1988. Amsterdam: Elsevier Science Publishers; 1989a. Metabolism of deuterium-labeled linolenic, linoleic, oleic, stearic, and palmitic acid in human subjects; pp. 713–716. [Google Scholar]
  136. Emken EA, Adlof RO, Rakoff H, Rohwedder WK. Metabolism of deuterium-labeled linolenic, linoleic, oleic, stearic, and palmitic acid in human subjects, Vol. Amsterdam: Elsevier Science Publishers; 1989b. [Google Scholar]
  137. Eskelinen MH, et al. Fat intake at midlife and cognitive impairment later in life: a population-based CAIDE study. International journal of geriatric psychiatry. 2008;23:741–747. doi: 10.1002/gps.1969. [DOI] [PubMed] [Google Scholar]
  138. Esposito G, et al. Imaging signal transduction via arachidonic acid in the human brain during visual stimulation, by means of positron emission tomography. NeuroImage. 2007a;34:1342–1351. doi: 10.1016/j.neuroimage.2006.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Esposito G, et al. Imaging signal transduction via arachidonic acid in the human brain during visual stimulation, by means of positron emission tomography. Neuroimage. 2007b;34:1342–1351. doi: 10.1016/j.neuroimage.2006.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Esposito G, et al. Imaging neuroinflammation in Alzheimer's disease with radiolabeled arachidonic acid and PET. J Nucl Med. 2008;49:1414–1421. doi: 10.2967/jnumed.107.049619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Evans DR, et al. Red blood cell membrane essential fatty acid metabolism in early psychotic patients following antipsychotic drug treatment. Prostaglandins Leukot Essent Fatty Acids. 2003a;69:393–399. doi: 10.1016/j.plefa.2003.08.010. [DOI] [PubMed] [Google Scholar]
  142. Evans DR, et al. Red blood cell membrane essential fatty acid metabolism in early psychotic patients following antipsychotic drug treatment. Prostaglandins, leukotrienes, and essential fatty acids. 2003b;69:393–399. doi: 10.1016/j.plefa.2003.08.010. [DOI] [PubMed] [Google Scholar]
  143. Fan YY, et al. Dietary (n-3) polyunsaturated fatty acids remodel mouse T-cell lipid rafts. The Journal of nutrition. 2003;133:1913–1920. doi: 10.1093/jn/133.6.1913. [DOI] [PubMed] [Google Scholar]
  144. Fan YY, et al. Dietary docosahexaenoic acid suppresses T cell protein kinase C theta lipid raft recruitment and IL-2 production. Journal of immunology. 2004;173:6151–6160. doi: 10.4049/jimmunol.173.10.6151. [DOI] [PubMed] [Google Scholar]
  145. Febbraio M, et al. The impact of overexpression and deficiency of fatty acid translocase (FAT)/CD36. Molecular and cellular biochemistry. 2002;239:193–197. [PubMed] [Google Scholar]
  146. Fernstrom JD. Effects of dietary polyunsaturated fatty acids on neuronal function. Lipids. 1999;34:161–169. doi: 10.1007/s11745-999-0350-3. [DOI] [PubMed] [Google Scholar]
  147. Ferrante A, et al. Neutrophil migration inhibitory properties of polyunsaturated fatty acids. The role of fatty acid structure, metabolism, and possible second messenger systems. The Journal of clinical investigation. 1994;93:1063–1070. doi: 10.1172/JCI117056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Fielding BA, et al. Postprandial lipemia: the origin of an early peak studied by specific dietary fatty acid intake during sequential meals. The American journal of clinical nutrition. 1996;63:36–41. doi: 10.1093/ajcn/63.1.36. [DOI] [PubMed] [Google Scholar]
  149. Fielding PE, Fielding CJ. Dynamics of lipoprotein transport in the human circulatory system. Biochemistry of Lipids, Lipoproteins, and Membranes. 2008:533–553. [Google Scholar]
  150. Frasure-Smith N, Lesperance F, Julien P. Major depression is associated with lower omega-3 fatty acid levels in patients with recent acute coronary syndromes. Biol Psychiatry. 2004;55:891–896. doi: 10.1016/j.biopsych.2004.01.021. [DOI] [PubMed] [Google Scholar]
  151. Fukaya T, et al. Arachidonic acid preserves hippocampal neuron membrane fluidity in senescent rats. Neurobiology of aging. 2007;28:1179–1186. doi: 10.1016/j.neurobiolaging.2006.05.023. [DOI] [PubMed] [Google Scholar]
  152. Gawrisch K, Eldho NV, Holte LL. The structure of DHA in phospholipid membranes. Lipids. 2003;38:445–452. doi: 10.1007/s11745-003-1082-0. [DOI] [PubMed] [Google Scholar]
  153. Gazzah N, et al. Decrease of brain phospholipid synthesis in free-moving n-3 fatty acid deficient rats. J Neurochem. 1995;64:908–918. doi: 10.1046/j.1471-4159.1995.64020908.x. [DOI] [PubMed] [Google Scholar]
  154. Genedani S, et al. Computer-assisted image analysis of caveolin-1 involvement in the internalization process of adenosine A2A-dopamine D2 receptor heterodimers. J Mol Neurosci. 2005;26:177–184. doi: 10.1385/JMN:26:2-3:177. [DOI] [PubMed] [Google Scholar]
  155. Gerster H. Can adults adequately convert alpha-linolenic acid (18:3n-3) to eicosapentaenoic acid (20:5n-3) and docosahexaenoic acid (22:6n-3)? International journal for vitamin and nutrition research. Internationale Zeitschrift fur Vitamin-und Ernahrungsforschung. Journal international de vitaminologie et de nutrition. 1998;68:159–173. [PubMed] [Google Scholar]
  156. Geyeregger R, et al. Polyunsaturated fatty acids interfere with formation of the immunological synapse. J Leukoc Biol. 2005;77:680–688. doi: 10.1189/jlb.1104687. [DOI] [PubMed] [Google Scholar]
  157. Gibbons GF, et al. Extracellular fatty acids are not utilized directly for the synthesis of very-low-density lipoprotein in primary cultures of rat hepatocytes. Biochem J. 1992;287(Pt 3):749–753. doi: 10.1042/bj2870749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Gillingham LG, et al. Dietary oils and FADS1-FADS2 genetic variants modulate [13C]alpha-linolenic acid metabolism and plasma fatty acid composition. Am J Clin Nutr. 2013;97:195–207. doi: 10.3945/ajcn.112.043117. [DOI] [PubMed] [Google Scholar]
  159. Giovacchini G, et al. Brain incorporation of [11C]arachidonic acid in young healthy humans measured with positron emission tomography. J Cereb Blood Flow Metab. 2002;22:1453–1462. doi: 10.1097/01.WCB.0000033209.60867.7A. [DOI] [PubMed] [Google Scholar]
  160. Giovacchini G, et al. Brain incorporation of 11C-arachidonic acid, blood volume, and blood flow in healthy aging: a study with partial-volume correction. J Nucl Med. 2004;45:1471–1479. [PubMed] [Google Scholar]
  161. Glaser C, Heinrich J, Koletzko B. Role of FADS1 and FADS2 polymorphisms in polyunsaturated fatty acid metabolism. Metabolism. 2010;59:993–999. doi: 10.1016/j.metabol.2009.10.022. [DOI] [PubMed] [Google Scholar]
  162. Glatz JF, van der Vusse GJ. Cellular fatty acid-binding proteins: their function and physiological significance. Progress in lipid research. 1996;35:243–282. doi: 10.1016/s0163-7827(96)00006-9. [DOI] [PubMed] [Google Scholar]
  163. Glatz JF, et al. Cellular lipid binding proteins as facilitators and regulators of lipid metabolism. Molecular and cellular biochemistry. 2002;239:3–7. [PubMed] [Google Scholar]
  164. Gordon JI, et al. The nucleotide sequence of rat liver fatty acid binding protein mRNA. The Journal of biological chemistry. 1983;258:3356–3363. [PubMed] [Google Scholar]
  165. Green JT, Orr SK, Bazinet RP. The emerging role of group VI calcium-independent phospholipase A2 in releasing docosahexaenoic acid from brain phospholipids. Journal of lipid research. 2008;49:939–944. doi: 10.1194/jlr.R700017-JLR200. [DOI] [PubMed] [Google Scholar]
  166. Green P, Yavin E. Elongation, desaturation, and esterification of essential fatty acids by fetal rat brain in vivo. Journal of lipid research. 1993;34:2099–2107. [PubMed] [Google Scholar]
  167. Green P, et al. Red cell membrane omega-3 fatty acids are decreased in nondepressed patients with social anxiety disorder. Eur Neuropsychopharmacol. 2006;16:107–113. doi: 10.1016/j.euroneuro.2005.07.005. [DOI] [PubMed] [Google Scholar]
  168. Grimm MO, et al. Docosahexaenoic acid reduces amyloid beta production via multiple pleiotropic mechanisms. The Journal of biological chemistry. 2011;286:14028–14039. doi: 10.1074/jbc.M110.182329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Guo J, et al. Selective transport of long-chain fatty acids by FAT/CD36 in skeletal muscle of broilers. Animal : an international journal of animal bioscience. 2013;7:422–429. doi: 10.1017/S1751731112001619. [DOI] [PubMed] [Google Scholar]
  170. Gurzell EA, et al. DHA-enriched fish oil targets B cell lipid microdomains and enhances ex vivo and in vivo B cell function. J Leukoc Biol. 2013;93:463–470. doi: 10.1189/jlb.0812394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Gustafsson PA, et al. Breastfeeding, very long polyunsaturated fatty acids (PUFA) and IQ at 6 1/2 years of age. Acta paediatrica. 2004;93:1280–1287. [PubMed] [Google Scholar]
  172. Hague TA, Christoffersen BO. Effect of dietary fats on arachidonic acid and eicosapentaenoic acid biosynthesis and conversion to C22 fatty acids in isolated liver cells. Biochim Biophys Acta. 1984a;796:205–217. doi: 10.1016/0005-2760(84)90349-7. [DOI] [PubMed] [Google Scholar]
  173. Hague TA, Christoffersen BO. Effect of dietary fats on arachidonic acid and eicosapentaenoic acid biosynthesis and conversion to C22 fatty acids in isolated liver cells. Biochim Biophys Acta. 1984b;796:205–217. doi: 10.1016/0005-2760(84)90349-7. [DOI] [PubMed] [Google Scholar]
  174. Hague TA, Christoffersen BO. Evidence for peroxisomal retrocon-version of adrenic acid (22:4n6) and docosahexaenoic acid (22:6n3) in isolated liver cells. Biochim Biophys Acta. 1986;875:165–173. doi: 10.1016/0005-2760(86)90165-7. [DOI] [PubMed] [Google Scholar]
  175. Hamilton JA, et al. Fatty acid transport: the diffusion mechanism in model and biological membranes. Journal of molecular neuroscience : MN. 2001;16:99–108. doi: 10.1385/JMN:16:2-3:99. discussion 151-7. [DOI] [PubMed] [Google Scholar]
  176. Hamilton JA, Guo W, Kamp F. Mechanism of cellular uptake of long-chain fatty acids: Do we need cellular proteins? Molecular and cellular biochemistry. 2002;239:17–23. [PubMed] [Google Scholar]
  177. Hamilton JA. New insights into the roles of proteins and lipids in membrane transport of fatty acids. Prostaglandins, leukotrienes, and essential fatty acids. 2007;77:355–361. doi: 10.1016/j.plefa.2007.10.020. [DOI] [PubMed] [Google Scholar]
  178. Hamilton JA, Brunaldi K. A model for fatty acid transport into the brain. Journal of molecular neuroscience : MN. 2007;33:12–17. doi: 10.1007/s12031-007-0050-3. [DOI] [PubMed] [Google Scholar]
  179. Hamosh M, et al. Pharyngeal lipase and digestion of dietary triglyceride in man. The Journal of clinical investigation. 1975;55:908–913. doi: 10.1172/JCI108019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Hamosh M. Lingual and gastric lipases. Nutrition. 1990;6:421–428. [PubMed] [Google Scholar]
  181. Hanhoff T, Lücke C, Spener F. Insights into binding of fatty acids by fatty acid binding proteins. Molecular and Cellular Biochemistry. 2002;239:45–54. [PubMed] [Google Scholar]
  182. Hansen JB, et al. Effects of highly purified eicosapentaenoic acid and docosahexaenoic acid on fatty acid absorption, incorporation into serum phospholipids and postprandial triglyceridemia. Lipids. 1998;33:131–138. doi: 10.1007/s11745-998-0188-8. [DOI] [PubMed] [Google Scholar]
  183. Harauma A, Moriguchi T. Dietary n-3 fatty acid deficiency in mice enhances anxiety induced by chronic mild stress. Lipids. 2011;46:409–416. doi: 10.1007/s11745-010-3523-z. [DOI] [PubMed] [Google Scholar]
  184. Harslof LB, et al. FADS genotype and diet are important determinants of DHA status: a cross-sectional study in Danish infants. Am J Clin Nutr. 2013;97:1403–1410. doi: 10.3945/ajcn.113.058685. [DOI] [PubMed] [Google Scholar]
  185. Havel RJ. Lipoproteins and lipid transport. Advances in experimental medicine and biology. 1975;63:37–59. doi: 10.1007/978-1-4684-3258-9_3. [DOI] [PubMed] [Google Scholar]
  186. Helland IB, et al. Maternal supplementation with very-long-chain n-3 fatty acids during pregnancy and lactation augments children's IQ at 4 years of age. Pediatrics. 2003;111:e39–e44. doi: 10.1542/peds.111.1.e39. [DOI] [PubMed] [Google Scholar]
  187. Herz J, Bock HH. Lipoprotein receptors in the nervous system. Annual review of biochemistry. 2002;71:405–434. doi: 10.1146/annurev.biochem.71.110601.135342. [DOI] [PubMed] [Google Scholar]
  188. Hesse C, et al. Measurement of apolipoprotein E (apoE) in cerebrospinal fluid. Neurochemical research. 2000;25:511–517. doi: 10.1023/a:1007516210548. [DOI] [PubMed] [Google Scholar]
  189. Heude B, Ducimetiere P, Berr C. Cognitive decline and fatty acid composition of erythrocyte membranes--The EVA Study. The American journal of clinical nutrition. 2003;77:803–808. doi: 10.1093/ajcn/77.4.803. [DOI] [PubMed] [Google Scholar]
  190. Hibbeln JR. Fish consumption and major depression. Lancet. 1998;351:1213. doi: 10.1016/S0140-6736(05)79168-6. [DOI] [PubMed] [Google Scholar]
  191. Hibbeln JR. Seafood consumption, the DHA content of mothers' milk and prevalence rates of postpartum depression: a cross-national, ecological analysis. J Affect Disord. 2002a;69:15–29. doi: 10.1016/s0165-0327(01)00374-3. [DOI] [PubMed] [Google Scholar]
  192. Hibbeln JR. Seafood consumption, the DHA content of mothers' milk and prevalence rates of postpartum depression: a cross-national, ecological analysis. Journal of affective disorders. 2002b;69:15–29. doi: 10.1016/s0165-0327(01)00374-3. [DOI] [PubMed] [Google Scholar]
  193. Hodson L, Skeaff CM, Fielding BA. Fatty acid composition of adipose tissue and blood in humans and its use as a biomarker of dietary intake. Prog Lipid Res. 2008;47:348–380. doi: 10.1016/j.plipres.2008.03.003. [DOI] [PubMed] [Google Scholar]
  194. Hohoff C, Spener F. Fatty acid binding protein and mammary-derived growth inhibitor. Fett (Lipid) 1998:6. doi: 10.1074/jbc.271.33.19943. [DOI] [PubMed] [Google Scholar]
  195. Hollander D, Chow SL, Dadufalza VD. Intestinal absorption of free oleic acid in the unanesthetized rat: evidence for a saturable component? Canadian journal of physiology and pharmacology. 1984;62:1136–1140. doi: 10.1139/y84-190. [DOI] [PubMed] [Google Scholar]
  196. Holt PR. Fats and bile salts. J. Am. Diet. Assoc. . 1971;60:491–498. [PubMed] [Google Scholar]
  197. Honigmann G, Schimke E, Beitz J, Mest HJ, Schliack V. Influence of a diet rich in linolenic acid on lipids, thrombocyte aggregation, and prostaglandins in type I (insulin-dependent) diabetes. Diabetologia. 1982;23:175. (abstract) [Google Scholar]
  198. Horrobin DF, Glen AI, Vaddadi K. The membrane hypothesis of schizophrenia. Schizophrenia research. 1994;13:195–207. doi: 10.1016/0920-9964(94)90043-4. [DOI] [PubMed] [Google Scholar]
  199. Hostetler HA, et al. L-FABP directly interacts with PPARalpha in cultured primary hepatocytes. Journal of lipid research. 2009;50:1663–1675. doi: 10.1194/jlr.M900058-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Hostetler HA, et al. Glucose regulates fatty acid binding protein interaction with lipids and peroxisome proliferator-activated receptor alpha. Journal of lipid research. 2010;51:3103–3116. doi: 10.1194/jlr.M005041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Hou TY, et al. n-3 polyunsaturated fatty acids suppress phosphatidylinositol 4,5-bisphosphate-dependent actin remodelling during CD4+ T-cell activation. Biochem J. 2012;443:27–37. doi: 10.1042/BJ20111589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Howe P, et al. Dietary intake of long-chain omega-3 polyunsaturated fatty acids: contribution of meat sources. Nutrition. 2006;22:47–53. doi: 10.1016/j.nut.2005.05.009. [DOI] [PubMed] [Google Scholar]
  203. Huan M, et al. Suicide attempt and n-3 fatty acid levels in red blood cells: a case control study in China. Biol Psychiatry. 2004;56:490–496. doi: 10.1016/j.biopsych.2004.06.028. [DOI] [PubMed] [Google Scholar]
  204. Huang TL, et al. Benefits of fatty fish on dementia risk are stronger for those without APOE epsilon4. Neurology. 2005;65:1409–1414. doi: 10.1212/01.wnl.0000183148.34197.2e. [DOI] [PubMed] [Google Scholar]
  205. Hultin M, et al. Intravenous lipid emulsions: removal mechanisms as compared to chylomicrons. Journal of lipid research. 1995;36:2174–2184. [PubMed] [Google Scholar]
  206. Huster D, Arnold K, Gawrisch K. Influence of docosahexaenoic acid and cholesterol on lateral lipid organization in phospholipid mixtures. Biochemistry. 1998;37:17299–17308. doi: 10.1021/bi980078g. [DOI] [PubMed] [Google Scholar]
  207. Ibrahimi A, et al. Expression of the CD36 homolog (FAT) in fibroblast cells: effects on fatty acid transport. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:2646–2651. doi: 10.1073/pnas.93.7.2646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Igarashi M, et al. Low liver conversion rate of alpha-linolenic to docosahexaenoic acid in awake rats on a high-docosahexaenoate-containing diet. J Lipid Res. 2006;47:1812–1822. doi: 10.1194/jlr.M600030-JLR200. [DOI] [PubMed] [Google Scholar]
  209. Igarashi M, et al. Docosahexaenoic acid synthesis from alpha-linolenic acid by rat brain is unaffected by dietary n-3 PUFA deprivation. Journal of lipid research. 2007a;48:1150–1158. doi: 10.1194/jlr.M600549-JLR200. [DOI] [PubMed] [Google Scholar]
  210. Igarashi M, et al. Docosahexaenoic acid synthesis from alpha-linolenic acid by rat brain is unaffected by dietary n-3 PUFA deprivation. J Lipid Res. 2007b;48:1150–1158. doi: 10.1194/jlr.M600549-JLR200. [DOI] [PubMed] [Google Scholar]
  211. Igarashi M, et al. Dietary n-3 PUFA deprivation for 15 weeks upregulates elongase and desaturase expression in rat liver but not brain. J Lipid Res. 2007c;48:2463–2470. doi: 10.1194/jlr.M700315-JLR200. [DOI] [PubMed] [Google Scholar]
  212. Igarashi M, et al. Rat heart cannot synthesize docosahexaenoic acid from circulating alpha-linolenic acid because it lacks elongase-2. Journal of lipid research. 2008;49:1735–1745. doi: 10.1194/jlr.M800093-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Igarashi M, et al. Kinetics of eicosapentaenoic acid in brain, heart and liver of conscious rats fed a high n-3 PUFA containing diet. Prostaglandins Leukot Essent Fatty Acids. 2013;89:403–412. doi: 10.1016/j.plefa.2013.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Ikeda I, et al. Digestion and lymphatic transport of eicosapentaenoic and docosahexaenoic acids given in the form of triacylglycerol, free acid and ethyl ester in rats. Biochimica et biophysica acta. 1995;1259:297–304. doi: 10.1016/0005-2760(95)00180-8. [DOI] [PubMed] [Google Scholar]
  215. Ikeda I, et al. Effects of dietary alpha-linolenic, eicosapentaenoic and docosahexaenoic acids on hepatic lipogenesis and beta-oxidation in rats. Bioscience, biotechnology, and biochemistry. 1998;62:675–680. doi: 10.1271/bbb.62.675. [DOI] [PubMed] [Google Scholar]
  216. Ikeda I, et al. Reduced hepatic triglyceride secretion in rats fed docosahexaenoic acid-rich fish oil suppresses postprandial hypertriglyceridemia. The Journal of nutrition. 2001;131:1159–1164. doi: 10.1093/jn/131.4.1159. [DOI] [PubMed] [Google Scholar]
  217. Ikemoto A, et al. Reversibility of n-3 fatty acid deficiency-induced alterations of learning behavior in the rat: level of n-6 fatty acids as another critical factor. Journal of lipid research. 2001;42:1655–1663. [PubMed] [Google Scholar]
  218. Ilegems E, et al. Ligand binding transmits conformational changes across the membrane-spanning region to the intracellular side of the 5-HT3 serotonin receptor. Chembiochem. 2005;6:2180–2185. doi: 10.1002/cbic.200500191. [DOI] [PubMed] [Google Scholar]
  219. Indu M, Ghafoorunissa N-3 fatty acids in Indian diets-comparison of the effects of precursor (alpha-linolenic acid) vs product (long chain n-3 polyunsaturated fatty acids) Nutr Res. 1992;12:569–582. [Google Scholar]
  220. Innis SM. Dietary (n-3) fatty acids and brain development. J Nutr. 2007;137:855–859. doi: 10.1093/jn/137.4.855. [DOI] [PubMed] [Google Scholar]
  221. Irvine RF, Letcher AJ, Dawson RM. Fatty acid stimulation of membrane phosphatidylinositol hydrolysis by brain phosphatidylinositol phosphodiesterase. The Biochemical journal. 1979;178:497–500. doi: 10.1042/bj1780497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Jayanthi LD, Samuvel DJ, Ramamoorthy S. Regulated internalization and phosphorylation of the native norepinephrine transporter in response to phorbol esters. Evidence for localization in lipid rafts and lipid raft-mediated internalization. The Journal of biological chemistry. 2004;279:19315–19326. doi: 10.1074/jbc.M311172200. [DOI] [PubMed] [Google Scholar]
  223. Jia Z, et al. The fatty acid transport protein (FATP) family: very long chain acyl-CoA synthetases or solute carriers? J Mol Neurosci. 2007;33:25–31. doi: 10.1007/s12031-007-0038-z. [DOI] [PubMed] [Google Scholar]
  224. Jonas A, Phillips MC. Biochemistry of Lipids, Lipoproteins and Membranes. 5th Edition. Elsevier Science; 2008. Lipoprotein structure; pp. 485–506. [Google Scholar]
  225. Jones CR, et al. Preferential in vivo incorporation of [3H]arachidonic acid from blood in rat brain synaptosomal fractions before and after cholinergic stimulation. Journal of neurochemistry. 1996;67:822–829. doi: 10.1046/j.1471-4159.1996.67020822.x. [DOI] [PubMed] [Google Scholar]
  226. Jones KT, Zhen J, Reith ME. Importance of cholesterol in dopamine transporter function. J Neurochem. 2012;123:700–715. doi: 10.1111/jnc.12007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Jones PJ, Pencharz PB, Clandinin MT. Absorption of 13C-labeled stearic, oleic, and linoleic acids in humans: application to breath tests. The Journal of laboratory and clinical medicine. 1985;105:647–652. [PubMed] [Google Scholar]
  228. Jorgensen MH, et al. Is there a relation between docosahexaenoic acid concentration in mothers' milk and visual development in term infants? Journal of pediatric gastroenterology and nutrition. 2001;32:293–296. doi: 10.1097/00005176-200103000-00011. [DOI] [PubMed] [Google Scholar]
  229. Jump DB, et al. Coordinate regulation of glycolytic and lipogenic gene expression by polyunsaturated fatty acids. J Lipid Res. 1994;35:1076–1084. [PubMed] [Google Scholar]
  230. Jump DB. Dietary polyunsaturated fatty acids and regulation of gene transcription. Curr Opin Lipidol. 2002;13:155–164. doi: 10.1097/00041433-200204000-00007. [DOI] [PubMed] [Google Scholar]
  231. Jump DB. N-3 polyunsaturated fatty acid regulation of hepatic gene transcription. Current opinion in lipidology. 2008;19:242–247. doi: 10.1097/MOL.0b013e3282ffaf6a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Jump DB, et al. Docosahexaenoic acid (DHA) and hepatic gene transcription. Chem Phys Lipids. 2008a;153:3–13. doi: 10.1016/j.chemphyslip.2008.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Jump DB, et al. Docosahexaenoic acid (DHA) and hepatic gene transcription. Chemistry and physics of lipids. 2008b;153:3–13. doi: 10.1016/j.chemphyslip.2008.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Jumpsen J, Clandinin MT. Brain Development. Champaign, IL: AOCS Press; 1995. Brain Lipids. [Google Scholar]
  235. Kaddurah-Daouk R, et al. Metabolomic mapping of atypical antipsychotic effects in schizophrenia. Mol Psychiatry. 2007;12:934–945. doi: 10.1038/sj.mp.4002000. [DOI] [PubMed] [Google Scholar]
  236. Kalipatnapu S, Chattopadhyay A. Membrane organization of the human serotonin(1A) receptor monitored by detergent insolubility using GFP fluorescence. Molecular membrane biology. 2005;22:539–547. doi: 10.1080/09687860500421738. [DOI] [PubMed] [Google Scholar]
  237. Kalmijn S, et al. Polyunsaturated fatty acids, antioxidants, and cognitive function in very old men. American journal of epidemiology. 1997a;145:33–41. doi: 10.1093/oxfordjournals.aje.a009029. [DOI] [PubMed] [Google Scholar]
  238. Kalmijn S, et al. Dietary fat intake and the risk of incident dementia in the Rotterdam Study. Annals of neurology. 1997b;42:776–782. doi: 10.1002/ana.410420514. [DOI] [PubMed] [Google Scholar]
  239. Kalmijn S, et al. Dietary intake of fatty acids and fish in relation to cognitive performance at middle age. Neurology. 2004;62:275–280. doi: 10.1212/01.wnl.0000103860.75218.a5. [DOI] [PubMed] [Google Scholar]
  240. Kampf JP, Cupp D, Kleinfeld AM. Different mechanisms of free fatty acid flip-flop and dissociation revealed by temperature and molecular species dependence of transport across lipid vesicles. The Journal of biological chemistry. 2006;281:21566–21574. doi: 10.1074/jbc.M602067200. [DOI] [PubMed] [Google Scholar]
  241. Kane JP. Apolipoprotein B: structural and metabolic heterogeneity. Annual review of physiology. 1983;45:637–650. doi: 10.1146/annurev.ph.45.030183.003225. [DOI] [PubMed] [Google Scholar]
  242. Kaur G, et al. Short-term docosapentaenoic acid (22:5 n-3) supplementation increases tissue docosapentaenoic acid, DHA and EPA concentrations in rats. The British journal of nutrition. 2010;103:32–37. doi: 10.1017/S0007114509991334. [DOI] [PubMed] [Google Scholar]
  243. Kaur G, et al. Docosapentaenoic acid (22:5n-3): a review of its biological effects. Progress in lipid research. 2011;50:28–34. doi: 10.1016/j.plipres.2010.07.004. [DOI] [PubMed] [Google Scholar]
  244. Kearns SD, Haag M. The effect of omega-3 fatty acids on Ca-ATPase in rat cerebral cortex. Prostaglandins, leukotrienes, and essential fatty acids. 2002;67:303–308. doi: 10.1054/plef.2002.0433. [DOI] [PubMed] [Google Scholar]
  245. Kiecolt-Glaser JK, et al. Depressive symptoms, omega-6:omega-3 fatty acids, and inflammation in older adults. Psychosomatic medicine. 2007;69:217–224. doi: 10.1097/PSY.0b013e3180313a45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Kim HW, et al. Regulation of rat brain polyunsaturated fatty acid (PUFA) metabolism during graded dietary n-3 PUFA deprivation. Prostaglandins Leukot Essent Fatty Acids. 2011;85:361–368. doi: 10.1016/j.plefa.2011.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Kim W, et al. n-3 polyunsaturated fatty acids suppress the localization and activation of signaling proteins at the immunological synapse in murine CD4+ T cells by affecting lipid raft formation. Journal of immunology. 2008a;181:6236–6243. doi: 10.4049/jimmunol.181.9.6236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Kim W, et al. n-3 polyunsaturated fatty acids suppress the localization and activation of signaling proteins at the immunological synapse in murine CD4+ T cells by affecting lipid raft formation. J Immunol. 2008b;181:6236–6243. doi: 10.4049/jimmunol.181.9.6236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Kim W, et al. Regulatory activity of polyunsaturated fatty acids in T-cell signaling. Prog Lipid Res. 2010;49:250–261. doi: 10.1016/j.plipres.2010.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Kliewer SA, et al. Peroxisome proliferator-activated receptors: from genes to physiology. Recent progress in hormone research. 2001;56:239–263. doi: 10.1210/rp.56.1.239. [DOI] [PubMed] [Google Scholar]
  251. Kobe F, et al. Stimulation-and palmitoylation-dependent changes in oligomeric conformation of serotonin 5-HT1A receptors. Biochimica et biophysica acta. 2008;1783:1503–1516. doi: 10.1016/j.bbamcr.2008.02.021. [DOI] [PubMed] [Google Scholar]
  252. Koletzko B, et al. Genetically determined variation in polyunsaturated Fatty Acid metabolism may result in different dietary requirements. Nestle Nutr Workshop Ser Pediatr Program. 2008;62:35–49. doi: 10.1159/000146246. [DOI] [PubMed] [Google Scholar]
  253. Kopecky J, et al. n-3 PUFA: bioavailability and modulation of adipose tissue function. Proc Nutr Soc. 2009;68:361–369. doi: 10.1017/S0029665109990231. [DOI] [PubMed] [Google Scholar]
  254. Kozak KR, et al. Amino acid determinants in cyclooxygenase-2 oxygenation of the endocannabinoid 2-arachidonylglycerol. The Journal of biological chemistry. 2001;276:30072–30077. doi: 10.1074/jbc.M104467200. [DOI] [PubMed] [Google Scholar]
  255. Kuperstein F, et al. Overexpression of dopamine receptor genes and their products in the postnatal rat brain following maternal n-3 fatty acid dietary deficiency. J Neurochem. 2005;95:1550–1562. doi: 10.1111/j.1471-4159.2005.03513.x. [DOI] [PubMed] [Google Scholar]
  256. Kuperstein F, Eilam R, Yavin E. Altered expression of key dopaminergic regulatory proteins in the postnatal brain following perinatal n-3 fatty acid dietary deficiency. J Neurochem. 2008;106:662–671. doi: 10.1111/j.1471-4159.2008.05418.x. [DOI] [PubMed] [Google Scholar]
  257. Lagarde M, Bernoud N, Brossard N, Lemaitre-Delaunay D, Thies F, Croset M, Lecerf J. Lysophosphatidylcholine as a preferred carrier form of docosahexaenoic acid to the brain. J. Mol. Neurosci. 2001;16:2–3. doi: 10.1385/JMN:16:2-3:201. [DOI] [PubMed] [Google Scholar]
  258. Lalovic A, et al. Altered expression of lipid metabolism and immune response genes in the frontal cortex of suicide completers. J Affect Disord. 2010;120:24–31. doi: 10.1016/j.jad.2009.04.007. [DOI] [PubMed] [Google Scholar]
  259. Lalush I, Bar H, Zakaria I, Eichler S, Shimoni E. Utilization of Amylose-Lipid Complexes as Molecular Nanocapsules for Conjugated Linoleic Acid. Biomacromolecules. 2005;6:121–130. doi: 10.1021/bm049644f. [DOI] [PubMed] [Google Scholar]
  260. Langelier B, et al. Long chain-polyunsaturated fatty acids modulate membrane phospholipid composition and protein localization in lipid rafts of neural stem cell cultures. Journal of cellular biochemistry. 2010;110:1356–1364. doi: 10.1002/jcb.22652. [DOI] [PubMed] [Google Scholar]
  261. Larque E, Demmelmair H, Koletzko B. Perinatal supply and metabolism of long-chain polyunsaturated fatty acids: importance for the early development of the nervous system. Annals of the New York Academy of Sciences. 2002;967:299–310. doi: 10.1111/j.1749-6632.2002.tb04285.x. [DOI] [PubMed] [Google Scholar]
  262. Laugerette F, et al. CD36 involvement in orosensory detection of dietary lipids, spontaneous fat preference, and digestive secretions. The Journal of clinical investigation. 2005;115:3177–3184. doi: 10.1172/JCI25299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Lauritzen L, et al. The essentiality of long chain n-3 fatty acids in relation to development and function of the brain and retina. Prog Lipid Res. 2001;40:1–94. doi: 10.1016/s0163-7827(00)00017-5. [DOI] [PubMed] [Google Scholar]
  264. Lee H, et al. Topiramate does not Alter the Kinetics of Arachidonic or Docosahexaenoic Acid in Brain Phospholipids of the Unanesthetized Rat. Neurochem Res. 2005;30:677–683. doi: 10.1007/s11064-005-2756-3. [DOI] [PubMed] [Google Scholar]
  265. Lee H, et al. Chronic fluoxetine increases cytosolic phospholipase A2 activity and arachidonic acid turnover in brain phospholipids of the unanesthetized rat. Psychopharmacology. 2006;190:103–115. doi: 10.1007/s00213-006-0582-1. [DOI] [PubMed] [Google Scholar]
  266. Lee H, et al. Chronic Administration of Lamotrigine Downregulates COX-2 mRNA and Protein in Rat Frontal Cortex. Neurochem Res. 2008;33:861–866. doi: 10.1007/s11064-007-9526-3. [DOI] [PubMed] [Google Scholar]
  267. Lee HJ, et al. Chronic imipramine but not bupropion increases arachidonic acid signaling in rat brain: is this related to 'switching' in bipolar disorder? Molecular psychiatry. 2010;15:602–614. doi: 10.1038/mp.2008.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Levant B, Ozias MK, Carlson SE. Diet (n-3) polyunsaturated fatty acid content and parity interact to alter maternal rat brain phospholipid fatty acid composition. J Nutr. 2006;136:2236–2242. doi: 10.1093/jn/136.8.2236. [DOI] [PubMed] [Google Scholar]
  269. Levant B, et al. Decreased brain docosahexaenoic acid content produces neurobiological effects associated with depression: Interactions with reproductive status in female rats. Psychoneuroendocrinology. 2008;33:1279–1292. doi: 10.1016/j.psyneuen.2008.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Lewis MD, et al. Suicide deaths of active-duty US military and omega-3 fatty-acid status: a case-control comparison. J Clin Psychiatry. 2011 doi: 10.4088/JCP.11m06879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Li Q, et al. Docosahexaenoic acid changes lipid composition and interleukin-2 receptor signaling in membrane rafts. J Lipid Res. 2005;46:1904–1913. doi: 10.1194/jlr.M500033-JLR200. [DOI] [PubMed] [Google Scholar]
  272. Li Q, et al. Eicosapentaenoic acid modifies lipid composition in caveolae and induces translocation of endothelial nitric oxide synthase. Biochimie. 2007;89:169–177. doi: 10.1016/j.biochi.2006.10.009. [DOI] [PubMed] [Google Scholar]
  273. Li Q, et al. n-3 polyunsaturated fatty acids prevent disruption of epithelial barrier function induced by proinflammatory cytokines. Mol Immunol. 2008;45:1356–1365. doi: 10.1016/j.molimm.2007.09.003. [DOI] [PubMed] [Google Scholar]
  274. Lim GP, et al. A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model. J Neurosci. 2005a;25:3032–3040. doi: 10.1523/JNEUROSCI.4225-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Lim SY, Hoshiba J, Salem N., Jr An extraordinary degree of structural specificity is required in neural phospholipids for optimal brain function: n-6 docosapentaenoic acid substitution for docosahexaenoic acid leads to a loss in spatial task performance. J Neurochem. 2005b;95:848–857. doi: 10.1111/j.1471-4159.2005.03427.x. [DOI] [PubMed] [Google Scholar]
  276. Lin PY, Huang SY, Su KP. A meta-analytic review of polyunsaturated fatty acid compositions in patients with depression. Biological psychiatry. 2010;68:140–147. doi: 10.1016/j.biopsych.2010.03.018. [DOI] [PubMed] [Google Scholar]
  277. Lin PY, et al. Are omega-3 fatty acids antidepressants or just mood-improving agents? The effect depends upon diagnosis, supplement preparation, and severity of depression. Mol Psychiatry. 2012;17:1161–1163. doi: 10.1038/mp.2012.111. author reply 11637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Ling KY, Lee HY, Hollander D. Mechanisms of linoleic acid uptake by rabbit small intestinal brush border membrane vesicles. Lipids. 1989;24:51–55. doi: 10.1007/BF02535264. [DOI] [PubMed] [Google Scholar]
  279. Lister MD, et al. Kinetic analysis of the Ca2+-dependent, membrane-bound, macrophage phospholipase A2 and the effects of arachidonic acid. The Journal of biological chemistry. 1988;263:7506–7513. [PubMed] [Google Scholar]
  280. Little TJ, et al. Free fatty acids have more potent effects on gastric emptying, gut hormones, and appetite than triacylglycerides. Gastroenterology. 2007;133:1124–1131. doi: 10.1053/j.gastro.2007.06.060. [DOI] [PubMed] [Google Scholar]
  281. Liu JJ, et al. Omega-3 Polyunsaturated Fatty Acid (PUFA) Status in Major Depressive Disorder With Comorbid Anxiety Disorders. J Clin Psychiatry. 2013;74:732–738. doi: 10.4088/JCP.12m07970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Liu Y, et al. Elevated delta-6 desaturase (FADS2) expression in the postmortem prefrontal cortex of schizophrenic patients: relationship with fatty acid composition. Schizophr Res. 2009a;109:113–120. doi: 10.1016/j.schres.2008.12.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Liu Y, et al. Elevated delta-6 desaturase (FADS2) expression in the postmortem prefrontal cortex of schizophrenic patients: relationship with fatty acid composition. Schizophrenia research. 2009b;109:113–120. doi: 10.1016/j.schres.2008.12.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Liu Y, McNamara RK. Elevated Delta-6 desaturase (FADS2) gene expression in the prefrontal cortex of patients with bipolar disorder. J Psychiatr Res. 2011a;45:269–272. doi: 10.1016/j.jpsychires.2010.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Liu Y, McNamara RK. Elevated Delta-6 desaturase (FADS2) gene expression in the prefrontal cortex of patients with bipolar disorder. Journal of psychiatric research. 2011b;45:269–272. doi: 10.1016/j.jpsychires.2010.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Lopez LB, Kritz-Silverstein D, Barrett Connor E. High dietary and plasma levels of the omega-3 fatty acid docosahexaenoic acid are associated with decreased dementia risk: the Rancho Bernardo study. The journal of nutrition, health & aging. 2011;15:25–31. doi: 10.1007/s12603-011-0009-5. [DOI] [PubMed] [Google Scholar]
  287. Lucarelli M, et al. Expression of receptors for native and chemically modified low-density lipoproteins in brain microvessels. FEBS letters. 1997;401:53–58. doi: 10.1016/s0014-5793(96)01435-4. [DOI] [PubMed] [Google Scholar]
  288. Luchtman DW, Song C. Cognitive enhancement by omega-3 fatty acids from child-hood to old age: findings from animal and clinical studies. Neuropharmacology. 2013;64:550–565. doi: 10.1016/j.neuropharm.2012.07.019. [DOI] [PubMed] [Google Scholar]
  289. Luiken JJ, Turcotte LP, Bonen A. Protein-mediated palmitate uptake and expression of fatty acid transport proteins in heart giant vesicles. Journal of lipid research. 1999;40:1007–1016. [PubMed] [Google Scholar]
  290. Luiken JJ, et al. Changes in fatty acid transport and transporters are related to the severity of insulin deficiency. American journal of physiology. Endocrinology and metabolism. 2002a;283:E612–E621. doi: 10.1152/ajpendo.00011.2002. [DOI] [PubMed] [Google Scholar]
  291. Luiken JJ, Bonen A, Glatz JF. Cellular fatty acid uptake is acutely regulated by membrane-associated fatty acid-binding proteins. Prostaglandins, leukotrienes, and essential fatty acids. 2002b;67:73–78. doi: 10.1054/plef.2002.0401. [DOI] [PubMed] [Google Scholar]
  292. Lukiw WJ, et al. A role for docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and Alzheimer disease. J Clin Invest. 2005;115:2774–2783. doi: 10.1172/JCI25420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Luxon BA. Inhibition of binding to fatty acid binding protein reduces the intracellular transport of fatty acids. The American journal of physiology. 1996;271:G113–G120. doi: 10.1152/ajpgi.1996.271.1.G113. [DOI] [PubMed] [Google Scholar]
  294. Ma DW, et al. n-3 PUFA alter caveolae lipid composition and resident protein localization in mouse colon. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2004;18:1040–1042. doi: 10.1096/fj.03-1430fje. [DOI] [PubMed] [Google Scholar]
  295. Maatman RG, et al. Expression of human liver fatty acid-binding protein in Escherichia coli and comparative analysis of its binding characteristics with muscle fatty acid-binding protein. Biochimica et biophysica acta. 1994;1214:1–10. doi: 10.1016/0005-2760(94)90002-7. [DOI] [PubMed] [Google Scholar]
  296. Maes M, et al. Fatty acid composition in major depression: decreased omega 3 fractions in cholesteryl esters and increased C20: 4 omega 6/C20:5 omega 3 ratio in cholesteryl esters and phospholipids. Journal of affective disorders. 1996;38:35–46. doi: 10.1016/0165-0327(95)00092-5. [DOI] [PubMed] [Google Scholar]
  297. Maes M, et al. Lowered omega3 polyunsaturated fatty acids in serum phospholipids and cholesteryl esters of depressed patients. Psychiatry research. 1999a;85:275–291. doi: 10.1016/s0165-1781(99)00014-1. [DOI] [PubMed] [Google Scholar]
  298. Maes M, et al. Lowered omega3 polyunsaturated fatty acids in serum phospholipids and cholesteryl esters of depressed patients. Psychiatry Res. 1999b;85:275–291. doi: 10.1016/s0165-1781(99)00014-1. [DOI] [PubMed] [Google Scholar]
  299. Magnani F, et al. Partitioning of the serotonin transporter into lipid microdomains modulates transport of serotonin. The Journal of biological chemistry. 2004;279:38770–38778. doi: 10.1074/jbc.M400831200. [DOI] [PubMed] [Google Scholar]
  300. Mahadik SP, et al. Utilization of precursor essential fatty acids in culture by skin fibroblasts from schizophrenic patients and normal controls. Prostaglandins Leukot Essent Fatty Acids. 1996;55:65–70. doi: 10.1016/s0952-3278(96)90147-7. [DOI] [PubMed] [Google Scholar]
  301. Makinen PI, et al. Silencing of either SR-A or CD36 reduces atherosclerosis in hyperlipidaemic mice and reveals reciprocal upregulation of these receptors. Cardiovascular research. 2010;88:530–538. doi: 10.1093/cvr/cvq235. [DOI] [PubMed] [Google Scholar]
  302. Malerba G, et al. SNPs of the FADS gene cluster are associated with polyunsaturated fatty acids in a cohort of patients with cardiovascular disease. Lipids. 2008;43:289–299. doi: 10.1007/s11745-008-3158-5. [DOI] [PubMed] [Google Scholar]
  303. Mamalakis G, et al. Depression and adipose polyunsaturated fatty acids in an adolescent group. Prostaglandins, leukotrienes, and essential fatty acids. 2004;71:289–294. doi: 10.1016/j.plefa.2004.04.002. [DOI] [PubMed] [Google Scholar]
  304. Marcheselli VL, et al. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. The Journal of biological chemistry. 2003;278:43807–43817. doi: 10.1074/jbc.M305841200. [DOI] [PubMed] [Google Scholar]
  305. Martin C, et al. CD36 as a lipid sensor. Physiology & behavior. 2011a;105:36–42. doi: 10.1016/j.physbeh.2011.02.029. [DOI] [PubMed] [Google Scholar]
  306. Martin C, et al. The lipid-sensor candidates CD36 and GPR120 are differentially regulated by dietary lipids in mouse taste buds: impact on spontaneous fat preference. PloS one. 2011b;6:e24014. doi: 10.1371/journal.pone.0024014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Martin-Nizard F, et al. Interactions of high-density lipoprotein 3 with brain capillary endothelial cells. Biochimica et biophysica acta. 1989;1005:201–208. doi: 10.1016/0005-2760(89)90038-6. [DOI] [PubMed] [Google Scholar]
  308. Martins JG. EPA but not DHA appears to be responsible for the efficacy of omega-3 long chain polyunsaturated fatty acid supplementation in depression: evidence from a meta-analysis of randomized controlled trials. J Am Coll Nutr. 2009;28:525–542. doi: 10.1080/07315724.2009.10719785. [DOI] [PubMed] [Google Scholar]
  309. Martins JG, Bentsen H, Puri BK. Eicosapentaenoic acid appears to be the key omega-3 fatty acid component associated with efficacy in major depressive disorder: a critique of Bloch and Hannestad and updated meta-analysis. Mol Psychiatry. 2012;17:1144–1149. doi: 10.1038/mp.2012.25. [DOI] [PubMed] [Google Scholar]
  310. Masoodi M, et al. Lipid signaling in adipose tissue: Connecting inflammation & metabolism. Biochim Biophys Acta. 2014 doi: 10.1016/j.bbalip.2014.09.023. [DOI] [PubMed] [Google Scholar]
  311. Masson CJ, et al. Fatty acid-and cholesterol transporter protein expression along the human intestinal tract. PLoS One. 2010;5:e10380. doi: 10.1371/journal.pone.0010380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Mathieu G, et al. Synergistic effects of stress and omega-3 fatty acid deprivation on emotional response and brain lipid composition in adult rats. Prostaglandins Leukot Essent Fatty Acids. 2008;78:391–401. doi: 10.1016/j.plefa.2008.05.003. [DOI] [PubMed] [Google Scholar]
  313. Mathieu G, et al. Dietary n-3 polyunsaturated fatty acid deprivation together with early maternal separation increases anxiety and vulnerability to stress in adult rats. Prostaglandins, leukotrienes, and essential fatty acids. 2011;85:129–136. doi: 10.1016/j.plefa.2011.07.001. [DOI] [PubMed] [Google Scholar]
  314. Mattes RD. Fat taste and lipid metabolism in humans. Physiology & behavior. 2005;86:691–697. doi: 10.1016/j.physbeh.2005.08.058. [DOI] [PubMed] [Google Scholar]
  315. Mattes RD. Oral detection of short-, medium-, and long-chain free fatty acids in humans. Chemical senses. 2009;34:145–150. doi: 10.1093/chemse/bjn072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Matthies HJ, et al. Subcellular localization of the antidepressant-sensitive norepinephrine transporter. BMC Neurosci. 2009;10:65. doi: 10.1186/1471-2202-10-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. Mayes PA. Harper's biochemistry. London: Prentice Hall; 1996. Metabolism of unsaturated fatty acids and eicosanoids; pp. 236–244. [Google Scholar]
  318. McIntosh AL, et al. Liver type fatty acid binding protein (L-FABP) gene ablation reduces nuclear ligand distribution and peroxisome proliferator-activated receptor-alpha activity in cultured primary hepatocytes. Archives of biochemistry and biophysics. 2009;485:160–173. doi: 10.1016/j.abb.2009.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. McNamara RK, et al. Selective deficits in erythrocyte docosahexaenoic acid composition in adult patients with bipolar disorder and major depressive disorder. J Affect Disord. 2010;126:303–311. doi: 10.1016/j.jad.2010.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. McNamara RK, et al. Differential effects of antipsychotic medications on polyunsaturated fatty acid biosynthesis in rats: Relationship with liver delta6-desaturase expression. Schizophr Res. 2011;129:57–65. doi: 10.1016/j.schres.2011.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. McNamara RK, Liu Y. Reduced expression of fatty acid biosynthesis genes in the prefrontal cortex of patients with major depressive disorder. Journal of affective disorders. 2011;129:359–363. doi: 10.1016/j.jad.2010.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. McNamara RK, Strawn JR. Role of Long-Chain Omega-3 Fatty Acids in Psychiatric Practice. PharmaNutrition. 2013;1:41–49. doi: 10.1016/j.phanu.2012.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. McPhail LC, Clayton CC, Snyderman R. A potential second messenger role for arachidonic acid: activation of Ca2+-dependent protein kinase. Transactions of the Association of American Physicians. 1984a;97:222–231. [PubMed] [Google Scholar]
  324. McPhail LC, Clayton CC, Snyderman R. A potential second messenger role for unsaturated fatty acids: activation of Ca2+-dependent protein kinase. Science. 1984b;224:622–625. doi: 10.1126/science.6231726. [DOI] [PubMed] [Google Scholar]
  325. Meex SJ, et al. Up-regulation of CD36/FAT in preadipocytes in familial combined hyperlipidemia. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2005;19:2063–2065. doi: 10.1096/fj.04-2403fje. [DOI] [PubMed] [Google Scholar]
  326. Melton EM, et al. Human fatty acid transport protein 2a/very long chain acyl-CoA synthetase 1 (FATP2a/Acsvl1) has a preference in mediating the channeling of exogenous n-3 fatty acids into phosphatidylinositol. J Biol Chem. 2011;286:30670–30679. doi: 10.1074/jbc.M111.226316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Meyer BJ, et al. Dietary intakes and food sources of omega-6 and omega-3 polyunsaturated fatty acids. Lipids. 2003;38:391–398. doi: 10.1007/s11745-003-1074-0. [DOI] [PubMed] [Google Scholar]
  328. Mialet-Perez J, et al. Serotonin 5-HT2A receptor-mediated hypertrophy is negatively regulated by caveolin-3 in cardiomyoblasts and neonatal cardiomyocytes. J Mol Cell Cardiol. 2012;52:502–510. doi: 10.1016/j.yjmcc.2011.07.019. [DOI] [PubMed] [Google Scholar]
  329. Mills JD, Hadley K, Bailes JE. Dietary supplementation with the omega-3 fatty acid docosahexaenoic acid in traumatic brain injury. Neurosurgery. 2011;68:474–481. doi: 10.1227/NEU.0b013e3181ff692b. discussion 481. [DOI] [PubMed] [Google Scholar]
  330. Milte CM, et al. Polyunsaturated fatty acids, cognition and literacy in children with ADHD with and without learning difficulties. Journal of child health care : for professionals working with children in the hospital and community. 2011;15:299–311. doi: 10.1177/1367493511403953. [DOI] [PubMed] [Google Scholar]
  331. Miquilena-Colina ME, et al. Hepatic fatty acid translocase CD36 upregulation is associated with insulin resistance, hyperinsulinaemia and increased steatosis in non-alcoholic steatohepatitis and chronic hepatitis. C. Gut. 2011;60:1394–1402. doi: 10.1136/gut.2010.222844. [DOI] [PubMed] [Google Scholar]
  332. Mitchell RW, et al. Fatty acid transport protein expression in human brain and potential role in fatty acid transport across human brain microvessel endothelial cells. Journal of neurochemistry. 2011;117:735–746. doi: 10.1111/j.1471-4159.2011.07245.x. [DOI] [PubMed] [Google Scholar]
  333. Modi HR, et al. Chronic clozapine reduces rat brain arachidonic acid metabolism by reducing plasma arachidonic acid availability. J Neurochem. 2013;124:376–387. doi: 10.1111/jnc.12078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  334. Moore SA, Yoder E, Spector AA. Role of the blood-brain barrier in the formation of long-chain omega-3 and omega-6 fatty acids from essential fatty acid precursors. Journal of neurochemistry. 1990;55:391–402. doi: 10.1111/j.1471-4159.1990.tb04150.x. [DOI] [PubMed] [Google Scholar]
  335. Moore SA, et al. Astrocytes, not neurons, produce docosahexaenoic acid (22:6 omega-3) and arachidonic acid (20:4 omega-6) Journal of neurochemistry. 1991;56:518–524. doi: 10.1111/j.1471-4159.1991.tb08180.x. [DOI] [PubMed] [Google Scholar]
  336. Moore SA. Polyunsaturated fatty acid synthesis and release by brain-derived cells in vitro. Journal of molecular neuroscience : MN. 2001;16:195–200. doi: 10.1385/JMN:16:2-3:195. discussion 215-21. [DOI] [PubMed] [Google Scholar]
  337. Moriguchi T, et al. Reversal of docosahexaenoic acid deficiency in the rat brain, retina, liver, and serum. Journal of lipid research. 2001;42:419–427. [PubMed] [Google Scholar]
  338. Moriguchi T. Effects of an n-3-deficient diet on brain, retina, and liver fatty acyl composition in artificially reared rats. The Journal of Lipid Research. 2004;45:1437–1445. doi: 10.1194/jlr.M400087-JLR200. [DOI] [PubMed] [Google Scholar]
  339. Morris MC, et al. Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Archives of neurology. 2003;60:940–946. doi: 10.1001/archneur.60.7.940. [DOI] [PubMed] [Google Scholar]
  340. Morris MC, et al. Fish consumption and cognitive decline with age in a large community study. Archives of neurology. 2005;62:1849–1853. doi: 10.1001/archneur.62.12.noc50161. [DOI] [PubMed] [Google Scholar]
  341. Morrisett JD, Pownall HJ, Jackson RL, Segura R, Gotto AM, Jr, Taunton OD. Effects of polyunsaturated and saturated fat diets on the chemical composition and thermotropic properties of human plasma lipoproteins, Vol. Champaign, IL: American Oil Chemists' Society: 1976. [Google Scholar]
  342. Muldoon MF, Manuck SB, Matthews KA. Lowering cholesterol concentrations and mortality: a quantitative review of primary prevention trials. BMJ. 1990;301:309–314. doi: 10.1136/bmj.301.6747.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Muldoon MF, et al. Cholesterol reduction and non-illness mortality: meta-analysis of randomised clinical trials. Bmj. 2001;322:11–15. doi: 10.1136/bmj.322.7277.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  344. Murakami H, et al. Enhanced response of T lymphocytes from Pgap3 knockout mouse: Insight into roles of fatty acid remodeling of GPI anchored proteins. Biochem Biophys Res Commun. 2012;417:1235–1241. doi: 10.1016/j.bbrc.2011.12.116. [DOI] [PubMed] [Google Scholar]
  345. Murphy EJ. L-FABP and I-FABP expression increase NBD-stearate uptake and cytoplasmic diffusion in L cells. The American journal of physiology. 1998;275:G244–G249. doi: 10.1152/ajpgi.1998.275.2.G244. [DOI] [PubMed] [Google Scholar]
  346. Murphy EJ, et al. Isolation and characterization of two distinct forms of liver fatty acid binding protein from the rat. Biochimica et biophysica acta. 1999;1436:413–425. doi: 10.1016/s0005-2760(98)00150-7. [DOI] [PubMed] [Google Scholar]
  347. Murphy EJ, et al. Brain arachidonic acid incorporation is decreased in heart fatty acid binding protein gene-ablated mice. Biochemistry. 2005;44:6350–6360. doi: 10.1021/bi047292r. [DOI] [PubMed] [Google Scholar]
  348. Murphy MG. Membrane fatty acids, lipid peroxidation and adenylate cyclase activity in cultured neural cells. Biochemical and biophysical research communications. 1985;132:757–763. doi: 10.1016/0006-291x(85)91197-0. [DOI] [PubMed] [Google Scholar]
  349. Musselman DL, Evans DL, Nemeroff CB. The relationship of depression to cardiovascular disease: epidemiology, biology, and treatment. Arch Gen Psychiatry. 1998;55:580–592. doi: 10.1001/archpsyc.55.7.580. [DOI] [PubMed] [Google Scholar]
  350. Naik P. Essentials of Biochemistry. New Delhi, India: Jaypee Brothers Medical Publishers (P) Ltd; 2011. Lipid Metabolism; pp. 190–222. [Google Scholar]
  351. Nariai T, et al. In vivo brain incorporation of [1-14C]arachidonate in awake rats, with or without cholinergic stimulation, following unilateral lesioning of nucleus basalis magnocellularis. Brain Res. 1991;559:1–9. doi: 10.1016/0006-8993(91)90279-5. [DOI] [PubMed] [Google Scholar]
  352. National Research Council. Building a knowledge network for biomedical research and a new taxonomy of disease, Vol. Washington, DC: The National Academies Press; 2011. [PubMed] [Google Scholar]
  353. Ng KH, et al. Dietary PUFA intakes in children with attention-deficit/ hyperactivity disorder symptoms. Br J Nutr. 2009;102:1635–1641. doi: 10.1017/S0007114509990821. [DOI] [PubMed] [Google Scholar]
  354. Nguyen LN, et al. Mfsd2a is a transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature. 2014;509:503–506. doi: 10.1038/nature13241. [DOI] [PubMed] [Google Scholar]
  355. Nickerson JG, et al. Greater transport efficiencies of the membrane fatty acid transporters FAT/CD36 and FATP4 compared with FABPpm and FATP1 and differential effects on fatty acid esterification and oxidation in rat skeletal muscle. The Journal of biological chemistry. 2009;284:16522–16530. doi: 10.1074/jbc.M109.004788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Nicolas C, et al. Dietary (n-6) polyunsaturated fatty acids affect beta-adrenergic receptor binding and adenylate cyclase activity in pig adipocyte plasma membrane. The Journal of nutrition. 1991;121:1179–1186. doi: 10.1093/jn/121.8.1179. [DOI] [PubMed] [Google Scholar]
  357. Noaghiul S, Hibbeln JR. Cross-national comparisons of seafood consumption and rates of bipolar disorders. Am J Psychiatry. 2003;160:2222–2227. doi: 10.1176/appi.ajp.160.12.2222. [DOI] [PubMed] [Google Scholar]
  358. Nothdurfter C, et al. Impact of lipid raft integrity on 5-HT3 receptor function and its modulation by antidepressants. Neuropsychopharmacology. 2010;35:1510–1519. doi: 10.1038/npp.2010.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Nothdurfter C, et al. Modulation of ligand-gated ion channels as a novel pharmacological principle. Pharmacopsychiatry. 2011;44(Suppl 1):S27–S34. doi: 10.1055/s-0031-1271704. [DOI] [PubMed] [Google Scholar]
  360. Nozaki S, et al. Reduced uptake of oxidized low density lipoproteins in monocyte-derived macrophages from CD36-deficient subjects. The Journal of clinical investigation. 1995;96:1859–1865. doi: 10.1172/JCI118231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Ntambi JM, Bene H. Polyunsaturated fatty acid regulation of gene expression. Journal of molecular neuroscience : MN. 2001;16:273–278. doi: 10.1385/JMN:16:2-3:273. discussion 279-84. [DOI] [PubMed] [Google Scholar]
  362. Nwankwo JO, Spector AA, Domann FE. A nucleotide insertion in the transcriptional regulatory region of FADS2 gives rise to human fatty acid delta-6-desaturase deficiency. J Lipid Res. 2003;44:2311–2319. doi: 10.1194/jlr.M300273-JLR200. [DOI] [PubMed] [Google Scholar]
  363. Nyalala JO, et al. Hypertriglyceridemia and hypercholesterolemia: effects of drug treatment on fatty acid composition of plasma lipids and membranes. Prostaglandins Leukot Essent Fatty Acids. 2008;78:271–280. doi: 10.1016/j.plefa.2008.03.003. [DOI] [PubMed] [Google Scholar]
  364. O'Banion MK. Cyclooxygenase-2: molecular biology, pharmacology, and neurobiology. Critical reviews in neurobiology. 1999;13:45–82. doi: 10.1615/critrevneurobiol.v13.i1.30. [DOI] [PubMed] [Google Scholar]
  365. Ockner RK, Pittman JP, Yager JL. Differences in the intestinal absorption of saturated and unsaturated long chain fatty acids. Gastroenterology. 1972;62:981–992. [PubMed] [Google Scholar]
  366. Ollis TE, Meyer BJ, Howe PR. Australian food sources and intakes of omega-6 and omega-3 polyunsaturated fatty acids. Annals of nutrition & metabolism. 1999;43:346–355. doi: 10.1159/000012803. [DOI] [PubMed] [Google Scholar]
  367. Orr SK, et al. Unesterified docosahexaenoic acid is protective in neuroinflammation. J Neurochem. 2013 doi: 10.1111/jnc.12392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  368. Ouellet M, et al. Diffusion of docosahexaenoic and eicosapentaenoic acids through the blood-brain barrier: An in situ cerebral perfusion study. Neurochemistry international. 2009;55:476–482. doi: 10.1016/j.neuint.2009.04.018. [DOI] [PubMed] [Google Scholar]
  369. Owada Y. Fatty acid binding protein: localization and functional significance in the brain. The Tohoku journal of experimental medicine. 2008;214:213–220. doi: 10.1620/tjem.214.213. [DOI] [PubMed] [Google Scholar]
  370. Panel on Macronutrients, Panel on the Definition of Dietary Fiber, Subcommittee on Upper Reference Levels of Nutrients, Subcommittee on Interpretation and Uses of Dietary Reference Intakes, and the Standing Committee on the Scientific Evaluation of Dietary Reference Intakes. Dietary Reference Intakes for Energy, Carbohydrate, Fiber, Fat, Fatty Acids, Cholesterol, Protein, and Amino Acids (Macronutrients) Washington, DC: Food and Nutrition Board, Institute of Medicine of the National Academies; 2002. pp. 422–541. [Google Scholar]
  371. Pardridge WM, Mietus LJ. Palmitate and cholesterol transport through the blood-brain barrier. J Neurochem. 1980;34:463–466. doi: 10.1111/j.1471-4159.1980.tb06621.x. [DOI] [PubMed] [Google Scholar]
  372. Peet M, et al. Depleted red cell membrane essential fatty acids in drug-treated schizophrenic patients. Journal of psychiatric research. 1995;29:227–232. doi: 10.1016/0022-3956(95)00001-l. [DOI] [PubMed] [Google Scholar]
  373. Peet M, et al. Depletion of omega-3 fatty acid levels in red blood cell membranes of depressive patients. Biological psychiatry. 1998;43:315–319. doi: 10.1016/s0006-3223(97)00206-0. [DOI] [PubMed] [Google Scholar]
  374. Petrescu AD, et al. Impact of L-FABP and glucose on polyunsaturated fatty acid induction of PPARalpha-regulated beta-oxidative enzymes. American journal of physiology. Gastrointestinal and liver physiology. 2013;304:G241–G256. doi: 10.1152/ajpgi.00334.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Phan CT, Tso P. Intestinal lipid absorption and transport. Frontiers in bioscience : a journal and virtual library. 2001;6:D299–D319. doi: 10.2741/phan. [DOI] [PubMed] [Google Scholar]
  376. Pichika R, et al. The synthesis and in vivo pharmacokinetics of fluorinated arachidonic acid: implications for imaging neuroinflammation. J Nucl Med. 2012;53:1383–1391. doi: 10.2967/jnumed.112.105734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  377. Pifferi F, et al. (n-3) polyunsaturated fatty acid deficiency reduces the expression of both isoforms of the brain glucose transporter GLUT1 in rats. J Nutr. 2005;135:2241–2246. doi: 10.1093/jn/135.9.2241. [DOI] [PubMed] [Google Scholar]
  378. Pifferi F, et al. n-3 Fatty acids modulate brain glucose transport in endothelial cells of the blood-brain barrier. Prostaglandins Leukot Essent Fatty Acids. 2007;77:279–286. doi: 10.1016/j.plefa.2007.10.011. [DOI] [PubMed] [Google Scholar]
  379. Pike LJ. Lipid rafts: bringing order to chaos. Journal of lipid research. 2003;44:655–667. doi: 10.1194/jlr.R200021-JLR200. [DOI] [PubMed] [Google Scholar]
  380. Pike LJ. Rafts defined: a report on the Keystone Symposium on Lipid Rafts and Cell Function. J Lipid Res. 2006;47:1597–1598. doi: 10.1194/jlr.E600002-JLR200. [DOI] [PubMed] [Google Scholar]
  381. Piomelli D, et al. Inhibition of Ca2+/calmodulin-dependent protein kinase II by arachidonic acid and its metabolites. Proceedings of the National Academy of Sciences of the United States of America. 1989;86:8550–8554. doi: 10.1073/pnas.86.21.8550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Piomelli D. Eicosanoids in synaptic transmission. Critical reviews in neurobiology. 1994;8:65–83. [PubMed] [Google Scholar]
  383. Pitas RE, et al. Lipoproteins and their receptors in the central nervous system. Characterization of the lipoproteins in cerebrospinal fluid and identification of apolipoprotein B,E(LDL) receptors in the brain. The Journal of biological chemistry. 1987;262:14352–14360. [PubMed] [Google Scholar]
  384. Plourde M, et al. Plasma incorporation, apparent retroconversion and beta-oxidation of 13C-docosahexaenoic acid in the elderly. Nutr Metab (Lond) 2011;8:5. doi: 10.1186/1743-7075-8-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Potter BJ, et al. Isolation and partial characterization of plasma membrane fatty acid binding proteins from myocardium and adipose tissue and their relationship to analogous proteins in liver and gut. Biochemical and biophysical research communications. 1987;148:1370–1376. doi: 10.1016/s0006-291x(87)80283-8. [DOI] [PubMed] [Google Scholar]
  386. Pownall HJ. Cellular transport of nonesterified fatty acids. Journal of molecular neuroscience : MN. 2001;16:109–115. doi: 10.1385/JMN:16:2-3:109. discussion 151-7. [DOI] [PubMed] [Google Scholar]
  387. Price PT, Nelson CM, Clarke SD. Omega-3 polyunsaturated fatty acid regulation of gene expression. Current opinion in lipidology. 2000;11:3–7. doi: 10.1097/00041433-200002000-00002. [DOI] [PubMed] [Google Scholar]
  388. Pucadyil TJ, Chattopadhyay A. Cholesterol modulates ligand binding and G-protein coupling to serotonin(1A) receptors from bovine hippocampus. Biochimica et biophysica acta. 2004;1663:188–200. doi: 10.1016/j.bbamem.2004.03.010. [DOI] [PubMed] [Google Scholar]
  389. Purdon D, Arai T, Rapoport SI. No evidence for direct incorporation of esterified palmitate from plasma into brain lipids of awake adult rats. J. Lipid Res. 1997;38:526–530. [PubMed] [Google Scholar]
  390. Raclot T, Groscolas R. Individual fish-oil n-3 polyunsaturated fatty acid deposition and mobilization rates for adipose tissue of rats in a nutritional steady state. The American journal of clinical nutrition. 1994;60:72–78. doi: 10.1093/ajcn/60.1.72. [DOI] [PubMed] [Google Scholar]
  391. Raclot T, Oudart H. Selectivity of fatty acids on lipid metabolism and gene expression. The Proceedings of the Nutrition Society. 1999;58:633–646. doi: 10.1017/s002966519900083x. [DOI] [PubMed] [Google Scholar]
  392. Raeder MB, et al. Associations between cod liver oil use and symptoms of depression: the Hordaland Health Study. Journal of Affective Disorders. 2007;101:245–249. doi: 10.1016/j.jad.2006.11.006. [DOI] [PubMed] [Google Scholar]
  393. Rahman T, et al. The very low density lipoprotein receptor is not necessary for maintaining brain polyunsaturated fatty acid concentrations. Prostaglandins Leukot Essent Fatty Acids. 2010;82:141–145. doi: 10.1016/j.plefa.2009.11.003. [DOI] [PubMed] [Google Scholar]
  394. Ramirez M, Amate L, Gil A. Absorption and distribution of dietary fatty acids from different sources. Early human development. 2001;65(Suppl):S95–S101. doi: 10.1016/s0378-3782(01)00211-0. [DOI] [PubMed] [Google Scholar]
  395. Ramsden CE, et al. Targeted alteration of dietary n-3 and n-6 fatty acids for the treatment of chronic headaches: A randomized trial. Pain. 2013 doi: 10.1016/j.pain.2013.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Rao JS, et al. Dietary n-3 PUFA deprivation alters expression of enzymes of the arachidonic and docosahexaenoic acid cascades in rat frontal cortex. Mol Psychiatry. 2007a;12:151–157. doi: 10.1038/sj.mp.4001887. [DOI] [PubMed] [Google Scholar]
  397. Rao JS, et al. Dietary n-3 PUFA deprivation alters expression of enzymes of the arachidonic and docosahexaenoic acid cascades in rat frontal cortex. Molecular psychiatry. 2007b;12:151–157. doi: 10.1038/sj.mp.4001887. [DOI] [PubMed] [Google Scholar]
  398. Rao JS, et al. n-3 polyunsaturated fatty acid deprivation in rats decreases frontal cortex BDNF via a p38 MAPK-dependent mechanism. Mol Psychiatry. 2007c;12:36–46. doi: 10.1038/sj.mp.4001888. [DOI] [PubMed] [Google Scholar]
  399. Rapoport S. In vivo fatty acid incorporation into brain phospholipids in relation to plasma availablity, signal transduction and membrane remodeling. J Mol Neurosci. 2001;16:243–261. 279–284. doi: 10.1385/JMN:16:2-3:243. [DOI] [PubMed] [Google Scholar]
  400. Rapoport SI, Pettigrew KD. A heterogenous, pore-vesicle membrane model for protein transfer from blood to cerebrospinal fluid at the choroid plexus. Microvasc Res. 1979;18:105–119. doi: 10.1016/0026-2862(79)90020-7. [DOI] [PubMed] [Google Scholar]
  401. Rapoport SI. In vivo fatty acid incorporation into brain phospholipids in relation to signal transduction and membrane remodeling. Neurochemical research. 1999;24:1403–1415. doi: 10.1023/a:1022584707352. [DOI] [PubMed] [Google Scholar]
  402. Rapoport SI, Chang MC, Spector AA. Delivery and turnover of plasma-derived essential PUFAs in mammalian brain. Journal of lipid research. 2001;42:678–685. [PubMed] [Google Scholar]
  403. Rapoport SI, Igarashi M. Can the rat liver maintain normal brain DHA metabolism in the absence of dietary DHA? Prostaglandins Leukot Essent Fatty Acids. 2009;81:119–123. doi: 10.1016/j.plefa.2009.05.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Rapoport SI, Igarashi M, Gao F. Quantitative contributions of diet and liver synthesis to docosahexaenoic acid homeostasis. Prostaglandins, leukotrienes, and essential fatty acids. 2010;82:273–276. doi: 10.1016/j.plefa.2010.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  405. Rapoport SI, Ramadan E, Basselin M. Docosahexaenoic acid (DHA) incorporation into the brain from plasma, as an in vivo biomarker of brain DHA metabolism and neurotransmission. Prostaglandins Other Lipid Mediat. 2011;96:109–113. doi: 10.1016/j.prostaglandins.2011.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  406. Rapoport SI. Translational studies on regulation of brain docosahexaenoic acid (DHA) metabolism in vivo. Prostaglandins Leukot Essent Fatty Acids. 2013;88:79–85. doi: 10.1016/j.plefa.2012.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  407. Reese TS, Karnovsky MJ. Fine structural localization of a blood-brain barrier to exogenous peroxidase. The Journal of cell biology. 1967;34:207–217. doi: 10.1083/jcb.34.1.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Renner U, et al. Localization of the mouse 5-hydroxytryptamine(1A) receptor in lipid microdomains depends on its palmitoylation and is involved in receptor-mediated signaling. Molecular pharmacology. 2007;72:502–513. doi: 10.1124/mol.107.037085. [DOI] [PubMed] [Google Scholar]
  409. Reubsaet FA, et al. The involvement of fatty acid binding protein in peroxisomal fatty acid oxidation. FEBS letters. 1990;267:229–230. doi: 10.1016/0014-5793(90)80931-8. [DOI] [PubMed] [Google Scholar]
  410. Reynolds CM, Roche HM. Conjugated linoleic acid and inflammatory cell signalling. Prostaglandins Leukot Essent Fatty Acids. 2010;82:199–204. doi: 10.1016/j.plefa.2010.02.021. [DOI] [PubMed] [Google Scholar]
  411. Ribarik Coe N, Bernlohr DA. Physiological properties and functions of intracellular fatty acid-binding proteins. Biochim. Biophys. Acta. 1998;1391:287–306. doi: 10.1016/s0005-2760(97)00205-1. [DOI] [PubMed] [Google Scholar]
  412. Richieri GV, et al. Fatty acid binding proteins from different tissues show distinct patterns of fatty acid interactions. Biochemistry. 2000;39:7197–7204. doi: 10.1021/bi000314z. [DOI] [PubMed] [Google Scholar]
  413. Riemer S, et al. Lowered omega-3 PUFAs are related to major depression, but not to somatization syndrome. J Affect Disord. 2010;123:173–180. doi: 10.1016/j.jad.2009.08.004. [DOI] [PubMed] [Google Scholar]
  414. Rizzi TS, et al. Genetic Variance in Combination with Fatty Acid Intake Might Alter Composition of the Fatty Acids in Brain. PLoS One. 2013;8:e68000. doi: 10.1371/journal.pone.0068000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  415. Robertson MD, et al. Mobilisation of enterocyte fat stores by oral glucose in humans. Gut. 2003;52:834–839. doi: 10.1136/gut.52.6.834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  416. Robinson PJ, et al. A quantitative method for measuring regional in vivo fatty-acid incorporation into and turnover within brain phospholipids: review and critical analysis. Brain Res Brain Res Rev. 1992;17:187–214. doi: 10.1016/0165-0173(92)90016-f. [DOI] [PubMed] [Google Scholar]
  417. Rockett BD, et al. Fish oil increases raft size and membrane order of B cells accompanied by differential effects on function. J Lipid Res. 2012;53:674–685. doi: 10.1194/jlr.M021782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  418. Rogers KR, et al. Docosahexaenoic acid alters epidermal growth factor receptor-related signaling by disrupting its lipid raft association. Carcinogenesis. 2010;31:1523–1530. doi: 10.1093/carcin/bgq111. [DOI] [PubMed] [Google Scholar]
  419. Ross BM, Seguin J, Sieswerda LE. Omega-3 fatty acids as treatments for mental illness: which disorder and which fatty acid? Lipids Health Dis. 2007;6:21. doi: 10.1186/1476-511X-6-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Ross BM, Maxwell R, Glen I. Increased breath ethane levels in medicated patients with schizophrenia and bipolar disorder are unrelated to erythrocyte omega-3 fatty acid abundance. Prog Neuropsychopharmacol Biol Psychiatry. 2010 doi: 10.1016/j.pnpbp.2010.11.032. [DOI] [PubMed] [Google Scholar]
  421. Russell KL, Berman NE, Levant B. Low brain DHA content worsens sensorimotor outcomes after TBI and decreases TBI-induced Timp1 expression in juvenile rats. Prostaglandins Leukot Essent Fatty Acids. 2013;89:97–105. doi: 10.1016/j.plefa.2013.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. Ruth MR, Proctor SD, Field CJ. Feeding long-chain n-3 polyunsaturated fatty acids to obese leptin receptor-deficient JCR:LA-cp rats modifies immune function and lipid-raft fatty acid composition. Br J Nutr. 2009;101:1341–1350. doi: 10.1017/S0007114508076277. [DOI] [PubMed] [Google Scholar]
  423. Samuvel DJ, et al. A role for p38 mitogen-activated protein kinase in the regulation of the serotonin transporter: evidence for distinct cellular mechanisms involved in transporter surface expression. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2005;25:29–41. doi: 10.1523/JNEUROSCI.3754-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  424. Sanchez-Font MF, et al. Overexpression of FABP7 in Down syndrome fetal brains is associated with PKNOX1 gene-dosage imbalance. Nucleic Acids Res. 2003;31:2769–2777. doi: 10.1093/nar/gkg396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  425. Sanchez-Villegas A, et al. Long chain omega-3 fatty acids intake, fish consumption and mental disorders in the SUN cohort study. Eur J Nutr. 2007;46:337–346. doi: 10.1007/s00394-007-0671-x. [DOI] [PubMed] [Google Scholar]
  426. Schaefer EJ, et al. Plasma phosphatidylcholine docosahexaenoic acid content and risk of dementia and Alzheimer disease: the Framingham Heart Study. Archives of neurology. 2006;63:1545–1550. doi: 10.1001/archneur.63.11.1545. [DOI] [PubMed] [Google Scholar]
  427. Schaeffer L, et al. Common genetic variants of the FADS1 FADS2 gene cluster and their reconstructed haplotypes are associated with the fatty acid composition in phospholipids. Hum Mol Genet. 2006a;15:1745–1756. doi: 10.1093/hmg/ddl117. [DOI] [PubMed] [Google Scholar]
  428. Schaeffer L, et al. Common genetic variants of the FADS1 FADS2 gene cluster and their reconstructed haplotypes are associated with the fatty acid composition in phospholipids. Hum Mol Genet. 2006b;15:1745–1756. doi: 10.1093/hmg/ddl117. [DOI] [PubMed] [Google Scholar]
  429. Schins A, et al. Altered omega-3 polyunsaturated fatty acid status in depressed post-myocardial infarction patients. Acta psychiatrica Scandinavica. 2007;115:35–40. doi: 10.1111/j.1600-0447.2006.00830.x. [DOI] [PubMed] [Google Scholar]
  430. Schley PD, Brindley DN, Field CJ. (n-3) PUFA alter raft lipid composition and decrease epidermal growth factor receptor levels in lipid rafts of human breast cancer cells. J Nutr. 2007;137:548–553. doi: 10.1093/jn/137.3.548. [DOI] [PubMed] [Google Scholar]
  431. Schoonjans K, Staels B, Auwerx J. The peroxisome proliferator activated receptors (PPARS) and their effects on lipid metabolism and adipocyte differentiation. Biochimica et biophysica acta. 1996;1302:93–109. doi: 10.1016/0005-2760(96)00066-5. [DOI] [PubMed] [Google Scholar]
  432. Schumann J, et al. Fatty acid and peptide profiles in plasma membrane and membrane rafts of PUFA supplemented RAW264.7 macrophages. PLoS One. 2011;6:e24066. doi: 10.1371/journal.pone.0024066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Schwieterman W, et al. Uptake of oleate by isolated rat adipocytes is mediated by a 40-kDa plasma membrane fatty acid binding protein closely related to that in liver and gut. Proceedings of the National Academy of Sciences of the United States of America. 1988;85:359–363. doi: 10.1073/pnas.85.2.359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Scott BL, Bazan NG. Membrane docosahexaenoate is supplied to the developing brain and retina by the liver. Proc Natl Acad Sci USA. 1989;86:2903–2907. doi: 10.1073/pnas.86.8.2903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  435. Scott BL, Bazan NG. Membrane docosahexaenoic acid is supplied to the developing brain and retina by the liver. Proc. Natl. Acad. Sci. USA. 1989;86:2903–2907. doi: 10.1073/pnas.86.8.2903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  436. Seidelin KN. Fatty acid composition of adipose tissue in humans. Implications for the dietary fat-serum cholesterol-CHD issue. Progress in lipid research. 1995;34:199–217. doi: 10.1016/0163-7827(95)00004-j. [DOI] [PubMed] [Google Scholar]
  437. Serhan CN, et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med. 2002;196:1025–1037. doi: 10.1084/jem.20020760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  438. Sessler AM, Ntambi JM. Polyunsaturated fatty acid regulation of gene expression. The Journal of nutrition. 1998;128:923–926. doi: 10.1093/jn/128.6.923. [DOI] [PubMed] [Google Scholar]
  439. Shafrir E, Gatt S, Khasis S. Partition of Fatty Acids of 20-24 Carbon Atoms between Serum Albumin and Lipoproteins. Biochimica et biophysica acta. 1965;98:365–371. doi: 10.1016/0005-2760(65)90129-3. [DOI] [PubMed] [Google Scholar]
  440. Shaikh SR, et al. Monounsaturated PE does not phase-separate from the lipid raft molecules sphingomyelin and cholesterol: role for polyunsaturation? Biochemistry. 2002;41:10593–10602. doi: 10.1021/bi025712b. [DOI] [PubMed] [Google Scholar]
  441. Shaikh SR, et al. Interaction of cholesterol with a docosahexaenoic acid-containing phosphatidylethanolamine: trigger for microdomain/raft formation? Biochemistry. 2003;42:12028–12037. doi: 10.1021/bi034931+. [DOI] [PubMed] [Google Scholar]
  442. Shaikh SR, et al. Oleic and docosahexaenoic acid differentially phase separate from lipid raft molecules: a comparative NMR, DSC, AFM, and detergent extraction study. Biophys J. 2004;87:1752–1766. doi: 10.1529/biophysj.104.044552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  443. Shen BW, Scanu AM, Kezdy FJ. Structure of human serum lipoproteins inferred from compositional analysis. Proceedings of the National Academy of Sciences of the United States of America. 1977;74:837–841. doi: 10.1073/pnas.74.3.837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  444. Shimshoni JA, et al. Valproate uncompetitively inhibits arachidonic acid acylation by rat acyl-CoA synthetase 4: relevance to valproate's efficacy against bipolar disorder. Biochim Biophys Acta. 2011;1811:163–169. doi: 10.1016/j.bbalip.2010.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  445. Shinitzky M, Barenholz Y. Fluidity parameters of lipid regions determined by fluorescence polarization. Biochim Biophys Acta. 1978;515:367–394. doi: 10.1016/0304-4157(78)90010-2. [DOI] [PubMed] [Google Scholar]
  446. Shirai K, et al. Existence of lipoprotein lipase in rat brain microvessels. The Tohoku journal of experimental medicine. 1986;149:449–450. doi: 10.1620/tjem.149.449. [DOI] [PubMed] [Google Scholar]
  447. Simopoulos AP. Is insulin resistance influenced by dietary linoleic acid and trans fatty acids? Free radical biology & medicine. 1994;17:367–372. doi: 10.1016/0891-5849(94)90023-x. [DOI] [PubMed] [Google Scholar]
  448. Simopoulos AP. Essential fatty acids in health and chronic disease. Am J Clin Nutr. 1999;70:560S–569S. doi: 10.1093/ajcn/70.3.560s. [DOI] [PubMed] [Google Scholar]
  449. Simopoulos AP. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Experimental biology and medicine. 2008;233:674–688. doi: 10.3181/0711-MR-311. [DOI] [PubMed] [Google Scholar]
  450. Simopoulos AP. Evolutionary aspects of diet: the omega-6/omega-3 ratio and the brain. Mol Neurobiol. 2011;44:203–215. doi: 10.1007/s12035-010-8162-0. [DOI] [PubMed] [Google Scholar]
  451. Sinclair AJ, Attar-Bashi NM, Li D. What is the role of alpha-linolenic acid for mammals? Lipids. 2002;37:1113–1123. doi: 10.1007/s11745-002-1008-x. [DOI] [PubMed] [Google Scholar]
  452. Singer P, et al. Defective desaturation and elongation of n-6 and n-3 fatty acids in hypertensive patients. Prostaglandins, leukotrienes, and medicine. 1984;15:159–165. doi: 10.1016/0262-1746(84)90173-2. [DOI] [PubMed] [Google Scholar]
  453. Singh M. Essential fatty acids, DHA and human brain. Indian journal of pediatrics. 2005;72:239–242. [PubMed] [Google Scholar]
  454. Sjogren B, Hamblin MW, Svenningsson P. Cholesterol depletion reduces serotonin binding and signaling via human 5-HT(7(a)) receptors. Eur J Pharmacol. 2006;552:1–10. doi: 10.1016/j.ejphar.2006.08.069. [DOI] [PubMed] [Google Scholar]
  455. Sjogren B, Svenningsson P. Depletion of the lipid raft constituents, sphingomyelin and ganglioside, decreases serotonin binding at human 5-HT7(a) receptors in HeLa cells. Acta Physiol (Oxf) 2007a;190:47–53. doi: 10.1111/j.1365-201X.2007.01687.x. [DOI] [PubMed] [Google Scholar]
  456. Sjogren B, Svenningsson P. Caveolin-1 affects serotonin binding and cell surface levels of human 5-HT7(a) receptors. FEBS Lett. 2007b;581:5115–5121. doi: 10.1016/j.febslet.2007.09.059. [DOI] [PubMed] [Google Scholar]
  457. Sjogren B, Csoregh L, Svenningsson P. Cholesterol reduction attenuates 5-HT1A receptor-mediated signaling in human primary neuronal cultures. Naunyn Schmiedebergs Arch Pharmacol. 2008;378:441–446. doi: 10.1007/s00210-008-0323-6. [DOI] [PubMed] [Google Scholar]
  458. Skeaff CM, Hodson L, McKenzie JE. Dietary-induced changes in fatty acid composition of human plasma, platelet, and erythrocyte lipids follow a similar time course. J Nutr. 2006;136:565–569. doi: 10.1093/jn/136.3.565. [DOI] [PubMed] [Google Scholar]
  459. Smart EJ, et al. Caveolins, liquid-ordered domains, and signal transduction. Molecular and cellular biology. 1999;19:7289–7304. doi: 10.1128/mcb.19.11.7289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  460. Smink W, et al. Linoleic and alpha-linolenic acid as precursor and inhibitor for the synthesis of long-chain polyunsaturated fatty acids in liver and brain of growing pigs. Animal : an international journal of animal bioscience. 2012;6:262–270. doi: 10.1017/S1751731111001479. [DOI] [PubMed] [Google Scholar]
  461. Smith QR, Nagura H. Fatty acid uptake and incorporation in brain: studies with the perfusion model. Journal of molecular neuroscience : MN. 2001a;16:167–172. doi: 10.1385/JMN:16:2-3:167. discussion 215-21. [DOI] [PubMed] [Google Scholar]
  462. Smith QR, Nagura H. Fatty acid uptake and incorporation in brain: studies with the perfusion model. J Mol Neurosci. 2001b;16:167–172. doi: 10.1385/JMN:16:2-3:167. discussion 215-21. [DOI] [PubMed] [Google Scholar]
  463. Smith WL. Prostanoid biosynthesis and mechanisms of action. The American journal of physiology. 1992;263:F181–F191. doi: 10.1152/ajprenal.1992.263.2.F181. [DOI] [PubMed] [Google Scholar]
  464. Smith WL. Cyclooxygenases, peroxide tone and the allure of fish oil. Curr Opin Cell Biol. 2005;17:174–182. doi: 10.1016/j.ceb.2005.02.005. [DOI] [PubMed] [Google Scholar]
  465. Solakivi T, et al. Delta-6-desaturase gene polymorphism is associated with lipoprotein oxidation in vitro. Lipids Health Dis. 2013;12:80. doi: 10.1186/1476-511X-12-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  466. Sommer B, et al. Extraction of membrane cholesterol disrupts caveolae and impairs serotonergic (5-HT2A) and histaminergic (H1) responses in bovine airway smooth muscle: role of Rho-kinase. Can J Physiol Pharmacol. 2009;87:180–195. doi: 10.1139/y08-114. [DOI] [PubMed] [Google Scholar]
  467. Song BJ, et al. Genetic ablation of CD36 does not alter mouse brain polyunsaturated fatty acid concentrations. Lipids. 2010;45:291–299. doi: 10.1007/s11745-010-3398-z. [DOI] [PubMed] [Google Scholar]
  468. Soni SP, et al. Docosahexaenoic acid enhances segregation of lipids between raft and nonraft domains: 2H-NMR study. Biophys J. 2008;95:203–214. doi: 10.1529/biophysj.107.123612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  469. Sorrentino D, et al. Oleate uptake by cardiac myocytes is carrier mediated and involves a 40-kD plasma membrane fatty acid binding protein similar to that in liver, adipose tissue, and gut. The Journal of clinical investigation. 1988;82:928–935. doi: 10.1172/JCI113700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  470. Sorrentino D, et al. Hepatocellular 22Na+ uptake: effect of oleate. The American journal of physiology. 1991;261:G1024–G1029. doi: 10.1152/ajpgi.1991.261.6.G1024. [DOI] [PubMed] [Google Scholar]
  471. Soupene E, Kuypers FA. Mammalian long-chain acyl-CoA synthetases. Exp Biol Med (Maywood) 2008;233:507–521. doi: 10.3181/0710-MR-287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  472. Soutar A. Does dietary fat influence plasma lipoprotein structure? Nature. 1978;273:11–12. doi: 10.1038/273011a0. [DOI] [PubMed] [Google Scholar]
  473. Spector AA. Plasma albumin as a lipoprotein, Vol. New York, NY: Marcel Dekker; 1986. [Google Scholar]
  474. Spector AA. Essentiality of fatty acids. Lipids. 1999;34(Suppl):S1–S3. doi: 10.1007/BF02562220. [DOI] [PubMed] [Google Scholar]
  475. Spector AA. Plasma free fatty acid and lipoproteins as sources of polyunsaturated fatty acid for the brain. Journal of molecular neuroscience : MN. 2001;16:159–165. doi: 10.1385/JMN:16:2-3:159. discussion 215-21. [DOI] [PubMed] [Google Scholar]
  476. Speizer LA, Watson MJ, Brunton LL. Differential effects of omega-3 fish oils on protein kinase activities in vitro. The American journal of physiology. 1991;261:E109–E114. doi: 10.1152/ajpendo.1991.261.1.E109. [DOI] [PubMed] [Google Scholar]
  477. Stables MJ, Gilroy DW. Old and new generation lipid mediators in acute inflammation and resolution. Prog Lipid Res. 2011;50:35–51. doi: 10.1016/j.plipres.2010.07.005. [DOI] [PubMed] [Google Scholar]
  478. Staggers JE, Fernando-Warnakulasuriya GJ, Wells MA. Studies on fat digestion, absorption, and transport in the suckling rat. II. Triacylglycerols: molecular species, stereospecific analysis, and specificity of hydrolysis by lingual lipase. Journal of lipid research. 1981;22:675–679. [PubMed] [Google Scholar]
  479. Stevens LJ, et al. Essential fatty acid metabolism in boys with attention-deficit hyperactivity disorder. The American journal of clinical nutrition. 1995;62:761–768. doi: 10.1093/ajcn/62.4.761. [DOI] [PubMed] [Google Scholar]
  480. Stillwell W, et al. Docosahexaenoic acid affects cell signaling by altering lipid rafts. Reprod. Nutr. Dev. 2005a;45:559–579. doi: 10.1051/rnd:2005046. [DOI] [PubMed] [Google Scholar]
  481. Stillwell W, et al. Docosahexaenoic acid affects cell signaling by altering lipid rafts. Reprod Nutr Dev. 2005b;45:559–579. doi: 10.1051/rnd:2005046. [DOI] [PubMed] [Google Scholar]
  482. Storch J, Corsico B. The emerging functions and mechanisms of mammalian fatty acid-binding proteins. Annual review of nutrition. 2008;28:73–95. doi: 10.1146/annurev.nutr.27.061406.093710. [DOI] [PubMed] [Google Scholar]
  483. Stremmel W, et al. Identification, isolation, and partial characterization of a fatty acid binding protein from rat jejunal microvillous membranes. The Journal of clinical investigation. 1985a;75:1068–1076. doi: 10.1172/JCI111769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  484. Stremmel W, et al. Isolation and partial characterization of a fatty acid binding protein in rat liver plasma membranes. Proceedings of the National Academy of Sciences of the United States of America. 1985b;82:4–8. doi: 10.1073/pnas.82.1.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  485. Stremmel W, Strohmeyer G, Berk PD. Hepatocellular uptake of oleate is energy dependent, sodium linked, and inhibited by an antibody to a hepatocyte plasma membrane fatty acid binding protein. Proceedings of the National Academy of Sciences of the United States of America. 1986;83:3584–3588. doi: 10.1073/pnas.83.11.3584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  486. Stremmel W. Fatty acid uptake by isolated rat heart myocytes represents a carrier-mediated transport process. The Journal of clinical investigation. 1988a;81:844–852. doi: 10.1172/JCI113393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  487. Stremmel W. Uptake of fatty acids by jejunal mucosal cells is mediated by a fatty acid binding membrane protein. The Journal of clinical investigation. 1988b;82:2001–2010. doi: 10.1172/JCI113820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  488. Strokin M, Sergeeva M, Reiser G. Docosahexaenoic acid and arachidonic acid release in rat brain astrocytes is mediated by two separate isoforms of phospholipase A2 and is differently regulated by cyclic AMP and Ca2+ British journal of pharmacology. 2003;139:1014–1022. doi: 10.1038/sj.bjp.0705326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  489. Sublette ME, et al. Omega-3 polyunsaturated essential fatty acid status as a predictor of future suicide risk. Am J Psychiatry. 2006;163:1100–1102. doi: 10.1176/ajp.2006.163.6.1100. [DOI] [PubMed] [Google Scholar]
  490. Sublette ME, et al. Plasma free polyunsaturated fatty acid levels are associated with symptom severity in acute mania. Bipolar Disord. 2007;9:759–765. doi: 10.1111/j.1399-5618.2007.00387.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  491. Sublette ME, et al. Plasma polyunsaturated fatty acids and regional cerebral glucose metabolism in major depression. Prostaglandins, leukotrienes, and essential fatty acids. 2009;80:57–64. doi: 10.1016/j.plefa.2008.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  492. Sublette ME, et al. Meta-analysis of the effects of eicosapentaenoic acid (EPA) in clinical trials in depression. J Clin Psychiatry. 2011 doi: 10.4088/JCP.10m06634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  493. Sublette ME, et al. Polyunsaturated fatty acid associations with dopaminergic indices in major depressive disorder. Int J Neuropsychopharmacol. 2014;17:383–391. doi: 10.1017/S1461145713001399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  494. Summers LK, et al. Uptake of individual fatty acids into adipose tissue in relation to their presence in the diet. The American journal of clinical nutrition. 2000;71:1470–1477. doi: 10.1093/ajcn/71.6.1470. [DOI] [PubMed] [Google Scholar]
  495. Surette ME, et al. Evidence for mechanisms of the hypotriglyceridemic effect of n-3 polyunsaturated fatty acids. Biochimica et biophysica acta. 1992;1126:199–205. doi: 10.1016/0005-2760(92)90291-3. [DOI] [PubMed] [Google Scholar]
  496. Szentandrassy N, et al. Protein kinase A is activated by the n-3 polyunsaturated fatty acid eicosapentaenoic acid in rat ventricular muscle. The Journal of physiology. 2007;582:349–358. doi: 10.1113/jphysiol.2007.132753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  497. Tai ES, et al. Polyunsaturated fatty acids interact with the PPARA-L162V polymorphism to affect plasma triglyceride and apolipoprotein C-III concentrations in the Framingham Heart Study. The Journal of nutrition. 2005;135:397–403. doi: 10.1093/jn/135.3.397. [DOI] [PubMed] [Google Scholar]
  498. Tan NS, et al. Selective cooperation between fatty acid binding proteins and peroxisome proliferator-activated receptors in regulating transcription. Molecular and cellular biology. 2002;22:5114–5127. doi: 10.1128/MCB.22.14.5114-5127.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  499. Tan ZS, et al. Red blood cell omega-3 fatty acid levels and markers of accelerated brain aging. Neurology. 2012;78:658–664. doi: 10.1212/WNL.0b013e318249f6a9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  500. Tanaka T, et al. Genome-wide association study of plasma polyunsaturated fatty acids in the InCHIANTI Study. PLoS Genet. 2009;5:e1000338. doi: 10.1371/journal.pgen.1000338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  501. Tanskanen A, et al. Fish consumption and depressive symptoms in the general population in Finland. Psychiatr Serv. 2001;52:529–531. doi: 10.1176/appi.ps.52.4.529. [DOI] [PubMed] [Google Scholar]
  502. Tchantchou F, et al. The fatty acid amide hydrolase inhibitor PF-3845 promotes neuronal survival, attenuates inflammation and improves functional recovery in mice with traumatic brain injury. Neuropharmacology. 2014;85:427–439. doi: 10.1016/j.neuropharm.2014.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  503. Teague H, et al. Dendritic cell activation, phagocytosis,and CD69 expression on cognate T cells are suppressed by n-3 long chain polyunsaturated fatty acids. Immunology. 2013 doi: 10.1111/imm.12088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  504. Tence M, et al. Synergistic effects of acetylcholine and glutamate on the release of arachidonic acid from cultured striatal neurons. Journal of neurochemistry. 1995;64:1605–1613. doi: 10.1046/j.1471-4159.1995.64041605.x. [DOI] [PubMed] [Google Scholar]
  505. Thambisetty M, et al. The utility of (11)C-arachidonate PET to study in vivo dopaminergic neurotransmission in humans. J Cereb Blood Flow Metab. 2012;32:676–684. doi: 10.1038/jcbfm.2011.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  506. Thies F, et al. Preferential incorporation of sn-2 lysoPC DHA over unesterified DHA in the young rat brain. The American journal of physiology. 1994;267:R1273–R1279. doi: 10.1152/ajpregu.1994.267.5.R1273. [DOI] [PubMed] [Google Scholar]
  507. Thompson J, Reese-Wagoner A, Banaszak L. Liver fatty acid binding protein: species variation and the accommodation of different ligands. Biochimica et biophysica acta. 1999;1441:117–130. doi: 10.1016/s1388-1981(99)00146-8. [DOI] [PubMed] [Google Scholar]
  508. Toorop AI, Romsos DR, Leveille GA. The metabolic fate of dietary 1-[14C]linoleate and 1-[14C]palmitate in meal-fed rats. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine. 1979;160:312–316. doi: 10.3181/00379727-160-40440. [DOI] [PubMed] [Google Scholar]
  509. Trebunova A, et al. The influence of omega-3 polyunsaturated fatty acids feeding on composition of fatty acids in fatty tissues and eggs of laying hens. DTW. Deutsche tierarztliche Wochenschrift. 2007;114:275–279. [PubMed] [Google Scholar]
  510. Triboulot C, et al. Dietary (n-3) polyunsaturated fatty acids exert antihypertensive effects by modulating calcium signaling in T cells of rats. J Nutr. 2001;131:2364–2369. doi: 10.1093/jn/131.9.2364. [DOI] [PubMed] [Google Scholar]
  511. Tso P. Intestinal lipid absoprtion, Vol. New York, NY: Raven Press; 1994. [Google Scholar]
  512. Turcotte LP, et al. Muscle palmitate uptake and binding are saturable and inhibited by antibodies to FABP(PM) Molecular and cellular biochemistry. 2000;210:53–63. doi: 10.1023/a:1007046929776. [DOI] [PubMed] [Google Scholar]
  513. Turk HF, Chapkin RS. Membrane lipid raft organization is uniquely modified by n-3 polyunsaturated fatty acids. Prostaglandins Leukot Essent Fatty Acids. 2012 doi: 10.1016/j.plefa.2012.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  514. U.S. Department of Agriculture: Agricultural Research Service. Nutrient Intakes from Food: Mean Amounts and Percentages of Calories from Protein, Carbohydrate, Fat, and Alcohol, One Day, 2005–2006. 2008 Vol., ed.^eds. [Google Scholar]
  515. Umhau JC, et al. Imaging incorporation of circulating docosahexaenoic acid into the human brain using positron emission tomography. J Lipid Res. 2009;50:1259–1268. doi: 10.1194/jlr.M800530-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  516. Umhau JC, et al. Brain Docosahexaenoic Acid [DHA] Incorporation and Blood Flow Are Increased in Chronic Alcoholics: A Positron Emission Tomography Study Corrected for Cerebral Atrophy. PLoS One. 2013;8:e75333. doi: 10.1371/journal.pone.0075333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  517. van Gelder BM, et al. Fish consumption, n-3 fatty acids, and subsequent 5-y cognitive decline in elderly men: the Zutphen Elderly Study. The American journal of clinical nutrition. 2007;85:1142–1147. doi: 10.1093/ajcn/85.4.1142. [DOI] [PubMed] [Google Scholar]
  518. Van Laethem F, Leo O. Membrane lipid rafts: new targets for immunoregulation. Curr Mol Med. 2002;2:557–570. doi: 10.2174/1566524023362122. [DOI] [PubMed] [Google Scholar]
  519. van Raalte DH, et al. Peroxisome proliferator-activated receptor (PPAR)-alpha: a pharmacological target with a promising future. Pharm Res. 2004;21:1531–1538. doi: 10.1023/b:pham.0000041444.06122.8d. [DOI] [PubMed] [Google Scholar]
  520. Vancassel S, et al. n-3 Polyunsaturated fatty acid supplementation reverses stress-induced modifications on brain monoamine levels in mice. J Lipid Res. 2008;49:340–348. doi: 10.1194/jlr.M700328-JLR200. [DOI] [PubMed] [Google Scholar]
  521. Vance JE, Vance DE. Lipoprotein assembly and secretion by hepatocytes. Annu Rev Nutr. 1990;10:337–356. doi: 10.1146/annurev.nu.10.070190.002005. [DOI] [PubMed] [Google Scholar]
  522. Veena SR, et al. Infant feeding practice and childhood cognitive performance in South India. Archives of disease in childhood. 2010;95:347–354. doi: 10.1136/adc.2009.165159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  523. Veerkamp JH, Peeters RA, Maatman RG. Structural and functional features of different types of cytoplasmic fatty acid-binding proteins. Biochimica et biophysica acta. 1991;1081:1–24. doi: 10.1016/0005-2760(91)90244-c. [DOI] [PubMed] [Google Scholar]
  524. Veerkamp JH, Zimmerman AW. Fatty acid-binding proteins of nervous tissue. Journal of molecular neuroscience : MN. 2001;16:133–142. doi: 10.1385/JMN:16:2-3:133. discussion 151-7. [DOI] [PubMed] [Google Scholar]
  525. Veerkamp JH, Maatman RGHJ. Cytoplasmic fatty acid-binding proteins: their structure and genes. Prog. Lipid Res. 1995;34:17–52. doi: 10.1016/0163-7827(94)00005-7. [DOI] [PubMed] [Google Scholar]
  526. Vial D, Piomelli D. Dopamine D2 receptors potentiate arachidonate release via activation of cytosolic, arachidonate-specific phospholipase A2. Journal of neurochemistry. 1995;64:2765–2772. doi: 10.1046/j.1471-4159.1995.64062765.x. [DOI] [PubMed] [Google Scholar]
  527. Wang ZJ, et al. Neuroprotective effects of arachidonic acid against oxidative stress on rat hippocampal slices. Chemico-biological interactions. 2006;163:207–217. doi: 10.1016/j.cbi.2006.08.005. [DOI] [PubMed] [Google Scholar]
  528. Washizaki K, et al. Brain arachidonic acid incorporation and precursor pool specific activity during intravenous infusion of unesterified [3H]arachidonate in the anesthetized rat. Journal of neurochemistry. 1994;63:727–736. doi: 10.1046/j.1471-4159.1994.63020727.x. [DOI] [PubMed] [Google Scholar]
  529. Wassall SR, et al. Order from disorder, corralling cholesterol with chaotic lipids. The role of polyunsaturated lipids in membrane raft formation. Chem Phys Lipids. 2004;132:79–88. doi: 10.1016/j.chemphyslip.2004.09.007. [DOI] [PubMed] [Google Scholar]
  530. Watanabe A, et al. Fabp7 maps to a quantitative trait locus for a schizophrenia endophenotype. PLoS Biol. 2007;5:e297. doi: 10.1371/journal.pbio.0050297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  531. Webb Y, Hermida-Matsumoto L, Resh MD. Inhibition of protein palmitoylation, raft localization, and T cell signaling by 2-bromopalmitate and polyunsaturated fatty acids. J Biol Chem. 2000;275:261–270. doi: 10.1074/jbc.275.1.261. [DOI] [PubMed] [Google Scholar]
  532. Weisiger RA, Zucker SD. Transfer of fatty acids between intracellular membranes: roles of soluble binding proteins, distance, and time. American journal of physiology. Gastrointestinal and liver physiology. 2002;282:G105–G115. doi: 10.1152/ajpgi.00238.2001. [DOI] [PubMed] [Google Scholar]
  533. Weisinger RA. Cytoplasmic transport of lipids: role of binding proteins. Comp. Biochem. Physiol. 1996;115B:319–331. [Google Scholar]
  534. Wenk M. The emerging field of lipidomics. Nat Rev Drug Discov. 2005;4:594–610. doi: 10.1038/nrd1776. [DOI] [PubMed] [Google Scholar]
  535. Widstrom RL, Norris AW, Spector AA. Binding of cytochrome P450 monooxygenase and lipoxygenase pathway products by heart fatty acid-binding protein. Biochemistry. 2001;40:1070–1076. doi: 10.1021/bi001602y. [DOI] [PubMed] [Google Scholar]
  536. Wieland SJ, Fletcher JE, Gong QH. Differential modulation of a sodium conductance in skeletal muscle by intracellular and extracellular fatty acids. The American journal of physiology. 1992;263:C308–C312. doi: 10.1152/ajpcell.1992.263.2.C308. [DOI] [PubMed] [Google Scholar]
  537. Williams JA, et al. Docosahexaenoic and Eicosapentaenoic Acids Segregate Differently between Raft and Nonraft Domains. Biophys J. 2012;103:228–237. doi: 10.1016/j.bpj.2012.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  538. Williams WM, et al. In vivo incorporation from plasma of radiolabeled palmitate and arachidonate into rat brain microvessels. Microvasc Res. 1997;53:163–166. doi: 10.1006/mvre.1996.1984. [DOI] [PubMed] [Google Scholar]
  539. Wolfrum C, et al. Binding of fatty acids and peroxisome proliferators to orthologous fatty acid binding proteins from human, murine, and bovine liver. Biochemistry. 2000;39:14363. doi: 10.1021/bi0051187. [DOI] [PubMed] [Google Scholar]
  540. Wong SH, et al. The adaptive effects of dietary fish and safflower oil on lipid and lipoprotein metabolism in perfused rat liver. Biochimica et biophysica acta. 1984;792:103–109. doi: 10.1016/0005-2760(84)90209-1. [DOI] [PubMed] [Google Scholar]
  541. Wong SW, et al. Fatty acids modulate Toll-like receptor 4 activation through regulation of receptor dimerization and recruitment into lipid rafts in a reactive oxygen species-dependent manner. J Biol Chem. 2009a;284:27384–27392. doi: 10.1074/jbc.M109.044065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  542. Wong SW, et al. Fatty acids modulate Toll-like receptor 4 activation through regulation of receptor dimerization and recruitment into lipid rafts in a reactive oxygen species-dependent manner. The Journal of biological chemistry. 2009b;284:27384–27392. doi: 10.1074/jbc.M109.044065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  543. Wosilait WD, Soler-Argilaga C. A theoretical analysis of the multiple binding of palmitate by bovine serum albumin: the relationship to uptake of free fatty acids by tissues. Life sciences. 1975;17:159–166. doi: 10.1016/0024-3205(75)90252-0. [DOI] [PubMed] [Google Scholar]
  544. Wu A, Ying Z, Gomez-Pinilla F. Dietary omega-3 fatty acids normalize BDNF levels, reduce oxidative damage, and counteract learning disability after traumatic brain injury in rats. J Neurotrauma. 2004;21:1457–1467. doi: 10.1089/neu.2004.21.1457. [DOI] [PubMed] [Google Scholar]
  545. Xiao YF, et al. Suppression of voltage-gated L-type Ca2+ currents by polyunsaturated fatty acids in adult and neonatal rat ventricular myocytes. Proceedings of the National Academy of Sciences of the United States of America. 1997;94:4182–4187. doi: 10.1073/pnas.94.8.4182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  546. Xu LZ, et al. Ligand specificity of brain lipid-binding protein. The Journal of biological chemistry. 1996;271:24711–24719. doi: 10.1074/jbc.271.40.24711. [DOI] [PubMed] [Google Scholar]
  547. Xu S, et al. CD36 enhances fatty acid uptake by increasing the rate of intracellular esterification but not transport across the plasma membrane. Biochemistry. 2013;52:7254–7261. doi: 10.1021/bi400914c. [DOI] [PubMed] [Google Scholar]
  548. Yamakawa-Kobayashi K, et al. A Val227Ala polymorphism in the peroxisome proliferator activated receptor alpha (PPARalpha) gene is associated with variations in serum lipid levels. Journal of medical genetics. 2002;39:189–191. doi: 10.1136/jmg.39.3.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  549. Yaqoob P, Shaikh SR. The nutritional and clinical significance of lipid rafts. Current opinion in clinical nutrition and metabolic care. 2010;13:156–166. doi: 10.1097/MCO.0b013e328335725b. [DOI] [PubMed] [Google Scholar]
  550. Ye S, et al. Polyunsaturated docosahexaenoic acid suppresses oxidative stress induced endothelial cell calcium influx by altering lipid composition in membrane caveolar rafts. Prostaglandins Leukot Essent Fatty Acids. 2010;83:37–43. doi: 10.1016/j.plefa.2010.02.002. [DOI] [PubMed] [Google Scholar]
  551. Yog R, et al. n-3 polyunsaturated fatty acids suppress mitochondrial translocation to the immunologic synapse and modulate calcium signaling in T cells. Journal of immunology. 2010;184:5865–5873. doi: 10.4049/jimmunol.0904102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  552. Yoshida H, et al. Effect of dietary seal and fish oils on triacylglycerol metabolism in rats. Journal of nutritional science and vitaminology. 1999;45:411–421. doi: 10.3177/jnsv.45.411. [DOI] [PubMed] [Google Scholar]
  553. Youdim KA, Martin A, Joseph JA. Essential fatty acids and the brain: possible health implications. International journal of developmental neuroscience : the official journal of the International Society for Developmental Neuroscience. 2000;18:383–399. doi: 10.1016/s0736-5748(00)00013-7. [DOI] [PubMed] [Google Scholar]
  554. Young G, Conquer J. Omega-3 fatty acids and neuropsychiatric disorders. Reproduction, nutrition, development. 2005;45:1–28. doi: 10.1051/rnd:2005001. [DOI] [PubMed] [Google Scholar]
  555. Yuan ZX, et al. Identification and profiling of targeted oxidized linoleic acid metabolites in rat plasma by quadrupole time-of-flight mass spectrometry. Biomed Chromatogr. 2013;27:422–432. doi: 10.1002/bmc.2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  556. Zeyda M, et al. LAT displacement from lipid rafts as a molecular mechanism for the inhibition of T cell signaling by polyunsaturated fatty acids. J Biol Chem. 2002;277:28418–28423. doi: 10.1074/jbc.M203343200. [DOI] [PubMed] [Google Scholar]
  557. Zilversmit DB. The composition and structure of lymph chylomicrons in dog, rat, and man. J Clin Invest. 1965;44:1610–1622. doi: 10.1172/JCI105267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  558. Zimmer L, et al. Chronic n-3 polyunsaturated fatty acid diet-deficiency acts on dopamine metabolism in the rat frontal cortex: a microdialysis study. Neurosci Lett. 1998;240:177–181. doi: 10.1016/s0304-3940(97)00938-5. [DOI] [PubMed] [Google Scholar]
  559. Zimmer L, et al. n-3 polyunsaturated fatty acid deficiency and dopamine metabolism in the rat frontal cortex. Lipids. 1999;34(Suppl):S251. doi: 10.1007/BF02562309. [DOI] [PubMed] [Google Scholar]
  560. Zimmer L, et al. Modification of dopamine neurotransmission in the nucleus accumbens of rats deficient in n-3 polyunsaturated fatty acids. J Lipid Res. 2000a;41:32–40. [PubMed] [Google Scholar]
  561. Zimmer L, et al. Chronic n-3 polyunsaturated fatty acid deficiency alters dopamine vesicle density in the rat frontal cortex. Neurosci Lett. 2000b;284:25–28. doi: 10.1016/s0304-3940(00)00950-2. [DOI] [PubMed] [Google Scholar]
  562. Zimmer L, et al. Chronic n-3 polyunsaturated fatty acid deficiency alters dopamine vesicle density in the rat frontal cortex. Neuroscience Letters. 2000c;284:25–28. doi: 10.1016/s0304-3940(00)00950-2. [DOI] [PubMed] [Google Scholar]
  563. Zucker SD. Kinetic model of protein-mediated ligand transport: influence of soluble binding proteins on the intermembrane diffusion of a fluorescent fatty acid. Biochemistry. 2001;40:977–986. doi: 10.1021/bi001277e. [DOI] [PubMed] [Google Scholar]

RESOURCES