Abstract
Over the past three decades a body of translational evidence has implicated dietary deficiency in long-chain omega-3 (LCn-3) fatty acids, including eicosapenaenoic acid (EPA) and docosahexaenoic acid (DHA), in the pathophysiology and etiology of major depressive disorder (MDD). Cross-national and cross-sectional data suggest that greater habitual intake of preformed EPA+DHA is associated with reduced risk for developing depressive symptoms and syndromal MDD. Erythrocyte EPA and DHA composition is highly correlated with habitual fish or fish oil intake, and case-control studies have consistently observed lower erythrocyte EPA and/or DHA levels in patients with MDD. Low erythrocyte EPA+DHA composition may also be associated with increased risk for suicide and cardiovascular disease, two primary causes of excess premature mortality in MDD. While controversial, dietary EPA+DHA supplementation may have antidepressant properties and may augment the therapeutic efficacy of antidepressant medications. Neuroimaging and rodent neurodevelopmental studies further suggest that low LCn-3 fatty acid intake or biostatus can recapitulate central pathophysiological features associated with MDD. Prospective findings suggest that low LCn-3 fatty acid biostatus increases risk for depressive symptoms in part by augmenting pro-inflammatory responsivity. When taken collectively, these translational findings provide a strong empirical foundation in support of dietary LCn-3 fatty acid deficiency as a modifiable risk factor for MDD. This review provides an overview of this translational evidence and then discusses future directions including strategies to translate this evidence into routine clinical screening and treatment algorithms.
Keywords: Major depressive disorder, Long-chain omega-3 fatty acids, Eicosapentaenoic acid (EPA), Docosahexaenoic acid (DHA)
Introduction
Major depressive disorder (MDD) is a leading cause of disability globally. In the United States (U.S.) severe forms of MDD are estimated to affect between 2-7% of the population and up to 16-20% suffer from milder forms (Kessler et al., 2007). The initial onset of MDD frequently occurs during adolescence and young adulthood (Kessler et al., 2005), and is ∼2-fold more prevalent in females after puberty (Kessler, 2003). Outcomes data indicate that MDD is associated with excess premature mortality primarily attributable to suicide and cardiovascular-related disorders (Angst et al., 2002; Osby et al., 2001). Bipolar disorder is also associated with recurrent episodes of depression, and prodromal MDD is a risk factor for mania in at-risk youth (Axelson et al., 2015; Egeland et al., 2000; Howes et al., 2011). The first-line treatment for MDD in adolescents and adults is typically a selective serotonin reuptake inhibitor (SSRI). However, approximately 30-40 percent of adolescent MDD patients exhibit residual symptoms following standard SSRI treatment (Kennard et al., 2006; Emslie et al., 1997), and SSRI treatment may precipitate suicidality and mania in at-risk youth (Hammad et al., 2006; Martin et al., 2004; Strawn et al., 2014). These and other data highlight an urgent need to identify modifiable risk and resilience mechanisms associated with the etiology of MDD to inform improvements in treatment and ultimately prevention strategies.
While aggressive efforts have been devoted to the identification of genetic risk factors associated with psychiatric disorders including MDD, it has become apparent that both genetic and environmental factors confer vulnerability (Ehringer et al., 2006; Merikangas et al., 2002). For example, a meta-analysis of community-based twin studies of MDD yielded a heritability estimate of .37, indicating that approximately two thirds of the liability is attributable to environmental factors (Sullivan et al., 2000). Moreover, environmental factors can regulate gene expression through epigenetic effects (i.e., DNA methylation) independent of DNA sequence polymorphisms (Perroud et al., 2014; Zhang et al., 2013). Environmental factors can also interact with DNA polymorphisms to increase risk for developing psychiatric disorders (Caspi & Moffitt, 2006). Accordingly, aggressive efforts also need to be devoted to the identification of environmental risk factors, particularly in view of their amenability to modification and prevention.
Over the last three decades a body of translational evidence has emerged which suggests that the habitual diet is relevant to the etiology of MDD. Specifically, evidence has implicated dietary deficiency in essential long-chain omega-3 (LCn-3) fatty acids, including eicosapenaenoic acid (EPA) and docosahexaenoic acid (DHA), in the pathophysiology and etiology of MDD. This is supported by converging evidence from cross-national and cross-sectional epidemiological surveys, case-control LCn-3 fatty acid biostatus studies, prospective observational and LCn-3 fatty acid intervention studies, rodent neurodevelopmental studies, and recent neuroimaging findings. Additionally, accumulating evidence suggests that LCn-3 fatty acid deficiency may increase risk for suicide and cardiovascular disease, two primary causes of excess premature mortality in patients with MDD. This review provides an overview of translational evidence implicating LCn-3 fatty acid deficiency in the pathophysiology and etiology of MDD, and then discusses future directions including strategies to translate this evidence into routine clinical screening and treatment algorithms.
LCn-3 fatty acid biosynthesis and biostatus
As background, omega-3 (n-3) and omega-6 (n-6) fatty acids are members of the polyunsaturated fatty acid (PUFA) family. Primary dietary sources of the short-chain n-3 fatty acid precursor α-linolenic acid (ALA, 18:3n-3) include flaxseed, linseed, canola, soy, and perilla oils, and primary dietary sources of the short-chain n-6 fatty acid precursor linoleic acid (18:2n-6) include safflower, soy, and corn oils. These PUFAs are considered ‘essential’ because mammals are entirely dependent on dietary sources to procure and maintain adequate concentrations in peripheral and central tissues. The biosynthesis of long-chain n-3 (LCn-3) fatty acids, including EPA (20:5n-3) and DHA (22:6n-3), from their short-chain precursors require a series of common and competitive microsomal desaturation and elongation reactions (Fig. 1)(Reardon & Brenna, 2013). The rate-limiting enzymes regulating LC-PUFA biosynthesis include delta-6 desaturase (delta6-desaturase, FADS2) and delta-5 desaturase (delta5-desaturase, FADS1), as well as elongases (e.g., ELOVL5), and the final synthesis of DHA is catalyzed by β-oxidation within peroxisomes (Wanders, 2013). FASD1 and FADS2 genes are primarily expressed in the liver and brain and are co-localized to human chromosome 11q12-11q13.1 (Marquardt et al., 2000). Desaturase enzymes are regulated by several factors including gonadal hormones (Bakewell et al., 2006; Burdge & Wootten, 2002; Childs et al., 2010; Giltay et al., 2004; McNamara et al., 2009a), insulin (Brenner, 2003), as well as single nucleotide polymorphisms within FADS2 and/or FASD1 genes (Lattka et al., 2010). Recent evidence further suggests that epigenetic effects (i.e., DNA methylation) are associated with delta5/6-desaturase enzyme activity (Howard et al., 2014) and that PUFA intake can induce DNA methylation resulting in reduced expression of FADS2 and ELOVL5 (Hoile et al., 2014). Therefore, PUFA homeostasis is ultimately governed by both environmental (i.e., diet) and genetic factors.
In healthy adult subjects ALA→EPA biosynthesis is extremely limited and ALA→DHA and EPA→DHA biosynthesis is negligible (Brenna et al., 2009). For example, 12-week supplementation with up to 3.6 g/d of flaxseed oil, a rich source of ALA, resulted in moderate increases in erythrocyte (red blood cell) EPA but did not significantly increase erythrocyte DHA levels in healthy adult subjects (Barcelo-Coblijn et al., 2008). Similarly, 4-week supplementation with flaxseed oil resulted in moderate increases in erythrocyte and breast milk EPA but did not significantly increase erythrocyte or breast milk DHA (Francois et al., 2003). This limitation in biosynthesis may be due in part to competition with high levels of linoleic acid (18:2n-6) in the diet (Taha et al., 2014). Unlike flaxseed oil, supplementation with fish oil robustly increases erythrocyte and breastmilk EPA+DHA biostatus (Flock et al., 2013; Arterburn et al., 2006; Barcelo-Coblijn et al., 2008). Primary sources of preformed LCn-3 fatty acids (EPA+DHA) include fatty cold water fish, including salmon, trout, tuna, as well as fish oil and algal-derived supplements. Accordingly, cross-national evidence has found that habitual fish intake is positively correlated with breastmilk (Brenna et al., 2007) and blood (Sands et al., 2005; Itomura et al., 2008) EPA+DHA composition. Together, these findings highlight the limited efficiency of hepatic PUFA biosynthesis in healthy human subjects and suggest that LCn-3 fatty acid biostatus is highly dependent on the LCn-3 fatty acid composition of the habitual diet.
Relevance to depression
Epidemiology
Cross-national epidemiological surveys have observed a significant inverse correlation between per capita fish or seafood consumption (primary dietary sources of preformed EPA+DHA) and lifetime prevalence rates of MDD (Hibbeln, 1998; Peet, 2004), postpartum depression (Hibbeln et al., 2002), and bipolar spectrum disorders (Noaghiul & Hibbeln, 2003). Several cross-sectional studies have investigated the relationship between habitual fish or fish oil intake and depression rates in the general population. For example, a cross-sectional survey of 21,835 adult and elderly subjects from Norway found that subjects who ingested cod liver oil on a daily basis (EPA: ∼300-600 mg/d; DHA: ∼300-600 mg/d) were 30 percent less likely to have depressive symptoms than non-users after adjusting for multiple possible confounding factors (Raeder et al., 2007). A meta-analyses of thirteen cross-sectional studies found that higher intake of fish (as well as fruit, vegetables, and whole grains) was significantly associated with a reduced depression risk (Lai et al., 2014). Because the initial onset of MDD frequently occurs during adolescence, it is relevant that lower dietary LCn-3 fatty acid intake in adolescents is associated with elevated depressive symptoms as well as cardiovascular risk factors (Allen et al., 2013; Murakami et al., 2010; Oddy et al., 2011; O'Sullivan et al., 2011; Swenne et al., 2011). Although these findings provide indirect support for an inverse association between fish intake frequency and MDD prevalence, numerous cultural and genetic variables may also contribute to this association.
It has been estimated that there has been a sharp increase in the consumption of linoleic acid (18:2n-6), and a reciprocal decline in α-linolenic acid (18:3n-3) and LCn-3 fatty acids, in the U.S. over the last century (Blasbalg et al., 2011). While it is unclear whether this relative decline in LCn-3 fatty acids has been associated with a change in the prevalence rates of depression in the U.S., a retrospective study found that shifts away from fish-based to Western diets in Arctic communities was associated with increased rates of seasonal affective disorder, depression, suicide, and cardiovascular disease (McGrath-Hanna et al., 2003). Moreover, case-control studies find that patients with mood disorders are more likely to consume diets with lower amounts of LCn-3 fatty acids and ALA compared with healthy controls and/or recommended intake levels (Clayton et al., 2008; Edwards et al., 1998; Evans et al., 2014; Davison & Kaplan, 2012; Jacka et al., 2011). It is also relevant that MDD is associated with excess premature mortality primarily attributable to suicide and cardiovascular disease (Angst et al., 2002; Osby et al., 2001), and cross-sectional epidemiological evidence suggests that higher LCn-3 fatty acid intake may be protective against suicidality (Tanskanen et al., 2001) and cardiovascular disease (Whelton et al., 2004). Together these findings suggest that habitual diets containing lower amounts of LCn-3 fatty acids may increase risk for developing depressive symptoms, suicidality, and co-morbid cardiovascular disease.
LCn-3 fatty acid biosynthesis and biostatus
Extant evidence from genome-wide association studies does not support an association between polymorphisms in delta-6 (FASD2) and delta-5 desaturase (FADS1) genes in the etiology of MDD (Levinson, 2006). A recent genotyping study did not observe an association between common single-nucleotide polymorphisms in FADS1 or FASD2 genes and depression or suicidality (Sublette et al., 2015). However, DNA methylation in the Elovl5 gene was found to be associated with depression and suicidality (Haghighi et al., 2015). Moreover, FADS1 mRNA expression was significantly lower in the postmortem prefrontal cortex of MDD patients relative to controls, and there were trends for lower expression of FADS2 and ELOVL5 (McNamara & Liu, 2011). A microarray study similarly found that FADS1 expression was reduced in postmortem prefrontal cortices of male MDD patients that committed suicide (Lalovic et al., 2010). It is also notable that antidepressant medications upregulate the expression of sterol regulatory element-binding protein (SREBP) (Raeder et al., 2006) which positively regulates FADS1 and FADS2 transcription (Matsuzaka et al., 2002). While these findings must be viewed as preliminary, they suggest that the dysregulation in LCn-3 fatty acid homeostasis in MDD may be mediated in part by epigenetic modifications of biosynthetic enzymes.
Several independent cross-sectional studies conducted in different countries have observed significantly lower erythrocyte membrane or plasma phospholipid EPA+DHA levels in adult MDD patients compared with healthy controls. A meta-analysis of 14 cross-sectional fatty acid composition studies found significant deficits in EPA and DHA, but not arachidonic acid (AA), in MDD patients (Lin et al., 2010). Other cross-sectional studies have similarly found that pediatric and adolescent patients with MDD exhibit erythrocyte EPA+DHA deficits compared with healthy youth (Clayton et al., 2008; Pottala et al., 2013; McNamara et al., 2014a). Some studies (Clayton et al., 2008; Adams et al., 1996; Edwards et al., 1998), but not others (Assies et al., 2010; Liu et al., 2013; McNamara et al., 2010a), have observed an inverse correlation between blood EPA+DHA levels, or a positive correlation between the AA/EPA ratio, and depression symptom severity. It is notable that erythrocyte EPA and/or DHA deficits are not unique to MDD, and have also been observed in patients with bipolar I disorder (McNamara & Welge, 2016), anxiety disorders (Green et al., 2006), and schizophrenia (van der Kemp et al., 2012). While there may be different etiological factors contributing to lower blood EPA+DHA levels observed in MDD patients, dietary fish oil supplementation, but not flaxseed oil supplementation (Gracious et al., 2010), is sufficient to robustly increase patient blood EPA+DHA levels (McNamara et al., 2014a). In view of evidence that erythrocyte EPA+DHA composition is highly correlated with habitual dietary fish intake and/or fish oil supplementation (Flock et al., 2013; Sands et al., 2005; Itomura et al., 2008), these data suggest that dietary EPA+DHA insufficiency represents a modifiable risk factor for EPA+DHA deficiency observed in MDD.
Several lines of evidence also suggest that increasing LCn-3 fatty acid status may reduce risk of suicide, a primary cause of excess premature mortality in MDD (Angst et al., 2002; Osby et al., 2001). A prospective longitudinal study found that low baseline plasma DHA composition was a significant predictor of future suicidal attempts in medication-free MDD patients (Sublette et al., 2006). In two case-control studies, erythrocyte or plasma LCn-3 fatty acid composition was found to be significantly reduced in suicidal patients (Garland et al., 2007; Huan et al., 2004). Two controlled trials found that dietary LCn-3 fatty acid supplementation reduced suicidality in MDD patients (Hallahan et al., 2007; Peet & Horrobin, 2002). However, prospective cohort studies have not observed an association between LCn-3 fatty acid intake and completed suicide in the general population (Tsai et al., 2014). Therefore, while extant evidence suggests that increasing LCn-3 fatty acid status in patients with psychiatric illness may be protective against suicidality, additional research is needed to evaluate whether depressed mood mediates this effect.
Excess premature mortality in patients with MDD is also attributable to cardiovascular-related diseases (Angst et al., 2002; Osby et al., 2001). Cross-sectional and prospective longitudinal studies suggest that low erythrocyte EPA+DHA (‘omega-3 index’) biostatus increases risk for cardiometabolic risk factors (Farzaneh-Far et al., 2009; Kelly et al., 2009; McKenney & Sica, 2007) and sudden cardiac arrest (Albert et al., 2002; Hu et al., 2002; Siscovick et al., 1995; Harris & Von Schacky, 2004). Moreover, LCn-3 fatty acid intake is associated with methylation of genes regulating immune-inflammatory and lipid homeostasis (Aslibekyan et al., 2014; Lee et al., 2013), and the low erythrocyte EPA+DHA levels observed in patients with MDD are associated with elevated serum levels of triglycerides and pro-inflammatory molecules including C-reactive protein (Baek & Park, 2013; Baghai et al., 2011). Additionally, the low erythrocyte EPA+DHA levels observed in patients with MDD are similar to levels observed in patients suffering acute coronary syndrome (Block et al., 2008) and would be anticipated to increase their risk for sudden cardiac arrest (Harris & Von Schacky, 2004)(Fig. 2). While the effects of LCn-3 fatty acid supplementation on cardiovascular events and sudden cardiac arrest in patients with a history of cardiovascular disease have been equivocal (Kwak et al., 2012), primary prevention studies in subjects without a history of cardiovascular disease (Wang et al., 2006) or subjects at high-risk for cardiovascular disease (Einvik et al., 2010) suggest that increasing LCn-3 fatty acid biostatus may have protective benefits. While there have been no LCn-3 fatty acid primary prevention studies conducted in patients with MDD, existing evidence provides a rationale for identifying and treating LCn-3 fatty acid deficiency in patients presenting with other cardiovascular risk factors.
The primary LCn-3 fatty acid found in mammalian brain gray matter is DHA, which comprises approximately 15% of total fatty acid composition (Carver et al., 2001; Connor et al., 1990; McNamara et al., 2008). Although EPA (20:5n-3) crosses the blood-brain barrier, it is rapidly oxidized and consequently comprises <1% of total brain fatty acid composition (Chen et al., 2013). In general human erythrocyte and frontal cortex DHA composition are positively correlated (Carver et al., 2001), and non-human primate studies indicate that DHA recuperation occurs more rapidly in erythrocytes than cortical gray matter following dietary fish oil supplementation (Connor et al., 1990). Case-control studies have investigated the fatty acid composition of postmortem brain tissue from MDD patients and/or suicide victims. These studies have observed DHA deficits in the prefrontal cortex or anterior cingulate of adult patients with MDD (Conklin et al., 2010; McNamara et al., 2007, 2013a), but not in the prefrontal cortex of adolescent or adult suicide victims (Lalovic et al., 2007; McNamara et al., 2009b). Other postmortem studies have not observed significant DHA deficits in temporal lobe structures including the amygdala in patients with MDD (Hamazaki et al., 2012; McNamara et al., 2014b). While this evidence suggests that MDD may be associated with DHA deficits selective to prefrontal regions, in view of the limitations associated with the postmortem approach these findings should be viewed as preliminary (McNamara & Jandacek, 2010).
LCn-3 fatty acid supplementation studies
To date numerous small open-label or placebo-controlled studies have investigated the antidepressant effects of short-term LCn-3 fatty acid supplementation. Despite heterogeneity in study design in terms of daily dose, LCn-3 fatty acid intervention, EPA:DHA ratio, trial duration, concomitant medications, use of a bioactive placebo (i.e., olive oil), and baseline symptom severity, meta-analyses of controlled trials observed a significant, albeit modest, advantage of LCn-3 fatty acids over placebo for reducing depression symptom severity in patients with MDD (Appleton et al., 2015; Grosso et al., 2014; Mocking et al., 2016) or bipolar disorder (Sarris et al., 2012). Additional data suggests that interventions with a higher EPA to DHA ratio may have greater antidepressant efficacy (Sublette et al., 2011). Controlled and open-label trials have also found that LCn-3 fatty acid supplementation, administered either adjunctively or as monotherapy, significantly reduces depression symptom severity in pediatric and adolescent patients (Clayton et al., 2009b; McNamara et al., 2014a; Nemets et al., 2006; Wozniak et al., 2007). Controlled and open-label trials have also observed greater reductions in depressive symptoms by combining LCn-3 fatty acids with SSRI medications (Gertsik et al., 2012; Jazayeri et al., 2008; McNamara et al., 2014a; Peet & Horrobin, 2002). While this body of evidence suggests that dietary LCn-3 fatty acid supplementation may have acute ‘antidepressant’ effects, large-scale trials are warranted to confirm these findings.
In addition to acute antidepressant effects, emerging clinical evidence suggests that higher LCn-3 fatty acid status may be protective against the initial development of MDD. A prospective surveillance study found that lower baseline DHA levels were a significant predictor of depression development in human hepatitis C patients during treatment with the pro-inflammatory cytokine interferon-α (IFN-α)(Su et al., 2010). A second prospective surveillance study found that lower baseline DHA levels, or a higher baseline AA/EPA+DHA ratio, were significant predictors of depression development in initially non-depressed human hepatitis C patients during IFN-α treatment (Lotrich et al., 2013). It is notable that a higher baseline AA/EPA+DHA ratio was also associated with greater increases in the pro-inflammatory cytokine interleukin-6 (IL-6) during IFN-α treatment (Lotrich et al., 2013a). A controlled supplementation trial found that pretreatment with EPA alone, which increased both erythrocyte EPA and DHA levels, but not DHA alone decreased the incidence of depression during IFN-α treatment in hepatitis C patients (Su et al., 2014). In view of evidence that MDD patients exhibit elevated indices of peripheral (Dowlati et al., 2010) and central (Setiawan et al., 2015) inflammation, and LCn-3 fatty acids and their bioactive metabolites have anti-inflammatory and inflammation-resolving properties (Dalli et al., 2015; Groeger et al., 2010; Serhan, 2010), these prospective findings may have broader implications for understanding the role of low LCn-3 fatty acid biostatus and pro-inflammatory signaling cascades in the pathoetiology of MDD.
Neuroimaging studies
The initial onset of MDD frequently occurs during adolescence (Kessler et al., 2005), a developmental period associated with a sharp increase in frontal cortex DHA levels (Carver et al., 2001) and rapid and dynamic changes in frontal cortex functional and structural connectivity with limbic structures that regulate mood (Giedd et al., 1999, 2009; Paus et al., 1999). Youth and adults with MDD exhibit decreased frontal white matter integrity and reduced connectivity within frontal lobe cortical networks (Connolly et al., 2013; Dickstein et al., 2010; Ho et al., 2014). It is relevant, therefore, that recent neuroimaging studies found that perinatal n-3 fatty acid deficiency in monkeys (Grayson et al., 2014) or low erythrocyte DHA biostatus in typically developing children (Almeida et al., 2016) are associated with reduced functional connectivity within prefrontal cortical networks. Moreover, a recent intervention study found that fish oil supplementation increased white matter microstructural integrity in MDD patients in association with reductions in depression symptom severity (Chhetr et al., 2016). Furthermore, hippocampal grey matter volume deficits are among the most consistent and robust neurostructural abnormality observed in MDD (Kempton et al., 2011), and greater habitual dietary LCn-3 fatty acid intake (Conklin et al., 2007) and erythrocyte EPA+DHA composition (Pottala et al., 2014) are associated with larger hippocampal volumes among healthy adults. It is also notable that higher blood IL-6 levels were associated with smaller hippocampal grey matter volume among healthy adults (Marsland et al., 2008), and decreased corticostriatal functional connectivity in MDD patients (Felger et al., 2015). While additional research is required, these preliminary neuroimaging findings suggest that low EPA+DHA biostatus and associated increases in pro-inflammatory signaling may be linked with abnormalities in cortical structure and function implicated in MDD.
Rodent studies
Animal studies have provided critical insight into the role of dietary LCn-3 fatty acids in normal brain development. The advantage of animal feeding studies is the ability to systematically and selectively control n-3 fatty acid intake during development, and perform invasive investigations of brain neurochemistry, neuroanatomy, and/or gene expression. In general, feeding studies have demonstrated that brain DHA accrual during perinatal development is required for normal cortical neurogenesis (Beltz et al., 2003; Coti Bertrand et al., 2006; Kawakita et al., 2006), neuroblast migration (Yavin et al., 2009), neuronal differentiation and arborization (Calderon & Kim, 2004), neurotrophic factor expression (Ikemoto et al., 2000; Rao et al., 2007), nerve growth factor-induced neurite outgrowth and synaptogenesis (Cao et al., 2009; Innis et al., 2001), and synaptic pruning (de Velasco et al., 2012). Additionally, LCn-3 fatty acids deficiency during development is associated with systemic inflammation (Madore et al., 2014; McNamara et al., 2010b), and increased vulnerability to neurodegenerative processes associated with inflammation (Orr et al., 2013) and lipid peroxidation (Green et al., 2001; Yavin et al., 2002). These and other data suggest that there are optimal LCn-3 fatty acids levels required for normal brain maturation and resilience.
Additional evidence from animal studies suggests that deficits in dietary LCn-3 fatty acids during perinatal brain development significantly impacts neurotransmitter systems implicated in MDD including serotonin (5-hydroxytryptamine, 5-HT) and dopamine. Maternal dietary fish oil fortification significantly increases serotonin concentrations in the frontal cortex (Chalon et al., 1998) and attenuates reductions in frontal cortex serotonin content in response to chronic stress (Vancassel et al., 2008) in young adulthood. Conversely, perinatal deficits in cortical DHA accrual are associated with impaired fenfluramine-induced elevations in serotonin release which are reversible with early (P0-P14), but not later (P21), postnatal n-3 fatty acid supplementation (Kodas et al., 2004). Perinatal deficits in cortical DHA accrual are also associated with reductions in midbrain expression of tryptophan hydroxylase-2, the rate-limiting enzyme in serotonin biosynthesis (McNamara et al., 2009c), and elevations in 5-HT2A receptor binding density in the rat frontal cortex (Delion et al., 1996). The 5-HIAA/5-HT ratio, an index of serotonin turnover, is significantly elevated in the regional brain of adult rats fed n-3-deficient diets, and this increase was positively correlated with plasma IL-6 levels and prevented by early normalization of n-3 fatty acid status (McNamara et al., 2010). Importantly, the increase in the 5-HIAA/5-HT ratio observed in the perinatal DHA-deficient rat brain is opposite to that produced by the SSRI antidepressant fluoxetine (McNamara et al., 2013b). Together this evidence suggests that LCn-3 fatty acid status during development has an enduring impact on central serotonin neurotransmission in young adult rats.
Consistent with clinical evidence implicating a dysregulation in serotonin neurotransmission in the pathophysiology of depression and aggression (Arango et al., 2002; Coccaro et al., 1997), post-weaning deficits in cortical DHA accrual are associated with elevated behavioral indices of depression and aggression in rats (DeMar et al., 2006). Importantly, dietary fish oil fortification significantly decreases depression-like behavior similar to SSRI medications in the forced swimming test, an effect that may be mediated by changes in 5-HT1A receptor function (Carlezon et al., 2005; Huang et al., 2008; Vines et al., 2012). Combining dietary fish oil supplementation with fluoxetine is significantly more effective than fluoxetine alone for reducing depression-like behavior in the forced swim test (Laino et al., 2010; Lakhwani et al., 2007). Although post-weaning deficits in cortical DHA accrual are not associated with diminished SSRI efficacy in female rats in the forced swim test (McNamara et al., 2013b), it is associated with abnormal behavioral activation in male rats (Able et al., 2014). It is also relevant that the Flinders Sensitive Line rats, an inbred rat model of depression, exhibit constitutive increases in regional brain AA/DHA ratio (Green et al., 2005). Together these findings suggest that DHA biostatus is associated with serotonin-regulated behaviors including depression and aggression, and that fish oil supplementation has antidepressant-like effects similar to SSRI medications.
A deficit in mesolimbic dopamine neurotransmission has been implicated in anhedonia, a core feature of depression (Nestler & Carlezon, 2006; Stein, 2008). Rat studies have demonstrated that deficits in brain DHA accrual during perinatal development are associated with a significant loss of dopamine neurons in the ventral tegmental area, the source of mesolimbic and mesocortical dopamine projections (Ahmad et al., 2008). Perinatal deficits in brain DHA accrual is associated with enduring deficits in mesocortical and mesolimbic dopamine neurotransmission in young adult rats that are reversible with early (P0-P14), but not later (P21), postnatal n-3 fatty acid supplementation (Kodas et al., 2002 Zimmer et al., 1998, 2002). Adolescent rats subjected to perinatal deficits in brain DHA accrual also exhibit increased expression of tyrosine hydroxylase, the rate limiting enzyme in dopamine biosynthesis, in the dorsal striatum (Bondi et al., 2014). Maternal dietary fish oil supplementation throughout gestation and lactation significantly increases dopamine concentrations in the frontal cortex of adult offspring (Chalon et al., 1998). These findings suggest that early perinatal brain DHA accrual is critical for the functional maturation of mesocortical and mesolimbic dopamine systems.
Future Directions – Clinical implementation
Together the reviewed body of translational evidence strongly suggests that LCn-3 fatty acid deficiency, particularly during perinatal development, may represent a plausible and modifiable risk factor for MDD. Among the clinical findings, meta-analyses of independent case-control studies demonstrate that MDD patients exhibit significantly lower blood EPA and/or DHA levels, which are correlated with fish or fish oil intake, compared with demographically similar healthy controls. Additionally, cross-sectional evidence and meta-analyses of controlled fish oil intervention studies suggest that increasing EPA+DHA biostatus mitigates risk for depressive symptoms, and potentially suicidality and cardiovascular disease. Therefore, translating this evidence into clinical practice by implementing routine screening and treatment of low blood EPA+DHA levels in patients with MDD represents an important future direction. Below we briefly discuss existing resources and general guidelines required for routine screening and treatment of low blood EPA+DHA levels in clinical practice.
Screening for LCn-3 fatty acid deficiency
There are currently several laboratory facilities that routinely perform blood fatty acid analyses by gas-liquid chromatography. For example, OmegaQuant, LLC is a Clinical Laboratory Improvement Amendments (CLIA)-certified lab that specializes in determining the blood fatty acid composition (www.omegaquant.com). For this procedure, whole blood (∼25 uL) is obtained from a finger prick and is spotted and dried onto anti-oxidant treated card which is then shipped at ambient temperature. Analogous to routine cholesterol testing, this approach can provide a valid, reliable, and relatively non-invasive measure of a patient's EPA+DHA biostatus. Erythrocyte EPA+DHA composition (‘omega-3 index’) has been widely characterized as a risk biomarker in the context of coronary heart disease (Harris, 2008). Based in part on prospective longitudinal evidence, erythrocyte EPA+DHA composition of ≤4% of total fatty acid composition is thought to place one at ‘high risk’ for sudden cardiac death, whereas >8% is protective (Harris & Von Schacky, 2004). Because MDD is associated with excess premature mortality attributable in part to cardiovascular-related diseases (Angst et al., 2002; Osby et al., 2001), and the erythrocyte EPA+DHA deficits consistently observed in MDD patients (Lin et al., 2010) are similar to patients suffering acute coronary syndrome (Block et al., 2008)(Fig. 1), the same risk categories may be appropriate within the context of clinical practice. It is also notable that erythrocyte EPA+DHA composition of ≤4% is highly prevalent in youth with MDD, and may therefore aid in the identification of youth that may be at elevated risk for developing MDD. For example, our study found that 90 percent of adolescents with SSRI-resistant MDD exhibited erythrocyte EPA+DHA composition of ≤4% (McNamara et al., 2014a). Collectively, these data support the idea that erythrocyte or whole blood EPA+DHA composition of ≤4% can be considered to be a ‘state of deficiency’ that requires corrective intervention.
Treating LCn-3 fatty acid deficiency
The U.S. Food and Drug Administration (FDA) considers LCn-3 fatty acid doses up to 3 g/d to be ‘generally regarded as safe’, and the European Food Safety Authority (EFSA) considers doses up to 5 g/d to be safe. The American Psychiatric Association has adopted the consensus recommendations of the American Heart Association for an EPA+DHA dose of 1 g/d in patients with MDD (Freeman et al., 2006). The American Heart Association also recommends 3 g/d EPA+DHA for reducing elevated triglyceride levels. Prescription ethyl ester EPA+DHA (Lovaza® in the US, Omacor® in Europe, GlaxoSmithKline), purified ethyl ester EPA containing no DHA (Vascepa®, Amarin Corporation), and a free versus ethyl ester EPA+DHA formulation (Epanova®, AstraZeneca) have been approved by the U.S. FDA for the treatment of hypertriglyceridemia (≥500 mg/dL). More recently a generic version of Lovaza has become available (Teva Pharmaceuticals USA, Inc.). Over-the-counter fish oil supplements containing similar ethyl ester EPA+DHA concentrations are also widely available. It is important to note, however, that no LCn-3 fatty acid formulation is currently approved by the FDA for the treatment of any psychiatric disorder, and reimbursement for off-label use is ultimately at the discretion of the insurance provider.
Controlled intervention studies suggest that daily EPA+DHA doses of 1-2 g are sufficient to increase erythrocyte EPA+DHA composition to levels ≥4% (Flock et al., 2013). EPA+DHA doses in the range of 1-4 g/d in a 2:1 EPA to DHA ratio are efficacious for the treatment of depressive symptoms (Grosso et al., 2014; Sublette et al., 2011). Lower initial starting doses may be appropriate for children. For example, a daily dose of 600 mg fish oil monotherapy significantly reduced depression symptom severity in children with MDD (Nemets et al., 2006). As with other psychotropic medications, upward dose titration may be required as clinically indicated. For example, in an open-label flexible dosing study LCn-3 fatty acid monotherapy led to a statistically significant reduction in depression (and manic) symptom severity scores in pediatric bipolar patients (Wozniak et al., 2007). In this study the starting dose was 1.3 g/d of EPA+DHA, the maximum dose was 4.3 g/d, and the mean dose was 2.6 g/d. While there is a need for additional dose-titrating secondary prevention trials to elucidate optimal LCn-3 fatty acid dosing strategies, existing evidence suggests that a 1 g/d starting dose of EPA+DHA is safe and well-tolerated in pediatric, adolescent, and adult psychiatric patients.
Fish oil and LCn-3 fatty acids have an established long-term safety record in the general population. Potential adverse events associated with LCn-3 fatty acid supplementation include gastrointestinal disturbances, including nausea, diarrhea, gastroesophageal reflux, eructation, and less commonly emesis. In double-blind clinical trials of adult patients, the principal adverse events reported after chronic (8-12 weeks) treatment were gastrointestinal problems, and were considered mild and reported as frequently in patients receiving the placebo (Freeman et al., 2006). In studies conducted in pediatric and adolescent patients, no clinically-significant treatment-emergent adverse events were reported at doses up to 4.3 g/d (Clayton et al., 2009; Nemets et al., 2006; Wozniak et al., 2007). In adults, treatment with LCn-3 fatty acid doses up to 7.5 g/d for 6 months were found to be well-tolerated (Yee et al., 2010).To minimize the gastrointestinal adverse events associated with LCn-3 fatty acids, patients should be instructed to take their capsules with meals. Although taking fish oil at high doses (>3 g/d) has been associated in isolated cases with increased bleeding time in subjects also taking anticoagulant medications (Buckley et al., 2004), controlled clinical trials have found that chronic high dose EPA+DHA alone or in combination with aspirin does not increase risk for clinically-significant increases in bleeding time (Eritsland et al., 1995; Mueller et al., 1992; Harris, 2007). Another safety consideration involves the potential threat of contamination of fish and seafood with methyl mercury, PCBs, and other environmental pollutants. However, most over-the-counter fish oil supplements are highly purified and cannot exceed U.S. FDA limits for PCBs. As with all medications, patients should be informed of potential risks associated with LCn-3 fatty acids, and patients with an allergy to shellfish or seafood should be closely monitored.
Conclusions
Major advances in the treatment and prevention of MDD will be accelerated by the identification of modifiable pathogenic factors conferring vulnerability. Evidence emerging over the past three decades suggests that habitual dietary LCn-3 fatty acid insufficiency, particularly during perinatal development, may represent a modifiable risk factor for MDD. Cross-sectional fatty acid composition studies provide strong evidence that adolescent and adult patients with MDD exhibit significant blood LCn-3 fatty acid deficits compared with healthy controls. Dietary supplementation with fish oil is sufficient to correct blood LCn-3 fatty acid deficits in MDD patients, and controlled trials suggest that LCn-3 fatty acid supplementation may reduce depressive symptoms in MDD patients. While controversial, evidence also suggests that LCn-3 fatty acid deficiency may increase risk for suicide and cardiovascular disease, two principle causes of excess premature mortality in patients with MDD. Recent prospective evidence further suggests that higher peripheral LCn-3 fatty acid status is protective against the initial development of MDD in response to inflammation, and suggest that low LCn-3 fatty acid biostatus may increase risk for depressive symptoms by augmenting pro-inflammatory responsivity. Additionally, neuroimaging and rodent neurodevelopmental studies provide evidence that LCn-3 fatty acid insufficiency impacts brain development in a manner relevant to the pathophysiology and treatment of MDD. When taken collectively, these translational findings provide a strong empirical foundation in support of dietary LCn-3 fatty acid deficiency being a modifiable risk factor for MDD.
While this body of evidence provides a compelling rationale to screen and treat LCn-3 fatty acid deficiency in patients with MDD, it has been slow impact conventional psychiatric training and practice. This may be due in part to a general lack of nutritional training in psychiatry, though the field is slowing evolving and nutritional medicine is gaining credibility (Sarris et al., 2015). While several independent meta-analysis have found that LCn-3 fatty acids are efficacious for reducing depression symptom severity antidepressant, it is important to recognize that neurostructural and neurochemical perturbations resulting from LCn-3 fatty acid deficiency during development may not be reversible with short-term treatment. This is directly supported by rodent studies finding that enduring impairments in serotonin and dopamine neurotransmission resulting from perinatal n-3 fatty acid deficiency are reversible with early but not later n-3 fatty acid supplementation despite normalization of LCn-3 fatty acid biostatus. Therefore, early detection and treatment of LCn-3 fatty acid deficiency may be required to exert maximal protection against the initial development of MDD. Indeed, because LCn-3 fatty acid monotherapy is safe and well-tolerated it is ideally suited as a prodromal intervention for youth at increased risk for developing MDD. This approach is supported by the observation that fish oil supplementation prevented or delayed the onset of psychosis in ultra-high risk youth (Amminger et al., 2010, 2015). Additionally, within a ‘clinical staging’ framework LCn-3 fatty acid monotherapy would also represent a safe first-line intervention in youth with MDD, particularly those at risk for SSRI-associated adverse events.
While there is a need for additional research to optimize and standardize LCn-3 fatty acid screening and treatment approaches, current evidence and existing infrastructure support widespread implementation in psychiatric practice. As proof-of-concept, our group recently initiated a pilot program that routinely performs blood fatty acid testing in all patients admitted to an in-patient psychiatric clinic in suburban Cincinnati (Messamore & McNamara, 2016). To date we have performed whole blood fatty acid tests on over one hundred patients with different psychiatric disorders, including MDD. Consistent with prior cross-sectional studies, initial results suggest that the majority of patients exhibit whole blood EPA+DHA levels at ≤4. In several notable cases, treating EPA+DHA deficiency with either prescription or over-the-counter fish oil supplements resulted in remarkable and sustained improvements in mood symptoms. Although these data must be viewed as preliminary, they demonstrate the feasibility of implementing routine screening and treatment of EPA+DHA deficiency in psychiatric practice.
Highlights.
A body of translational evidence suggests that dietary long-chain omega-3 (LCn-3) fatty acid deficiency, particularly during development, is a modifiable risk factor for depression.
LCn-3 fatty acid deficiency is also a potential risk factor for suicide and cardiovascular disease, two primary causes of excess premature mortality in patients with MDD.
Low LCn-3 fatty acid biostatus may increase risk for depressive symptoms by augmenting pro-inflammatory signalling.
Future directions include implementing routine screening and treatment of LCn-3 fatty acid deficiency in clinical practice.
Acknowledgments
This work was supported in part by National Institutes of Health grants MH097818, MH107378, and DK097599 to R.K.M.
Disclosures: R.K.M. has received research support from NARSAD, Martek Biosciences Inc, Inflammation Research Foundation, Ortho-McNeil Janssen, AstraZeneca, Eli Lilly, and was a member of the Inflammation Research Foundation scientific advisory board.
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.
References
- Able JA, Liu Y, Jandacek R, Rider T, Tso P, McNamara RK. Omega-3 fatty acid deficient male rats exhibit abnormal behavioral activation in the forced swim test following chronic fluoxetine treatment: association with altered 5-HT1A and alpha2A adrenergic receptor expression. J Psychiatr Res. 2014;50:42–50. doi: 10.1016/j.jpsychires.2013.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adams PB, Lawson S, Sanigorski A, Sinclair AJ. Arachidonic acid to eicosapentaenoic acid ratio in blood correlates positively with clinical symptoms of depression. Lipids. 1996;31:S157–161. doi: 10.1007/BF02637069. [DOI] [PubMed] [Google Scholar]
- Ahmad SO, Park JH, Radel JD, Levant B. 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]
- Albert CM, Campos H, Stampfer MJ, Ridker PM, Manson JE, Willett WC, Ma J. Blood levels of long-chain n-3 fatty acids and the risk of sudden death. N Engl J Med. 2002;346:1113–1118. doi: 10.1056/NEJMoa012918. [DOI] [PubMed] [Google Scholar]
- Allen KL, Mori TA, Beilin L, Byrne SM, Hickling S, Oddy WH. Dietary intake in population-based adolescents: support for a relationship between eating disorder symptoms, low fatty acid intake and depressive symptoms. J Hum Nutr Diet. 2013;26:459–469. doi: 10.1111/jhn.12024. [DOI] [PubMed] [Google Scholar]
- Almeida DM, Jandacek RJ, Weber WA, McNamara RK. Docosahexaenoic acid biostatus is associated with event-related functional connectivity in cortical attention networks of typically developing children. Nutr Neurosci. 2016 doi: 10.1179/1476830515Y.0000000046. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- Amminger GP, Schäfer MR, Papageorgiou K, Klier CM, Cotton SM, Harrigan SM, Mackinnon A, McGorry PD, Berger GE. 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]
- Amminger GP, Schäfer MR, Schlögelhofer M, Klier CM, McGorry PD. Longer-term outcome in the prevention of psychotic disorders by the Vienna omega-3 study. Nat Commun. 2015;6:7934. doi: 10.1038/ncomms8934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Angst F, Stassen HH, Clayton PJ, Angst J. Mortality of patients with mood disorders: follow-up over 34-38 years. J Affect Disord. 2002;68:167–181. doi: 10.1016/s0165-0327(01)00377-9. [DOI] [PubMed] [Google Scholar]
- Appleton KM, Sallis HM, Perry R, Ness AR, Churchill R. Omega-3 fatty acids for depression in adults. Cochrane Database Syst Rev. 2015;11:CD004692. doi: 10.1002/14651858.CD004692.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arango V, Underwood MD, Mann JJ. Serotonin brain circuits involved in major depression and suicide. Prog Brain Res. 2002;136:443–453. doi: 10.1016/s0079-6123(02)36037-0. [DOI] [PubMed] [Google Scholar]
- Arterburn LM, Hall EB, Oken H. Distribution, interconversion, and dose response of n-3 fatty acids in humans. Am J Clin Nutr. 2006;83:1467S–1476S. doi: 10.1093/ajcn/83.6.1467S. [DOI] [PubMed] [Google Scholar]
- Aslibekyan S, Wiener HW, Havel PJ, Stanhope KL, O'Brien DM, Hopkins SE, Absher DM, Tiwari HK, Boyer BB. DNA methylation patterns are associated with n-3 fatty acid intake in Yup'ik people. J Nutr. 2014;144:425–430. doi: 10.3945/jn.113.187203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Assies J, Pouwer F, Lok A, Mocking RJ, Bockting CL, Visser I, Abeling NG, Duran M, Schene AH. Plasma and erythrocyte fatty acid patterns in patients with recurrent depression: a matched case-control study. PLoS One. 2010;5:e10635. doi: 10.1371/journal.pone.0010635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Axelson D, Goldstein B, Goldstein T, Monk K, Yu H, Hickey MB, Sakolsky D, Diler R, Hafeman D, Merranko J, Iyengar S, Brent D, Kupfer D, Birmaher B. Diagnostic precursors to bipolar disorder in offspring of parents with bipolar disorder: A longitudinal study. Am J Psychiatry. 2015;172:638–646. doi: 10.1176/appi.ajp.2014.14010035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baek D, Park Y. Association between erythrocyte n-3 polyunsaturated fatty acids and biomarkers of inflammation and oxidative stress in patients with and without depression. Prostaglandins Leukot Essent Fatty Acids. 2013;89:291–296. doi: 10.1016/j.plefa.2013.09.008. [DOI] [PubMed] [Google Scholar]
- Baghai TC, Varallo-Bedarida G, Born C, Häfner S, Schüle C, Eser D, Rupprecht R, Bondy B, von Schacky C. Major depressive disorder is associated with cardiovascular risk factors and low Omega-3 Index. J Clin Psychiatry. 2011;72:1242–1247. doi: 10.4088/JCP.09m05895blu. [DOI] [PubMed] [Google Scholar]
- Bakewell L, Burdge GC, Calder PC. Polyunsaturated fatty acid concentrations in young men and women consuming their habitual diets. Br J Nutr. 2006;96:93–99. doi: 10.1079/bjn20061801. [DOI] [PubMed] [Google Scholar]
- Barceló-Coblijn G, Murphy EJ, Othman R, Moghadasian MH, Kashour T, Friel JK. Flaxseed oil and fish-oil capsule consumption alters human red blood cell n-3 fatty acid composition: a multiple-dosing trial comparing 2 sources of n-3 fatty acid. Am J Clin Nutr. 2008;88:801–809. doi: 10.1093/ajcn/88.3.801. [DOI] [PubMed] [Google Scholar]
- Beltz BS, Tlusty MF, Benton JL, Sandeman DC. Omega-3 fatty acids upregulate adult neurogenesis. Neurosci Lett. 2007;415:154–158. doi: 10.1016/j.neulet.2007.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blasbalg TL, Hibbeln JR, Ramsden CE, Majchrzak SF, Rawlings RR. Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century. Am J Clin Nutr. 2011;93:950–962. doi: 10.3945/ajcn.110.006643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Block RC, Harris WS, Reid KJ, Sands SA, Spertus JA. EPA and DHA in blood cell membranes from acute coronary syndrome patients and controls. Atherosclerosis. 2008;197:821–828. doi: 10.1016/j.atherosclerosis.2007.07.042. [DOI] [PubMed] [Google Scholar]
- Bondi CO, Taha AY, Tock JL, Totah NK, Cheon Y, Torres GE, Rapoport SI, Moghaddam B. Adolescent behavior and dopamine availability are uniquely sensitive to dietary omega-3 fatty acid deficiency. Biol Psychiatry. 2014;75:38–46. doi: 10.1016/j.biopsych.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brenna JT, Varamini B, Jensen RG, Diersen-Schade DA, Boettcher JA, Arterburn LM. Docosahexaenoic and arachidonic acid concentrations in human breast milk worldwide. Am J Clin Nutr. 2007;85:1457–1464. doi: 10.1093/ajcn/85.6.1457. [DOI] [PubMed] [Google Scholar]
- Brenna JT, Salem N, Jr, Sinclair AJ, Cunnane SC International Society for the Study of Fatty Acids and Lipids, ISSFAL. alpha-Linolenic acid supplementation and conversion to n-3 long-chain polyunsaturated fatty acids in humans. Prostaglandins Leukot Essent Fatty Acids. 2009;80:85–91. doi: 10.1016/j.plefa.2009.01.004. [DOI] [PubMed] [Google Scholar]
- Brenner RR, Rimoldi OJ, Lombardo YB, González MS, Bernasconi AM, Chicco A, Basabe JC. Desaturase activities in rat model of insulin resistance induced by a sucrose-rich diet. Lipids. 2003;38:733–742. doi: 10.1007/s11745-003-1121-x. [DOI] [PubMed] [Google Scholar]
- Bridge JA, Goldstein TR, Brent DA. Adolescent suicide and suicidal behavior. J Child Psychol Psychiatry. 2006;47:372–394. doi: 10.1111/j.1469-7610.2006.01615.x. [DOI] [PubMed] [Google Scholar]
- Buckley MS, Goff AD, Knapp WE. Fish oil interaction with warfarin. Ann Pharmacother. 2004;38:50–52. doi: 10.1345/aph.1D007. [DOI] [PubMed] [Google Scholar]
- Burdge GC, Wootton SA. Conversion of alpha-linolenic acid to eicosapentaenoic, docosapentaenoic and docosahexaenoic acids in young women. Br J Nutr. 2002;88:411–420. doi: 10.1079/BJN2002689. [DOI] [PubMed] [Google Scholar]
- Calderon F, Kim HY. Docosahexaenoic acid promotes neurite growth in hippocampal neurons. J Neurochem. 2004;90:979–988. doi: 10.1111/j.1471-4159.2004.02520.x. [DOI] [PubMed] [Google Scholar]
- Cao D, Kevala K, Kim J, Moon HS, Jun SB, Lovinger D, Kim HY. Docosahexaenoic acid promotes hippocampal neuronal development and synaptic function. J Neurochem. 2009;111:510–521. doi: 10.1111/j.1471-4159.2009.06335.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlezon WA, Jr, Mague SD, Parow AM, Stoll AL, Cohen BM, Renshaw PF. Antidepressant-like effects of uridine and omega-3 fatty acids are potentiated by combined treatment in rats. Biol Psychiatry. 2005;57:343–350. doi: 10.1016/j.biopsych.2004.11.038. [DOI] [PubMed] [Google Scholar]
- Carver JD, Benford VJ, Han B, Cantor AB. The relationship between age and the fatty acid composition of cerebral cortex and erythrocytes in human subjects. Brain Res Bull. 2001;56:79–85. doi: 10.1016/s0361-9230(01)00551-2. [DOI] [PubMed] [Google Scholar]
- Caspi A, Moffitt TE. Gene-environment interactions in psychiatry: joining forces with neuroscience. Nat Rev Neurosci. 2006;7:583–590. doi: 10.1038/nrn1925. [DOI] [PubMed] [Google Scholar]
- Chalon S. Omega-3 fatty acids and monoamine neurotransmission. Prostaglandins Leukot Essent Fatty Acids. 2006;75:259–269. doi: 10.1016/j.plefa.2006.07.005. [DOI] [PubMed] [Google Scholar]
- Chen CT, Domenichiello AF, Trépanier MO, Liu Z, Masoodi M, Bazinet RP. The low levels of eicosapentaenoic acid in rat brain phospholipids are maintained via multiple redundant mechanisms. J Lipid Res. 2013;54:2410–2422. doi: 10.1194/jlr.M038505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chhetry BT, Hezghia A, Miller JM, Lee S, Coopera TB, Oquendoa MA, Mann JJ, Sublette ME. Omega-3 polyunsaturated fatty acid supplementation and white matter changes in major depression. J Psychiatr Res. 2016 doi: 10.1016/j.jpsychires.2015.12.007. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Childs CE, Romeu-Nadal M, Burdge GC, Calder PC. Gender differences in the n-3 fatty acid content of tissues. Proc Nutr Soc. 2008;67:19–27. doi: 10.1017/S0029665108005983. [DOI] [PubMed] [Google Scholar]
- Clayton EH, Hanstock TL, Hirneth SJ, Kable CJ, Garg ML, Hazell PL. Long-chain omega-3 polyunsaturated fatty acids in the blood of children and adolescents with juvenile bipolar disorder. Lipids. 2008;43:1031–1038. doi: 10.1007/s11745-008-3224-z. [DOI] [PubMed] [Google Scholar]
- Clayton EH, Hanstock TL, Hirneth SJ, Kable CJ, Garg ML, Hazell PL. Reduced mania and depression in juvenile bipolar disorder associated with long-chain omega-3 polyunsaturated fatty acid supplementation. Eur J Clin Nutr. 2009;63:1037–1040. doi: 10.1038/ejcn.2008.81. [DOI] [PubMed] [Google Scholar]
- Coccaro EF, Kavoussi RJ, Trestman RL, Gabriel SM, Cooper TB, Siever LJ. Serotonin function in human subjects: intercorrelations among central 5-HT indices and aggressiveness. Psychiatry Res. 1997;73:1–14. doi: 10.1016/s0165-1781(97)00108-x. [DOI] [PubMed] [Google Scholar]
- Conklin SM, Gianaros PJ, Brown SM, Yao JK, Hariri AR, Manuck SB, Muldoon MF. Long-chain omega-3 fatty acid intake is associated positively with corticolimbic gray matter volume in healthy adults. Neurosci Lett. 2007;421:209–212. doi: 10.1016/j.neulet.2007.04.086. [DOI] [PubMed] [Google Scholar]
- Conklin SM, Runyan CA, Leonard S, Reddy RD, Muldoon MF, Yao JK. Age-related changes of n-3 and n-6 polyunsaturated fatty acids in the anterior cingulate cortex of individuals with major depressive disorder. Prostaglandins Leukot Essent Fatty Acids. 2010;82:111–119. doi: 10.1016/j.plefa.2009.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Connolly CG, Wu J, Ho TC, Hoeft F, Wolkowitz O, Eisendrath S, Frank G, Hendren R, Max JE, Paulus MP, Tapert SF, Banerjee D, Simmons AN, Yang TT. Resting-state functional connectivity of subgenual anterior cingulate cortex in depressed adolescents. Biol Psychiatry. 2013;74:898–907. doi: 10.1016/j.biopsych.2013.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Connor WE, Neuringer M, Lin DS. Dietary effects on brain fatty acid composition: the reversibility of n-3 fatty acid deficiency and turnover of docosahexaenoic acid in the brain, erythrocytes, and plasma of rhesus monkeys. J Lipid Res. 1990;31:237–247. [PubMed] [Google Scholar]
- Coti Bertrand P, O'Kusky JR, Innis SM. Maternal dietary (n-3) fatty acid deficiency alters neurogenesis in the embryonic rat brain. J Nutr. 2006;136:1570–1575. doi: 10.1093/jn/136.6.1570. [DOI] [PubMed] [Google Scholar]
- Dalli J, Chiang N, Serhan CN. Elucidation of novel 13-series resolvins that increase with atorvastatin and clear infections. Nat Med. 2015;21:1071–1075. doi: 10.1038/nm.3911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davison KM, Kaplan BJ. Food intake and blood cholesterol levels of community-based adults with mood disorders. BMC Psychiatry. 2012;12:10. doi: 10.1186/1471-244X-12-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Velasco PC, Mendonça HR, Borba JM, Andrade da Costa BL, Guedes RC, Navarro DM, Santos GK, Faria-Melibeu Ada C, Campello Costa P, Serfaty CA. Nutritional restriction of omega-3 fatty acids alters topographical fine tuning and leads to a delay in the critical period in the rodent visual system. Exp Neurol. 2012;234:220–229. doi: 10.1016/j.expneurol.2011.12.032. [DOI] [PubMed] [Google Scholar]
- Delion S, Chalon S, Guilloteau D, Besnard JC, Durand G. alpha-Linolenic acid dietary deficiency alters age-related changes of dopaminergic and serotoninergic neurotransmission in the rat frontal cortex. J Neurochem. 1996;66:1582–1591. doi: 10.1046/j.1471-4159.1996.66041582.x. [DOI] [PubMed] [Google Scholar]
- DeMar JC, Jr, Ma K, Bell JM, Igarashi M, Greenstein D, Rapoport SI. One generation of n-3 polyunsaturated fatty acid deprivation increases depression and aggression test scores in rats. J Lipid Res. 2006;47:172–180. doi: 10.1194/jlr.M500362-JLR200. [DOI] [PubMed] [Google Scholar]
- Dickstein DP, Gorrostieta C, Ombao H, Goldberg LD, Brazel AC, Gable CJ, Kelly C, Gee DG, Zuo XN, Castellanos FX, Milham MP. Fronto-temporal spontaneous resting state functional connectivity in pediatric bipolar disorder. Biol Psychiatry. 2010;68:839–846. doi: 10.1016/j.biopsych.2010.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, Lanctôt KL. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–457. doi: 10.1016/j.biopsych.2009.09.033. [DOI] [PubMed] [Google Scholar]
- Edwards R, Peet M, Shay J, Horrobin D. Omega-3 polyunsaturated fatty acid levels in the diet and in red blood cell membranes of depressed patients. J Affect Disord. 1998;48:149–155. doi: 10.1016/s0165-0327(97)00166-3. [DOI] [PubMed] [Google Scholar]
- Egeland JA, Hostetter AM, Pauls DL, Sussex JN. Prodromal symptoms before onset of manic-depressive disorder suggested by first hospital admission histories. J Am Acad Child Adolesc Psychiatry. 2000;39:1245–1252. doi: 10.1097/00004583-200010000-00011. [DOI] [PubMed] [Google Scholar]
- Ehringer MA, Rhee SH, Young S, Corley R, Hewitt JK. Genetic and environmental contributions to common psychopathologies of childhood and adolescence: a study of twins and their siblings. J Abnorm Child Psychol. 2006;34:1–17. doi: 10.1007/s10802-005-9000-0. [DOI] [PubMed] [Google Scholar]
- Einvik G, Klemsdal TO, Sandvik L, Hjerkinn EM. A randomized clinical trial on n-3 polyunsaturated fatty acids supplementation and all-cause mortality in elderly men at high cardiovascular risk. Eur J Cardiovasc Prev Rehabil. 2010;17:588–592. doi: 10.1097/HJR.0b013e328339cc70. [DOI] [PubMed] [Google Scholar]
- Emslie GJ, Rush AJ, Weinberg WA, Kowatch RA, Hughes CW, Carmody T, Rintelmann J. A doubleblind, randomized placebo-controlled trial of fluoxetine in depressed children and adolescents with depression. Arch Gen Psychiatry. 1997;54:1031–1037. doi: 10.1001/archpsyc.1997.01830230069010. [DOI] [PubMed] [Google Scholar]
- Eritsland J, Arnesen H, Seljeflot I, Kierulf P. Long-term effects of n-3 polyunsaturated fatty acids on haemostatic variables and bleeding episodes in patients with coronary artery disease. Blood Coagul Fibrinolysis. 1995;6:17–22. doi: 10.1097/00001721-199502000-00003. [DOI] [PubMed] [Google Scholar]
- Evans SJ, Ringrose RN, Harrington GJ, Mancuso P, Burant CF, McInnis MG. Dietary intake and plasma metabolomic analysis of polyunsaturated fatty acids in bipolar subjects reveal dysregulation of linoleic acid metabolism. J Psychiatr Res. 2014;57:58–64. doi: 10.1016/j.jpsychires.2014.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farzaneh-Far R, Harris WS, Garg S, Na B, Whooley MA. Inverse association of erythrocyte n-3 fatty acid levels with inflammatory biomarkers in patients with stable coronary artery disease: The Heart and Soul Study. Atherosclerosis. 2009;205:538–543. doi: 10.1016/j.atherosclerosis.2008.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Felger JC, Li Z, Haroon E, Woolwine BJ, Jung MY, Hu X, Miller AH. Inflammation is associated with decreased functional connectivity within corticostriatal reward circuitry in depression. Mol Psychiatry. 2015 doi: 10.1038/mp.2015.168. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flock MR, Skulas-Ray AC, Harris WS, Etherton TD, Fleming JA, Kris-Etherton PM. Determinants of erythrocyte omega-3 fatty acid content in response to fish oil supplementation: a dose-response randomized controlled trial. J Am Heart Assoc. 2013;2:e000513. doi: 10.1161/JAHA.113.000513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Francois CA, Connor SL, Bolewicz LC, Connor WE. Supplementing lactating women with flaxseed oil does not increase docosahexaenoic acid in their milk. Am J Clin Nutr. 2003;77:226–233. doi: 10.1093/ajcn/77.1.226. [DOI] [PubMed] [Google Scholar]
- Freeman MP, Hibbeln JR, Wisner KL, Davis JM, Mischoulon D, Peet M, Keck PE, Jr, Marangell LB, Richardson AJ, Lake J, Stoll AL. Omega-3 fatty acids: evidence basis for treatment and future research in psychiatry. J Clin Psychiatry. 2006;67:1954–1967. doi: 10.4088/jcp.v67n1217. [DOI] [PubMed] [Google Scholar]
- Garland MR, Hallahan B, McNamara M, Carney PA, Grimes H, Hibbeln JR, Harkin A, Conroy RM. Lipids and essential fatty acids in patients presenting with self-harm. Br J Psychiatry. 2007;190:112–117. doi: 10.1192/bjp.bp.105.019562. [DOI] [PubMed] [Google Scholar]
- Gertsik L, Poland RE, Bresee C, Rapaport MH. Omega-3 fatty acid augmentation of citalopram treatment for patients with major depressive disorder. J Clin Psychopharmacol. 2012;32:61–64. doi: 10.1097/JCP.0b013e31823f3b5f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giedd JN, Blumenthal J, Jeffries NO, Castellanos FX, Liu H, Zijdenbos A, Paus T, Evans AC, Rapoport JL. Brain development during childhood and adolescence: a longitudinal MRI study. Nat Neurosci. 1999;2:861–863. doi: 10.1038/13158. [DOI] [PubMed] [Google Scholar]
- Giedd JN, Lalonde FM, Celano MJ, White SL, Wallace GL, Lee NR, Lenroot RK. Anatomical brain magnetic resonance imaging of typically developing children and adolescents. J Am Acad Child Adolesc Psychiatry. 2009;48:465–470. doi: 10.1097/CHI.0b013e31819f2715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giltay EJ, Gooren LJ, Toorians AW, Katan MB, Zock PL. Docosahexaenoic acid concentrations are higher in women than in men because of estrogenic effects. Am J Clin Nutr. 2004;80:1167–1174. doi: 10.1093/ajcn/80.5.1167. [DOI] [PubMed] [Google Scholar]
- Gracious BL, Chirieac MC, Costescu S, Finucane TL, Youngstrom EA, Hibbeln JR. Randomized, placebo-controlled trial of flax oil in pediatric bipolar disorder. Bipolar Disord. 2010;12:142–154. doi: 10.1111/j.1399-5618.2010.00799.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grayson DS, Kroenke CD, Neuringer M, Fair DA. Dietary omega-3 fatty acids modulate large-scale systems organization in the rhesus macaque brain. J Neurosci. 2014;34:2065–2074. doi: 10.1523/JNEUROSCI.3038-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green P, Glozman S, Weiner L, Yavin E. Enhanced free radical scavenging and decreased lipid peroxidation in the rat fetal brain after treatment with ethyl docosahexaenoate. Biochim Biophys Acta. 2001;1532:203–212. doi: 10.1016/s1388-1981(01)00132-9. [DOI] [PubMed] [Google Scholar]
- Green P, Gispan-Herman I, Yadid G. Increased arachidonic acid concentration in the brain of Flinders Sensitive Line rats, an animal model of depression. J Lipid Res. 2005;46:1093–1096. doi: 10.1194/jlr.C500003-JLR200. [DOI] [PubMed] [Google Scholar]
- Green P, Hermesh H, Monselise A, Marom S, Presburger G, Weizman A. 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]
- Groeger AL, Cipollina C, Cole MP, Woodcock SR, Bonacci G, Rudolph TK, Rudolph V, Freeman BA, Schopfer FJ. Cyclooxygenase-2 generates anti-inflammatory mediators from omega-3 fatty acids. Nat Chem Biol. 2010;6:433–441. doi: 10.1038/nchembio.367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grosso G, Pajak A, Marventano S, Castellano S, Galvano F, Bucolo C, Drago F, Caraci F. Role of omega-3 Fatty acids in the treatment of depressive disorders: a comprehensive meta-analysis of randomized clinical trials. PLoS One. 2014;9:e96905. doi: 10.1371/journal.pone.0096905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haghighi F, Galfalvy H, Chen S, Huang YY, Cooper TB, Burke AK, Oquendo MA, Mann JJ, Sublette ME. DNA methylation perturbations in genes involved in polyunsaturated Fatty Acid biosynthesis associated with depression and suicide risk. Front Neurol. 2015;6:92. doi: 10.3389/fneur.2015.00092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hallahan B, Hibbeln JR, Davis JM, Garland MR. Omega-3 fatty acid supplementation in patients with recurrent self-harm. Single-centre double-blind randomised controlled trial. Br J Psychiatry. 2007;190:118–122. doi: 10.1192/bjp.bp.106.022707. [DOI] [PubMed] [Google Scholar]
- Hamazaki K, Hamazaki T, Inadera H. Fatty acid composition in the postmortem amygdala of patients with schizophrenia, bipolar disorder, and major depressive disorder. J Psychiatr Res. 2012;46:1024–1028. doi: 10.1016/j.jpsychires.2012.04.012. [DOI] [PubMed] [Google Scholar]
- Hammad TA, Laughren T, Racoosin J. Suicidality in pediatric patients treated with antidepressant drugs. Arch Gen Psychiatry. 2006;63:332–339. doi: 10.1001/archpsyc.63.3.332. [DOI] [PubMed] [Google Scholar]
- Harris WS, Von Schacky C. The Omega-3 Index: a new risk factor for death from coronary heart disease? Prev Med. 2004;39:212–220. doi: 10.1016/j.ypmed.2004.02.030. [DOI] [PubMed] [Google Scholar]
- Harris WS. Expert opinion: omega-3 fatty acids and bleeding-cause for concern? Am J Cardiol. 2007;99(6A):44C–46C. doi: 10.1016/j.amjcard.2006.11.021. [DOI] [PubMed] [Google Scholar]
- Harris WS. The omega-3 index as a risk factor for coronary heart disease. Am J Clin Nutr. 2008;87:1997S–2002S. doi: 10.1093/ajcn/87.6.1997S. [DOI] [PubMed] [Google Scholar]
- Hibbeln JR. Fish consumption and major depression. Lancet. 1998;351:1213. doi: 10.1016/S0140-6736(05)79168-6. [DOI] [PubMed] [Google Scholar]
- Hibbeln JR. Seafood consumption, the DHA content of mothers' milk and prevalence rates of postpartum depression: a cross-national, ecological analysis. J Affect Disord. 2002;69:15–29. doi: 10.1016/s0165-0327(01)00374-3. [DOI] [PubMed] [Google Scholar]
- Ho TC, Yang G, Wu J, Cassey P, Brown SD, Hoang N, Chan M, Connolly CG, Henje-Blom E, Duncan LG, Chesney MA, Paulus MP, Max JE, Patel R, Simmons AN, Yang TT. Functional connectivity of negative emotional processing in adolescent depression. J Affect Disord. 2014;155:65–74. doi: 10.1016/j.jad.2013.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoile SP, Irvine NA, Kelsall CJ, Sibbons C, Feunteun A, Collister A, Torrens C, Calder PC, Hanson MA, Lillycrop KA, Burdge GC. Maternal fat intake in rats alters 20:4n-6 and 22:6n-3 status and the epigenetic regulation of Fads2 in offspring liver. J Nutr Biochem. 2013;24:1213–1220. doi: 10.1016/j.jnutbio.2012.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoile SP, Clarke-Harris R, Huang RC, Calder PC, Mori TA, Beilin LJ, Lillycrop KA, Burdge GC. Supplementation with N-3 long-chain polyunsaturated fatty acids or olive oil in men and women with renal disease induces differential changes in the DNA methylation of FADS2 and ELOVL5 in peripheral blood mononuclear cells. PLoS One. 2014;9:e109896. doi: 10.1371/journal.pone.0109896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howard TD, Mathias RA, Seeds MC, Herrington DM, Hixson JE, Shimmin LC, Hawkins GA, Sellers M, Ainsworth HC, Sergeant S, Miller LR, Chilton FH. DNA methylation in an enhancer region of the FADS cluster is associated with FADS activity in human liver. PLoS One. 2014;9:e97510. doi: 10.1371/journal.pone.0097510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howes OD, Lim S, Theologos G, Yung AR, Goodwin GM, McGuire P. A comprehensive review and model of putative prodromal features of bipolar affective disorder. Psychol Med. 2011;41:1567–1577. doi: 10.1017/S0033291710001790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu FB, Bronner L, Willett WC, Stampfer MJ, Rexrode KM, Albert CM, Hunter D, Manson JE. Fish and omega-3 fatty acid intake and risk of coronary heart disease in women. JAMA. 2002;287:1815–1821. doi: 10.1001/jama.287.14.1815. [DOI] [PubMed] [Google Scholar]
- Huan M, Hamazaki K, Sun Y, Itomura M, Liu H, Kang W, Watanabe S, Terasawa K, Hamazaki T. 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]
- Huang SY, Yang HT, Chiu CC, Pariante CM, Su KP. Omega-3 fatty acids on the forced-swimming test. J Psychiatr Res. 2008;42:58–63. doi: 10.1016/j.jpsychires.2006.09.004. [DOI] [PubMed] [Google Scholar]
- Ikemoto A, Nitta A, Furukawa S, Ohishi M, Nakamura A, Fujii Y, Okuyama H. Dietary n-3 fatty acid deficiency decreases nerve growth factor content in rat hippocampus. Neurosci Lett. 2000;285:99–102. doi: 10.1016/s0304-3940(00)01035-1. [DOI] [PubMed] [Google Scholar]
- Innis SM, de La Presa Owens S. Dietary fatty acid composition in pregnancy alters neurite membrane fatty acids and dopamine in newborn rat brain. J Nutr. 2001;131:118–122. doi: 10.1093/jn/131.1.118. [DOI] [PubMed] [Google Scholar]
- Itomura M, Fujioka S, Hamazaki K, Kobayashi K, Nagasawa T, Sawazaki S, Kirihara Y, Hamazaki T. Factors influencing EPA+DHA levels in red blood cells in Japan. In Vivo. 2008;22:131–135. [PubMed] [Google Scholar]
- Jacka FN, Mykletun A, Berk M, Bjelland I, Tell GS. The association between habitual diet quality and the common mental disorders in community-dwelling adults: the Hordaland Health study. Psychosom Med. 2011;73:483–490. doi: 10.1097/PSY.0b013e318222831a. [DOI] [PubMed] [Google Scholar]
- Jazayeri S, Tehrani-Doost M, Keshavarz SA, Hosseini M, Djazayery A, Amini H, Jalali M, Peet M. Comparison of therapeutic effects of omega-3 fatty acid eicosapentaenoic acid and fluoxetine, separately and in combination, in major depressive disorder. Aust N Z J Psychiatry. 2008;42:192–198. doi: 10.1080/00048670701827275. [DOI] [PubMed] [Google Scholar]
- Kawakita E, Hashimoto M, Shido O. Docosahexaenoic acid promotes neurogenesis in vitro and in vivo. Neuroscience. 2006;139:991–997. doi: 10.1016/j.neuroscience.2006.01.021. [DOI] [PubMed] [Google Scholar]
- Kelley DS, Siegel D, Fedor DM, Adkins Y, Mackey BE. DHA supplementation decreases serum C-reactive protein and other markers of inflammation in hypertriglyceridemic men. J Nutr. 2009;139:495–501. doi: 10.3945/jn.108.100354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kempton MJ, Salvador Z, Munafò MR, Geddes JR, Simmons A, Frangou S, Williams SC. Structural neuroimaging studies in major depressive disorder. Meta-analysis and comparison with bipolar disorder. Arch Gen Psychiatry. 2011;68:675–690. doi: 10.1001/archgenpsychiatry.2011.60. [DOI] [PubMed] [Google Scholar]
- Kennard B, Silva S, Vitiello B, Curry J, Kratochvil C, Simons A, Hughes J, Feeny N, Weller E, Sweeney M, Reinecke M, Pathak S, Ginsburg G, Emslie G, March J TADS Team. Remission and residual symptoms after short-term treatment in the Treatment of Adolescents with Depression Study (TADS) J Am Acad Child Adolesc Psychiatry. 2006;45:1404–1411. doi: 10.1097/01.chi.0000242228.75516.21. [DOI] [PubMed] [Google Scholar]
- Kessler RC. Epidemiology of women and depression. J Affect Disord. 2003;74:5–13. doi: 10.1016/s0165-0327(02)00426-3. [DOI] [PubMed] [Google Scholar]
- Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:593–602. doi: 10.1001/archpsyc.62.6.593. [DOI] [PubMed] [Google Scholar]
- Kessler RC, Merikangas KR, Wang PS. Prevalence, comorbidity, and service utilization for mood disorders in the United States at the beginning of the twenty-first century. Annu Rev Clin Psychol. 2007;3:137–158. doi: 10.1146/annurev.clinpsy.3.022806.091444. [DOI] [PubMed] [Google Scholar]
- Kodas E, Vancassel S, Lejeune B, Guilloteau D, Chalon S. Reversibility of n-3 fatty acid deficiency-induced changes in dopaminergic neurotransmission in rats: critical role of developmental stage. J Lipid Res. 2002;43:1209–1219. [PubMed] [Google Scholar]
- Kodas E, Galineau L, Bodard S, Vancassel S, Guilloteau D, Besnard JC, Chalon S. Serotoninergic neurotransmission is affected by n-3 polyunsaturated fatty acids in the rat. J Neurochem. 2004;89:695–702. doi: 10.1111/j.1471-4159.2004.02401.x. [DOI] [PubMed] [Google Scholar]
- Kwak SM, Myung SK, Lee YJ, Seo HG Korean Meta-analysis Study Group. Efficacy of omega-3 fatty acid supplements (eicosapentaenoic acid and docosahexaenoic acid) in the secondary prevention of cardiovascular disease: a meta-analysis of randomized, double-blind, placebo-controlled trials. Arch Intern Med. 2012;172:686–694. doi: 10.1001/archinternmed.2012.262. [DOI] [PubMed] [Google Scholar]
- Lai JS, Hiles S, Bisquera A, Hure AJ, McEvoy M, Attia J. A systematic review and meta-analysis of dietary patterns and depression in community-dwelling adults. Am J Clin Nutr. 2014;99:181–197. doi: 10.3945/ajcn.113.069880. [DOI] [PubMed] [Google Scholar]
- Laino CH, Fonseca C, Sterin-Speziale N, Slobodianik N, Reinés A. Potentiation of omega-3 fatty acid antidepressant-like effects with low non-antidepressant doses of fluoxetine and mirtazapine. Eur J Pharmacol. 2010;648:117–126. doi: 10.1016/j.ejphar.2010.08.047. [DOI] [PubMed] [Google Scholar]
- Lakhwani L, Tongia SK, Pal VS, Agrawal RP, Nyati P, Phadnis P. Omega-3 fatty acids have antidepressant activity in forced swimming test in Wistar rats. Acta Pol Pharm. 2007;64:271–276. [PubMed] [Google Scholar]
- Lalovic A, Levy E, Canetti L, Sequeira A, Montoudis A, Turecki G. Fatty acid composition in postmortem brains of people who completed suicide. J Psychiatry Neurosci. 2007;32:363–370. [PMC free article] [PubMed] [Google Scholar]
- Lalovic A, Klempan T, Sequeira A, Luheshi G, Turecki G. 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]
- Lattka E, Illig T, Koletzko B, Heinrich J. Genetic variants of the FADS1 FADS2 gene cluster as related to essential fatty acid metabolism. Curr Opin Lipidol. 2010;21:64–69. doi: 10.1097/MOL.0b013e3283327ca8. [DOI] [PubMed] [Google Scholar]
- Lee HS, Barraza-Villarreal A, Hernandez-Vargas H, Sly PD, Biessy C, Ramakrishnan U, Romieu I, Herceg Z. Modulation of DNA methylation states and infant immune system by dietary supplementation with ω-3 PUFA during pregnancy in an intervention study. Am J Clin Nutr. 2013;98:480–487. doi: 10.3945/ajcn.112.052241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levinson DF. The genetics of depression: a review. Biol Psychiatry. 2006;60:84–92. doi: 10.1016/j.biopsych.2005.08.024. [DOI] [PubMed] [Google Scholar]
- Lin PY, Huang SY, Su KP. A meta-analytic review of polyunsaturated fatty acid compositions in patients with depression. Biol Psychiatry. 2010;68:140–147. doi: 10.1016/j.biopsych.2010.03.018. [DOI] [PubMed] [Google Scholar]
- Liu JJ, Galfalvy HC, Cooper TB, Oquendo MA, Grunebaum MF, Mann JJ, Sublette ME. 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]
- Lotrich FE, Sears B, McNamara RK. Elevated ratio of arachidonic acid to long-chain omega-3 fatty acids predicts depression development following interferon-alpha treatment: Relationship with interleukin-6. Brain Behav Immun. 2013a;31:48–53. doi: 10.1016/j.bbi.2012.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madore C, Nadjar A, Delpech JC, Sere A, Aubert A, Portal C, Joffre C, Layé S. Nutritional n-3 PUFAs deficiency during perinatal periods alters brain innate immune system and neuronal plasticity-associated genes. Brain Behav Immun. 2014;41:22–31. doi: 10.1016/j.bbi.2014.03.021. [DOI] [PubMed] [Google Scholar]
- Marquardt A, Stöhr H, White K, Weber BH. cDNA cloning, genomic structure, and chromosomal localization of three members of the human fatty acid desaturase family. Genomics. 2000;66:175–183. doi: 10.1006/geno.2000.6196. [DOI] [PubMed] [Google Scholar]
- Marsland AL, Gianaros PJ, Abramowitch SM, Manuck SB, Hariri AR. Interleukin-6 covaries inversely with hippocampal grey matter volume in middle-aged adults. Biol Psychiatry. 2008;64:484–490. doi: 10.1016/j.biopsych.2008.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin A, Young C, Leckman JF, Mukonoweshuro C, Rosenheck R, Leslie D. Age effects on antidepressant-induced manic conversion. Arch Pediatr Adolesc Med. 2004;158:773–780. doi: 10.1001/archpedi.158.8.773. [DOI] [PubMed] [Google Scholar]
- Matsuzaka T, Shimano H, Yahagi N, Amemiya-Kudo M, Yoshikawa T, Hasty AH, Tamura Y, Osuga J, Okazaki H, Iizuka Y, Takahashi A, Sone H, Gotoda T, Ishibashi S, Yamada N. Dual regulation of mouse Delta(5)- and Delta(6)-desaturase gene expression by SREBP-1 and PPARalpha. J Lipid Res. 2002;43:107–114. [PubMed] [Google Scholar]
- McGrath-Hanna NK, Greene DM, Tavernier RJ, Bult-Ito A. Diet and mental health in the Arctic: is diet an important risk factor for mental health in circumpolar peoples? A review. Int J Circumpolar Health. 2003;62:228–241. doi: 10.3402/ijch.v62i3.17560. [DOI] [PubMed] [Google Scholar]
- McKenney JM, Sica D. Role of prescription omega-3 fatty acids in the treatment of hypertriglyceridemia. Pharmacotherapy. 2007;27:715–728. doi: 10.1592/phco.27.5.715. [DOI] [PubMed] [Google Scholar]
- McNamara RK, Hahn CG, Jandacek R, Rider T, Tso P, Stanford K, Richtand NM. Selective deficits in the omega-3 fatty acid docosahexaenoic acid in the postmortem orbitofrontal cortex of patients with major depressive disorder. Biol Psychiatry. 2007;62:17–24. doi: 10.1016/j.biopsych.2006.08.026. [DOI] [PubMed] [Google Scholar]
- McNamara RK, Liu Y, Jandacek R, Rider T, Tso P. The aging human orbitofrontal cortex: Decreasing polyunsaturated fatty acid composition and associated increases in lipogenic gene expression and stearoyl-CoA desaturase activity. Prostaglandins Leukot Essent Fatty Acids. 2008;78:293–304. doi: 10.1016/j.plefa.2008.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Able JA, Liu Y, Jandacek R, Rider T, Tso P. Gender differences in rat erythrocyte and brain docosahexaenoic acid composition: Role of ovarian hormones and dietary omega-3 fatty acid composition. Psychoneuroendocrinology. 2009a;34:532–539. doi: 10.1016/j.psyneuen.2008.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Jandacek RJ, Rider T, Tso P, Dwivedi Y, Roberts RC, Conley RR, Pandey GN. Fatty acid composition of the postmortem prefrontal cortex of male and female adolescent suicide victims. Prostaglandins Leukot Essent Fatty Acids. 2009b;80:19–26. doi: 10.1016/j.plefa.2008.10.002. [DOI] [PubMed] [Google Scholar]
- McNamara RK, Able JA, Liu Y, Jandacek R, Rider T, Tso P, Lipton JW. Omega-3 fatty acid deficiency during perinatal development increases serotonin turnover in the prefrontal cortex and decreases midbrain tryptophan hydroxylase-2 expression in adult female rats: Dissociation from estrogenic effects. J Psychiatr Res. 2009c;43:656–663. doi: 10.1016/j.jpsychires.2008.09.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Jandacek R, Rider T, Tso P, Cole-Strauss A, Lipton JW. Omega-3 fatty acid deficiency increases constitutive pro-inflammatory cytokine production in rats: relationship with central serotonin turnover. Prostaglandins Leukot Essent Fatty Acids. 2010;83:185–191. doi: 10.1016/j.plefa.2010.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Jandacek RJ. Investigation of postmortem brain polyunsaturated fatty acid composition in psychiatric disorders: Limitations, challenges, and future directions. J Psychiatr Res. 2011;45:44–46. doi: 10.1016/j.jpsychires.2010.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Liu Y. Reduced fatty acid biosynthesis gene expression in the prefrontal cortex of patients with major depressive disorder. J Affect Disord. 2011;129:359–363. doi: 10.1016/j.jad.2010.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Jandacek R, Tso P, Dwivedi Y, Ren X, Pandey GN. Lower docosahexaenoic acid concentrations in the postmortem prefrontal cortex of adult depressed suicide victims compared with controls without cardiovascular disease. J Psychiatr Res. 2013a;47:1187–1191. doi: 10.1016/j.jpsychires.2013.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Able JA, Liu Y, Jandacek RJ, Rider T, Tso P, Lipton JW. Omega-3 fatty acid deficiency does not alter the effects of chronic fluoxetine treatment on central serotonin turnover or behavior in the forced swim test in female rats. Pharmacol Biochem Behav. 2013b;114:1–8. doi: 10.1016/j.pbb.2013.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Strimpfel J, Jandacek R, Rider T, Tso P, Welge JA, Strawn JR, DelBello MP. Detection and treatment of long-chain omega-3 fatty acid deficiency in adolescents with SSRI-resistant major depressive disorder. PharmaNutrition. 2014a;2:38–46. doi: 10.1016/j.phanu.2014.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Rider T, Jandacek R, Tso P. Abnormal fatty acid pattern in the superior temporal gyrus distinguishes bipolar disorder from major depression and schizophrenia and resembles multiple sclerosis. Psychiatry Res. 2014b;215:560–567. doi: 10.1016/j.psychres.2013.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McNamara RK, Welge JA. Meta-analysis of erythrocyte polyunsaturated fatty acid biostatus in bipolar disorder. Bipolar Disord. 2016 doi: 10.1111/bdi.12386. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merikangas KR, Chakravarti A, Moldin SO, Araj H, Blangero JC, Burmeister M, Crabbe J, Jr, Depaulo JR, Jr, Foulks E, Freimer NB, Koretz DS, Lichtenstein W, Mignot E, Reiss AL, Risch NJ, Takahashi JS. Future of genetics of mood disorders research. Biol Psychiatry. 2002;52:457–477. doi: 10.1016/s0006-3223(02)01471-3. [DOI] [PubMed] [Google Scholar]
- Messamore E, McNamara RK. Detection and treatment of omega-3 fatty acid deficiency in psychiatric practice: Rationale and implementation. Lipids Health Dis. 2016;15:1–13. doi: 10.1186/s12944-016-0196-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mocking RJ, Harmsen I, Assies J, Koeter MW, Ruhé HG, Schene AH. Meta-analysis and meta-regression of omega-3 polyunsaturated fatty acid supplementation for major depressive disorder. Transl Psychiatry. 2016;6:e756. doi: 10.1038/tp.2016.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mueller BA, Talbert RL, Tegeler CH, Prihoda TJ. The bleeding time effects of a single dose of aspirin in subjects receiving omega-3 fatty acid dietary supplementation. J Clin Pharmacol. 1991;31:185–190. doi: 10.1002/j.1552-4604.1991.tb03706.x. [DOI] [PubMed] [Google Scholar]
- Murakami K, Miyake Y, Sasaki S, Tanaka K, Arakawa M. Fish and n-3 polyunsaturated fatty acid intake and depressive symptoms: Ryukyus Child Health Study. Pediatrics. 2010;126:623–630. doi: 10.1542/peds.2009-3277. [DOI] [PubMed] [Google Scholar]
- Nemets H, Nemets B, Apter A, Bracha Z, Belmaker RH. Omega-3 treatment of childhood depression: a controlled, double-blind pilot study. Am J Psychiatry. 2006;163:1098–1100. doi: 10.1176/ajp.2006.163.6.1098. [DOI] [PubMed] [Google Scholar]
- Nestler EJ, Carlezon WA., Jr The mesolimbic dopamine reward circuit in depression. Biol Psychiatry. 2006;59:1151–1159. doi: 10.1016/j.biopsych.2005.09.018. [DOI] [PubMed] [Google Scholar]
- Niculescu MD, Lupu DS, Craciunescu CN. Perinatal manipulation of α-linolenic acid intake induces epigenetic changes in maternal and offspring livers. FASEB J. 2013;27:350–358. doi: 10.1096/fj.12-210724. [DOI] [PubMed] [Google Scholar]
- 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]
- Oddy WH, Hickling S, Smith MA, O'Sullivan TA, Robinson M, de Klerk NH, Beilin LJ, Mori TA, Syrette J, Zubrick SR, Silburn SR. Dietary intake of omega-3 fatty acids and risk of depressive symptoms in adolescents. Depress Anxiety. 2011;28:582–588. doi: 10.1002/da.20822. [DOI] [PubMed] [Google Scholar]
- Orr SK, Palumbo S, Bosetti F, Mount HT, Kang JX, Greenwood CE, Ma DW, Serhan CN, Bazinet RP. Unesterified docosahexaenoic acid is protective in neuroinflammation. J Neurochem. 2013;127:378–393. doi: 10.1111/jnc.12392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Osby U, Brandt L, Correia N, Ekbom A, Sparén P. Excess mortality in bipolar and unipolar disorder in Sweden. Arch Gen Psychiatry. 2001;58:844–850. doi: 10.1001/archpsyc.58.9.844. [DOI] [PubMed] [Google Scholar]
- O'Sullivan TA, Ambrosini GL, Mori TA, Beilin LJ, Oddy WH. Omega-3 Index correlates with healthier food consumption in adolescents and with reduced cardiovascular disease risk factors in adolescent boys. Lipids. 2011;46:59–67. doi: 10.1007/s11745-010-3499-8. [DOI] [PubMed] [Google Scholar]
- Paus T, Zijdenbos A, Worsley K, Collins DL, Blumenthal J, Giedd JN, Rapoport JL, Evans AC. Structural maturation of neural pathways in children and adolescents: in vivo study. Science. 1999;283:1908–1911. doi: 10.1126/science.283.5409.1908. [DOI] [PubMed] [Google Scholar]
- Peet M, Horrobin DF. A dose-ranging study of the effects of ethyl-eicosapentaenoate in patients with ongoing depression despite apparently adequate treatment with standard drugs. Arch Gen Psychiatry. 2002;59:913–919. doi: 10.1001/archpsyc.59.10.913. [DOI] [PubMed] [Google Scholar]
- Peet M. International variations in the outcome of schizophrenia and the prevalence of depression in relation to national dietary practices: an ecological analysis. Br J Psychiatry. 2004;184:404–408. doi: 10.1192/bjp.184.5.404. [DOI] [PubMed] [Google Scholar]
- Perroud N, Dayer A, Piguet C, Nallet A, Favre S, Malafosse A, Aubry JM. Childhood maltreatment and methylation of the glucocorticoid receptor gene NR3C1 in bipolar disorder. Br J Psychiatry. 2014;204:30–35. doi: 10.1192/bjp.bp.112.120055. [DOI] [PubMed] [Google Scholar]
- Pottala JV, Talley JA, Churchill SW, Lynch DA, von Schacky C, Harris WS. Red blood cell fatty acids are associated with depression in a case-control study of adolescents. Prostaglandins Leukot Essent Fatty Acids. 2012;86:161–165. doi: 10.1016/j.plefa.2012.03.002. [DOI] [PubMed] [Google Scholar]
- Pottala JV, Yaffe K, Robinson JG, Espeland MA, Wallace R, Harris WS. Higher RBC EPA + DHA corresponds with larger total brain and hippocampal volumes: WHIMS-MRI study. Neurology. 2014;82:435–442. doi: 10.1212/WNL.0000000000000080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raeder MB, Fernø J, Glambek M, Stansberg C, Steen VM. Antidepressant drugs activate SREBP and up-regulate cholesterol and fatty acid biosynthesis in human glial cells. Neurosci Lett. 2006;395:185–190. doi: 10.1016/j.neulet.2005.10.096. [DOI] [PubMed] [Google Scholar]
- Raeder MB, Steen VM, Vollset SE, Bjelland I. Associations between cod liver oil use and symptoms of depression: the Hordaland Health Study. J Affect Disord. 2007;101:245–249. doi: 10.1016/j.jad.2006.11.006. [DOI] [PubMed] [Google Scholar]
- Rao JS, Ertley RN, Lee HJ, DeMar JC, Jr, Arnold JT, Rapoport SI, Bazinet RP. n-3 polyunsaturated fatty acid deprivation in rats decreases frontal cortex BDNF via a p38 MAPK-dependent mechanism. Mol Psychiatry. 2007;12:36–46. doi: 10.1038/sj.mp.4001888. [DOI] [PubMed] [Google Scholar]
- Reardon HT, Brenna JT. Microsomal biosynthesis of omega-3 fatty acids. In: McNamara RK, editor. The Omega-3 Fatty Acid Deficiency Syndrome: Opportunities for Disease Prevention. Nova Science Publishers, Inc; U.S.A: 2013. pp. 3–17. [Google Scholar]
- Sands SA, Reid KJ, Windsor SL, Harris WS. The impact of age, body mass index, and fish intake on the EPA and DHA content of human erythrocytes. Lipids. 2005;40:343–347. doi: 10.1007/s11745-006-1392-2. [DOI] [PubMed] [Google Scholar]
- Sarris J, Mischoulon D, Schweitzer I. Omega-3 for bipolar disorder: meta-analyses of use in mania and bipolar depression. J Clin Psychiatry. 2012;73:81–86. doi: 10.4088/JCP.10r06710. [DOI] [PubMed] [Google Scholar]
- Sarris J, Logan AC, Akbaraly TN, Amminger GP, Balanzá-Martínez V, Freeman MP, Hibbeln J, Matsuoka Y, Mischoulon D, Mizoue T, Nanri A, Nishi D, Ramsey D, Rucklidge JJ, Sanchez-Villegas A, Scholey A, Su KP, Jacka FN International Society for Nutritional Psychiatry Research. Nutritional medicine as mainstream in psychiatry. Lancet Psychiatry. 2015;2:271–274. doi: 10.1016/S2215-0366(14)00051-0. [DOI] [PubMed] [Google Scholar]
- Serhan CN. Novel lipid mediators and resolution mechanisms in acute inflammation: to resolve or not? Am J Pathol. 2010;177:1576–1591. doi: 10.2353/ajpath.2010.100322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Setiawan E, Wilson AA, Mizrahi R, Rusjan PM, Miler L, Rajkowska G, Suridjan I, Kennedy JL, Rekkas PV, Houle S, Meyer JH. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry. 2015;72:268–275. doi: 10.1001/jamapsychiatry.2014.2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siscovick DS, Raghunathan TE, King I, Weinmann S, Wicklund KG, Albright J, Bovbjerg V, Arbogast P, Smith H, Kushi LH, et al. Dietary intake and cell membrane levels of long-chain n-3 polyunsaturated fatty acids and the risk of primary cardiac arrest. JAMA. 1995;274:1363–1367. doi: 10.1001/jama.1995.03530170043030. [DOI] [PubMed] [Google Scholar]
- Stein DJ. Depression, anhedonia, and psychomotor symptoms: the role of dopaminergic neurocircuitry. CNS Spectr. 2008;13:561–565. doi: 10.1017/s1092852900016837. [DOI] [PubMed] [Google Scholar]
- Strawn JR, Adler CM, McNamara RK, Welge JA, Bitter SM, Mills NP, Barzman DH, Cerullo MA, Chang KD, Strakowski SM, DelBello MP. Antidepressant tolerability in anxious and depressed youth at high risk for bipolar disorder: A prospective naturalistic treatment study. Bipolar Disord. 2014;16:523–530. doi: 10.1111/bdi.12113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su KP, Huang SY, Peng CY, Lai HC, Huang CL, Chen YC, Aitchison KJ, Pariante CM. Phospholipase A2 and cyclooxygenase 2 genes influence the risk of interferon-alpha-induced depression by regulating polyunsaturated fatty acids levels. Biol Psychiatry. 2010;67:550–557. doi: 10.1016/j.biopsych.2009.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Su KP, Lai HC, Yang HT, Su WP, Peng CY, Chang JP, Chang HC, Pariante CM. Omega-3 fatty acids in the prevention of interferon-alpha-induced depression: Results from a randomized, controlled trial. Biol Psychiatry. 2014;76:559–566. doi: 10.1016/j.biopsych.2014.01.008. [DOI] [PubMed] [Google Scholar]
- Sublette ME, Hibbeln JR, Galfalvy H, Oquendo MA, Mann JJ. 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]
- Sublette ME, Ellis SP, Geant AL, Mann JJ. Meta-analysis of the effects of eicosapentaenoic acid (EPA) in clinical trials in depression. J Clin Psychiatry. 2011;72:1577–1584. doi: 10.4088/JCP.10m06634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sublette ME, Vaquero C, Baca-Garcia E, Pachano G, Huang YY, Oquendo MA, Mann JJ. Lack of association of SNPs from the FADS1-FADS2 gene cluster with major depression or suicidal behavior. Psychiatr Genet. 2015 doi: 10.1097/YPG.0000000000000111. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sullivan PF, Neale MC, Kendler KS. Genetic epidemiology of major depression: review and meta-analysis. Am J Psychiatry. 2000;157:1552–1562. doi: 10.1176/appi.ajp.157.10.1552. [DOI] [PubMed] [Google Scholar]
- Swenne I, Rosling A, Tengblad S, Vessby B. Omega-3 polyunsaturated essential fatty acids are associated with depression in adolescents with eating disorders and weight loss. Acta Paediatr. 2011;100:1610–1615. doi: 10.1111/j.1651-2227.2011.02400.x. [DOI] [PubMed] [Google Scholar]
- Taha AY, Cheon Y, Faurot KF, Macintosh B, Majchrzak-Hong SF, Mann JD, Hibbeln JR, Ringel A, Ramsden CE. Dietary omega-6 fatty acid lowering increases bioavailability of omega-3 polyunsaturated fatty acids in human plasma lipid pools. Prostaglandins Leukot Essent Fatty Acids. 2014;90:151–157. doi: 10.1016/j.plefa.2014.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanskanen A, Hibbeln JR, Hintikka J, Haatainen K, Honkalampi K, Viinamäki H. Fish consumption, depression, and suicidality in a general population. Arch Gen Psychiatry. 2001;58:512–513. doi: 10.1001/archpsyc.58.5.512. [DOI] [PubMed] [Google Scholar]
- Tsai AC, Lucas M, Okereke OI, O'Reilly EJ, Mirzaei F, Kawachi I, Ascherio A, Willett WC. Suicide mortality in relation to dietary intake of n-3 and n-6 polyunsaturated fatty acids and fish: equivocal findings from 3 large US cohort studies. Am J Epidemiol. 2014;179:1458–1466. doi: 10.1093/aje/kwu086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Kemp WJ, Klomp DW, Kahn RS, Luijten PR, Hulshoff Pol HE. A meta-analysis of the polyunsaturated fatty acid composition of erythrocyte membranes in schizophrenia. Schizophr Res. 2012;141:153–161. doi: 10.1016/j.schres.2012.08.014. [DOI] [PubMed] [Google Scholar]
- Vancassel S, Leman S, Hanonick L, Denis S, Roger J, Nollet M, Bodard S, Kousignian I, Belzung C, Chalon S. 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]
- Vines A, Delattre AM, Lima MM, Rodrigues LS, Suchecki D, Machado RB, Tufik S, Pereira SI, Zanata SM, Ferraz AC. The role of 5-HT1A receptors in fish oil-mediated increased BDNF expression in the rat hippocampus and cortex: a possible antidepressant mechanism. Neuropharmacology. 2012;62:184–191. doi: 10.1016/j.neuropharm.2011.06.017. [DOI] [PubMed] [Google Scholar]
- Wanders RJA. Peroxisomal biosynthesis of omega-3 fatty acids and human peroxisomal diseases. In: McNamara RK, editor. The Omega-3 Fatty Acid Deficiency Syndrome: Opportunities for Disease Prevention. Nova Science Publishers, Inc; U.S.A: 2013. pp. 19–30. [Google Scholar]
- Wang C, Harris WS, Chung M, Lichtenstein AH, Balk EM, Kupelnick B, Jordan HS, Lau J. n-3 Fatty acids from fish or fish-oil supplements, but not alpha-linolenic acid, benefit cardiovascular disease outcomes in primary- and secondary-prevention studies: a systematic review. Am J Clin Nutr. 2006;84:5–17. doi: 10.1093/ajcn/84.1.5. [DOI] [PubMed] [Google Scholar]
- Whelton SP, He J, Whelton PK, Muntner P. Meta-analysis of observational studies on fish intake and coronary heart disease. Am J Cardiol. 2004;93:1119–1123. doi: 10.1016/j.amjcard.2004.01.038. [DOI] [PubMed] [Google Scholar]
- Wozniak J, Biederman J, Mick E, Waxmonsky J, Hantsoo L, Best C, Cluette-Brown JE, Laposata M. Omega-3 fatty acid monotherapy for pediatric bipolar disorder: a prospective open-label trial. Eur Neuropsychopharmacol. 2007;17:440–447. doi: 10.1016/j.euroneuro.2006.11.006. [DOI] [PubMed] [Google Scholar]
- Yavin E, Brand A, Green P. Docosahexaenoic acid abundance in the brain: a biodevice to combat oxidative stress. Nutr Neurosci. 2002;5:149–157. doi: 10.1080/10284150290003159. [DOI] [PubMed] [Google Scholar]
- Yavin E, Himovichi E, Eilam R. Delayed cell migration in the developing rat brain following maternal omega 3 alpha linolenic acid dietary deficiency. Neuroscience. 2009;162:1011–1122. doi: 10.1016/j.neuroscience.2009.05.012. [DOI] [PubMed] [Google Scholar]
- Yee LD, Lester JL, Cole RM, Richardson JR, Hsu JC, Li Y, Lehman A, Belury MA, Clinton SK. Omega-3 fatty acid supplements in women at high risk of breast cancer have dose-dependent effects on breast adipose tissue fatty acid composition. Am J Clin Nutr. 2010;91:1185–1194. doi: 10.3945/ajcn.2009.29036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang TY, Labonté B, Wen XL, Turecki G, Meaney MJ. Epigenetic mechanisms for the early environmental regulation of hippocampal glucocorticoid receptor gene expression in rodents and humans. Neuropsychopharmacology. 2013;38:111–123. doi: 10.1038/npp.2012.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimmer L, Delion-Vancassel S, Durand G, Guilloteau D, Bodard S, Besnard JC, Chalon S. Modification of dopamine neurotransmission in the nucleus accumbens of rats deficient in n-3 polyunsaturated fatty acids. J Lipid Res. 2000;41:32–40. [PubMed] [Google Scholar]
- Zimmer L, Vancassel S, Cantagrel S, Breton P, Delamanche S, Guilloteau D, Durand G, Chalon S. The dopamine mesocorticolimbic pathway is affected by deficiency in n-3 polyunsaturated fatty acids. Am J Clin Nutr. 2002;75:662–667. doi: 10.1093/ajcn/75.4.662. [DOI] [PubMed] [Google Scholar]