Next Article in Journal
Mitochondrial Dysfunction in Diabetic Cardiomyopathy: The Possible Therapeutic Roles of Phenolic Acids
Next Article in Special Issue
An Update on Vitamin D Metabolism
Previous Article in Journal
Revealing Metabolic Perturbation Following Heavy Methamphetamine Abuse by Human Hair Metabolomics and Network Analysis
Previous Article in Special Issue
Lactobacillus Strains Alleviated Hyperlipidemia and Liver Steatosis in Aging Rats via Activation of AMPK
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Omega 3 Fatty Acids on Main Dimensions of Psychopathology

1
Department of Neuroscience, Faculty of Medicine, University of Turin, 10126 Turin, Italy
2
Center for Personality Disorders, Psychiatric Clinic, 10126 Turin, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(17), 6042; https://doi.org/10.3390/ijms21176042
Submission received: 17 July 2020 / Revised: 11 August 2020 / Accepted: 19 August 2020 / Published: 21 August 2020
(This article belongs to the Special Issue Nutraceuticals in Chronic Diseases)

Abstract

:
The usefulness of polyunsaturated fatty acids on inflammatory, cardiovascular, and the nervous system was studied in the last decades, but the mechanisms underlying their benefic properties are still partially unknown. These agents seem to express their action on the membrane phospholipid composition and permeability and modulation of second messenger cascades. In psychiatry, the efficacy and tolerability of omega-3 fatty acids were investigated in several psychiatric disorders, including major depression, bipolar disorder, personality disorders, high-risk conditions to develop psychosis, attention-deficit hyperactivity disorder, and autism spectrum disorders. Initial findings in this field are promising, and some relevant questions need to be addressed. In particular, the effects of these agents on the main symptom dimensions have to be investigated in a trans-diagnostic perspective. The present systematic review is aimed to examine the available data on the efficacy of omega-3 fatty acids on domains of psychotic symptoms, affective symptoms, impulsivity, and aggressiveness, and harmful behaviors, and suicide risk.

1. Introduction

In the last decades, the scientific literature has highlighted an urgent need to identify new compounds to treat psychiatric disorders in order to improve and boost available medications that entail not insignificant side effects. [1] As the harmful effects of the western diet, the absence of polyunsaturated fatty acids (PUFAs), on immune memory and inflammation processes are recognized [2], several studies evaluated the relationships between the low plasma level of long-chain polyunsaturated fatty acids and several medical conditions. [3] The effects of omega-3 fatty acids (n-3 PUFAs), including α-linolenic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), and of omega-6 fatty acids (n-6 PUFAs), including linoleic and arachidonic acid on overall human health have been extensively studied [4,5,6].
Nowadays, a consistent body of literature supports the importance of polyunsaturated fatty acids in brain function and the effect of their supplementation in mental disorders, but the molecular mechanisms underpinning their action are not completely clear [7,8,9]. Omega-3 fatty acids are essential fatty acids that humans and other animals require for good health, but cannot be synthesized. So, these compounds are introduced into the organism with food and metabolic conversion. The lipid derivatives of omega-3 PUFAs include anti-inflammatory eicosanoids, such as resins and resolvins, which mediate the effects on immune function [10,11]. Long-chain PUFAs, including at least 20 carbon atoms, have important functional roles as components of phospholipids forming the cell membranes [12], and as signaling molecules in all tissues, including the brain [13,14]. DHA is an essential omega-3 long-chain polyunsaturated fatty acid involved in many cellular processes in mammalian cells [15], represents approximately 60% of PUFAs in neuronal membranes [16], and is a precursor for bioactive compounds that modulate cell signaling and gene expression [17]. It takes part in neuronal growth, development, and function, acting as a neurotrophic factor and modulating synaptic activity [18], and it has been found to reduce the deleterious effects of chronic activation of inflammatory signaling cascades in the brain and to ameliorate neurotoxic injuries implicated in neurodegenerative disorders [19].
Some investigations showed that cellular molecules such as resolvins, protectins, peroxisome proliferator-activated receptor γ (PPAR-γ), AMP-activated protein kinase (AMPK), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) are potential targets of the anti-inflammatory action of DHA [20,21,22]. Other authors observed that DHA supplementation in an aged rat model could reduce cerebral reactive oxygen species, prevent age-related cognitive dysfunction [23], and decrease oxidative stress in rats subjected to permanent cerebral ischemia [20]. Another potential mechanism of the action of DHA is the regulation of the retinoid X receptor (RXR) that is a crucial developmental and survival factor implicated in multiple transcription pathways, such as the dopaminergic system [24].
Supplementation with uridine, DHA, and choline resulted in enhanced levels of phosphatidylcholine (PC), as well as other membrane phospholipids in the brains of healthy adult rodents [25]. The same effect was observed when DHA was substituted with EPA, but not with the omega-6 PUFA arachidonic acid (AA) [26]. The combination of DHA and EPA also intensified membrane phospholipid levels in brains of pups born from rats supplemented during gestation and nursing and produced an increased number of dendritic spines per unit area and the improvement of synaptic activities [25,27]. These findings suggested that synaptogenesis is partially determined by the availability of phospholipid precursors into the brain. In fact, phospholipid precursors supplementation in animal models produced biochemical and structural modifications promoting the release of neurotransmitters, such as dopamine [28] and acetylcholine [29], and improvement in specific cognitive tasks [30].
Supplementation with phospholipid precursors not only enhanced synaptogenesis in normal conditions, but also provided benefit under degenerative conditions that are characterized by synaptic deficiency both in laboratory rodents and humans. The restorative effects of supplementation with uridine monophosphate (UMP), DHA, and choline were indicated by increasing striatal dopamine levels and tyrosine hydroxylase (TH) activity, and by enhancing levels of membrane phospholipids and synaptic proteins in a rat model of Parkinson’s disease [31].
On the basis of these preclinical findings [32], several studies have been performed to test the usefulness of these agents in clinical conditions. Recent reviews [7,8,9,10] and meta-analyses [33,34] supported the role of PUFAs in the treatment of psychiatric disorders. In particular, the efficacy and tolerability of omega-3 fatty acids (DHA and EPA) were investigated in major depression, bipolar disorder, personality disorders, high-risk conditions to develop psychosis, attention-deficit hyperactivity disorder, and autism spectrum disorders. An interesting perspective is to deal with the effects of these agents in a trans-diagnostic perspective and investigate their effects on the main psychiatric symptom dimensions. The present systematic review is aimed to examine available data on the efficacy of omega-3 fatty acids on domains of psychotic symptoms, affective symptoms, impulsivity, and harmful behaviors.

2. Psychotic Symptoms

Deficits in long-chain n-3 PUFAs are one of the mechanisms thought to contribute to the risk of psychosis [35]. In schizophrenia and other psychotic disorders, the phospholipid structure of the cell membrane is altered and these modifications may contribute to various features of the pathophysiology observed in psychotic disorders, including neurotransmission, immune activation, and antioxidative defense [36]. This hypothesis is supported by studies that showed a plasma n-3 PUFAs deficiency in patients with schizophrenia compared to controls. [37] Besides, some evidence has suggested that the onset of psychotic disorders arises during human brain development in which n-3 PUFAs play a central role [38]. So, n-3 PUFAs supplementation could have an effect in preventing transition to psychosis [39,40], in particular, in high-risk populations [41].
These initial promising findings allow us to take into consideration the use of non-pharmacological compounds for early interventions in young people at risk for psychosis [42]. The results obtained so far about PUFAs as an add-on strategy in the treatment of schizophrenia are controversial; while several studies [40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57] produced favorable data, some others [58,59,60,61] showed no significant differences in clinical outcomes.
The symptoms dimension of psychosis is characterized by abnormalities in five symptom domains: delusions, hallucinations, disorganized thinking, disorganized behaviors, and negative symptoms. Lower levels of n-3 PUFAs seem to correlate with more severe negative symptoms in ultra-high risk (UHR) patients for psychosis and could be adopted as a biomarker that predicts the conversion to psychosis in UHR subjects [43].
Some findings suggested a role of EPA as an add-on therapy (in combination with antipsychotic therapy) in general, positive, negative, and depressive symptoms [46,47,49,50,51] in patients with UHR and first episode of psychosis. EPA or DHA augmentation to antipsychotics reduced the deterioration of hippocampus tissues with a positive effect on negative symptoms [47], decreased the oxidative stress status of plasma with a positive effect on global and negative symptoms [50], and increased the telomerase levels in peripheral blood cells with a positive effect on the severity of illness [51].
Some authors suggested that in stable schizophrenia, omega-3 fatty acid supplementation had a beneficial effect on positive symptoms (delusions and hallucinations) [52,55]. Among PUFAs, EPA has been found superior than placebo and also than DHA in reducing positive [52,55] and negative symptoms [54]. Moreover, supplementation with EPA induced a less severe impairment of the course of psychosis [56].
Previous findings [62] showed that omega-3 fatty acids have beneficial effects on triglycerides in patients with psychotic symptoms and metabolic syndrome and may enhance the brain-derived neurotrophic factor (BDNF) levels through their anti-inflammatory properties with a reduction of cognitive dysfunction in these patients [63]. Probably, an appropriate dietary supplementation could play a partially therapeutic effect even in more severe patients, improving some behavioral aspects and reducing the cognitive deterioration [64]. In addition, EPA supplementation was found to be associated with a marked increase of glutathione, an antioxidant agent, in patients with first episode of psychosis. [46]
Further studies are needed to investigate the role of omega-3 fatty acids as a treatment for the specific clusters of psychotic symptoms in psychiatric disorders other than schizophrenia. In fact, there is lack of evidence regarding the efficacy of PUFAs on the psychotic cluster of symptoms in bipolar disorder, schizoaffective disorder, and schizotypal personality disorder. To our knowledge only one case report documented the benefits of omega-3 fatty acids supplementation (EPA 1.187 g/day + DHA 0.613 g/day) in association with cholecalciferol (vitamin D3) and mood stabilizers for psychotic features in bipolar disorder [65].
In apparent contrast with the expectations, higher scores on positive schizotypal trait measures in healthy adults were found to be associated with higher concentrations of omega-6 and omega-3 fatty acids in red cell membranes [66]. These findings seem to support the hypothesis that high blood levels of PUFAs may confer some protection against psychotic breakdown [67].
Main findings are displayed in Table 1.

3. Affective Symptoms

Affective symptoms include depression, hypomania, mania, and anxiety symptoms. Abnormalities in affectivity characterize mood disorders but are also frequently associated with other mental disorders, such as psychotic and personality disorders. The role of PUFAs in treating affective symptoms such as depression can be linked to the inflammation hypothesis of the pathogenesis of depression and to the effects of EPA and DHA in reducing the effects of chronic activation of the inflammatory cascade [68]. In addition, it has been found that patients with major depressive disorder (MDD) have a lower level of EPA and DHA in their peripheral tissues (plasma, serum, and red blood cells) than control subjects [69].
The antidepressant efficacy of PUFAs could be partly explained by their role in myelinization processes—several models have been proposed to explain oligodendroglia cell loss/dysfunction in MDD. Increased levels of circulating corticosterone linked to the over-activation of the hypothalamic, pituitary, adrenal (HPA) axis in stress conditions could be at the origin of oligodendroglia loss/dysfunction in depression [70]. Studies in rats show that lower omega-3 PUFAs intake causes abnormalities of myelin [71] and that omega-3 PUFAs administration stimulates the expression of myelin proteins through the activation of signaling pathways involved in brain development [72]. Some experimental brain injury studies in rodents support this association and conclude that the supplementation with n-3 PUFAs increases the degree of differentiation of oligodendroglia cells [73,74].
Furthermore, an unbalanced omega-3/omega-6 ratio could lead to a range of functional consequences in the monoamine transport system [75]. In fact, there is evidence that severe n-3 PUFA deficiency alters the dopaminergic and serotonergic transmission systems, inducing an increased vulnerability to mood disorders. The mesolimbic dopaminergic pathway was more active, whereas the mesocortical pathway was found to be less active in rats with n-3 PUFA deficiency. This imbalance in dopaminergic neurotransmission could be related to symptoms of depressive episodes, such as lack of motivation, decreased response to reward, and impairment in intellective abilities [76].
The inflammatory response and the stimulation of BDNF have been hypothesized as two potential mechanisms through which omega-3 fatty acids could provide their anxiolytic effect. In particular, as anxiety is associated with an increased production of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), the effect of n-3 PUFAs of decreasing inflammatory cytokines indirectly promote the reduction of anxious symptoms. Moreover, when BDNF is low, it fails to stimulate synaptic growth of serotonergic neurons in the brain, and low levels of serotonin are associated with anxiety. Thus, BDNF could reduce depression and anxiety by stimulating synaptic growth of serotonergic neurons in the brain [77,78].
A consistent number of studies [79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112] investigated the role of PUFAs in the treatment of major depressive disorder (MDD). Although some of them [79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96] found no significant data, several trials [97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112] showed an improvement of depressive symptoms in patients treated with n-3 PUFAs in combination with antidepressants. In particular, a combination of higher dose of EPA with lower dose of DHA (EPA 1.67 g/day + DHA 0.83 g/day), added to existing antidepressant treatment, revealed beneficial effects on mood [102,103]. On the contrary, lower doses of EPA with higher doses of DHA (EPA 0.6 g/day + DHA 2.2 g/day added to standard therapy) evidenced no benefits on depressive symptoms [85]. Some authors [108] stated that higher doses of n-3 PUFAs (EPA 2.1/day + DHA 2.5 g/day) are more efficacious on depressive symptoms [85,86]. Several studies [113,114,115,116,117,118,119,120] investigating the efficacy of n-3 PUFAs supplementation (2.5 g/day with EPA:DHA = 2:1) in patients who suffer from bipolar disorder treated with mood stabilizers confirmed the effect of these agents on depressive symptoms [120] but not on manic ones [8,9]. Unlike major depression, in the case of bipolar disorder, higher doses of EPA (4.4 g/day) and DHA (2.4 g/day) seem to be not useful to obtain a better improvement of affective symptoms [113]. The reviews [121,122,123,124] and meta-analyses [33,125] on this topic concluded that there is a pressing need for further investigations, especially concerning the manic symptoms.
With regard to anxious symptoms, observational and experimental studies have demonstrated that ω-3 supplementation could ameliorate anxiety symptoms. High ω-3 fatty acid intake is associated with reduced likelihood of meeting the diagnostic criteria for anxiety disorders [126,127] and lower levels of self-reported anxiety symptom severity [128,129]. However, the literature data on the efficacy of PUFAs on anxiety are still scarce and inconclusive. Two trials [130,131] suggested a positive effect of n-3 PUFAs (EPA 2.25 g/day + DHA 0.5 g/day) on anxiety in patients with substance abuse and one open-label study reported an improvement of anxious symptoms in patients with post-traumatic stress disorder [132].
Depressive and anxious symptoms often occur in patients with schizophrenia or other psychotic disorders. In particular, young people at UHR to develop psychosis who present affective symptoms (depression and anxiety) show lower plasma levels of omega-3 fatty acids and higher levels of omega-6 fatty acids then controls [41]. On this basis, some authors investigated the effects of EPA and DHA supplementation in patients with schizophrenia and depressive and anxious symptoms. The results showed that EPA at doses ranging between 1 and 4 g/day is efficacious in reducing the depressive symptoms [50,54] and anxious symptoms [54] of schizophrenia. Among personality disorders, n-3 PUFAs were found efficacious on depressive symptoms of patients with borderline personality disorder (BPD), both in monotherapy (EPA 1 g/day) [133] and in addition to standard pharmacotherapies (EPA 1.2 g/day + DHA 0.9 g/day) [134].
Main findings are displayed in Table 2.

4. Impulsive and Aggressive Symptoms

There is cross-sectional evidence that deficiency of omega-3 fatty acids is associated with hostility, aggressive behaviors, and impulsivity in both psychiatric and non-psychiatric populations [135]. The relationship between omega-3 PUFAs and aggression have also been explored in rodent studies, that have found an increase of aggressive behaviors with both omega-3 PUFAs deficiency [136] and high-omega-6 PUFAs intake [137]. Omega-3 administration seems to have beneficial effects in reducing aggression among the general population [138]. A study performed in a group of Australian inmates found that the n-3 PUFAs levels were inversely related to the degree of aggressive behavior and hostility [139]. The results of several studies collected in a meta-analysis [140] are encouraging for the use of omega-3 fatty acid supplementation to prevent and reduce aggressive behaviors in both children and adults. The relationship between abnormal PUFAs distribution and conditions characterized by a high-level of impulsivity could be partly explained by the effect of these compounds on the serotonin system and membrane stability. In particular, EPA influences serotonin release and DHA improves membrane-embedded serotonin receptor accessibility with an increase of cognitive function, propensity for prosocial behavior, and of impulsive behavior control [141].
Higher omega-6/omega-3 ratios have been found in subjects with anger, irritability, and aggressiveness, in particular during exposure to elevated inflammatory cytokines [142]. Thus, inflammation and inflammation-related genetic polymorphisms are two possible additional sources of variability that may contribute to understanding the biological substrates of aggressive states [143].
The relationship between omega-3 PUFAs and aggressive and impulsive behaviors have been observed in various psychiatric contexts. It is possible that omega-3 fatty acids may alter brain functionality prior to emergent or detectable behavioral changes [135].
Lower plasma levels of total omega-3 PUFAs and a trend toward a higher ratio of omega-6 to omega-3 PUFAs were observed in conditions with a high level of impulsivity. For example, among subjects with gambling disorder, a higher percentage composition of EPA and a lower AA/EPA ratio and AA/DHA ratio in the red blood cell membrane was observed in impulsive gamblers, compared with non-impulsive ones [144]. In a similar way, cocaine abusers with impulsive and aggressive behaviors showed an imbalance between omega-3 and omega-6 fatty acids [145,146], and violent males and impulsive offenders hospitalized in a forensic psychiatric unit presented a lower plasmatic DHA level than controls [147].
Some available evidence suggests a beneficial effect of omega-3 supplementation on aggressive and antisocial behavior in adolescence and adulthood [148]. The mechanisms underpinning these results is still unclear, although the upregulation of dysfunctional prefrontal regions is one candidate mediator [149,150].
Studies that evaluated the effects of PUFAs on impulsivity and aggressiveness in major psychiatric disorders indicated that the addition of rather low doses of omega-3 fatty acids (EPA 0.54 g/day + DHA 0.36 g/day) to antipsychotic treatment might reduce agitation and violent behaviors in inpatients with schizophrenia in the chronic phase [61]. Single therapy with omega -3 fatty acids (EPA 0.93 g/day + DHA 0.29 g/day) showed an improvement of impulsive dyscontrol and aggressiveness in patients affected by ADHD (attention deficit hyperactivity disorder) [151,152] and in patients affected by BPD [133,134]. Moreover, combined therapy with EPA (1–1.2 g/day) plus DHA (0.6–0.9 g/day) and valproic acid (800–1300 mg/day—plasma range: 50–100 µg/mL) was superior to single therapy with valproic acid on impulsive–behavioral dyscontrol and outbursts of anger in BPD patients [153]. Combined therapy with omega-3 fatty acids showed long-lasting effects at the end of 24 weeks of follow-up in terms of anger control [154].
Main findings are displayed in Table 3.

5. Self-harm Behaviors and Suicidal Conducts

Self-harm behaviors include self-injuries, such as cutting and self-mutilation, burning, scratching, or hitting body parts [155]. The desire to self-harm is a common symptom of some personality disorders, but is frequent also in patients with mood disorders, anxiety disorders, substance use disorders, eating disorders, post-traumatic stress disorder, and schizophrenia [156]. The classification challenge presented by repetitive, non-lethal, self-harm is reflected in the numerous terms used for it in the historical literature, including self-mutilation, focal suicide, parasuicide, suicide gesture, wrist-cutting syndrome, delicate self-cutting, deliberate self-harm, self-injury, and self-injurious behavior. Currently, DSM-5 [157] includes several types of repetitive, non-lethal self-injurious behaviors: nonsuicidal self-injury (NSSI), trichotillomania (hair-pulling disorder), excoriation (skin-picking) disorder, and suicidal behavior disorder.
A theoretical model indicates the pathways whereby cholesterol-lowering treatments could affect PUFAs and lipid rafts, leading to alterations in serotonergic neurotransmission, corticotrophic function, and inflammation, and thereby increase parasuicidal and suicidal risk. So, there could be a preventative intervention, with respect to suicide and parasuicide, in administering n-3 PUFA supplements to patients for whom lower cholesterol is medically important and who have or develop psychiatric vulnerabilities, for example, patients with cardiac diseases or metabolic syndrome [158,159].
Unfortunately, studies on the effect of PUFAs on self-harm behaviors and suicide are still sparse. Some of these were performed in subjects without a psychiatric diagnosis and showed that low n-3 PUFAs plasma levels were associated with an increased number of deaths for suicide compared with other causes of death in active duty US military personnel. Moreover, veterans treated with PUFAs reported a reduction in suicidal ideation after omega-3 fatty acid supplementation [160,161].
Other investigations found that low EPA and DHA percentages, elevated omega-6/omega-3 ratio, and higher AA concentration seem to be associated with increased self-harm and suicidal attempts in patients with mood disorders [162,163,164,165]. Having low levels of arachidonic acid and receiving a combination of high dose of EPA and lithium were found protective for suicide attempts and deliberate self-harm in the case of bipolar disorder [166].
In BPD patients, n-3 PUFAs supplementation (EPA 1–1.2 g/day + DHA 0.6–0.9 g/day), added to standard psychiatric therapies, showed a significant reduction of self-harming and parasuicidal attitudes [133,134,153,154]. In particular, the reduction of suicidal behaviors seems to be independent of change in the depression score [134].
Main findings are displayed in Table 4.

6. Conclusive Remarks

Recent evidence supports the importance of polyunsaturated fatty acids in brain functioning and the action of their supplementation in psychiatric disorders, but the underlying mechanisms of the potential preventive and therapeutic effect of PUFAs is still unclear.
Preclinical studies hypothesized that omega-3 fatty acids may attenuate stress-related changes in animals with depressive features, as well as in humans [167]. These agents seem to be involved in myelinization processes and synaptic pruning, that are fundamental processes during brain development. PUFAs have immune-modulatory, and anti-inflammatory properties through the modulation of omega-6 fatty acids and the promotion of resolvins, neuroprotective factors, and anti-inflammatory mediators. They are also involved in membrane fluidity producing an improvement of monoaminergic transmission.
In the last decades, the role of long-chain PUFAs in the treatment of several psychiatric disorders has gradually increased, as confirmed by the growing number of randomized controlled trials testing the efficacy of essential fatty acids, especially omega-3 supplementation. Nevertheless, an overall agreement about their efficacy is still lacking, and the results of most trials are controversial and inconclusive. Differences in methods, including sample size, selection criteria, choice, and dosage of fatty acids (i.e., EPA, or DHA, or a combination of the two), and the duration of supplementation, often make results not comparable.
The aim of this review is to evaluate, in a trans-diagnostic perspective, the efficacy of omega-3 fatty acids on the main psychiatric symptom dimensions, in particular on domains of psychotic symptoms, affective symptoms, impulsivity, and harmful behaviors.
Concerning psychosis, the available data on the effects of EPA and DHA needs to be replicated. However, initial findings are rather promising and show that these compounds, in addition to antipsychotics, may improve all symptom domains of psychosis, in particular negative symptoms. Evidence on the effects of PUFAs as single therapy for schizophrenic patients and on psychotic symptoms that are part of the clinical picture of psychiatric disorders other than schizophrenia are not sufficient.
The available literature reported promising data also on the role of n-3 PUFAs in the treatment of affective symptoms. In particular, these compounds, administered at high doses (1–2 g/day, with a ratio EPA:DHA = 2:1), seem able to improve depressive symptoms, both in monotherapy and in addition to antidepressants in major depression, in combination with mood stabilizers in bipolar depression, in addition to antipsychotics in schizophrenia. Evidence of the efficacy of manic symptoms is lacking, while the effects on anxiety need to be clarified and confirmed.
Concerning impulsivity and aggressiveness, the initial results on the beneficial effects of EPA and DHA supplementation are encouraging, both in single and combined interventions, and induce a decrease in the levels of impulsivity, outbursts of anger, and overt aggression. This area of psychopathology needs to be explored in depth by future investigations to allow us to draw more reliable conclusions. Another symptom domain that appears to be sensitive to the action of PUFAs is that of self-harm conduct. The effects of PUFAs were studied in patients with a high risk of harmful and suicidal behaviors, mainly in subjects with severe personality disorders, but also in patients with mood disorders. Additional important factors that require evaluation in this population include lipid-associated genetic variants and epigenetic markers that modulate the effects of lipid status and of PUFAs supplementation on self-harm and suicide risk.
In conclusion, polyunsaturated fatty acids are dietary supplements that can have a role on the basis of initial research data in treating several symptoms of psychiatric disorders, at least in combination with traditional medications. The lack of severe adverse effects is a significant reason to carefully consider the therapeutic potential of these agents. However, we have no sufficient evidence to draw final conclusions and to propose official indications for PUFAs in psychiatry. This is mainly due to serious limitations and considerable heterogeneity in the design and methods of available trials. As is suggested by the organization of this review, the authors’ opinion is that less controversial and discordant data could be obtained in incoming studies by focusing, not on specific categorical diagnoses, but on psychopathological dimensions and symptom domains observed from a trans-nosographical perspective. Such dimensions could be the direct expression of the pathophysiological and biochemical pathways on which PUFAs actually produce their beneficial effects.

Funding

This research was supported by the funds of the Italian Ministry of Health for the Departments of Excellence.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMPAdenosine monophosphate
EPAEicosapentaenoic acid;
DHADocosahexaenoic acid;
ethyl-EPAEthyl-eicosapentaenoic acid;
AAArachidonic acid;
α-LAα-lipoic acid;
UHRUltra-high risk;
FEPFirst episode of psychosis;
CDRSChildhood Depression Rating Scale;
CDIChildhood Depression Inventory;
EPDSEdinburgh Postnatal Depression Scale;
GDSGeriatric Depression Scale;
Decrease of;
Increase of;
MDDMajor depressive disorder;
HDRSHamilton Depression Rating Scale;
BDIBeck Depression Inventory;
PDSSPostpartum Depression Screening Scale;
PANSSPEPPositive and Negative Syndrome ScaleIndividual-Family Psychoeducational Psychotherapy;
BPDBorderline personality disorder;
PTSDPosttraumatic Stress Disorder;
PUFAsPolyunsaturated fatty acid;
> Superior of;
< Inferior of;
ADHDAttention Deficit Hyperactivity Disorder;
BDNFBrain-Derived Neurotrophic Factor

References

  1. Nemeroff, C.B. Psychopharmacology and the future of personalized treatment. Depress. Anxiety 2014, 31, 906–908. [Google Scholar] [CrossRef] [PubMed]
  2. Bazan, N.G. Docosanoids and elovanoids from omega-3 fatty acids are pro-homeostatic modulators of inflammatory responses, cell damage and neuroprotection. Mol. Aspects Med. 2018, 64, 18–33. [Google Scholar] [CrossRef] [PubMed]
  3. Bang, H.O.; Dyerberg, J.; Nielsen, A.B. Plasma lipid and lipoprotein pattern in Greenlandic West-coast Eskimos. Lancet 1971, 1, 1143–1145. [Google Scholar] [CrossRef]
  4. Simopoulos, A.P. Essential fatty acids in health and chronic disease. Am. J. Clin. Nutr. 1999, 70, 560–569. [Google Scholar] [CrossRef] [Green Version]
  5. Shahidi, F.; Ambigaipalan, P. Omega-3 Polyunsaturated Fatty Acids and Their Health Benefits. Annu. Rev. Food Sci. Technol. 2018, 9, 345–381. [Google Scholar] [CrossRef] [PubMed]
  6. Ruiz-León, A.M.; Lapuente, M.; Estruch, R.; Casas, R. Clinical advances in immunonutrition and atherosclerosis: A review. Front. Immunol. 2019, 10, 837. [Google Scholar] [CrossRef] [PubMed]
  7. Mischoulon, D.; Freeman, M.P. Omega-3 fatty acids in psychiatry. Psychiatr. Clin. N. Am. 2013, 36, 15–23. [Google Scholar] [CrossRef] [PubMed]
  8. Bozzatello, P.; Brignolo, E.; De Grandi, E.; Bellino, S. Supplementation with omega-3 fatty acids in psychiatric disorders: A review of literature data. J. Clin. Med. 2016, 5, 67. [Google Scholar] [CrossRef]
  9. Bozzatello, P.; Rocca, P.; Mantelli, E.; Bellino, S. Polyunsaturated Fatty Acids: What is Their Role in Treatment of Psychiatric Disorders? Int. J. Mol. Sci. 2019, 20, 5257. [Google Scholar] [CrossRef] [Green Version]
  10. Weylandt, K.H.; Chiu, C.Y.; Gomolka, B.; Waechter, S.F.; Wiedenmann, B. Omega-3 fatty acids and their lipid mediators: Towards an understanding of resolvin and protectin formation. Prostaglandins Other Lipid Mediat. 2012, 97, 73–82. [Google Scholar] [CrossRef]
  11. Calder, P.C. Marine omega-3 fatty acids and inflammatory processes: Effects, mechanisms and clinical relevance. Biochim. Biophys. Acta 2015, 1851, 469–484. [Google Scholar] [CrossRef] [PubMed]
  12. Dyall, S.C. Long-chain omega-3 fatty acids and the brain: A review of the independent and shared effects of EPA, DPA and DHA. Front. Aging Neurosci. 2015, 7, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Willatts, P.; Forsyth, J.S.; DiModugno, M.K.; Varma, S.; Colvin, M. Effect of long-chain polyunsaturated fatty acids in infant formula on problem solving at 10 months of age. Lancet 1998, 352, 688–691. [Google Scholar] [CrossRef]
  14. Birch, E.E.; Garfield, S.; Hoffman, D.R.; Uauy, R.; Birch, D.G. A randomized controlled trial of early dietary supply of long-chain polyunsaturated fatty acids and mental development in term infants. Dev. Med. Child Neurol. 2000, 42, 174–181. [Google Scholar] [CrossRef] [PubMed]
  15. Heras-Sandoval, D.; Pedraza-Chaverri, J.; Perez-Rojas, J.M. Role of docosahexaenoic acid in the modulation of glial cells in Alzheimer’s disease. J. Neuroinfl. 2016, 13, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Belkouch, M.; Hachem, M.; Elgot, A.; Lo Van, A.; Picq, M.; Guichardant, M. The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer’s disease. J. Nutr. Biochem. 2016, 38, 1–11. [Google Scholar] [CrossRef] [PubMed]
  17. Calder, P.C. Mechanisms of action of (n-3) fatty acids. J. Nutr. 2012, 142, 592S–599S. [Google Scholar] [CrossRef] [Green Version]
  18. Ruiz-Roso, M.B.; Olivares-Alvaro, E.; Quintela, J.C.; Ballesteros, S. Effects of Low Phytanic Acid-Concentrated DHA on Activated Microglial Cells: Comparison with a Standard Phytanic Acid- Concentrated DHA. NeuroMolecular. Med. 2018, 20, 328–342. [Google Scholar] [CrossRef]
  19. Lefkowitz, D.L.; Lefkowitz, S.S. Microglia and myeloperoxidase: A deadly partnership in neurodegenerative disease. Free Radic. Biol. Med. 2008, 45, 726–731. [Google Scholar] [CrossRef]
  20. Chang, C.Y.; Kuan, Y.H.; Li, J.R.; Chen, W.Y. Docosahexaenoic acid reduces cellular inflammatory response following permanent focal cerebral ischemia in rats. J. Nutr. Biochem. 2013, 24, 2127–2137. [Google Scholar] [CrossRef]
  21. Marcheselli, V.L.; Hong, S.; Lukiw, W.J.; Tian, X.H. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and proinflammatory gene expression. J. Biol. Chem. 2003, 278, 43807–43817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Zuniga, J.; Cancino, M.; Medina, F.; Varela, P. N-3 PUFA supplementation triggers PPAR-alpha activation and PPAR-alpha/NF-kappaB interaction: Anti-inflammatory implications in liver ischemia-reperfusion injury. PLoS ONE 2011, 6, e28502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hashimoto, M.; Katakura, M.; Tanabe, Y.; Al Mamun, A. n-3 fatty acids effectively improve the reference memory-related learning ability associated with increased brain docosahexaenoic acid-derived docosanoids in aged rats. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2015, 1851, 203–209. [Google Scholar] [CrossRef] [PubMed]
  24. Healy-Stoffel, M.; Levant, B. N-3 (Omega-3) fatty acids: Effects on brain dopamine system and potential role in the etiology and treatment of neuropsychiatric disorders. CNS Neurol. Disord. Drug Targets 2018, 17, 216–232. [Google Scholar] [CrossRef] [PubMed]
  25. Sakamoto, T.; Cansev, M.; Wurtman, R.J. Oral supplementation with docosahexaenoic acid and uridine 50-monophosphate increases dendritic spine density in adult gerbil hippocampus. Brain Res. 2007, 1182, 50–59. [Google Scholar] [CrossRef] [Green Version]
  26. Cansev, M.; Wurtman, R.J. Chronic administration of docosahexaenoic acid or eicosapentaenoic acid, but not arachidonic acid, alone or in combination with uridine, increases brain phosphatide and synaptic protein levels in gerbils. Neuroscience 2007, 148, 421–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Cansev, M.; Marzloff, G.; Sakamoto, T.; Ulus, I.H.; Wurtman, R.J. Giving uridine and/or docosahexaenoic acid orally to rat dams during gestation and nursing increases synaptic elements in brains of weanling pups. Dev. Neurosci. 2009, 11, 45–57. [Google Scholar] [CrossRef]
  28. Wang, L.; Pooler, A.M.; Albrecht, M.A.; Wurtman, R.J. Dietary uridine-50-monophosphate supplementation increases potassium-evoked dopamine release and promotes neurite outgrowth in aged rats. J. Mol. Neurosci. 2005, 27, 137–145. [Google Scholar] [CrossRef]
  29. Wang, L.; Albrecht, M.A.; Wurtman, R.J. Dietary supplementation with uridine-50- monophosphate (UMP), a membrane phosphatide precursor, increases acetylcholine level and release in striatum of aged rat. Brain Res. 2007, 1133, 42–48. [Google Scholar] [CrossRef] [Green Version]
  30. Holguin, S.; Huang, Y.; Liu, J.; Wurtman, R. Chronic administration of DHA and UMP improves the impaired memory of environmentally impoverished rats. Behav. Brain Res. 2008, 191, 11–16. [Google Scholar] [CrossRef] [Green Version]
  31. Cansev, M.; Ilcol, Y.O.; Yilmaz, M.S.; Hamurtekin, E.; Ulus, I.H. Peripheral administration of CDP-choline, phosphocholine or choline increases plasma adrenaline and noradrenaline concentrations. Auton. Autacoid Pharmacol. 2008, 28, 41–58. [Google Scholar] [CrossRef] [PubMed]
  32. Cansev, M. Synaptogenesis: Modulation by Availability of Membrane Phospholipid Precursors. Neuromol. Med. 2016, 18, 426–440. [Google Scholar] [CrossRef] [PubMed]
  33. Sublette, M.E.; Ellis, S.P.; Geant, A.L.; Mann, J.J. Meta-analysis of the effects of eicosapentaenoic acid (EPA) in clinical trials in depression. J. Clin. Psychiatry 2011, 72, 1577–1584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Amminger, G.P.; Berger, M.; Rice, S.M.; Davey, C.G. Novel biotherapies are needed in youth mental health. Australas. Psychiatry 2017, 25, 117–120. [Google Scholar] [CrossRef] [PubMed]
  36. Horrobin, D.F.; Glen, A.I.; Vaddadi, K. The membrane hypothesis of schizophrenia. Schizophr. Res. 1994, 13, 195–207. [Google Scholar] [CrossRef]
  37. Van Der Kemp, W.J.; Klomp, D.W.; Kahn, R.S.; Luijten, P.R. A meta-analysis of the polyunsaturated fatty acid composition of erythrocyte membranes in schizophrenia. Schizophr. Res. 2012, 141, 153–161. [Google Scholar] [CrossRef]
  38. Campoy, C.; Escolano-Margarit, M.V.; Anjos, T.; Szajewska, H. Omega 3 fatty acids on child growth, visual acuity and neurodevelopment. Br. J. Nutr. 2012, 107, S85–S106. [Google Scholar] [CrossRef]
  39. Agostoni, C.; Nobile, M.; Ciappolino, V.; Delvecchio, G.; Tesei, A. The Role of omega-3 fatty acids in developmental psychopathology: A systematic review on early psychosis, Autism, and ADHD. Int. J. Mol. Sci. 2017, 18, 2608. [Google Scholar] [CrossRef] [Green Version]
  40. Amminger, G.P.; Chanen, A.M.; Ohmann, S.; Klier, C.M.; Mossaheb, N.; Bechdolf, A.; Schäfer, M.R. Omega-3 Fatty Acid Supplementation in Adolescents with Borderline Personality Disorder and Ultra-High Risk Criteria for Psychosis: A Post Hoc Subgroup Analysis of a Double-Blind, Randomized Controlled Trial. Can J. Psychiatry 2013, 58, 402–408. [Google Scholar] [CrossRef] [Green Version]
  41. Clark, S.R.; Baune, B.T.; Schubert, K.O.; Lavoie, S. Prediction of transition from ultra-high risk to first-episode psychosis using a probabilistic model combining history, clinical assessment and fatty-acid biomarkers. Transl. Psychiatry 2016, 6, e897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Amminger, G.P.; Schäfer, M.R.; Schlögelhofer, M.; Klier, C. Longer-term outcome in the prevention of psychotic disorders by the Vienna omega-3 study. Nat. Commun. 2015, 6, 7934. [Google Scholar]
  43. Amminger, G.P.; Schäfer, M.R.; Papageorgiou, K.; Klier, C.M. Long-chain omega-3 fatty acids for indicated prevention of psychotic disorders: A randomized, placebo-controlled trial. Arch. Gen. Psychiatry 2010, 67, 146–154. [Google Scholar] [CrossRef] [PubMed]
  44. Smesny, S.; Milleit, B.; Hipler, U.C.; Milleit, C.; Schäfer, M.R.; Klier, C.M.; Holub, M.; Holzer, I.; Berger, G.E.; Otto, M.; et al. Omega-3 fatty acid supplementation changes intracellular phospholipase A2 activity and membrane fatty acid profiles in individuals at ultra-high risk for psychosis. Mol. Psychiatry 2014, 19, 317–324. [Google Scholar] [CrossRef] [PubMed]
  45. Berger, G.E.; Proffitt, T.M.; McConchie, M.; Yuen, H.; Wood, S.J.; Amminger, G.P.; Brewer, W.; McGorry, P.D. Ethyl-eicosapentaenoic acid in first-episode psychosis: A randomized, placebo-controlled trial. J. Clin. Psychiatry 2007, 68, 1867–1875. [Google Scholar] [CrossRef]
  46. Berger, G.E.; Wood, S.J.; Wellard, R.M.; Proffitt, T. Ethyl-eicosapentaenoic acid in first-episode psychosis. A 1H-MRS study. Neuropsychopharmacology 2008, 33, 2467–2473. [Google Scholar] [CrossRef] [Green Version]
  47. Wood, S.J.; Cocchi, L.; Proffitt, T.M.; McConchie, M.; Jackson, G.D.; Takahashi, T.; Pantelis, C.; Mc Gorny, P.D.; Berger, G.E. Neuroprotective effects of ethyl-eicosapentaenoic acid in first episode psychosis: A longitudinal T2 relaxometry pilot study. Psychiatry Res. Neuroimaging 2010, 182, 180–182. [Google Scholar] [CrossRef]
  48. Emsley, R.; Chiliza, B.; Asmal, L.; Emsley, R. A randomized, controlled trial of omega-3 fatty acids plus an antioxidant for relapse prevention after antipsychotic discontinuation in first-episode schizophrenia. Schizophr. Res. 2014, 158, 230–235. [Google Scholar] [CrossRef]
  49. Pawełczyk, T.; Grancow-Grabka, M.; Kotlicka-Antczak, M.; Trafalska, E.; Pawełczyk, A. A randomized controlled study of the efficacy of six-month supplementation with concentrated fish oil rich in omega-3 polyunsaturated fatty acids in first episode schizophrenia. J. Psychiatr. Res. 2016, 73, 34–44. [Google Scholar] [CrossRef]
  50. Pawelczyk, T.; Grancow-Grabka, M.; Trafalska, E.; Szemraj, J. Oxidative stress reduction related to the efficacy of n-3 polyunsaturated fatty acids in first episode schizophrenia: Secondary outcome analysis of the OFFER randomized trial. Prostaglandins Leukot. Essent. Fat Acids 2017, 121, 7–13. [Google Scholar] [CrossRef]
  51. Pawelczyk, T.; Grancow-Grabka, M.; Trafalska, E.; Szemraj, J. Telomerase level increase is related to n-3 polyunsaturated fatty acid efficacy in first episode schizophrenia: Secondary outcome analysis of the OFFER randomized clinical trial. Prog. Neuropsychopharm. Biol. Psychiatry 2018, 83, 142–148. [Google Scholar] [CrossRef] [PubMed]
  52. Peet, M.; Brind, J.; Ramchand, C.N.; Shah, S. Two double-blind placebo-controlled pilot studies of eicosapentaenoic acid in the treatment of schizophrenia. Schizophr. Res. 2001, 49, 243–251. [Google Scholar] [CrossRef]
  53. Peet, M.; Horrobin, D.F. Study Group. A dose-ranging exploratory study of the effects of ethyl-eicosapentaenoate in patients with persistent schizophrenic symptoms. J. Psychiatry Res. 2002, 36, 7–18. [Google Scholar] [CrossRef]
  54. Emsley, R.; Myburgh, C.; Oosthuizen, P.; van Rensburg, S.J. Randomized, placebo-controlled study of ethyl-eicosapentaenoic acid as supplemental treatment in schizophrenia. Am. J. Psychiatry 2002, 159, 1596–1598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Jamilian, H.; Solhi, H.; Jamilian, M. Randomized, placebo-controlled clinical trial of omega-3 as supplemental treatment in schizophrenia. Glob. J. Health Sci. 2014, 18, 103–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Bentsen, H.; Osnes, K.; Refsum, H. A randomized placebo-controlled trial of an omega-3 fatty acid and vitamins E + C in schizophrenia. Transl. Psychiatry 2013, 3, e335. [Google Scholar] [CrossRef] [Green Version]
  57. Robinson, D.G.; Schooler, N.R.; Rosenheck, R.A.; Lin, H. Predictors of hospitalization of individuals with first-episode psychosis: Data from a 2-year follow-up of the raise-ETP. Psychiatry Serv. 2019, 7, 569–577. [Google Scholar] [CrossRef]
  58. McGorry, P.D.; Nelson, B.; Markulev, C.; Yuen, H.; Schafer, M.R.; Mossaheb, N.; Schlögelhofer, M.; Smesny, S.; Hickie, I.B.; Berger, G.E.; et al. Effect of ω-3 Polyunsaturated Fatty Acids in Young People at Ultrahigh Risk for Psychotic Disorders. JAMA Psychiatry 2017, 74, 19. [Google Scholar] [CrossRef]
  59. Emsley, R.; Niehaus, D.J.; Koen, L.; Oosthuizen, P.P.; Turner, H.J.; Carey, P.; van Rensburg, S.J.; Maritz, J.S.; Murck, H. The effects of eicosapentaenoic acid in tardive dyskinesia: A randomized, placebo-controlled trial. Schizophr. Res. 2006, 84, 112–120. [Google Scholar] [CrossRef]
  60. Fenton, W.S.; Dickerson, F.; Boronow, J.; Hibbeln, J.R.; Knable, M. A placebo-controlled trial of omega-3 fatty acid (ethyl eicosapentaenoic acid) supplementation for residual symptoms and cognitive impairment in schizophrenia. Am. J. Psychiatry 2001, 158, 2071–2074. [Google Scholar] [CrossRef]
  61. Qiao, Y.; Mei, Y.; Han, H.; Liu, F.; Yang, X.M.; Shao, Y.; Xie, B.; Long, B. Effects of Omega-3 in the treatment of violent schizophrenia patients. Schizophr. Res. 2018, 195, 283–285. [Google Scholar] [CrossRef] [PubMed]
  62. Yonezawa, K.; Kusumoto, Y.; Kanchi, N.; Kinoshita, H.; Kanegae, S.; Yamaguchi, N.; Ozawa, H. Recent trends in mental illness and omega-3 fatty acids. J. Neural Transm. 2020, 1–9. [Google Scholar] [CrossRef] [PubMed]
  63. Xu, F.; Fan, W.; Wang, W.; Tang, W. Effects of omega-3 fatty acids on metabolic syndrome in patients with schizophrenia: A 12-week randomized placebo-controlled trial. Psychopharmacology 2019, 236, 1273–1279. [Google Scholar] [CrossRef] [PubMed]
  64. Tang, W.; Wang, Y.; Xu, F.; Fan, W.; Zhang, Y.; Fan, K.; Wang, W.; Zhang, Y.; Zhang, C. Omega-3 fatty acids ameliorate cognitive dysfunction in schizophrenia patients with metabolic syndrome [published online ahead of print, 2020 Apr 15]. Brain Behav. Immun. 2020, 88, 529–534. [Google Scholar] [CrossRef] [PubMed]
  65. Marano, G.; Traversi, G.; Nannarelli, C.; Mazza, S.; Mazza, M. Omega-3 fatty acids and schizophrenia: Evidences and recommendations. Clin. Ter. 2013, 164, e529–e537. [Google Scholar] [CrossRef]
  66. Vesco, A.T.; Lehmann, J.; Gracious, B.L.; Arnold, L.E.; Young, A.S.; Fristad, M.A. Omega-3 Supplementation for Psychotic Mania and Comorbid Anxiety in Children. J. Child Adolesc. Pharmacol. 2015, 25, 7. [Google Scholar] [CrossRef] [Green Version]
  67. Richardson, A.J.; Cyhlarova, E.; Ross, M.A. Omega-3 and omega-6 fatty acid concentrations in red blood cell membranes relate to schizotypal traits in healthy adults. Prostaglandins Leukot. Essent. Fat. Acids 2003, 69, 46–466. [Google Scholar] [CrossRef]
  68. Amodeo, G.; Trusso, M.A.; Fagiolini, A. Depression and inflammation: Disentangling a clear yet complex and multifaceted link. Neuropsychiatry 2017, 7, 448–457. [Google Scholar] [CrossRef]
  69. Lin, P.Y.; Huang, S.Y.; Su, K.P. A meta-analytic review of polyunsaturated fatty acid compositions in patients with depression. Biol. Psychiatry 2010, 68, 140–147. [Google Scholar] [CrossRef]
  70. Boda, E. Myelin and oligodendrocyte lineage cell dysfunctions: New players in the etiology and treatment of depression and stress-related disorders. Eur. J. Neurosci. 2019. [Google Scholar] [CrossRef]
  71. Trapp, B.D.; Bernsohn, J. Essential fatty acid deficiency and CNS myelin. Biochemical and morphological observations. J. Neurol. Sci. 1978, 37, 149–266. [Google Scholar] [CrossRef]
  72. Salvati, S.; Natali, F.; Attorri, L.; Di Benedetto, R.; Leonardi, F.; Di Biase, A.; Ferri, F.; Fortuna, S.; Lorenzini, P.; Sanchez, M.; et al. Eicosapentaenoic acid stimulates the expression of myelin proteins in rat brain. J. Neurosci. Res. 2008, 86, 776–784. [Google Scholar] [CrossRef] [PubMed]
  73. Ward, R.E.; Huang, W.; Curran, O.E.; Priestley, J.V.; Michael-Titus, A.T. Docosahexaenoic acid prevents white matter damage after spinal cord injury. J. Neurotr. 2010, 27, 1769–1780. [Google Scholar] [CrossRef] [PubMed]
  74. Mills, J.D.; Hadley, K.; Bailes, J.E. Dietary supplementation with the omega-3 fatty acid docosahexaenoic acid in traumatic brain injury. Neurosurgery 2011, 68, 474–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Logan, A.C. Neurobehavioral aspects of omega-3 fatty acids: Possible mechanisms and therapeutic value in major depression. Altern. Med. Rev. 2003, 8, 410–425. [Google Scholar] [PubMed]
  76. Chalon, S. Omega-3 fatty acids and monoamine neurotransmission. Prostaglandins Leukot. Essent. Fat. Acids 2006, 75, 259–269. [Google Scholar] [CrossRef]
  77. Mamounas, L.A.; Blue, M.E.; Siuciak, J.A.; Altar, C.A. Brain-derived neurotrophic factor promotes the survival and sprouting of serotonergic axons in rat brain. J. Neurosci. 1995, 5, 7929–7939. [Google Scholar] [CrossRef]
  78. Mattson, M.P.; Maudsley, S.; Martin, B. BDNF and 5-HT: A dynamic duo in age-related neuronal plasticity and neurodegenerative disorders. Trends Neurosci. 2004, 27, 589–594. [Google Scholar] [CrossRef]
  79. Llorente, A.M.; Jensen, C.L.; Voigt, R.G.; Fraley, J.K.; Berretta, M.C.; Heird, W.C. Effect of maternal docosahexaenoic acid supplementation on postpartum depression and information processing. Am. J. Obs. Gynecol. 2003, 188, 1348–1353. [Google Scholar] [CrossRef]
  80. Marangell, L.B.; Martinez, J.M.; Zboyan, H.A.; Kertz, B.; Kim, H.F.; Puryear, L.J. A double-blind, placebo-controlled study of the omega-3 fatty acid docosahexaenoic acid in the treatment of major depression. Am. J. Psychiatry 2003, 160, 996–998. [Google Scholar] [CrossRef]
  81. Silvers, K.M.; Woolley, C.C.; Hamilton, F.C.; Watts, P.M.; Watson, R.A. Randomised double-blind placebo-controlled trial of fish oil in the treatment of depression. Prostaglandins Leukot. Essent. Fat. Acids 2005, 72, 211–218. [Google Scholar] [CrossRef] [PubMed]
  82. Grenyer, B.F.; Crowe, T.; Meyer, B.; Owen, A.J. Fish oil supplementation in the treatment of major depression: A randomised double- blind placebo-controlled trial. Prog. Neuropsychopharmacol. Biol. Psychiatry 2007, 31, 1393–1396. [Google Scholar] [CrossRef] [PubMed]
  83. Freeman, M.P.; Davis, M.; Sinha, P.; Wisner, K.L.; Hibbeln, J.R.; Gelenberg, A.J. Omega-3 fatty acids and supportive psychotherapy for perinatal depression: A randomized placebo-controlled study. J. Affect Disord. 2008, 110, 142–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Jazayeri, S.; Tehrani-Doost, M.; Keshavarz, S.A.; Hosseini, 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. [Google Scholar] [CrossRef]
  85. Rees, A.M.; Austin, M.P.; Parker, G.B. Omega-3 fatty acids as a treatment for perinatal depression: Randomized double-blind placebo-controlled trial. Aust. N. Z. J. Psychiatry 2008, 42, 199–205. [Google Scholar] [CrossRef]
  86. Rogers, P.J.; Appleton, K.M.; Kessler, D.; Peters, T.J.; Gunnel, D.; Hayward, R.C.; Heatherley, S.V.; Christian, L.M.; McNaughton, S.A.; Ness, A.R. No effect of n-3 long-chain polyunsaturated fatty acid (EPA and DHA) supplementation on depressed mood and cognitive function: A randomised controlled trial. Br. J. Nutr. 2008, 99, 421–431. [Google Scholar] [CrossRef] [Green Version]
  87. Doornbos, B.; Van Goor, S.A.; Dijck-Brouwer, D.A.; Schaafsma, A.; Muskiet, F.A. Supplementation of a low dose of DHA or DHA+AA does not prevent peripartum depressive symptoms in a small population based sample. Prog. Neuropsychopharmacol. Biol. Psychiatry 2008, 33, 49–52. [Google Scholar] [CrossRef]
  88. Lucas, M.; Asselin, G.; Merette, C.; Poulin, M.J.; Dodin, S. Ethyleicosapentaenoic acid for the treatment of psychological distress and depressive symptoms in middle- aged women: A double-blind, placebo-controlled, randomized clinical trial. Am. J. Clin. Nutr. 2009, 89, 641–651. [Google Scholar] [CrossRef] [Green Version]
  89. Makrides, M.; Gibson, R.A.; McPhee, A.J.; Yelland, L. DOMInO Investigative Team, Effect of DHA supplementation during pregnancy on maternal depression and neurodevelopment of young children: A randomized controlled trial. JAMA 2010, 304, 1675–1683. [Google Scholar] [CrossRef] [Green Version]
  90. Antypa, N.; Smelt, A.H.; Strengholt, A.; Van der Does, A.J. Effects of omega-3 fatty acid supplementation on mood and emotional information processing in recovered depressed individuals. J. Psychopharmacol. 2012, 26, 738–743. [Google Scholar] [CrossRef]
  91. Mozurkewich, E.L.; Clinton, C.M.; Chilimigras, J.L.; Hamilton, S.E.; Allbaugh, L.J.; Berman, D.R.; Marcus, S.M.; Romero, V.C.; Treadwell, M.C.; Keeton, K.L.; et al. The mothers, Omega-3, and mental health study: A double-blind, randomized controlled trial. Am. J. Obs. Gynecol. 2013, 313, 1–9. [Google Scholar]
  92. Mischoulon, D.; Nierenberg, A.A.; Schettler, P.J.; Kinkead, B.L.; Fehling, K.; Martinson, M.A.; Hyman Rapaport, M. A double-blind, randomized controlled clinical trial comparing eicosapentaenoic acid versus docosahexaenoic acid for depression. J. Clin. Psychiatry 2015, 76, 54–61. [Google Scholar] [CrossRef] [PubMed]
  93. Park, Y.; Park, Y.S.; Kim, S.H.; Oh, D.H. Supplementation of n-3 polyunsaturated fatty acids for major depressive disorder: A randomized, double-blind, 12-week, placebo-controlled trial in Korea. Ann. Nutr. Metab. 2015, 66, 141–148. [Google Scholar] [CrossRef] [PubMed]
  94. Young, A.S.; Arnold, L.E.; Wolfson, H.L.; Fristad, M.A. Psychoeducational psychotherapy and omega-3 supplementation improve co-occurring behavioral problems in youth with depression: Results from a pilot RCT. J. Abnorm. Child Psychol. 2017, 45, 1025–1037. [Google Scholar] [CrossRef]
  95. Gabbay, V.; Freed, R.D.; Alonso, C.M.; Senger, S.; Stadterman, J.; Davidson, B.A.; Klein, R.G. A Double-blind placebo-controlled trial of omega-3 fatty acids as a monotherapy for adolescent depression. J. Clin. Psychiatry 2018, 26, 79. [Google Scholar] [CrossRef]
  96. Tayama, J.; Ogawa, S.; Nakaya, N.; Sone, T. Omega-3 polyunsaturated fatty acids and psychological intervention for workers with mild to moderate depression: A double-blind randomized controlled trial. J. Affect Disord. 2019, 245, 364–370. [Google Scholar] [CrossRef]
  97. Nemets, B.; Stahl, Z.; Belmaker, R.H. Addition of omega-3 fatty acid to maintenance medication treatment for recurrent unipolar depressive disorder. Am. J. Psychiatry 2002, 159, 477–479. [Google Scholar] [CrossRef]
  98. Peet, M.; Horrobin, D.F. 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. [Google Scholar] [CrossRef]
  99. Su, K.P.; Huang, S.Y.; Chiu, C.C.; Shen, W.W. Omega-3 fatty acids in major depressive disorder. A preliminary double-blind, placebo-controlled trial. Eur. Neuropsychopharmacol. 2003, 13, 267–271. [Google Scholar] [CrossRef]
  100. Nemets, H.; Nemets, B.; Apter, A.; Brach, Z. Omega-3 treatment of childhood depression: A controlled, double-blind pilot study. Am. J. Psychiatry 2006, 163, 1098–1100. [Google Scholar] [CrossRef]
  101. Mischoulon, D.; Papakostas, G.I.; Dording, C.M.; Farabaugh, A.H.; Sonawalla, S.B.; Agoston, A.M.; Smith, J.; Beaumont, E.C.; Dahan, L.E.; Alpert, J.E.; et al. A double-blind, randomized controlled trial of ethyl- eicosapentaenoate for major depressive disorder. J. Clin. Psychiatry 2009, 70, 1636–1644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Rondanelli, M.; Giacosa, A.; Opizzi, A.; Pelucchi, C. Effect of omega-3 fatty acids supplementation on depressive symptoms and on health-related quality of life in the treatment of elderly women with depression: A double-blind, placebo-controlled, randomized clinical trial. J. Am. Coll. Nutr. 2010, 29, 55–64. [Google Scholar] [CrossRef] [PubMed]
  103. Rondanelli, M.; Giacosa, A.; Opizzi, A.; Pelucchi, C. Long chain omega 3 polyunsaturated fatty acids supplementation in the treatment of elderly depression: Effects on depressive symptoms, on phospholipids fatty acids profile and on health-related quality of life. J. Nutr. Health Aging 2011, 15, 37–44. [Google Scholar] [CrossRef] [PubMed]
  104. Lespérance, F.; Frasure-Smith, N.; St-André, E.; Lespérance, F.; Frasure-Smith, N.; Turecki, G.; Wisniewski, S.R. The efficacy of omega-3 supplementation for major depression: A randomized controlled trial. J. Clin. Psychiatry 2011, 72, 1054–1062. [Google Scholar] [CrossRef] [PubMed]
  105. Tajalizadekhoob, Y.; Sharifi, F.; Fakhrzadeh, H.; Mirarefin, M. The effect of low-dose omega 3 fatty acids on the treatment of mild to moderate depression in the elderly: A double-blind, randomized, placebocontrolled study. Eur. Arch. Psychiatry Clin. Neurosci. 2011, 261, 539–549. [Google Scholar] [CrossRef]
  106. Gertsik, L.; Poland, R.E.; Bresee, C.; Rapaport, M.H. Omega-3 fatty acid augmentation of citalopram treatment for patients with major depressive disorder. J. Clin. Psychopharmacol. 2012, 32, 61–64. [Google Scholar] [CrossRef] [Green Version]
  107. Krawczyk, K.; Rybakowski, J. Augmentation of antidepressants with unsaturated fatty acids omega-3 in drug-resistant depression. Psychiatry Pol. 2012, 46, 585–598. [Google Scholar]
  108. Rizzo, A.M.; Corsetto, P.A.; Montorfano, G.; Opizzi, A. Comparison between the AA/EPA ratio in depressed and non depressed elderly females: Omega-3 fatty acid supplementation correlates with improved symptoms but does not change immunological parameters. Nutr. J. 2012, 11, 82. [Google Scholar] [CrossRef] [Green Version]
  109. Mozaffari-Khosravi, H.; Yassini-Ardakani, M.; Karamati, M.; Shariati-Bafghi, S.E. Eicosapentaenoic acid versus docosahexaenoic acid in mild-to-moderate depression: A randomized, doubleblind, placebo-controlled trial. Eur. Neuropsychopharmacol. 2013, 23, 636–644. [Google Scholar] [CrossRef]
  110. Judge, M.P.; Beck, C.T.; Durham, H.; McKelvey, M.M.; Lammi-Keefe, C.J. Pilot trial evaluating maternal docosahexaenoic acid consumption during pregnancy: Decreased postpartum depressive symptomatology. Int. J. Nurs. Sci. 2014, 1, 339–345. [Google Scholar] [CrossRef] [Green Version]
  111. Ginty, A.T.; Conklin, S.M. Short-term supplementation of acute long-chain omega-3 polyunsaturated fatty acids may alter depression status and decrease symptomology among young adults with depression: A preliminary randomized and placebo controlled trial. Psychiatry Res. 2015, 229, 485–489. [Google Scholar] [CrossRef] [PubMed]
  112. Jahangard, L.; Sadeghi, A.; Ahmadpanah, M.; Holsboer-Trachsler, E.; Sadeghi-Bahmani, D.; Haghighi, M.; Brand, S. Influence of adjuvant omega-3-polyunsaturated fatty acids on depression, sleep, and emotion regulation among outpatients with major depressive disorders—Results from a double-blind, randomized and placebo-controlled clinical trial. J. Psychiatry Res. 2018, 107, 48–56. [Google Scholar] [CrossRef] [PubMed]
  113. Chiu, C.C.; Huang, S.Y.; Chen, C.C.; Su, K.P. Omega-3 fatty acids are more beneficial in the depressive phase than in the manic phase in patients with bipolar I disorder. J. Clin. Psychiatry 2005, 66, 1613–1614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. McNamara, R.K.; Jandacek, R.; Rider, T.; Tso, P.; Stanford, K.E.; Hahn, C.G.; Richtand, N.M. Deficits in docosahexaenoic acid and associated elevations in the metabolism of arachidonic acid and saturated fatty acids in the postmortem orbitofrontal cortex of patients with bipolar disorder. Psychiatry Res. 2008, 160, 285–299. [Google Scholar] [CrossRef] [Green Version]
  115. Hirashima, F.; Parow, A.M.; Stoll, A.L.; Demopulos, C.M.; Damico, K.E.; Rohan, M.L.; Eskesen, J.G.; Zuo, C.S.; Cohen, B.M.; Renshaw, P.F. Omega-3 fatty acid treatment and T (2) whole brain relaxation times in bipolar disorder. Am. J. Psychiatry 2019, 161, 1922–1924. [Google Scholar] [CrossRef]
  116. Keck, P.E., Jr.; Mintz, J.; McElroy, S.L.; Freeman, M.P. Double-blind, randomized, placebo-controlled trials of ethyl-eicosapentanoate in the treatment of bipolar depression and rapid cycling bipolar disorder. Biol. Psychiatry 2006, 60, 1020–1022. [Google Scholar] [CrossRef]
  117. Frangou, S.; Lewis, M.; McCrone, P. Efficacy of ethyl-eicosapentaenoic acid in bipolar depression: Randomised double-blind placebo-controlled study. Br. J. Psychiatry 2006, 188, 46–50. [Google Scholar] [CrossRef] [Green Version]
  118. Murphy, B.L.; Stoll, A.L.; Harris, P.Q.; Ravichandran, C.; Babb, S.M.; Carlezon, W.A.; Cohen, B.M. Omega-3 fatty acid treatment, with or without cytidine, fails to show therapeutic properties in bipolar disorder: A double-blind, randomized add-on clinical trial. J. Clin. Psychopharmacol. 2012, 32, 699–703. [Google Scholar] [CrossRef]
  119. Stoll, A.L.; Severus, W.E.; Freeman, M.P.; Rueter, S. Omega 3 fatty acids in bipolar disorder: A preliminary double-blind, placebo-controlled trial. Arch. Gen. Psychiatry 1999, 56, 407–412. [Google Scholar] [CrossRef]
  120. Gracious, B.L.; Chirieac, M.C.; Costescu, S.; Finucane, T.L. Randomized, placebo-controlled trial of flax oil in pediatric bipolar disorder. Bipolar. Disord. 2010, 12, 142–154. [Google Scholar] [CrossRef] [Green Version]
  121. Saunders, E.F.; Ramsden, C.E.; Sherazy, M.S.; Gelenberg, A.J. Omega-3 and omega-6 polyunsaturated fatty acids in bipolar disorder: A review of biomarker and treatment studies. J. Clin. Psychiatry 2016, 77, 1301–1308. [Google Scholar] [CrossRef] [PubMed]
  122. Turnbull, T.; Cullen-Drill, M.; Smaldone, A. Efficacy of omega-3 fatty acid supplementation on improvement of bipolar symptoms: A systematic review. Arch. Psychiatry Nurs. 2008, 22, 305–311. [Google Scholar] [CrossRef] [PubMed]
  123. Montgomery, P.; Richardson, A.J. Omega-3 fatty acids for bipolar disorder. Cochrane Database Syst. Rev. 2008, 2, CD005169. [Google Scholar] [CrossRef] [PubMed]
  124. Kraguljac, N.V.; Montori, V.M.; Pavuluri, M.; Chai, H.S.; Wilson, B.S.; Unal, S.S. Efficacy of omega-3 fatty acids in mood disorders—A systematic review and metaanalysis. Psychopharmacol. Bull. 2009, 42, 39–54. [Google Scholar] [PubMed]
  125. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Jacka, F.N.; Ystrom, E.; Brantsaeter, A.L.; Karevold, E.; Roth, C.; Haugen, M.; Meltzer, H.M.; Schjolberg, S.; Berk, M. Maternal and early postnatal nutrition and mental health of offspring by age 5 years: A prospective cohort study. J. Am. Acad. Child Adolesc. Psychiatry 2013, 52, 1038–1047. [Google Scholar] [CrossRef] [PubMed]
  127. Verly-Miguel, M.V.; Farias, D.R.; Pinto Tde, J.; Lepsch, J.; Nardi, A.E.; Kac, G. Serum docosahexaenoic acid (DHA) is inversely associated with anxiety disorders in early pregnancy. J. Anxiety Disord. 2015, 30, 34–40. [Google Scholar] [CrossRef] [PubMed]
  128. Dos Santos, L.F.; Loureiro, S.R.; Crippa, J.A.; de Lima Osório, F. Adaptation and initial psychometric study of the self-report version of Liebowitz Social Anxiety Scale (LSAS-SR). Int. J. Psychiatry Clin. Pract. 2013, 17, 139–143. [Google Scholar] [CrossRef]
  129. Green, P.; Hermesh, H.; Monselise, A.; Marom, S. Red cell membrane omega-3 fatty acids are decreased in non depressed patients with social anxiety disorder. Eur. Neuropsychopharmacol. 2006, 16, 107–113. [Google Scholar] [CrossRef]
  130. Buydens-Branchey, L.; Branchey, M. n-3 polyunsaturated fatty acids decrease anxiety feelings in a population of substance abusers. J. Clin. Psychopharmacol. 2006, 26, 661–665. [Google Scholar] [CrossRef]
  131. Buydens-Branchey, L.; Branchey, M.; Hibbeln, J.R. Associations between increases in plasma n-3 polyunsaturated fatty acids following supplementation and decreases in anger and anxiety in substance abusers. Prog. Neuropsychopharmacol. Biol. Psychiatry 2008, 32, 568–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Okuyama, J.; Funakoshi, S.; Tomita, H.; Yamaguchi, T.; Matsuoka, H. School-Based Interventions Aimed at the Prevention and Treatment of Adolescents Affected by the 2011 Great East Japan Earthquake: A Three-Year Longitudinal Study. Tohoku J. Exp. Med. 2017, 42, 203–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zanarini, M.C.; Frankenburg, F.R. Omega-3 fatty acid treatment of women with borderline personality disorder: A double blind, placebo-controlled pilot study. Am. J. Psychiatry 2003, 160, 167–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Hallahan, B.; Hibblen, J.R.; Davi, J.M.; Garland, M.R. Omega-3 fatty acids supplementation in patients with recurrent self-harm: Single center double bind randomized controlled trial. Br. J. Psychiatry 2007, 190, 118–122. [Google Scholar] [CrossRef] [Green Version]
  135. Ginty, A.T.; Muldoon, M.F.; Kuan, D.C.H. Omega-3 Supplementation and the Neural Correlates of Negative Affect and Impulsivity: A Double-Blind, Randomized, Placebo-Controlled Trial in Midlife Adults. Psychosom. Med. 2017, 79, 549–556. [Google Scholar] [CrossRef]
  136. DeMar, J.C., Jr.; Ma, K.; Bell, J.M.; Igarashi, M. One generation of n-3 polyunsaturated fatty acid deprivation increases depression and aggression test scores in rats. J. Lipid Res. 2006, 47, 172–180. [Google Scholar] [CrossRef] [Green Version]
  137. Raygada, M.; Cho, E.; Hilakivi-Clarke, L. High maternal intake of polyunsaturated fatty acids during pregnancy in mice alters offsprings’ aggressive behavior, immobility in the swim test, locomotor activity and brain protein kinase C activity. J. Nutr. 1998, 128, 2505–2511. [Google Scholar] [CrossRef]
  138. Bègue, L.; Zaalberg, A.; Shankland, R.; Duke, A.; Jacquet, J.; Kaliman, P.; Pennel, L.; Chanove, M.; Arvers, P.; Bushman, B.J. Omega-3 supplements reduce self-reported physical aggression in healthy adults. Psychiatry Res. 2018, 261, 307–311. [Google Scholar] [CrossRef] [PubMed]
  139. Meyer, B.J.; Byrne, M.K.; Collier, C.; Parletta, N.; Crawford, D.; Winberg, P.C.; Webster, D.; Chapman, K.; Thomas, G.; Dally, J.; et al. Baseline omega-3 index correlates with aggressive and attention deficit disorder behaviours in adult prisoners. PLoS ONE 2018, 13, e0197231. [Google Scholar] [CrossRef]
  140. Gajos, J.M.; Beaver, K.M. The effect of omega-3 fatty acids on aggression: A meta-analysis. Neurosci. Biobehav. Rev. 2016, 69, 147–158. [Google Scholar] [CrossRef]
  141. Patrick, R.P.; Ames, B.N. Vitamin D and the omega-3 fatty acids control serotonin synthesis and action, part 2: Relevance for ADHD, bipolar disorder, schizophrenia, and impulsive behavior. FASEB J. 2015, 29, 2207–2222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Lotrich, F.E. Inflammatory cytokine-associated depression. Brain Res. 2015, 1617, 113–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Beier, A.M.; Lauritzen, L.; Galfalvy, H.C.; Cooper, T.B.; Oquendo, M.A.; Grunebaum, M.F.; Mann, J.J.; Sublette, M.E. Low plasma eicosapentaenoic acid levels are associated with elevated trait aggression and impulsivity in major depressive disorder with a history of comorbid substance use disorder. J. Psychiatry Res. 2014, 57, 133–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Sanchez-Paez, P.; Perez-Templado, J.; Saiz-Ruiz, J.; Pastor, O.; Ibanez, A. Essential fatty acids and Barratt impulsivity in gambling disorder. BMC Psychiatry 2020, 20, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Buydens-Branchey, L.; Branchey, M.; McMakin, D.L.; Hibbeln, J.R. Polyunsaturated fatty acid status and aggression in cocaine addicts. Drug Alcohol. Depend. 2003, 71, 319–323. [Google Scholar] [CrossRef]
  146. Buydens-Branchey, L.; Branchey, M.; McMakin, D.L.; Hibbeln, J.R. Polyunsaturated fatty acid status and relapse vulnerability in cocaine addicts. Psychiatry Res. 2003, 120, 29–35. [Google Scholar] [CrossRef]
  147. Virkkunen, M.E.; Horrobin, D.F.; Jenkins, D.K.; Manku, M.S. Plasma phospholipid essential fatty acids and prostaglandins in alcoholic, habitually violent, and impulsive offenders. Biol. Psychiatry 1987, 22, 1087–1096. [Google Scholar] [CrossRef]
  148. Choy, O.; Raine, A. Omega-3 Supplementation as a Dietary Intervention to Reduce Aggressive and Antisocial Behavior. Curr. Psychiatry Rep. 2018, 20, 32. [Google Scholar] [CrossRef]
  149. Kidd, P.M. Omega-3 DHA and EPA for cognition, behavior, and mood: Clinical findings and structural-functional synergies with cell membrane phospholipids. Altern. Med. Rev. 2007, 12, 207–227. [Google Scholar]
  150. Yang, Y.; Raine, A. Prefrontal structural and functional brain imaging findings in antisocial, violent, and psychopathic individuals: A meta-analysis. Psychiatry Res. 2009, 174, 81–88. [Google Scholar] [CrossRef] [Green Version]
  151. Sinn, N.; Bryan, J.; Wilson, C. Cognitive effects of polyunsaturated fatty acids in children with attention deficit hyperactivity disorder symptoms: A randomised controlled trial. Prostaglandins Leukort. Essent. Fat. Acids 2008, 78, 311–326. [Google Scholar] [CrossRef] [PubMed]
  152. Perera, H.; Jeewandara, K.C.; Seneviratne, S.; Guruge, C. Combined n-3 and n-6 supplementation in children with attention-deficit hyperactivity disorder (ADHD) refractory to methylphenidate treatment. J. Child. Neurol. 2012, 27, 747–753. [Google Scholar] [CrossRef] [PubMed]
  153. Bellino, S.; Bozzatello, P.; Rocca, G.; Bogetto, F. Efficacy of omega-3fatty acids in the treatment of borderline personality disorder: A study of the association with valproic acid. J. Psychopharmacol. 2014, 28, 125–132. [Google Scholar] [CrossRef] [PubMed]
  154. Bozzatello, P.; Rocca, P.; Bellino, S. Combination of omega-3 fatty acids and valproic acid in treatment of borderline personality disorder: A follow-up study. Clin. Drug Investig. 2018, 38, 367–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Skegg, K. Self-harm. Lancet 2005, 366, 1471–1483. [Google Scholar] [CrossRef]
  156. Lenkiewicz, K.; Racicka, E.; Bryńska, A. Self-injury - placement in mental disorders classifications, risk factors and primary mechanisms. Review of the literature. Psychiatry Pol. 2017, 51, 323–334. [Google Scholar] [CrossRef] [PubMed]
  157. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; American Psychiatric Association: Arlington, VA, USA, 2013. [Google Scholar]
  158. Daray, F.M.; Mann, J.J.; Sublette, M.E. How lipids may affect risk for suicidal behavior. J. Psychiatr. Res. 2018, 104, 16–23. [Google Scholar] [CrossRef] [Green Version]
  159. Hibbeln, J.R. Depression, suicide and deficiencies of omega-3 essential fatty acids in modern diets. World Rev. Nutr. Diet. 2009, 99, 17–30. [Google Scholar] [CrossRef]
  160. Marriott, B.P.; Hibbeln, J.R.; Killeen, T.K.; Magruder, K.M.; Holes-Lewis, K.; Tolliver, B.K.; Turner, T.H. BRAVO Group Design and methods for the Better Resiliency among Veterans and non-Veterans with Omega-3′s (BRAVO) study: A double blind, placebo-controlled trial of omega-3 fatty acid supplementation among adult individuals at risk of suicide. Contemp. Clin. Trials 2016, 47, 325–333. [Google Scholar] [CrossRef]
  161. Lewis, M.D.; Hibbeln, J.R.; Johnson, J.E.; Lin, Y.H. Suicide deaths of active-duty US military and omega-3 fatty-acid status: A case-control comparison. J. Clin. Psychiatry 2011, 72, 1585–1590. [Google Scholar] [CrossRef] [Green Version]
  162. Sublette, M.E.; Hibbeln, J.R.; Galfalvy, H.; Oquendo, M.A.; Mann, J.J. Omega-3 polyunsaturated essential fatty acid status as a predictor of future suicide risk. Am. J. Psychiatry 2006, 163, 1100–1102. [Google Scholar] [CrossRef] [PubMed]
  163. Tanskanen, A.; Hibbeln, J.R.; Hintikka, J.; Haatainen, K. Fish consumption, depression, and suicidality in a general population. Arch. Gen. Psychiatry 2001, 58, 512–513. [Google Scholar] [CrossRef] [PubMed]
  164. Vaz, J.S.; Kac, G.; Nardi, A.E.; Hibbeln, J.R. Omega-6 fatty acids and greater likelihood of suicide risk and major depression in early pregnancy. J. Affect. Disord. 2014, 152–154, 76–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Gallagher, A.T.; Byrne, F.; Murray, I.T.; Lally, J.; Davis, J.M. Lipids and polyunsaturated fatty acid levels in deliberate self-harm: A 10-year follow-up study. Ir. J. Psychol. Med. 2017, 34, 117–126. [Google Scholar] [CrossRef]
  166. Kurosawa, K.; Terao, T.; Kanehisa, M.; Shiotsuki, I.; Ishii, N.; Takenaka, R.; Sakamoto, T.; Matsukawa, T.; Yokoyama, K.; Ando, S.; et al. Naturally absorbed polyunsaturated fatty acids, lithium, and suicide-related behaviors: A case-controlled study. J. Affect. Disord. 2018, 241, 200–205. [Google Scholar] [CrossRef]
  167. Berger, G. Comments on Bozzatello et al. Supplementation with Omega-3 Fatty Acids in Psychiatric Disorders: A Review of Literature Data. J. Clin. Med. 2016, 5, 69. [Google Scholar] [CrossRef] [Green Version]
Table 1. Randomized controlled trials and open trials of omega-3 PUFAs on psychotic symptoms.
Table 1. Randomized controlled trials and open trials of omega-3 PUFAs on psychotic symptoms.
Study (Year) [Ref.]Interventional Arm(s)Comparison Arm(s)SampleTreatment DurationResults
Amminger et al., 2013 [40]EPA
700 mg/day + DHA
480 mg/day +
7.6 mg vitamin E
placebo81 young
individuals at
UHR
12 weeks↓ positive symptoms, negative symptoms and
general symptoms,
↑ level of functioning
Amminger et al., 2015 [42]EPA
700 mg/day + DHA
480 mg/day
placebo81 young
individuals at
UHR
6.7 years follow-up↓ risk of progression to psychotic disorder and psychiatric morbidity
Amminger et al., 2010 [43]EPA
700 mg/day +
DHA
480 mg/day
Placebo81 individuals at UHR12 weeks↓ progression in psychosis in young UHR
patients
Smesny et al.,
2014 [44]
EPA
700 mg/day +
DHA
480 mg/day
Placebo81 young
individuals at
UHR
12 weeksnormalizing PLA2 activity and
d-6-desaturase-mediated metabolism of o-3 and
o-6 PUFAs
Berger et al.,
2007 [45]
Ethyl-EPA
3 g/day +
antipsychotics
Placebo +
antipsychotics
69 patients at FEP12 weeksaccelerated treatment response
Berger et al., 2008 [46]EPA 2 g/day +
Antipsychotics
Placebo +
antipsychotics
24 patients at FEP12 weeks↓ of negative symptoms
Wood et al., 2010 [47]Ethyl-EPA
2 g/day +
antipsychotics
Placebo +
antipsychotics
17 patients at FEP12 weeksincreased water in hippocampal tissues and
positive effect on negative symptoms
Emsley et al., 2014 [48]EPA 2 g/day + DHA 1
g/day + α-LA 300 mg/day
placebo33 patients after FEP on antipsychotic discontinuation2 yearsrelapse prevention of psychotic symptoms
Pawelzcyk et al., 2016 [49]EPA + DHA
2.2 g/day
Placebo 71 patients at FEP26 weeks↓ psychotic symptoms measured with PANSS
↓ depressive symptoms
↑ level of functioning
Pawelzcyk et al., 2017 [50]EPA + DHA
2.2 g/day
placebo71 patients at FEP26 weeksimproved PANSS negative and general
symptoms, along with global functioning
Peet et al.,2001 [52]EPA or DHA
2 g/day +
antipsychotics
Placebo +
Antipsychotics
45 patients with stable schizophrenia12 weeks↓ psychotic symptoms measured with PANSS in
the group treated with EPA
Peet et al., 2002 [53]Ethyl-EPA
1–4 g/day +
antipsychotics
Placebo +
antipsychotics
115 patients with stable schizophrenia12 weeks↓ positive symptoms measured with PANSS,
↓ depressive symptoms
Emsley et al.,
2002 [54]
ethyl-EPA
3 g/day +
antipsychotics
Placebo +
antipsychotics
40 patients with stable schizophrenia12 weeks↓ positive symptoms
and negative symptoms
measured with PANSS
Jamilian et al., 2014 [55]ethyl-EPA
1 g/day +
antipsychotics
Placebo +
antipsychotics
60 patients with stable schizophrenia8 weeks↓ psychotic symptoms measured with PANSS
Bentsen et al., 2013 [56]ethyl-EPA
2 g/day +
antipsychotics
Placebo +
antipsychotics
99 patients with stable schizophrenia16 weeks ↓ impairment of the course of psychosis
Robinson et al., 2019 [57]EPA 740 mg and DHA
400 mg/day +
antipsychotics
Placebo+
Antipsychotics

50 patients with stable schizophrenia16 weeks↓ confusion, anxiety, depression, irritability and
tiredness/fatigue
Emsley et al., 2006 [59]ethyl-EPA
2 g/day +
antipsychotics
Placebo + antipsychotics77 patients with stable schizophrenia12 weeksno efficacy on specific psychotic symptoms
Fenton et al., 2001 [60]ethyl-EPA
3 g/day +
antipsychotics
Placebo +
antipsychotics
87 patients with stable schizophrenia16 weeksno significant differences in positive, negative
symptoms, mood or cognition
Table 2. Randomized controlled trials and open trials of omega-3 PUFAs on affective symptoms.
Table 2. Randomized controlled trials and open trials of omega-3 PUFAs on affective symptoms.
Study (Year) [Ref.]Interventional Arm(s)Comparison Arm(s)SampleTreatment Duration Results
Pawelzcyk et al., 2018 [51]EPA + DHA
2.2 g/day
placebo71 patients at FEP26 weeks↑ level of telomerase in peripheral blood cells
with ↓ depressive symptoms
Llorente et al., 2003 [79]DHA 0.2 g/day monotherapyPlacebo99 healthy pregnant
women
16 weeksno effect on postpartum depression
Marangell et al., 2003 [80]add on to standard therapy DHA 2 g/day monotherapyStandard therapy36 patients with MDD12 weeksno significant differences
Silvers et al., 2005 [81]EPA 0.6 g/day + DHA 2.4 g/day
added to standard therapy
Standard therapy77 patients with MDD12 weeksno evidence that n-3PUFAs improved mood
compared to placebo.
Mood improved in both groups within the first 2 weeks
of the study
Greyner et al., 2007 [82]EPA 0.6 g/day + DHA 2.2 g/day add
to standard therapy
Standard therapy83 patients with MDD16 weeksno significant differences
Hallahan et al., 2007 [134]EPA (1.2 g/day) + DHA (0.9 g/day)Placebo49 patients with self-harm behaviors (35 BPD)12 weeksImprovement of depression, suicidality and reaction to daily stress
Freeman et al., 2008 [83]EPA 1.1 g/day + DHA 0.8 g/dayplacebo59 women8 weeksno benefit on perinatal depressive symptoms
Jazayeri et al.,
2008 [84]
EPA 1 g/day fluoxetine 20 mg/day60 patients with MDD8 weeks↓ depressive symptoms in both groups
Rees et al., 2008 [85]ethyl-EPA 0.4 g/day + DHA 1.6 g/dayplacebo26 pregnant patients6 weeksno benefits on depressive symptoms
Rogers et al., 2008 [86]EPA 0.63 g/day + DHA 0.85 g/day
monotherapy
placebo218 mild to moderate depressed patients untreated12 weeksn-3PUFAs not have beneficial or harmful effects on mood in mild to moderate depression.
Doornbos et al., 2009 [87]DHA 0.22 g/day or DHA 0.22 g/day +
AA (0.22 g/day arachidonic acid)
monotherapy
placebo119 healthy pregnant women28 weeksred blood cell DHA, AA and DHA/AA ratio did not correlate with EPDS or blues scores
Lucas et al., 2009 [88]EPA 1.05 g/day + DHA 0.25 g/day
mono-therapy
placebo120 patients with psychological distress
with or without MDD in comorbidity
8 weeksno significant differences
Makrides et al., 2010 [89]DHA-rich tuna oil capsules 0.5 g/day
monotherapy
placebo2399 healthy pregnant
women at 21 weeks’
gestation
women received assigned capsules daily, from study entry until birth of their childDHA during pregnancy did not lower levels of
postpartum depression
Antypa et al., 2012 [90]EPA 1.74 g/day+ DHA 0.25 g/day
added to standard therapy
Standard therapy71 patients with history of at least one MDD4 weeksno significant effects on memory, attention,
cognitive reactivity and depressive symptoms
Mozurkewich et al., 2013 [91]EPA 1.06 g/day+ DHA 0.27 g/day monotherapyEPA 0.18 g/day + DHA 0.9 g/day
monotherapy
126 healthy pregnant women6–8 weeksno differences between groups in BDI scores or
other depression endpoints
Mischoulon et al., 2009 [92]EPA 1 g/day + (+ 0.2% dL
alphatocopherol) monotherapy
placebo57 patients with MDD8 weeks↓ depressive symptoms assessed with HDRS, but no statistical significance
Park et al., 2015 [93]EPA 1140 g/day + DHA 0.6 g/day add to standard therapyStandard therapy35 patients with MDD12 weeksno significant differences
Young et al., 2017 [94]PEP + EPA 1.4 g/day + DHA 0.2 g/day + 0.4 g/day otherplacebo72 depressed patients 7–14 years old12 weeks↓ co-occurring behavior symptoms in youth with depression.
Gabbay et al., 2018 [95]2:1 ratio of EPA to DHA: Initial dose
of 1.2 g/day. Doses were raised in
increments of 0.6 g/day every 2 weeks
(maximum possible dose of
3.6 g/day, combined EPA 2.4 g + DHA 1.2 g)
placebo51 psychotropic medication-free adolescents with MDD aged 12–19 years old10 weeksn-3PUFAs do not appear to be superior to placebo.
Tayama et al., 2019 [96]DHA 500 mg/day + EPA 1000 mg/dayplacebo20 patients with mild-to-moderate depression12 weeksno significant differences
Nemets et al., 2006 [97]ethyl-EPA 0.4 g/day + DHA 0.2 g/dayplacebo20 patients 6–12 years-old16 weeks↓ depressive symptoms measured with CDRS, CDI and CGI
Peet & Horrobin 2002 [98]Ethyl-EPA 1/2/4 g/day + Standard therapyPlacebo + Standard therapy 70 patients with persistent depression despite ongoing treatment 12 weeksEthyl-EPA 1 g/day group > placebo group
no significant differences in the Ethyl-EPA 2 and 4 g/day groups
Su et al., 2003 [99]ethyl-EPA
4.4 g/day + DHA 2.2 g/day
add-on existing antidepressant
treatment
Placebo + existing antidepressant
treatment
22 patients with MDD8 weeks↓ depressive symptoms
measured with HDRS
Nemets et al., 2006 [100]ethyl-EPA
0.4 g/day + DHA 0.2 g/day
Placebo20 depressed patients 6–12 years-old16 weeks↓ depressive symptoms measured with CDRS, CDI and CGI
Mischoulon et al. (2009) [101]EPA 1 g/day + (+0.2% dL
alphatocopherol) monotherapy
Placebo57 MDD patients8 weeks↓ depressive symptoms assessed with HDRS, but no statistical significance
Rondanelli et al., 2010,2011 [102,103]EPA 1.67 g/day + DHA 0.83 g/day added to existing antidepressant treatmentPlacebo +
existing antidepressant
treatment
46 elderly female residents in a nursing
home
8 weeks↓ depressive symptoms assessed with GDS,
improvement of phospholipids fatty acids
profile
Lespérance et al., 2011
[104]
EPA 1.05 g/day + DHA 0.15 g/day + existing antidepressant
treatment
Placebo + existing antidepressant treatment432 patients with a major depressive episode8 weeks↓ depressive symptoms only for patients without comorbid anxiety disorders
Tajalizadekhoob et al., 2011 [105]EPA 0.18 g/day + DHA 0.12 g/day
add to standard therapy (55 patients)
or in monotherapy (11 patients)
Placebo66 patients with mild-to moderate depression aged > 66 years24 weekslow-dose n-3PUFAs have some efficacy in mild to moderate depression
Gertsik et al., 2012 [106]EPA 0.9 g/day + DHA 0.2 g/day + other n-3 PUFAs (0.1 g/day) added to
citalopram
Placebo added to citalopram42 MDD patients taking citalopram9 weekssignificantly greater improvement in HDRS
scores
Krawczyk et al., 2012 [107]EPA 2.2 g/day + DHA 0.7 g/day +
GLA (0.24 g/day) + vit. E added to
standard therapy
Standard therapy21 patients with severe episode of treatment resistant recurring depression8 weeksn-3PUFAs significantly improved HDRS scores
Rizzo et al., 2012
[108]
EPA/DHA 2.1/2.5 g of n3-PUFA
monotherapy
Placebo46 MMD patients (only women > 66 years old)8 weeksmean GDS score and AA/EPA ratio, in whole
blood and RBC membrane phospholipids, were
significantly lower
Mozaffari-Khosravi et al., 2012
[109]
EPA 1 g/day or DHA 1 g/day added
to standard therapy
Placebo + Standard therapy81 mild to moderate
depressed patients
12 weeks↓ HDRS score EPA > compared with those in the DHA or placebo groups
Judge et al., 2014
[110]
DHA 0.3 g/dayPlacebo42 healthy pregnant
women
8 weeks↓ depressive symptoms assessed with PDSS
Ginty et al., 2015
[111]
EPA + DHA 1.4 g/day monotherapyPlacebo23 depressed patients3 weeksn-3PUFAs group had a significant reduction in
BDI scores over time
Jahangard et al., 2018
[112]
n-3 PUFAs (1000 mg/day) + sertraline
(50–200 mg/day)
sertraline
(50–200 mg/day)
50 MDD outpatients12 weeks↓ depression, anxiety, sleep and patients’
competencies to regulate their emotions.
Chiu et al., 2005
[113]
4.4 g/day
EPA + 2.4 g/day DHA added on valproate 2 g/day
valproate 2 g/day and16 newly hospitalized
patients in the acute
manic phase of bipolar
disorder
4 weeksNo significant differences
Hirashima
et al.,
2004
[115]
High dose: EPA, 5.0–5.2 g/day;
DHA, 3.0–3.4 g/day; other, 0.3–1.7
g/day + standard therapy
Standard therapy21 patients with bipolar disorder4 weeksNo significant differences
Keck et al., 2006
[116]
EPA 6 g/day in addition to at least
one mood stabilizer
at least
one mood stabilizer
121 patients with bipolar
depression or rapid
cycling bipolar disorder
4 monthsNo significant differences
Frangou et al., 2006
[117]
ethyl-EPA 1 or 2 g/day added to
stable psychotropic medications
stable psychotropic medications75 patients with bipolar disorder12 weeks↓ depressive symptoms measured with HDRS
Murphy et al., 2012
[118]
omega-3 fatty acids plus cytidine or
omega-3 fatty acid plus placebo in addition to a
mood stabilizer
only placebo in addition to a
mood stabilizer
45 patients with type I
bipolar disorder
4 monthsno benefits of omega-3 fatty acids on affective
symptoms
Stoll et al., 1999
[119]
EPA 6.2 g/day + DHA 3.4 g/dayPlacebo30 patients with bipolar disorder16 weeks↓ depressive symptoms measured with HDRS
Gracious et al., 2010
[120]
ALA in addition to psychotropic
medication
Standard therapychildren and adolescent
with bipolar I or II
disorder
16 weekssignificant improvement of overall symptom severity compared with placebo
Zanarini & Franknburg, 2003
[133]
Ethyl-EPA 1 g/day (with no standard
psychiatric therapies)
placebo30 females with BPD8 weeks↓ depression
Buydens-Branchey et al., 2006, 2008
[130,131]
eicosapentaenoic acid + docosahexaenoic acid 3 g/dayPlacebo44 patients with anxiety disorder and substance abuse disorder3 months + 3 months after therapy discontinuation↓ anxiety symptoms > in PUFAs group than in placebo one, also after therapy discontinuation
Table 3. Randomized controlled trials and open trials of omega-3 PUFAs on impulsivity and aggression.
Table 3. Randomized controlled trials and open trials of omega-3 PUFAs on impulsivity and aggression.
Study (Year) [Ref.]Interventional Arm(s)Comparison Arm(s)SampleTreatment Duration Results
Qiao et al., 2018 [61]DHA 360 mg/day + EPA
540 mg/day +
antipsychotics
Placebo +
antipsychotics
50 patients with stable schizophrenia12 weeks↓ violence, but no improvement in positive and
negative symptoms
Zanarini & Franknburg, 2003
[133]
Ethyl-EPA 1 g/day (with no standard
psychiatric therapies)
placebo30 females with BPD8 weeks↓ aggression
Ginty et al., 2017
[135]
EPA 1 g/day + DHA 0.4 g/dayPlacebo272 healthy volunteers18 weeksNo significative differences on impulsive behaviors nor on corticolimbic and corticostriatal brain functionality
Bègue et al.,
2017
[150]
EPA 0.772 g/day +
DHA 0.638 g/day
Placebo194 Participants aged 18–45 from the general population6 weeksOmega-3 supplements significantly decreased self-reported aggressiveness
Sinn and Bryan,
2008
[151]
EPA 93 mg/day + DHA 29
mg/day +
gamma-linolenic acid
10 mg/day versus placebo.
No ADHD medications
Placebo132 children
(7 to 12 years)
with ADHD
15 weeksimproved in inattention, hyperactivity and
impulsivity in most ADHD scales in parents reports; no improvement in teachers reports Limits: No ADHD diagnosis (reported ADHD symptoms)
Perera et al., 2012
[152]
omega-3 + omega-6 with ADHD medicationsPlacebo with ADHD medications98 children
(6 to 12 years)
with ADHD
diagnosis
6 monthsimproved behavior and learning in restlessness,
aggressiveness, completing work and academic
performance, but not in inattention,
impulsiveness and cooperation with parents and
teachers
Bellino et al., 2014 [153]EPA (1.2 g/day) + DHA (0.8 g/day) + valproate (800–1300 mg/day)Valproate (800–1300 mg/day)43 BPD outpatients12 weeksNo differences with regard to global symptoms. Improvement of impulsivity, anger and self-mutilating conducts in omega-3 group
Bozzatello et al., 2018 [154]EPA (1.2 g/day) + DHA (0.8 g/day) + valproate (800–1300 mg/day)Valproate (800–1300 mg/day)43 BPD outpatients24 weeks follow-upCombined therapy with omega-3 fatty acids showed long-lasting effects after discontinuation in terms of anger control.
Table 4. Randomized controlled trials and open trials of omega-3 PUFAs on self-harm behaviors.
Table 4. Randomized controlled trials and open trials of omega-3 PUFAs on self-harm behaviors.
Study (Year) [Ref.]Interventional Arm(s)Comparison Arm(s)SampleTreatment Duration Results
Hallahan et al., 2007 [134]EPA (1.2 g/day) + DHA (0.9 g/day)Placebo49 patients with self-harm behaviors (35 BPD)12 weeksImprovement of self-harm behaviors, suicidality, and reaction to daily stress
Bellino et al., 2014 [153]EPA (1.2 g/day) + DHA (0.8 g/day) + valproate (800–1300 mg/day)Valproate (800–1300 mg/day)43 BPD outpatients12 weeksNo differences with regard to global symptoms. Improvement of impulsivity, anger and self-mutilating conducts in omega-3 group
Marriott et al., 2016
[160]
3300 mg of n-3 PUFAs/day with 1650 mg EPA plus 1650 mg DHA per day Placebo40 United States (U.S.) military Veterans and non-Veterans ages 18–90 years with suicide risk6 months↓ suicidal behaviors
Gallagher et al., 2017
[165]
n-3 PUFAs including
EPA or DHA
Placebo40 individuals who
self-harmed and 40
controls
10 years↓ self-harm, ↓depressive symptoms and
↓ impulsivity

Share and Cite

MDPI and ACS Style

Bozzatello, P.; De Rosa, M.L.; Rocca, P.; Bellino, S. Effects of Omega 3 Fatty Acids on Main Dimensions of Psychopathology. Int. J. Mol. Sci. 2020, 21, 6042. https://doi.org/10.3390/ijms21176042

AMA Style

Bozzatello P, De Rosa ML, Rocca P, Bellino S. Effects of Omega 3 Fatty Acids on Main Dimensions of Psychopathology. International Journal of Molecular Sciences. 2020; 21(17):6042. https://doi.org/10.3390/ijms21176042

Chicago/Turabian Style

Bozzatello, Paola, Maria Laura De Rosa, Paola Rocca, and Silvio Bellino. 2020. "Effects of Omega 3 Fatty Acids on Main Dimensions of Psychopathology" International Journal of Molecular Sciences 21, no. 17: 6042. https://doi.org/10.3390/ijms21176042

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop