Next Article in Journal
Effect of Probiotics in Breast Cancer: A Systematic Review and Meta-Analysis
Previous Article in Journal
Defense System of the Manila Clam Ruditapes philippinarum under High-Temperature and Hydrogen Sulfide Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unsaturated Fatty Acids and Their Immunomodulatory Properties

Department of Biological Sciences, Kean University, Union, NJ 07083, USA
*
Author to whom correspondence should be addressed.
Biology 2023, 12(2), 279; https://doi.org/10.3390/biology12020279
Submission received: 6 January 2023 / Revised: 7 February 2023 / Accepted: 7 February 2023 / Published: 9 February 2023
(This article belongs to the Section Immunology)

Abstract

:

Simple Summary

Diet can influence human health in both positive and negative ways. Ω-3 polyunsaturated fatty acids have an overall positive effect on humans. The sources of these fatty acids are primarily plant seeds and fish. Consumption of ω-3 fatty acids reduces inflammation and markers associated with certain diseases. Reduction in inflammation occurs through metabolites of ω-3 fatty acids and biophysical and biochemical changes in plasma membrane properties. In general, a diet high in ω-3 and low in ω-6 fats is considered favorable. This can be achieved by increasing fish and vegetable consumption while reducing animal fats in our diet.

Abstract

Oils are an essential part of the human diet and are primarily derived from plant (or sometimes fish) sources. Several of them exhibit anti-inflammatory properties. Specific diets, such as Mediterranean diet, that are high in ω-3 polyunsaturated fatty acids (PUFAs) and ω-9 monounsaturated fatty acids (MUFAs) have even been shown to exert an overall positive impact on human health. One of the most widely used supplements in the developed world is fish oil, which contains high amounts of PUFAs docosahexaenoic and eicosapentaenoic acid. This review is focused on the natural sources of various polyunsaturated and monounsaturated fatty acids in the human diet, and their role as precursor molecules in immune signaling pathways. Consideration is also given to their role in CNS immunity. Recent findings from clinical trials utilizing various fatty acids or diets high in specific fatty acids are reviewed, along with the mechanisms through which fatty acids exert their anti-inflammatory properties. An overall understanding of diversity of polyunsaturated fatty acids and their role in several molecular signaling pathways is useful in formulating diets that reduce inflammation and increase longevity.

1. Fatty Acids in Living Systems

Fats and oils are storage lipids used across almost all living organisms [1]. Per a suggested daily ration from the Dietary Guidelines for Americans, total fat should be limited to 20–35% of daily calories [2]. Fats and oils are derivatives of fatty acids, which are carboxylic acids with hydrocarbon chains ranging in length from four to 36 carbons (Figure 1). Fatty acids (FAs) are essential components not only of fats, but also membrane lipids—phospholipids, and sphingolipids—with the latter being abundant in neural tissues [1]. In saturated fatty acids, the hydrocarbon chains contain no double bonds, and in unsaturated fatty acids they contain one or more double bonds. Simple fatty acids’ nomenclature states the number of carbons in the chain and the number of bonds after a colon; the position of the bonds is designated by Δ and a number of carbons relative to the carboxylic carbon (=α-carbon). Polyunsaturated fatty acids (PUFAs), with a double bond between a third or fourth carbon or between sixth and seventh carbon from the methyl end of the chain (=ω-carbon), are especially important for human health, and are alternatively called ω-3 and ω-6 fatty acids to reflect the fact that the physiological role of these fatty acids is more related to the position of the double bond counting from the methyl end (Figure 1).
In order to produce necessary polyunsaturated fatty acids, humans require the ω-3 PUFA α-linolenic acid (ALA, 18:3Δ9,12,15) but lack enzymatic ability to synthesize it, and thus must obtain it from plant sources. From this essential fatty acid, humans can then synthesize more PUFAs, such as eicosapentaenoic acid (EPA, 20:5Δ5, 8, 11,14,17 or ω-3 group) and docosahexaenoic acid (DHA, 22:6Δ4,7,10,13,16,19, also ω-3 group) [1]. EPA and DHA can be also sourced from fish and seafood [3].

2. Biosynthesis of Essential Fatty Acids in Plants

Biosynthesis of fatty acids in plants is located in plastids, the photosynthetic organelles of plant cells. Fatty acids are then subsequently utilized as components of plastid and endoplasmic reticulum membrane phospholipids, storage lipids, or as extracellular waxes [4]. Major plastid lipids are first synthesized using 16:0 and 18:1 acyl groups, and additional double bonds are later added by fatty acid desaturases (FADs). Desaturases insert the double bonds into fatty acid hydrocarbon chains to moderate the fluidity of lipids and corresponding membranes [5]. Membranes with phospholipids that contain saturated fatty acids are more rigid, which creates physiological issues when the membrane solidifies as a result of cold action; unsaturated fatty acids make phospholipids more flexible and allow them to resist cold stress [6,7]. Accumulation of unsaturated fatty acids such as ALA in plant membranes is a common abiotic stress response that leads to an increase in membrane fluidity and resistance to membrane rigidification caused by chilling. In addition to the moderation of membrane fluidity, C18 PUFAs act as intrinsic antioxidants. The double bonds in unsaturated fatty acids make them susceptible to reactive oxygen species (ROS) that are usually produced as a result of stress. Overexpression of ω-3 fatty acid desaturases is a method of general defense in plants, to induce stress response. In the case of an extreme increase in free radicals (which actually could be a result of photosynthesis), C18 PUFAs suffer from peroxidation, resulting in accumulation of malondialdehyde (MDA), which at low levels plays a signaling role, facilitating stress perception; however, in instances of excessive peroxidation of PUFAs, high levels of MDA and ROS may result in a massive oxidative catastrophe and DNA damage. Thus, plants maintain an accurate unsaturated fatty acid and reactive species homeostasis [8,9].

Unsaturated Fatty Acids Synthesis

Oleic (OA, 18:1Δ9), linoleic (LA, 18:2Δ9,12), and α-linolenic (ALA, 18: 3Δ9,12,15) acids are key unsaturated fatty acids synthesized in plants [10]. They participate in stress responses and are precursors for plant hormones. FA synthesis is regulated by central phytohormones, abscisic acid, auxin and jasmonic acid, which organize plant growth, development and defense [11]. Hundreds of plant fatty acids, their structures and the related literature can be found using the PlantFAdb online resource [12].
Oleic acid is synthesized de novo in plastids from acetyl-coA; first, a saturated stearic acid (18:0) is produced by a fatty acid synthase (FAS) and acetyl-coA carboxylase, then stearoyl-ACP desaturase introduces a first double bond in the 9th position [10] (Figure 2). Humans can endogenously synthesize this acid and thus it is not essential [13]. As biosynthesis of C18 PUFAs in plants is coupled with synthesis of membrane lipids, oleic acid is then incorporated into a phospholipid, such as phosphatidic acid or phosphatidylcholine, for the subsequent desaturation either via prokaryotic (in chloroplasts) or eukaryotic (endoplasmic reticulum) pathways [14]. Biosynthesis of the successive linoleic and α-linolenic acids requires Δ12 (ω-6 group) and Δ15 (ω-3 group) FADs, which are present only in photosynthetic organisms. The genes for ω-6- and ω-3- FADs are found in genomes of many staple plants and in general, their expression is heavily induced by cold stress [15,16,17]. Humans lack these enzymes, thus making ω-3 and ω-6 fatty acids essential to the human diet [18].

3. Dietary Plant Sources of Fatty Acids

Plant oils and seeds are excellent dietary sources of essential fatty acids. ALA in high concentrations is present in flaxseed/linseed oils (Table 1). LA is found in large amounts in safflower oil. Recent genomic studies of safflower (Carthamus tinctorius) revealed tandem duplications of a ω-6 FAD being specifically expressed in seeds, resulting in a high content of linoleic acid [19]. In order to improve the production of 18:3 fatty acids, genetic modification has been attempted in model plants of tobacco and Arabidopsis [20,21], which resulted in improving their resistance to abiotic stress and increasing the production of 18:3 fatty acids. This can potentially lead to breeding of stress-tolerant plants with a higher nutritional quality of oils.
Oleic acid is not essential for humans, but is synthesized by many plants of nutritional value such as Olea europaea, Brassica species, Arachis species and some others (Table 1). Genome editing using CRISPR-Cas9 technique, which disrupts ω6- desaturase, has been performed in rice (Oryza sativa) and soybeans (Glycine max), resulting in oils rich in oleic acid [22,23]. High-oleic and high-stearic oils were also produced in cotton, using hairpin RNA-mediated post-transcriptional gene silencing of ω6- desaturase [24]. These high-stability cooking oils, after evaluation by food technologists, can potentially replace saturated fats and hydrogenated oils.
Many cooking oils contain a significant amount of saturated fats. For example, palmitic acid (16:0) makes up to 44% of palm oil, 26% of cocoa butter, and 8–20% of olive oil and 10–12% of soybean oil [25]. It is, however, possible to breed novel plant varieties with a higher content of PUFA and a lower content of saturated fatty acids. For example, genetic modification of soybean lines, which results in early termination of palmitoyl-acyl carrier protein thioesterase, has been shown to reduce levels of palmitic acid and can be instrumental to breeding soybean varieties with healthier oils [26].
In addition to seafood sources, plants can be instrumental in providing significant amounts of EPA and DHA. Microalgae cultivated on glucose sources as well as transgenic plants (e.g., Camelina) can be engineered to accumulate ω-3 EPA and DHA, presenting a sustainable alternative to fish [3,27,28].

4. Polyunsaturated Fatty Acids in Human Diet

The importance of food as a therapeutic tool was recognized by Hippocrates thousands of years ago with the well-known quote: “Let food by thy medicine and medicine by thy food”. It has, however, been a challenge to conclusively confirm that a particular diet can provide protection against disease. The case of PUFAs is no exception. They are capable of binding to a number of enzymes/proteins and induce a variety of effects (Table 2). A search of the PubMed database for fatty acids yields over 500,000 publications. The majority of these articles have an overall positive conclusion in terms of the benefit of unsaturated fatty acids for human health. In similar manner, in a search restricted to human clinical trials, the conclusion is also generally positive in terms of the health benefits of ω-3 polyunsaturated fatty acids [29,30,31,32,33,34]. The typical Western diet tends to be high in ω-6 polyunsaturated fatty acids due to the high consumption of meat and meat-derived products. The dominant fatty acids in meat are saturated and high in arachidonic acid (AA), an ω-6 fatty acid associated with inflammation. In general, inflammation is an immunological response seen during infection, cell injury or exposure to harmful chemical agents [35]. Furthermore, the dominance of corn and soy beans in Western diet inadvertently leads to a higher consumption of ω-6 fatty acids, as corn and soy beans are low in ω-3 fatty acids and high in ω-6. The dominant PUFA in corn and soy beans is an ω-6 linoleic acid. Seeds and their corresponding oils that are high in concentrations of ω-3 fatty acids, such as flaxseed, tend not to be common in Western diets. Any supplementation or consumption of foods high in ω-3 fatty acids favors restoration of the imbalance of ω-3 to ω-6 and likely yields health benefits.

5. Long Chain Polyunsaturated Fatty Acids (LC-PUFAs)

In the case of the long chain polyunsaturated ω-3 fatty acids (LC-PUFAs), docosahexaenoic (DHA) and eicosapentaenoic acid (EPA) are the most common and widely used as supplements. Common sources include fish oil, krill oil and ethyl esters. Consumption of fish yields significant health benefits to humans and is believed to be partially responsible for the health benefits of the Mediterranean diet [33,36,37]. Krill oil consumption leads to the highest incorporation of DHA and EPA into plasma phospholipids [38]. The ability of the human body to absorb DHA and EPA is also affected by the environment of the fatty acid [39]. Consumption of intact salmon yields a higher absorption and bioavailability when compared to fish oil. Nonetheless, ω-3 fatty acids have been shown to be incorporated into plasma membranes regardless of the source [40]. The mere substitution of saturated fatty acids with PUFAs in a diet increases bacterial family populations of Lachnospiraceae and Bifidobacterium spp. [41]. This phenomenon brings a different dimension with respect to the health benefits of PUFAs in the human diet. If alterations in specific microbiota populations can impart a positive effect on the hosts, PUFAs are undoubtedly capable of such an effect. It is striking that certain bacterial populations are associated with lower inflammation and that diet indirectly can exert anti-inflammatory properties [31,42,43]. Consumption of walnut, an excellent source of ALA, has been shown to alter the microbiota bacterial family Lachnospiraceae. While fatty acid supplementation in the form of oil capsules is very common and can convey similar benefits, it seems more advantageous to consume whole foods rich in specific PUFAs. Examples of such foods include salmon, sardines, walnuts and flaxseeds. Algal sources offer an attractive alternative to fish whose DHA content can vary depending on whether they are farmed or wild caught. The oxidative stability of fatty acids during digestion is another parameter that still requires further investigation. The nature of fatty acids makes them susceptible to peroxidation during digestion [44]. A considerable amount of peroxidation occurs in the gastric phase of digestion. Emulsification and encapsulation can provide protection in the gastric phase, and ultimately increase the bioaccessibility of PUFAs [45]. Food preservation involving smoking of salmon oils has been shown to reduce peroxidation of salmon oils, and provides a relatively low-tech means of preventing peroxidation of PUFAs in fish [46].

6. Human Clinical Trials Involving Polyunsaturated Fatty Acids

Numerous clinical trials involving DHA and EPA supplementation show an overall positive impact on a wide range of diseases such as Crohn’s disease, major depressive disorder, cardiovascular disease, autism, hypertension, arthritis and lupus [30,32,40,47,48,49,50,51]. It must be noted though that a significant number of clinical trials do not show any measurable health benefits [52,53,54,55]. Conflicting reports often lead to doubts as to the efficacy of PUFAs on human health. Single nucleotide polymorphisms (SNPs) at the level of the fatty acid desaturases 1, 2, 3 (FADs 1,2,3) and elongases (ELOVL 2,5) have been reported to interact with the overall production of PUFAs in the colostrum of pregnant women [56]. It is therefore reasonable to assume that the genetic makeup of individuals can have a positive or negative interaction on PUFA supplementation, and can partly explain why several studies yield conflicting results. In a study with Danish infants who were homozygous for the FADs minor allele rs1535, there was a 1.8% DHA increase. On the contrary, minor allele carriers of the rs174448 and rs174575, had a DHA reduction [57]. Interestingly, breastfeeding duration had a positive impact on DHA levels in these infants, regardless of genotype. In a study conducted in Mexico, the maternal rs174602 SNP had a positive enrichment on infant amino acid and amino sugar metabolic pathways, and decreased fatty acid metabolism [58]. Supplementation with EPA in females has been shown to result in higher plasma DHA levels when compared to males [59]. Polymorphism rs953413 of the ELOVL2 gene seems to exert an influence on DHA plasma levels. Collectively, these clinical data support the use of a genetic analysis of participants of studies involving long chain PUFA supplementation, particularly of the FADs and ELOVL genes.
ALA, a shorter ω-3 PUFA, has comparatively less pronounced health benefits with regard to DHA and EPA [60]. Despite supplementation of 67 healthy individuals with 3.6 g/day of ALA for 8 weeks, no significant improvements were observed in terms of oxidative stress, inflammation and blood pressure. In a study involving 59 untreated pre-hypertensive patients supplemented with 4.7 g/day ALA, a reduction in TNF-α and free fatty acids was observed, but not in any other vascular markers [61].
Oleic acid, which is found in high concentrations in olive oil and peanuts, is an ω-9 monounsaturated fatty acid. Consumption of olive oil and peanut oil has been associated with lower risk of developing asthma and improved glucose regulation [62,63]. However, in a study involving patients with stable coronary disease, intake of either extra virgin olive oil or pecans had no effect on plasma fatty acids [64]. In another study investigating inflammatory markers and oxidative status in obese men, no measurable reduction was observed after consuming either high oleic or conventional oleic peanuts [65].
Ω-6 polyunsaturated fatty acids such as LA and AA are considered less desirable in terms of health benefits. The overall consensus is that the ratio of ω-3 to ω-6 should be as high as possible. It is believed that humans evolved with an ω-3 to ω-6 ratio of 1 to 1, but currently Western diet is around 1 to 15 [66].

7. Anti-Inflammatory Properties of Polyunsaturated Fatty Acids

Ω-3 LC-PUFAs (DHA and EPA) tend to have a more consistent anti-inflammatory effect on immune cells in comparison to other PUFAs [63,67,68]. The anti-inflammatory effect is observed both in vitro and in vivo [49,69,70]. There are several pathways that have been elucidated which provide evidence as to how fatty acids induce an anti-inflammatory state. The peroxisome proliferator-activated receptors (PPARs) have been shown to bind to DHA and EPA and ultimately suppress the production of cytokines related to the NF-κB inflammatory master transcription factor. Another pathway through which fatty acids reduce inflammation is through resolvins and neuroprotectins [71,72]. Aspirin acetylation of the COX enzyme significantly increases production of resolvins and neuroprotectins in the presence of DHA and EPA. Fatty acids incorporated in the plasma membranes of cells are all susceptible to cleaving by phospholipases during inflammatory stress. AA is known to be involved in the production of various prostanoids, via the COX pathway, that have an overall inflammatory effect [73]. The proportion of AA to DHA and EPA is thought to affect inflammation. Specifically, in the presence of aspirin and subsequent acetylation of the COX-2 enzyme, production of resolvins is enhanced, thereby reducing the impact of the inflammatory prostaglandins generated by AA. Another mechanism through which PUFAs can exert anti-inflammation is by affecting the plasma membrane’s properties through lipid rafts. Lipid rafts are domains in plasma membranes that are characterized by higher levels of cholesterol, glycophospholipids and receptors [74]. They play a key role in several cellular activities, including endocytosis, cell signaling and exocytosis. DHA was shown to decrease levels of lipid rafts by as much as 30%. In cancer cells, this reduction in lipid rafts was associated with a reduction in cell proliferation. Both DHA and EPA incorporation increased levels of the antigen presenting molecule MHC I that is expressed on all nucleated cells [75]. Efficient antigen presentation via MHC I leads to faster immune resolution and reduction of chronic inflammatory conditions which are thought to support malignant cell proliferation. The increase in MHC I expression was not attributable to any conformational changes affecting antibody binding to MHC I, but rather to an increase in the plasma membrane. Surprisingly, even AA exhibits an immunomodulatory effect in some instances by preventing M2 polarization of macrophages [76]. Opposing this effect is PGE2, a by-product of AA, which enhances M2 polarization. In another study, AA clearly aggravated obesity and increased inflammatory microbiota [77]. Once again, the balance of ω-3 to ω-6 fatty acids seems to be the key factor in attaining an anti-inflammatory state.

7.1. PUFAs and Neuroinflammation—Effect on Brain Microglia

There is a substantial body of literature addressing the role of PUFAs in neuroinflammation [78]. The brain is a lipid-rich organ containing a great diversity of lipid species, especially PUFAs. The main source of PUFA in the brain is derived from the diet and needs to enter the brain [79]. Lipids in general have three primary mechanisms of entry into the brain across the blood–brain barrier (BBB): passive diffusion, transcytosis via receptor-mediated endocytic pathways, and transport using transmembrane proteins [80]. Conditions that disrupt neuronal homeostasis result in brain synthesized PUFAs. For example, bacterial endotoxin lipopolysaccharide (LPS, an inflammatory stimulus) treatment of astrocytes results in upregulated synthesis of AA and DHA [81].
Microglia are the resident immune cell type of the CNS, and make up roughly 15% of glial cells in the brain. Their hallmark feature is a branched and ramified morphology which facilitates sensing and response to brain injury and infection. Most studies which address neuroinflammation focus on the status of microglia, as they play a central role in most neurological disorders including Alzheimer’s disease (AD), multiple sclerosis (MS), ischemic stroke, traumatic brain injury (TBI) and even cancer. Microglia, like other tissue macrophages, can be polarized into “classical” pro-inflammatory phenotype and the “alternative” anti-inflammatory phenotype, often referred to as M1 and M2, respectively, although these likely represent extremes within a spectrum of responses. The M1 state is typically activated by TLR ligands such as LPS which induce the expression of cytokines such as TNF-α, IL-1β and IL-6. The M2 phenotype results from stimulation with cytokines such as IL-4 and IL-13, and promotes expression of cytokines and growth factors such as IL-10 and TGFb, which are immunosuppressive and promote tissue regeneration. With respect to neurological disorders, microglial expression of M1 markers correlates with poorer clinical outcomes. In multiple animal models, experimental manipulation, which produces M1 microglia (such as LPS injection), generally exacerbates the disease severity [82,83,84]. Conversely, the repolarization of microglia to the M2 anti-inflammatory state tends to have a beneficial effect. Therefore, therapeutic intervention which promotes the M2 polarization of microglia would be desirable for many neurological diseases. In this regard, PUFAs have gained attention as potential modulators of neuroinflammation.
In general, saturated FAs induce a pro-inflammatory phenotype in microglia, while MUFAs and PUFAs promote the M2 state. Saturated PA has been shown to stimulate expression of pro-inflammatory cytokine gene expression to a similar extent as LPS in cultured astrocytes and BV-2 microglial cells in a TLR4-dependent manner [85,86], whereas unsaturated OA has been shown to have the opposite effect [87]. In an in vitro model, it was recently shown that OA can mitigate the effects of PA-stimulated microglia neurotoxic effects in neuronal cocultures [88]. At the forefront of the role of lipids in neuroinflammation are the ω-3 long chain PUFAs and their derivatives, specialized pro-resolving lipid mediators (SPMs). This class of lipids have emerged as central players in limiting the M1 phenotype and dampening neuroinflammation. It is well established that ω-3 long chain PUFAs downregulate LPS-stimulated pro-inflammatory genes such as TNF-α and IL-6 in microglia, both in vitro and in vivo [87,89,90,91,92]. DHA is a potent M2 polarizing agent in microglia, and can reduce inflammation in several neuronal disease models [93,94,95]. Furthermore, mice deficient in DHA synthesis exhibit increased M1 inflammatory markers in the brain [96]. The ω-3 long chain PUFAs were discovered to work on multiple levels, including regulation of receptor activity, signaling kinases and gene expression. ALA, which is found in walnut extract, downregulates the expression of surface TLR4 and iNOS induction [97,98,99]. DHA and EPA were shown to enhance SIRT-1 deacetylase activity, which had the effect of blocking NF-kB activation of inflammatory genes in the MG6 murine microglial cell line [100]. The effect of DHA on BV-2 microglial cells was recently analyzed in depth using quantitative proteomics [101]. Confirming earlier studies, many signaling proteins in the NF-kB pathway, including sequestome-1, NOS and CD40 were found to be differentially expressed [102]. Interestingly, it was also discovered that DHA influences the pattern of protein expression involved in fatty acid metabolism and ribosome function, suggesting a more global mechanism of DHA regulation of microglial function. Much of the anti-inflammatory activity these dietary PUFAs is likely mediated by SPMs. One of the SPMs, resolvin RvD1, can enhance IL-4 induced M2 polarity of BV-2 microglia in vitro [103]. RvD1 can also promote this shift away from M1 in vivo [104].

7.2. PUFAs and Neuroinflammation—Effect on Brain Astrocytes

Astrocytes are the main glial cells of the brain and play essential roles in energy metabolism, maintenance of extracellular ion concentrations, formation of the blood–brain barrier and general CNS homeostasis. Astrocytes are also able to modulate inflammation both directly via release of soluble mediators and indirectly by influencing neighboring microglia [105,106]. Astrocytes can also respond to signals from damaged neurons, resulting in reactive astrogliosis, which is characteristic of neuroinflammation in general. LPS stimulation of astrocytes results in a release of DHA which likely promotes survival of neurons during neuroinflammation [81]. PUFAs can also act on astrocytes to attenuate inflammation. DHA and EPA-treated astrocytes show a decrease in NF-kB and immunoproteosome activity [107]. Fortasyn Connect©, a nutrient combination which includes DHA and EPA, is able to inhibit astrogliosis in an in vitro model [108].

8. PUFAs and Neurological Diseases

Alzheimer’s disease (AD) in an invariably fatal disease marked by a steady decline in cognitive abilities and neurodegeneration. The role for microglia in AD has recently been highlighted, as they carry out phagocytic removal of amyloid plaques [109,110]. The potential for using ω-3 long chain PUFAs to ameliorate AD symptoms and progression has gained much attention [111]. In a variety of animal models for AD, diets and formulations rich in ω-3 long chain PUFAs had anti-inflammatory effects and a positive impact on disease progression [112,113,114,115,116,117]. Amyloid aggregates have the ability to promote M1 neuroinflammation, and this is dampened in the presence of DHA [118]. EPA can protect against an amyloid injection AD model in rats [119,120,121]. Interestingly, DHA and EPA treatment was shown to simultaneously enhance phagocytosis of amyloid protein while promoting an M2 phenotype [122,123]. EPA was also shown to enhance neuroprotective factors produced by astrocytes in the rat hippocampus [93]. ALA can also mobilize microglia to phagocytose extracellular tau aggregates [124]. ALA has also been shown to act via astrocytes, as conditioned media collected from human astrocyte cultures protected SH-SY5Y cells from amyloid-induced cell death [125]. Fish oil was also shown to activate AQP4 on astrocytes, resulting in glymphatic clearance of amyloid from the brain [126]. SPMs and the pathways they govern are also being tested for their ability to treat AD in preclinical models. AD patients exhibit fewer SPMs in the hippocampus, and the maresin MaR1 can inhibit M1 gene expression in the human microglial cell line CHME3 [127]. MaR1 is also able to reduce amyloid induced cytokine stimulation, alongside inducing amyloid phagocytosis by macrophages in vitro [128]. MaR1 can prevent microglia, and astrocyte activation in an AD mouse model had substantial benefits in preventing cognitive decline as well [129]. Administration of resolvin RvE1 and lipoxin LXA4 into the intraperitoneum of 5XFAD mice decreased neuroinflammation and lowered the amyloid plaque burden [130]. Intranasal delivery of a mixture of SPMs including resolvins RvE1, RvD1, RvD2, maresin MaR1 and neuroprotectin D1 was able to reduce microglial activation and restore some brain function in an AD mouse model [131].
In addition to AD, the immunomodulatory effects of PUFAs have been shown to have beneficial effects in experimental models for several other neurological diseases, including multiple sclerosis (MS), traumatic brain injury (TBI), stroke and Parkinson’s disease (PD). In general, microglial activation and expression of M1 inflammatory genes tends to contribute to neuronal cell death. Conditioned media from THP-1 macrophages treated with PA, but not OA or LA, induces apoptosis of SH-SY5Y neuronal-like cells in culture [132]. Similarly, DHA treatment of LPS-activated BV-2 translates into a less cytotoxic effect on neurons in cell cocultures [91]. In a model of myelin-induced damage which approximates pathological features of multiple sclerosis (MS), DHA and EPA shift the microglial response away from M1 [133]. Metabolites of DHA are lower in the chronic model of MS [134]. Furthermore, in the experimental autoimmune encephalomyelitis model of MS, a triglyceride formulation of DHA is able to prevent inflammation and exert neuroprotective effects [135]. There is also evidence for PUFAs in neuroprotection from stroke and brain injury. Consistent with the studies cited above, ω-3 long chain PUFAs are able to prevent neuroinflammation associated with ischemic damage and promote an M2 phenotype in microglia associated with at the site of injury [136,137]. There are multiple reports which show that DHA, either as a single agent therapy or prepared in dietary formulations, can prevent inflammation and neuronal damage in a mouse model [138,139,140,141,142,143]. DHA treatment of rats with traumatic brain injury showed less microglial endoplasmic reticulum stress and autophagy [144,145]. The mechanism of action is in part via the ω-3 fatty acid receptor GPR120 [146]. SPMs are also likely to play a role in preventing neuroinflammation associated with neuronal damage [147]. These results are encouraging and have spurred the development of small molecule agonists of SPM receptors to treat neurological disorders involving brain injury [148].
Table 1. Content of important fatty acids in plant oils commonly used as food sources (% from all FA) [149,150,151,152].
Table 1. Content of important fatty acids in plant oils commonly used as food sources (% from all FA) [149,150,151,152].
Rich in ω-3 and ω-6
Sunflower OilRapeseed OilMustard OilPeanut OilOlive OilAvocado OilGrapeseed OilFlaxseed OilWalnut Oil
Palmitic acid (PA)5.943.9710.249.3715.1110.087.25.876.3
Oleic acid (OA)30–80 *63.6836.6555.3368.8560.719.917.4120.5
Linoleic acid (LA, ω-6)21–70 *17.4322.0623.698.511.868.115.7655.5
α-linolenic acid (ALA, ω-3)0.79---0.541.20.155.4014.8
* Content depends on a plant breed and industrial processing.
Table 2. PUFAs and their relevant metabolic enzymes/ligand targets.
Table 2. PUFAs and their relevant metabolic enzymes/ligand targets.
PUFATranscription Factor/EnzymeMetabolite/
Ligand
Inflammatory
Effect
Docosahexaenoic acid (DHA)COX-2Resolvin DAnti [71]
PLA2Protectin D1Anti [153]
PPAR-αLigandAnti [154]
PPAR-γLigandAnti [154]
Eicosapentaenoic acid (EPA)COX-2
PPAR-α
PPAR-γ
Resolvins E
Ligand
Ligand
Anti [72]

Anti [155]
Anti [156]
α-Linolenic acid (ALA)PPAR-α
PPAR-γ
Ligand
Ligand
Anti [157]
Anti [158]
Arachidonic acid (AA)COX-2
COX-2
PPAR-α
PPAR-δ
PGE2
PGI2
Ligand
Ligand
Pro [159]
Anti [160]
Anti [157]
Anti-Apoptotic [161]
Linoleic acid (LA)PPAR-αLigandEnergy Control
[157,162]
Oleic acid (OA) *TLX-NR2E1LigandNeurogenesis, Anti
[157,163]
* MUFA, monounsaturated fatty acid.

9. Conclusions

The anti-inflammatory properties of unsaturated fatty acids have been shown in multiple studies. PUFAs from the ω-3 group reduce inflammation in multiple tissues, including neural tissues. Increasing ω-3 PUFAs and ω-9 MUFAs in the human diet through enrichment with plant food sources such as flaxseed/linseed, walnut oil, olive oil, and fish products may be beneficial in decreasing the overall inflammatory response in the human body. As we gain more knowledge in terms of our understanding of how various PUFAs interact with each other, it is possible that improved formulations will arise. A better understanding of how individual genotypes influence the absorption and metabolism of PUFAs will also help to design better studies utilizing PUFAs. Lastly, alternative routes of administration, other than the typical oral administration of PUFAs, may yield more pronounced health benefits in the future.

Author Contributions

Conceptualization, E.V.; writing—original draft preparation, S.C., M.S. and E.V.; writing—review and editing, S.C., M.S. and E.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nelson, D.L.; Cox, M.M. Lehninger Principles of Biochemistry; W.H. Freeman and Company: New York, NY, USA, 2017; p. 1308. [Google Scholar]
  2. US Department of Health and Human Services and US Department of Agriculture. 2015–2020 Dietary Guidelines for Americans. Available online: http://health.gov/dietaryguidelines/2015/guidelines/ (accessed on 15 January 2023).
  3. Oliver, L.; Dietrich, T.; Maranon, I.; Villaran, M.C.; Barrio, R.J. Producing Omega-3 Polyunsaturated Fatty Acids: A Review of Sustainable Sources and Future Trends for the EPA and DHA Market. Resources 2020, 9, 148. [Google Scholar] [CrossRef]
  4. Zhukov, A.V.; Shumskaya, M. Very-long-chain fatty acids (VLCFAs) in plant response to stress. Funct. Plant Biol. 2020, 47, 695–703. [Google Scholar] [CrossRef] [PubMed]
  5. Cerone, M.; Smith, T.K. Desaturases: Structural and mechanistic insights into the biosynthesis of unsaturated fatty acids. Iubmb Life 2022, 74, 1036–1051. [Google Scholar] [CrossRef] [PubMed]
  6. Los, D.A.; Murata, N. Membrane fluidity and its roles in the perception of environmental signals. Biochim. Biophys Acta—Biomemb 2004, 1666, 142–157. [Google Scholar] [CrossRef]
  7. Murata, N.; Los, D.A. Membrane fluidity and temperature perception. Plant Physiol. 1997, 115, 875–879. [Google Scholar] [CrossRef]
  8. Zhang, W.H.; Wang, C.X.; Qin, C.B.; Wood, T.; Olafsdottir, G.; Welti, R.; Wang, X.M. The oleate-stimulated phospholipase D, PLD delta, and phosphatidic acid decrease H2O2-induced cell death in arabidopsis. Plant Cell 2003, 15, 2285–2295. [Google Scholar] [CrossRef]
  9. He, M.; He, C.Q.; Ding, N.Z. Abiotic Stresses: General Defenses of Land Plants and Chances for Engineering Multistress Tolerance. Front. Plant Sci. 2018, 9, 1771. [Google Scholar] [CrossRef]
  10. He, M.; Qin, C.-X.; Wang, X.; Ding, N.-Z. Plant Unsaturated Fatty Acids: Biosynthesis and Regulation. Front. Plant Sci. 2020, 11, 390. [Google Scholar] [CrossRef]
  11. Shahid, M.; Cai, G.Q.; Zu, F.; Zhao, Q.; Qasim, M.U.; Hong, Y.Y.; Fan, C.C.; Zhou, Y.M. Comparative Transcriptome Analysis of Developing Seeds and Silique Wall Reveals Dynamic Transcription Networks for Effective Oil Production in Brassica napus L. Int. J. Mol. Sci. 2019, 20, 1982. [Google Scholar] [CrossRef]
  12. Ohlrogge, J.; Thrower, N.; Mhaske, V.; Stymne, S.; Baxter, M.; Yang, W.L.; Liu, J.J.; Shaw, K.; Shorrosh, B.; Zhang, M.; et al. PlantFAdb: A resource for exploring hundreds of plant fatty acid structures synthesized by thousands of plants and their phylogenetic relationships. Plant J. 2018, 96, 1299–1308. [Google Scholar] [CrossRef] [Green Version]
  13. Piccinin, E.; Cariello, M.; De Santis, S.; Ducheix, S.; Sabba, C.; Ntambi, J.M.; Moschetta, A. Role of Oleic Acid in the Gut-Liver Axis: From Diet to the Regulation of Its Synthesis via Stearoyl-CoA Desaturase 1 (SCD1). Nutrients 2019, 11, 2283. [Google Scholar] [CrossRef]
  14. Wu, Q.; Liu, T.; Liu, H.; Zheng, G.C. Unsaturated fatty acid: Metabolism, synthesis and gene regulation. Afr. J. Biotechnol. 2009, 8, 1782–1785. [Google Scholar]
  15. Yu, C.; Wang, H.S.; Yang, S.; Tang, X.F.; Duan, M.; Meng, Q.W. Overexpression of endoplasmic reticulum omega-3 fatty acid desaturase gene improves chilling tolerance in tomato. Plant Physiol. Biochem. 2009, 47, 1102–1112. [Google Scholar] [CrossRef]
  16. Lee, S.H.; Ahn, S.J.; Im, Y.J.; Cho, K.; Chung, G.C.; Cho, B.H.; Han, O. Differential impact of low temperature on fatty acid unsaturation and lipoxygenase activity in figleaf gourd and cucumber roots. Biochem. Biophys. Res. Commun. 2005, 330, 1194–1198. [Google Scholar] [CrossRef]
  17. Roman, A.; Hernandez, M.L.; Soria-Garcia, A.; Lopez-Gomollon, S.; Lagunas, B.; Picorel, R.; Martinez-Rivas, J.M.; Alfonso, M. Non-redundant Contribution of the Plastidial FAD8 omega-3 Desaturase to Glycerolipid Unsaturation at Different Temperatures in Arabidopsis. Mol. Plant 2015, 8, 1599–1611. [Google Scholar] [CrossRef]
  18. Tvrzicka, E.; Kremmyda, L.S.; Stankova, B.; Zak, A. Fatty acids are biocompounds: Their role in human metabolism, health and disease—A review. Part 1: Classification, dietary sources and biological functions. Biomed. Pap. 2011, 155, 117–130. [Google Scholar] [CrossRef]
  19. Wu, Z.H.; Liu, H.; Zhan, W.; Yu, Z.C.; Qin, E.R.; Liu, S.; Yang, T.G.; Xiang, N.Y.; Kudrna, D.; Chen, Y.; et al. The chromosome-scale reference genome of safflower (Carthamus tinctorius) provides insights into linoleic acid and flavonoid biosynthesis. Plant Biotechnol. J. 2021, 19, 1725–1742. [Google Scholar] [CrossRef]
  20. Shi, Y.; Yue, X.; An, L. Integrated regulation triggered by a cryophyte omega-3 desaturase gene confers multiple-stress tolerance in tobacco. J. Exp. Bot. 2018, 69, 2131–2148. [Google Scholar] [CrossRef]
  21. Yin, Y.M.; Jiang, X.X.; Ren, M.Y.; Xue, M.; Nan, D.N.; Wang, Z.L.; Xing, Y.P.; Wang, M.Y. AmDREB2C, from Ammopiptanthus mongolicus, enhances abiotic stress tolerance and regulates fatty acid composition in transgenic Arabidopsis. Plant Physiol. Biochem. 2018, 130, 517–528. [Google Scholar] [CrossRef]
  22. Abe, K.; Araki, E.; Suzuki, Y.; Toki, S.; Saika, H. Production of high oleic/low linoleic rice by genome editing. Plant Physiol. Biochem. 2018, 131, 58–62. [Google Scholar] [CrossRef]
  23. Do, P.T.; Nguyen, C.X.; Bui, H.T.; Tran, L.T.N.; Stacey, G.; Gillman, J.D.; Zhang, Z.Y.J.; Stacey, M.G. Demonstration of highly efficient dual gRNA CRISPR/Cas9 editing of the homeologous GmFAD2-1A and GmFAD2-1B genes to yield a high oleic, low linoleic and alpha-linolenic acid phenotype in soybean. BMC Plant Biol. 2019, 19, 311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Liu, Q.; Singh, S.P.; Green, A.G. High-stearic and high-oleic cottonseed oils produced by hairpin RNA-mediated post-transcriptional gene silencing. Plant Physiol. 2002, 129, 1732–1743. [Google Scholar] [CrossRef] [PubMed]
  25. Carta, G.; Murru, E.; Banni, S.; Manca, C. Palmitic acid: Physiological role, metabolism and nutritional implications. Front. Physiol. 2017, 8, 902. [Google Scholar] [CrossRef] [PubMed]
  26. Carrero-Colon, M.; Hudson, K. Reduced palmitic acid content in soybean as a result of mutation in FATB1a. PLoS ONE 2022, 17, e0262327. [Google Scholar] [CrossRef]
  27. Valenzuela, B.A.; Sanhueza, C.J.; Valenzuela, B.R. Las microalgas: Una fuente renovable para la obtención de ácidos grasos omega-3 de cadena larga para la nutrición humana y animal. Rev. Chil. De Nutr. 2015, 42, 306–310. [Google Scholar] [CrossRef]
  28. Napier, J.A.; Usher, S.; Haslam, R.P.; Ruiz-Lopez, N.; Sayanova, O. Transgenic plants as a sustainable, terrestrial source of fish oils. Eur. J. Lipid Sci. Technol. 2015, 117, 1317–1324. [Google Scholar] [CrossRef]
  29. Van Name, M.A.; Savoye, M.; Chick, J.M.; Galuppo, B.T.; Feldstein, A.E.; Pierpont, B.; Johnson, C.; Shabanova, V.; Ekong, U.; Valentino, P.L.; et al. A Low ω-6 to ω-3 PUFA Ratio (n-6:n-3 PUFA) Diet to Treat Fatty Liver Disease in Obese Youth. J. Nutr. 2020, 150, 2314–2321. [Google Scholar] [CrossRef]
  30. Stańdo, M.; Piatek, P.; Namiecinska, M.; Lewkowicz, P.; Lewkowicz, N. Omega-3 polyunsaturated fatty acids EPA and DHA as an adjunct to non-surgical treatment of periodontitis: A randomized clinical trial. Nutrients 2020, 12, 2614. [Google Scholar] [CrossRef]
  31. Watson, H.; Mitra, S.; Croden, F.C.; Taylor, M.; Wood, H.M.; Perry, S.L.; Spencer, J.A.; Quirke, P.; Toogood, G.J.; Lawton, C.L.; et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut 2018, 67, 1974–1983. [Google Scholar] [CrossRef]
  32. Keim, S.A.; Gracious, B.; Boone, K.M.; Klebanoff, M.A.; Rogers, L.K.; Rausch, J.; Coury, D.L.; Sheppard, K.W.; Husk, J.; Rhoda, D.A. ω-3 and ω-6 fatty acid supplementation may reduce autism symptoms based on parent report in preterm toddlers. J. Nutr. 2018, 148, 227–235. [Google Scholar] [CrossRef]
  33. Davis, C.R.; Bryan, J.; Hodgson, J.M.; Woodman, R.; Murphy, K.J. A mediterranean diet reduces F(2)-isoprostanes and triglycerides among older Australian men and women after 6 months. J. Nutr. 2017, 147, 1348–1355. [Google Scholar] [CrossRef] [Green Version]
  34. Kalstad, A.A.; Myhre, P.L.; Laake, K.; Tveit, S.H.; Schmidt, E.B.; Smith, P.; Nilsen, D.W.T.; Tveit, A.; Fagerland, M.W.; Solheim, S.; et al. Effects of n-3 Fatty acid supplements in ederly patients after myocardial infarction: A randomized, controlled trial. Circulation 2021, 143, 528–539. [Google Scholar] [CrossRef]
  35. Medzhitov, R. Inflammation 2010: New Adventures of an Old Flame. Cell 2010, 140, 771–776. [Google Scholar] [CrossRef]
  36. Giroli, M.G.; Werba, J.P.; Risé, P.; Porro, B.; Sala, A.; Amato, M.; Tremoli, E.; Bonomi, A.; Veglia, F. Effects of Mediterranean diet or low-fat diet on blood fatty acids in patients with coronary heart disease. A randomized intervention study. Nutrients 2021, 13, 2389. [Google Scholar] [CrossRef]
  37. Murphy, K.J.; Dyer, K.A.; Hyde, B.; Davis, C.R.; Bracci, E.L.; Woodman, R.J.; Hodgson, J.M. Long-Term Adherence to a Mediterranean Diet 1-Year after Completion of the MedLey Study. Nutrients 2022, 14, 3098. [Google Scholar] [CrossRef]
  38. Schuchardt, J.P.; Schneider, I.; Meyer, H.; Neubronner, J.; von Schacky, C.; Hahn, A. Incorporation of EPA and DHA into plasma phospholipids in response to different omega-3 fatty acid formulations—A comparative bioavailability study of fish oil vs. krill oil. Lipids Health Dis. 2011, 10, 145. [Google Scholar] [CrossRef]
  39. Ahmed Nasef, N.; Zhu, P.; Golding, M.; Dave, A.; Ali, A.; Singh, H.; Garg, M. Salmon food matrix influences digestion and bioavailability of long-chain omega-3 polyunsaturated fatty acids. Food Funct. 2021, 12, 6588–6602. [Google Scholar] [CrossRef]
  40. Brennan Laing, B.; Cavadino, A.; Ellett, S.; Ferguson, L.R. Effects of an Omega-3 and Vitamin D Supplement on Fatty Acids and Vitamin D Serum Levels in Double-Blinded, Randomized, Controlled Trials in Healthy and Crohn’s Disease Populations. Nutrients 2020, 12, 1139. [Google Scholar] [CrossRef]
  41. Telle-Hansen, V.H.; Gaundal, L.; Bastani, N.; Rud, I.; Byfuglien, M.G.; Gjøvaag, T.; Retterstøl, K.; Holven, K.B.; Ulven, S.M.; Myhrstad, M.C.W. Replacing saturated fatty acids with polyunsaturated fatty acids increases the abundance of Lachnospiraceae and is associated with reduced total cholesterol levels-a randomized controlled trial in healthy individuals. Lipids Health Dis. 2022, 21, 92. [Google Scholar] [CrossRef]
  42. Tindall, A.M.; McLimans, C.J.; Petersen, K.S.; Kris-Etherton, P.M.; Lamendella, R. Walnuts and Vegetable Oils Containing Oleic Acid Differentially Affect the Gut Microbiota and Associations with Cardiovascular Risk Factors: Follow-up of a Randomized, Controlled, Feeding Trial in Adults at Risk for Cardiovascular Disease. J. Nutr. 2020, 150, 806–817. [Google Scholar] [CrossRef]
  43. Wan, Y.; Wang, F.; Yuan, J.; Li, J.; Jiang, D.; Zhang, J.; Li, H.; Wang, R.; Tang, J.; Huang, T.; et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: A 6-month randomised controlled-feeding trial. Gut 2019, 68, 1417–1429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Floros, S.; Toskas, A.; Pasidi, E.; Vareltzis, P. Bioaccessibility and Oxidative Stability of Omega-3 Fatty Acids in Supplements, Sardines and Enriched Eggs Studied Using a Static In Vitro Gastrointestinal Model. Molecules 2022, 2, 415. [Google Scholar] [CrossRef] [PubMed]
  45. Venugopalan, V.K.; Gopakumar, L.R.; Kumaran, A.K.; Chatterjee, N.S.; Soman, V.; Peeralil, S.; Mathew, S.; McClements, D.J.; Nagarajarao, R.C. Encapsulation and Protection of Omega-3-Rich Fish Oils Using Food-Grade Delivery Systems. Foods 2021, 10, 1566. [Google Scholar] [CrossRef] [PubMed]
  46. Bower, C.K.; Hietala, K.A.; Oliveira, A.C.; Wu, T.H. Stabilizing oils from smoked pink salmon (Oncorhynchus gorbuscha). J. Food Sci. 2009, 74, C248–C257. [Google Scholar] [CrossRef] [PubMed]
  47. Pisaniello, A.D.; Psaltis, P.J.; King, P.M.; Liu, G.; Gibson, R.A.; Tan, J.T.; Duong, M.; Nguyen, T.; Bursill, C.A.; Worthley, M.I.; et al. Omega-3 fatty acids ameliorate vascular inflammation: A rationale for their atheroprotective effects. Atherosclerosis 2021, 324, 27–37. [Google Scholar] [CrossRef]
  48. Mischoulon, D.; Dunlop, B.W.; Kinkead, B.; Schettler, P.J.; Lamon-Fava, S.; Rakofsky, J.J.; Nierenberg, A.A.; Clain, A.J.; Mletzko Crowe, T.; Wong, A.; et al. Omega-3 Fatty Acids for Major Depressive Disorder With High Inflammation: A Randomized Dose-Finding Clinical Trial. J. Clin. Psychiatry 2022, 83, 42432. [Google Scholar] [CrossRef]
  49. Dawczynski, C.; Dittrich, M.; Neumann, T.; Goetze, K.; Welzel, A.; Oelzner, P.; Völker, S.; Schaible, A.M.; Troisi, F.; Thomas, L.; et al. Docosahexaenoic acid in the treatment of rheumatoid arthritis: A double-blind, placebo-controlled, randomized cross-over study with microalgae vs. sunflower oil. Clin. Nutr. 2018, 37, 494–504. [Google Scholar] [CrossRef]
  50. Das, U.N. Beneficial effect of eicosapentaenoic and docosahexaenoic acids in the management of systemic lupus erythematosus and its relationship to the cytokine network. Prostaglandins Leukot. Essent. Fat. Acids 1994, 51, 207–213. [Google Scholar] [CrossRef]
  51. Alfaddagh, A.; Elajami, T.K.; Saleh, M.; Elajami, M.; Bistrian, B.R.; Welty, F.K. The effect of eicosapentaenoic and docosahexaenoic acids on physical function, exercise, and joint replacement in patients with coronary artery disease: A secondary analysis of a randomized clinical trial. J. Clin. Lipidol. 2018, 12, 937–947. [Google Scholar] [CrossRef]
  52. Albert, C.M.; Cook, N.R.; Pester, J.; Moorthy, M.V.; Ridge, C.; Danik, J.S.; Gencer, B.; Siddiqi, H.K.; Ng, C.; Gibson, H.; et al. Effect of Marine Omega-3 Fatty Acid and Vitamin D Supplementation on Incident Atrial Fibrillation: A Randomized Clinical Trial. JAMA 2021, 325, 1061–1073. [Google Scholar] [CrossRef]
  53. Chiva-Blanch, G.; Bratseth, V.; Laake, K.; Arnesen, H.; Solheim, S.; Schmidt, E.B.; Badimon, L.; Seljeflot, I. One year of omega 3 polyunsaturated fatty acid supplementation does not reduce circulating prothrombotic microvesicles in elderly subjects after suffering a myocardial infarction. Clin. Nutr. 2021, 40, 5674–5677. [Google Scholar] [CrossRef]
  54. De Borst, M.H.; Baia, L.C.; Hoogeveen, E.K.; Giltay, E.J.; Navis, G.; Bakker, S.J.L.; Geleijnse, J.M.; Kromhout, D.; Soedamah-Muthu, S.S. Effect of Omega-3 Fatty Acid Supplementation on Plasma Fibroblast Growth Factor 23 Levels in Post-Myocardial Infarction Patients with Chronic Kidney Disease: The Alpha Omega Trial. Nutrients 2017, 9, 1233. [Google Scholar] [CrossRef]
  55. Okereke, O.I.; Vyas, C.M.; Mischoulon, D.; Chang, G.; Cook, N.R.; Weinberg, A.; Bubes, V.; Copeland, T.; Friedenberg, G.; Lee, I.M.; et al. Effect of Long-term Supplementation With Marine Omega-3 Fatty Acids vs. Placebo on Risk of Depression or Clinically Relevant Depressive Symptoms and on Change in Mood Scores: A Randomized Clinical Trial. JAMA 2021, 326, 2385–2394. [Google Scholar] [CrossRef]
  56. Li, P.; Chen, Y.; Song, J.; Yan, L.; Tang, T.; Wang, R.; Fan, X.; Zhao, Y.; Qi, K. Maternal DHA-rich n-3 PUFAs supplementation interacts with FADS genotypes to influence the profiles of PUFAs in the colostrum among Chinese Han population: A birth cohort study. Nutr. Metab. 2022, 19, 48. [Google Scholar] [CrossRef]
  57. Harsløf, L.B.; Larsen, L.H.; Ritz, C.; Hellgren, L.I.; Michaelsen, K.F.; Vogel, U.; Lauritzen, L. FADS genotype and diet are important determinants of DHA status: A cross-sectional study in Danish infants. Am. J. Clin. Nutr. 2013, 97, 1403–1410. [Google Scholar] [CrossRef]
  58. Tandon, S.; Gonzalez-Casanova, I.; Barraza-Villarreal, A.; Romieu, I.; Demmelmair, H.; Jones, D.P.; Koletzko, B.; Stein, A.D.; Ramakrishnan, U. Infant Metabolome in Relation to Prenatal DHA Supplementation and Maternal Single-Nucleotide Polymorphism rs174602: Secondary Analysis of a Randomized Controlled Trial in Mexico. J. Nutr. 2021, 151, 3339–3349. [Google Scholar] [CrossRef]
  59. Metherel, A.H.; Irfan, M.; Klingel, S.L.; Mutch, D.M.; Bazinet, R.P. Higher Increase in Plasma DHA in Females Compared to Males Following EPA Supplementation May Be Influenced by a Polymorphism in ELOVL2: An Exploratory Study. Lipids 2021, 56, 211–228. [Google Scholar] [CrossRef]
  60. Burak, C.; Wolffram, S.; Zur, B.; Langguth, P.; Fimmers, R.; Alteheld, B.; Stehle, P.; Egert, S. Effect of alpha-linolenic acid in combination with the flavonol quercetin on markers of cardiovascular disease risk in healthy, non-obese adults: A randomized, double-blinded placebo-controlled crossover trial. Nutrition 2019, 58, 47–56. [Google Scholar] [CrossRef]
  61. Joris, P.J.; Draijer, R.; Fuchs, D.; Mensink, R.P. Effect of α-linolenic acid on vascular function and metabolic risk markers during the fasting and postprandial phase: A randomized placebo-controlled trial in untreated (pre-)hypertensive individuals. Clin. Nutr. 2020, 39, 2413–2419. [Google Scholar] [CrossRef]
  62. Cazzoletti, L.; Zanolin, M.E.; Spelta, F.; Bono, R.; Chamitava, L.; Cerveri, I.; Garcia-Larsen, V.; Grosso, A.; Mattioli, V.; Pirina, P.; et al. Dietary fats, olive oil and respiratory diseases in Italian adults: A population-based study. Clin. Exp. Allergy 2019, 49, 799–807. [Google Scholar] [CrossRef]
  63. Vassiliou, E.K.; Gonzalez, A.; Garcia, C.; Tadros, J.H.; Chakraborty, G.; Toney, J.H. Oleic acid and peanut oil high in oleic acid reverse the inhibitory effect of insulin production of the inflammatory cytokine TNF-alpha both in vitro and in vivo systems. Lipids Health Dis. 2009, 8, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. De Araújo, A.R.; Sampaio, G.R.; da Silva, L.R.; Portal, V.L.; Markoski, M.M.; de Quadros, A.S.; Rogero, M.M.; da Silva Torres, E.A.F.; Marcadenti, A. Effects of extra virgin olive oil and pecans on plasma fatty acids in patients with stable coronary artery disease. Nutrition 2021, 91–92, 111411. [Google Scholar] [CrossRef] [PubMed]
  65. Caldas, A.P.S.; Alves, R.D.M.; Hermsdorff, H.H.M.; de Oliveira, L.L.; Bressan, J. Effects of high-oleic peanuts within a hypoenergetic diet on inflammatory and oxidative status of overweight men: A randomised controlled trial. Br. J. Nutr. 2020, 123, 673–680. [Google Scholar] [CrossRef] [PubMed]
  66. Simopoulos, A.P. The importance of the ratio of omega-6/omega-3 essential fatty acids. Biomed. Pharmacother. 2002, 56, 365–379. [Google Scholar] [CrossRef] [PubMed]
  67. Kong, W.; Yen, J.H.; Vassiliou, E.; Adhikary, S.; Toscano, M.G.; Ganea, D. Docosahexaenoic acid prevents dendritic cell maturation and in vitro and in vivo expression of the IL-12 cytokine family. Lipids Health Dis. 2010, 9, 12. [Google Scholar] [CrossRef]
  68. Pettit, L.K.; Varsanyi, C.; Tadros, J.; Vassiliou, E. Modulating the inflammatory properties of activated microglia with Docosahexaenoic acid and Aspirin. Lipids Health Dis. 2013, 12, 16. [Google Scholar] [CrossRef]
  69. Borsini, A.; Nicolaou, A.; Camacho-Muñoz, D.; Kendall, A.C.; Di Benedetto, M.G.; Giacobbe, J.; Su, K.P.; Pariante, C.M. Omega-3 polyunsaturated fatty acids protect against inflammation through production of LOX and CYP450 lipid mediators: Relevance for major depression and for human hippocampal neurogenesis. Mol. Psychiatry 2021, 26, 6773–6788. [Google Scholar] [CrossRef]
  70. Naeini, Z.; Toupchian, O.; Vatannejad, A.; Sotoudeh, G.; Teimouri, M.; Ghorbani, M.; Nasli-Esfahani, E.; Koohdani, F. Effects of DHA-enriched fish oil on gene expression levels of p53 and NF-κB and PPAR-γ activity in PBMCs of patients with T2DM: A randomized, double-blind, clinical trial. Nutr. Metab. Cardiovasc. Dis. 2020, 30, 441–447. [Google Scholar] [CrossRef]
  71. Dalli, J.; Winkler, J.W.; Colas, R.A.; Arnardottir, H.; Cheng, C.Y.; Chiang, N.; Petasis, N.A.; Serhan, C.N. Resolvin D3 and aspirin-triggered resolvin D3 are potent immunoresolvents. Chem. Biol. 2013, 20, 188–201. [Google Scholar] [CrossRef]
  72. Sun, Y.P.; Oh, S.F.; Uddin, J.; Yang, R.; Gotlinger, K.; Campbell, E.; Colgan, S.P.; Petasis, N.A.; Serhan, C.N. Resolvin D1 and its aspirin-triggered 17R epimer. Stereochemical assignments, anti-inflammatory properties, and enzymatic inactivation. J. Biol. Chem. 2007, 282, 9323–9334. [Google Scholar] [CrossRef]
  73. Markworth, J.F.; D’Souza, R.F.; Aasen, K.M.M.; Mitchell, S.M.; Durainayagam, B.R.; Sinclair, A.J.; Peake, J.M.; Egner, I.M.; Raastad, T.; Cameron-Smith, D.; et al. Arachidonic acid supplementation transiently augments the acute inflammatory response to resistance exercise in trained men. J. Appl. Physiol. 2018, 125, 271–286. [Google Scholar] [CrossRef]
  74. Corsetto, P.A.; Cremona, A.; Montorfano, G.; Jovenitti, I.E.; Orsini, F.; Arosio, P.; Rizzo, A.M. Chemical-physical changes in cell membrane microdomains of breast cancer cells after omega-3 PUFA incorporation. Cell Biochem. Biophys. 2012, 64, 45–59. [Google Scholar] [CrossRef]
  75. Shaikh, S.R.; Rockett, B.D.; Salameh, M.; Carraway, K. Docosahexaenoic acid modifies the clustering and size of lipid rafts and the lateral organization and surface expression of MHC class I of EL4 cells. J. Nutr. 2009, 139, 1632–1639. [Google Scholar] [CrossRef]
  76. Xu, M.; Wang, X.; Li, Y.; Geng, X.; Jia, X.; Zhang, L.; Yang, H. Arachidonic Acid Metabolism Controls Macrophage Alternative Activation Through Regulating Oxidative Phosphorylation in PPARγ Dependent Manner. Front. Immunol. 2021, 12, 618501. [Google Scholar] [CrossRef]
  77. Zhuang, P.; Shou, Q.; Lu, Y.; Wang, G.; Qiu, J.; Wang, J.; He, L.; Chen, J.; Jiao, J.; Zhang, Y. Arachidonic acid sex-dependently affects obesity through linking gut microbiota-driven inflammation to hypothalamus-adipose-liver axis. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 2715–2726. [Google Scholar] [CrossRef]
  78. Joffre, C.; Rey, C.; Layé, S. N-3 Polyunsaturated Fatty Acids and the Resolution of Neuroinflammation. Front. Pharmacol. 2019, 10, 1022. [Google Scholar] [CrossRef]
  79. Garcia Corrales, A.V.; Haidar, M.; Bogie, J.F.J.; Hendriks, J.J.A. Fatty acid synthesis in glial cells of the cns. Int. J. Mol. Sci. 2021, 22, 8159. [Google Scholar] [CrossRef]
  80. Pifferi, F.; Laurent, B.; Plourde, M. Lipid Transport and Metabolism at the Blood-Brain Interface: Implications in Health and Disease. Front. Physiol. 2021, 12, 645646. [Google Scholar] [CrossRef]
  81. Aizawa, F.; Nishinaka, T.; Yamashita, T.; Nakamoto, K.; Koyama, Y.; Kasuya, F.; Tokuyama, S. Astrocytes release polyunsaturated fatty acids by lipopolysaccharide stimuli. Biol. Pharm. Bull. 2016, 39, 1100–1106. [Google Scholar] [CrossRef]
  82. Ji, A.; Diao, H.; Wang, X.; Yang, R.; Zhang, J.; Luo, W.; Cao, R.; Cao, Z.; Wang, F.; Cai, T. N-3 polyunsaturated fatty acids inhibit lipopolysaccharide-induced microglial activation and dopaminergic injury in rats. NeuroToxicology 2012, 33, 780–788. [Google Scholar] [CrossRef]
  83. Choi, J.Y.; Jang, J.S.; Son, D.J.; Im, H.S.; Kim, J.Y.; Park, J.E.; Choi, W.R.; Han, S.B.; Hong, J.T. Antarctic krill oil diet protects against lipopolysaccharide-induced oxidative stress, neuroinflammation and cognitive impairment. Int. J. Mol. Sci. 2017, 18, 2554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Tyrtyshnaia, A.; Konovalova, S.; Bondar, A.; Ermolenko, E.; Sultanov, R.; Manzhulo, I. Anti-inflammatory activity of N-docosahexaenoylethanolamine and n-eicosapentaenoylethanolamine in a mouse model of lipopolysaccharide-induced neuroinflammation. Int. J. Mol. Sci. 2021, 22, 10728. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, Z.; Liu, D.; Wang, F.; Liu, S.; Zhao, S.; Ling, E.A.; Hao, A. Saturated fatty acids activate microglia via Toll-like receptor 4/NF-κB signalling. Br. J. Nutr. 2012, 107, 229–241. [Google Scholar] [CrossRef] [PubMed]
  86. Gupta, S.; Knight, A.G.; Gupta, S.; Keller, J.N.; Bruce-Keller, A.J. Saturated long-chain fatty acids activate inflammatory signaling in astrocytes. J. Neurochem. 2012, 120, 1060–1071. [Google Scholar] [CrossRef] [PubMed]
  87. Moon, D.O.; Kim, K.C.; Jin, C.Y.; Han, M.H.; Park, C.; Lee, K.J.; Park, Y.M.; Choi, Y.H.; Kim, G.Y. Inhibitory effects of eicosapentaenoic acid on lipopolysaccharide-induced activation in BV2 microglia. Int. Immunopharmacol. 2007, 7, 222–229. [Google Scholar] [CrossRef]
  88. Beaulieu, J.; Costa, G.; Renaud, J.; Moitié, A.; Glémet, H.; Sergi, D.; Martinoli, M.G. The Neuroinflammatory and Neurotoxic Potential of Palmitic Acid Is Mitigated by Oleic Acid in Microglial Cells and Microglial-Neuronal Co-cultures. Mol. Neurobiol. 2021, 58, 3000–3014. [Google Scholar] [CrossRef]
  89. Lu, D.Y.; Tsao, Y.Y.; Leung, Y.M.; Su, K.P. Docosahexaenoic acid suppresses neuroinflammatory responses and induces heme oxygenase-1 expression in BV-2 microglia: Implications of antidepressant effects for omega-3 fatty acids. Neuropsychopharmacology 2010, 35, 2238–2248. [Google Scholar] [CrossRef]
  90. Hadad, N.; Levy, R. Combination of EPA with Carotenoids and Polyphenol Synergistically Attenuated the Transformation of Microglia to M1 Phenotype Via Inhibition of NF-κB. NeuroMolecular Med. 2017, 19, 436–451. [Google Scholar] [CrossRef]
  91. Liu, B.; Zhang, Y.; Yang, Z.; Liu, M.; Zhang, C.; Zhao, Y.; Song, C. ω-3 DPA Protected Neurons from Neuroinflammation by Balancing Microglia M1/M2 Polarizations through Inhibiting NF-κB/MAPK p38 Signaling and Activating Neuron-BDNF-PI3K/AKT Pathways. Mar. Drugs 2021, 19, 587. [Google Scholar] [CrossRef]
  92. Antonietta Ajmone-Cat, M.; Lavinia Salvatori, M.; de Simone, R.; Mancini, M.; Biagioni, S.; Bernardo, A.; Cacci, E.; Minghetti, L. Docosahexaenoic acid modulates inflammatory and antineurogenic functions of activated microglial cells. J. Neurosci. Res. 2012, 90, 575–587. [Google Scholar] [CrossRef]
  93. Dong, Y.; Xu, M.; Kalueff, A.V.; Song, C. Dietary eicosapentaenoic acid normalizes hippocampal omega-3 and 6 polyunsaturated fatty acid profile, attenuates glial activation and regulates BDNF function in a rodent model of neuroinflammation induced by central interleukin-1β administration. Eur. J. Nutr. 2018, 57, 1781–1791. [Google Scholar] [CrossRef]
  94. Manzhulo, O.; Tyrtyshnaia, A.; Kipryushina, Y.; Dyuizen, I.; Manzhulo, I. Docosahexaenoic acid induces changes in microglia/macrophage polarization after spinal cord injury in rats. Acta Histochem. 2018, 120, 741–747. [Google Scholar] [CrossRef]
  95. Chang, C.Y.; Wu, C.C.; Wang, J.D.; Li, J.R.; Wang, Y.Y.; Lin, S.Y.; Chen, W.Y.; Liao, S.L.; Chen, C.J. DHA attenuated Japanese Encephalitis virus infection-induced neuroinflammation and neuronal cell death in cultured rat Neuron/glia. Brain Behav. Immun. 2021, 93, 194–205. [Google Scholar] [CrossRef]
  96. Talamonti, E.; Sasso, V.; To, H.; Haslam, R.P.; Napier, J.A.; Ulfhake, B.; Pernold, K.; Asadi, A.; Hessa, T.; Jacobsson, A.; et al. Impairment of DHA synthesis alters the expression of neuronal plasticity markers and the brain inflammatory status in mice. FASEB J. 2020, 34, 2024–2040. [Google Scholar] [CrossRef]
  97. Willis, L.M.; Bielinski, D.F.; Fisher, D.R.; Matthan, N.R.; Joseph, J.A. Walnut extract inhibits LPS-induced activation of Bv-2 microglia via internalization of TLR4: Possible involvement of phospholipase D2. Inflammation 2010, 33, 325–333. [Google Scholar] [CrossRef]
  98. Carey, A.N.; Fisher, D.R.; Bielinski, D.F.; Cahoon, D.S.; Shukitt-Hale, B. Walnut-Associated Fatty Acids Inhibit LPS-Induced Activation of BV-2 Microglia. Inflammation 2020, 43, 241–250. [Google Scholar] [CrossRef]
  99. Lowry, J.R.; Marshall, N.; Wenzel, T.J.; Murray, T.E.; Klegeris, A. The dietary fatty acids α-linolenic acid (ALA) and linoleic acid (LA) selectively inhibit microglial nitric oxide production. Mol. Cell. Neurosci. 2020, 109, 103569. [Google Scholar] [CrossRef]
  100. Inoue, T.; Tanaka, M.; Masuda, S.; Ohue-Kitano, R.; Yamakage, H.; Muranaka, K.; Wada, H.; Kusakabe, T.; Shimatsu, A.; Hasegawa, K.; et al. Omega-3 polyunsaturated fatty acids suppress the inflammatory responses of lipopolysaccharide-stimulated mouse microglia by activating SIRT1 pathways. Biochim. Et Biophys. Acta—Mol. Cell Biol. Lipids 2017, 1862, 552–560. [Google Scholar] [CrossRef]
  101. Yang, Z.H.; Amar, M.; Sampson, M.; Courville, A.B.; Sorokin, A.V.; Gordon, S.M.; Aponte, A.M.; Stagliano, M.; Playford, M.P.; Fu, Y.P.; et al. Comparison of Omega-3 Eicosapentaenoic Acid Versus Docosahexaenoic Acid-Rich Fish Oil Supplementation on Plasma Lipids and Lipoproteins in Normolipidemic Adults. Nutrients 2020, 12, 749. [Google Scholar] [CrossRef]
  102. Yang, B.; Ren, X.L.; Li, Z.H.; Shi, M.Q.; Ding, F.; Su, K.P.; Guo, X.J.; Li, D. Lowering effects of fish oil supplementation on proinflammatory markers in hypertension: Results from a randomized controlled trial. Food Funct. 2020, 11, 1779–1789. [Google Scholar] [CrossRef]
  103. Li, L.; Wu, Y.; Wang, Y.; Wu, J.; Song, L.; Xian, W.; Yuan, S.; Pei, L.; Shang, Y. Resolvin D1 promotes the interleukin-4-induced alternative activation in BV-2 microglial cells. J. Neuroinflammation 2014, 11, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Krashia, P.; Cordella, A.; Nobili, A.; La Barbera, L.; Federici, M.; Leuti, A.; Campanelli, F.; Natale, G.; Marino, G.; Calabrese, V.; et al. Blunting neuroinflammation with resolvin D1 prevents early pathology in a rat model of Parkinson’s disease. Nat. Commun. 2019, 10, 3945. [Google Scholar] [CrossRef] [PubMed]
  105. Zamanian, J.L.; Xu, L.; Foo, L.C.; Nouri, N.; Zhou, L.; Giffard, R.G.; Barres, B.A. Genomic analysis of reactive astrogliosis. J. Neurosci. 2012, 32, 6391–6410. [Google Scholar] [CrossRef] [PubMed]
  106. Gotoh, M.; Miyamoto, Y.; Ikeshima-Kataoka, H. Astrocytic Neuroimmunological Roles Interacting with Microglial Cells in Neurodegenerative Diseases. Int. J. Mol. Sci. 2023, 24, 1599. [Google Scholar] [CrossRef]
  107. Zgorzynska, E.; Dziedzic, B.; Markiewicz, M.; Walczewska, A. Omega-3 pufas suppress il-1β-induced hyperactivity of immunoproteasomes in astrocytes. Int. J. Mol. Sci. 2021, 22, 5410. [Google Scholar] [CrossRef]
  108. Badia-Soteras, A.; de Vries, J.; Dykstra, W.; Broersen, L.M.; Verkuyl, J.M.; Smit, A.B.; Verheijen, M.H.G. High-Throughput Analysis of Astrocyte Cultures Shows Prevention of Reactive Astrogliosis by the Multi-Nutrient Combination Fortasyn Connect. Cells 2022, 11, 1428. [Google Scholar] [CrossRef]
  109. Wyss-Coray, T.; Rogers, J. Inflammation in Alzheimer disease-A brief review of the basic science and clinical literature. Cold Spring Harb. Perspect. Med. 2012, 2, a006346. [Google Scholar] [CrossRef]
  110. Villegas-Llerena, C.; Phillips, A.; Garcia-Reitboeck, P.; Hardy, J.; Pocock, J.M. Microglial genes regulating neuroinflammation in the progression of Alzheimer’s disease. Curr. Opin. Neurobiol. 2016, 36, 74–81. [Google Scholar] [CrossRef]
  111. Venigalla, M.; Sonego, S.; Gyengesi, E.; Sharman, M.J.; Münch, G. Novel promising therapeutics against chronic neuroinflammation and neurodegeneration in Alzheimer’s disease. Neurochem. Int. 2016, 95, 63–74. [Google Scholar] [CrossRef]
  112. Oksman, M.; Iivonen, H.; Hogyes, E.; Amtul, Z.; Penke, B.; Leenders, I.; Broersen, L.; Lütjohann, D.; Hartmann, T.; Tanila, H. Impact of different saturated fatty acid, polyunsaturated fatty acid and cholesterol containing diets on beta-amyloid accumulation in APP/PS1 transgenic mice. Neurobiol. Dis. 2006, 23, 563–572. [Google Scholar] [CrossRef]
  113. Van de Rest, O.; Berendsen, A.A.M.; Haveman-Nies, A.; de Groot, L.C.P.G.M. Dietary patterns, cognitive decline, and dementia: A systematic review. Adv. Nutr. 2015, 6, 154–168. [Google Scholar] [CrossRef] [Green Version]
  114. Ma, Q.L.; Zhu, C.; Morselli, M.; Su, T.; Pelligrini, M.; Lu, Z.; Jones, M.; Denver, P.; Castro, D.; Gu, X.; et al. The Novel Omega-6 Fatty Acid Docosapentaenoic Acid Positively Modulates Brain Innate Immune Response for Resolving Neuroinflammation at Early and Late Stages of Humanized APOE-Based Alzheimer’s Disease Models. Front. Immunol. 2020, 11, 558036. [Google Scholar] [CrossRef]
  115. Gao, J.; Wang, L.; Zhao, C.; Wu, Y.; Lu, Z.; Gu, Y.; Ba, Z.; Wang, X.; Wang, J.; Xu, Y. Peony seed oil ameliorates neuroinflammation-mediated cognitive deficits by suppressing microglial activation through inhibition of NF-kappa B pathway in presenilin 1/2 conditional double knockout mice. J. Leukoc. Biol. 2021, 110, 1005–1022. [Google Scholar] [CrossRef]
  116. Moura, E.L.R.; Dos Santos, H.; Celes, A.P.M.; Bassani, T.B.; Souza, L.C.; Vital, M.A.B.F. Effects of a Nutritional Formulation Containing Caprylic and Capric Acid, Phosphatidylserine, and Docosahexaenoic Acid in Streptozotocin-Lesioned Rats. J. Alzheimer’s Dis. Rep. 2020, 4, 353–363. [Google Scholar] [CrossRef]
  117. Fujita, Y.; Kano, K.; Kishino, S.; Nagao, T.; Shen, X.; Sato, C.; Hatakeyama, H.; Ota, Y.; Niibori, S.; Nomura, A.; et al. Dietary cis-9, trans-11-conjugated linoleic acid reduces amyloid β-protein accumulation and upregulates anti-inflammatory cytokines in an Alzheimer’s disease mouse model. Sci. Rep. 2021, 11, 9749. [Google Scholar] [CrossRef]
  118. Geng, X.; Yang, B.; Li, R.; Teng, T.; Ladu, M.J.; Sun, G.Y.; Greenlief, C.M.; Lee, J.C. Effects of Docosahexaenoic Acid and Its Peroxidation Product on Amyloid-β Peptide-Stimulated Microglia. Mol. Neurobiol. 2020, 57, 1085–1098. [Google Scholar] [CrossRef]
  119. Minogue, A.M.; Lynch, A.M.; Loane, D.J.; Herron, C.E.; Lynch, M.A. Modulation of amyloid-β-induced and age-associated changes in rat hippocampus by eicosapentaenoic acid. J. Neurochem. 2007, 103, 914–926. [Google Scholar] [CrossRef]
  120. Kelly, L.; Grehan, B.; Chiesa, A.D.; O’Mara, S.M.; Downer, E.; Sahyoun, G.; Massey, K.A.; Nicolaou, A.; Lynch, M.A. The polyunsaturated fatty acids, EPA and DPA exert a protective effect in the hippocampus of the aged rat. Neurobiol. Aging 2011, 32, e2311–e2318. [Google Scholar] [CrossRef]
  121. Lynch, A.M.; Loane, D.J.; Minogue, A.M.; Clarke, R.M.; Kilroy, D.; Nally, R.E.; Roche, Ó.J.; O’Connell, F.; Lynch, M.A. Eicosapentaenoic acid confers neuroprotection in the amyloid-β challenged aged hippocampus. Neurobiol. Aging 2007, 28, 845–855. [Google Scholar] [CrossRef]
  122. Ivkovic, S.; Major, T.; Mitic, M.; Loncarevic-Vasiljkovic, N.; Jovic, M.; Adzic, M. Fatty acids as biomodulators of Piezo1 mediated glial mechanosensitivity in Alzheimer’s disease. Life Sci. 2022, 297, 120470. [Google Scholar] [CrossRef]
  123. Hjorth, E.; Zhu, M.; Toro, V.C.; Vedin, I.; Palmblad, J.; Cederholm, T.; Freund-Levi, Y.; Faxen-Irving, G.; Wahlund, L.O.; Basun, H.; et al. Omega-3 fatty acids enhance phagocytosis of alzheimer’s disease-related amyloid-β42 by human microglia and decrease inflammatory markers. J. Alzheimer’s Dis. 2013, 35, 697–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Desale, S.E.; Chinnathambi, S. α– Linolenic acid modulates phagocytosis and endosomal pathways of extracellular Tau in microglia. Cell Adhes. Migr. 2021, 15, 84–100. [Google Scholar] [CrossRef] [PubMed]
  125. Litwiniuk, A.; Domańska, A.; Chmielowska, M.; Martyńska, L.; Bik, W.; Kalisz, M.; Mendonça Junior, F.J.B. The Effects of Alpha-Linolenic Acid on the Secretory Activity of Astrocytes and β Amyloid-Associated Neurodegeneration in Differentiated SH-SY5Y Cells: Alpha-Linolenic Acid Protects the SH-SY5Y cells against β Amyloid Toxicity. Oxidative Med. Cell. Longev. 2020, 2020, 8908901. [Google Scholar] [CrossRef] [PubMed]
  126. Ren, H.X.; Luo, C.M.; Feng, Y.Q.; Yao, X.L.; Shi, Z.; Liang, F.Y.; Kang, J.X.; Wan, J.B.; Pei, Z.; Su, H.X. Omega-3 polyunsaturated fatty acids promote amyloid-beta clearance from the brain through mediating the function of the glymphatic system. Faseb J. 2017, 31, 282–293. [Google Scholar] [CrossRef]
  127. Zhu, M.; Wang, X.; Hjorth, E.; Colas, R.A.; Schroeder, L.; Granholm, A.C.; Serhan, C.N.; Schultzberg, M. Pro-Resolving Lipid Mediators Improve Neuronal Survival and Increase Aβ42 Phagocytosis. Mol. Neurobiol. 2016, 53, 2733–2749. [Google Scholar] [CrossRef]
  128. Wang, Y.; Leppert, A.; Tan, S.; van der Gaag, B.; Li, N.; Schultzberg, M.; Hjorth, E. Maresin 1 attenuates pro-inflammatory activation induced by β-amyloid and stimulates its uptake. J. Cell. Mol. Med. 2021, 25, 434–447. [Google Scholar] [CrossRef]
  129. Yin, P.; Wang, X.; Wang, S.; Wei, Y.; Feng, J.; Zhu, M. Maresin 1 Improves Cognitive Decline and Ameliorates Inflammation in a Mouse Model of Alzheimer’s Disease. Front. Cell. Neurosci. 2019, 13, 466. [Google Scholar] [CrossRef]
  130. Kantarci, A.; Aytan, N.; Palaska, I.; Stephens, D.; Crabtree, L.; Benincasa, C.; Jenkins, B.G.; Carreras, I.; Dedeoglu, A. Combined administration of resolvin E1 and lipoxin A4 resolves inflammation in a murine model of Alzheimer’s disease. Exp. Neurol. 2018, 300, 111–120. [Google Scholar] [CrossRef]
  131. Emre, C.; Arroyo-García, L.E.; Do, K.V.; Jun, B.; Ohshima, M.; Alcalde, S.G.; Cothern, M.L.; Maioli, S.; Nilsson, P.; Hjorth, E.; et al. Intranasal delivery of pro-resolving lipid mediators rescues memory and gamma oscillation impairment in App NL-G-F/NL-G-F mice. Commun. Biol. 2022, 5, 245. [Google Scholar] [CrossRef]
  132. Little, J.P.; Madeira, J.M.; Klegeris, A. The saturated fatty acid palmitate induces human monocytic cell toxicity toward neuronal cells: Exploring a possible link between obesity-related metabolic impairments and neuroinflammation. J. Alzheimer’s Dis. 2012, 30, 179–183. [Google Scholar] [CrossRef]
  133. Chen, S.; Zhang, H.; Pu, H.; Wang, G.; Li, W.; Leak, R.K.; Chen, J.; Liou, A.K.; Hu, X. N-3 PUFA supplementation benefits microglial responses to myelin pathology. Sci. Rep. 2014, 4, 7458. [Google Scholar] [CrossRef] [Green Version]
  134. Poisson, L.M.; Suhail, H.; Singh, J.; Datta, I.; Deni, A.; Labuzek, K.; Hoda, N.; Shankar, A.; Kumar, A.; Cerghet, M.; et al. Untargeted plasma metabolomics identifies endogenous metabolite with drug-like properties in chronic animal model of multiple sclerosis. J. Biol. Chem. 2015, 290, 30697–30712. [Google Scholar] [CrossRef]
  135. Mancera, P.; Wappenhans, B.; Cordobilla, B.; Virgili, N.; Pugliese, M.; Rueda, F.; Espinosa-Parrilla, J.F.; Domingo, J.C. Natural docosahexaenoic acid in the triglyceride form attenuates in vitro microglial activation and ameliorates autoimmune encephalomyelitis in mice. Nutrients 2017, 9, 681. [Google Scholar] [CrossRef]
  136. Cai, W.; Liu, S.; Hu, M.; Sun, X.; Qiu, W.; Zheng, S.; Hu, X.; Lu, Z. Post-stroke DHA Treatment Protects Against Acute Ischemic Brain Injury by Skewing Macrophage Polarity Toward the M2 Phenotype. Transl. Stroke Res. 2018, 9, 669–680. [Google Scholar] [CrossRef]
  137. Jiang, W.; Whellan, D.J.; Adams, K.F.; Babyak, M.A.; Boyle, S.H.; Wilson, J.L.; Patel, C.B.; Rogers, J.G.; Harris, W.S.; O’Connor, C.M. Long-Chain Omega-3 Fatty Acid Supplements in Depressed Heart Failure Patients: Results of the OCEAN Trial. JACC Heart Fail. 2018, 6, 833–843. [Google Scholar] [CrossRef]
  138. Okabe, N.; Nakamura, T.; Toyoshima, T.; Miyamoto, O.; Lu, F.; Itano, T. Eicosapentaenoic acid prevents memory impairment after ischemia by inhibiting inflammatory response and oxidative damage. J. Stroke Cerebrovasc. Dis. 2011, 20, 188–195. [Google Scholar] [CrossRef]
  139. Lalancette-Hébert, M.; Julien, C.; Cordeau, P.; Bohacek, I.; Weng, Y.C.; Calon, F.; Kriz, J. Accumulation of dietary docosahexaenoic acid in the brain attenuates acute immune response and development of postischemic neuronal damage. Stroke 2011, 42, 2903–2909. [Google Scholar] [CrossRef]
  140. Eady, T.N.; Belayev, L.; Khoutorova, L.; Atkins, K.D.; Zhang, C.; Bazan, N.G. Docosahexaenoic Acid Signaling Modulates Cell Survival in Experimental Ischemic Stroke Penumbra and Initiates Long-Term Repair in Young and Aged Rats. PLoS ONE 2012, 7, e46151. [Google Scholar] [CrossRef]
  141. Eady, T.N.; Khoutorova, L.; Obenaus, A.; Mohd-Yusof, A.; Bazan, N.G.; Belayev, L. Docosahexaenoic acid complexed to albumin provides neuroprotection after experimental stroke in aged rats. Neurobiol. Dis. 2014, 62, 1–7. [Google Scholar] [CrossRef]
  142. Belayev, L.; Hong, S.H.; Freitas, R.S.; Menghani, H.; Marcell, S.J.; Khoutorova, L.; Mukherjee, P.K.; Reid, M.M.; Oria, R.B.; Bazan, N.G. DHA modulates MANF and TREM2 abundance, enhances neurogenesis, reduces infarct size, and improves neurological function after experimental ischemic stroke. CNS Neurosci. Ther. 2020, 26, 1155–1167. [Google Scholar] [CrossRef]
  143. Black, E.K.; Phillips, J.K.; Seminetta, J.; Bailes, J.; Lee, J.M.; Finan, J.D. The effect of dietary supplementation with high- or low-dose omega-3 fatty acid on inflammatory pathology after traumatic brain injury in rats. Transl. Neurosci. 2021, 12, 76–82. [Google Scholar] [CrossRef] [PubMed]
  144. Harvey, L.D.; Yin, Y.; Attarwala, I.Y.; Begum, G.; Deng, J.; Yan, H.Q.; Dixon, C.E.; Sun, D. Administration of DHA reduces endoplasmic reticulum stress-associated inflammation and alters microglial or macrophage activation in traumatic brain injury. ASN Neuro 2015, 7, 1759091415618969. [Google Scholar] [CrossRef] [PubMed]
  145. Yin, Y.; Li, E.; Sun, G.; Yan, H.Q.; Foley, L.M.; Andrzejczuk, L.A.; Attarwala, I.Y.; Hitchens, T.K.; Kiselyov, K.; Dixon, C.E.; et al. Effects of DHA on Hippocampal Autophagy and Lysosome Function After Traumatic Brain Injury. Mol. Neurobiol. 2018, 55, 2454–2470. [Google Scholar] [CrossRef] [PubMed]
  146. Ren, Z.; Chen, L.; Wang, Y.; Wei, X.; Zeng, S.; Zheng, Y.; Gao, C.; Liu, H. Activation of the Omega-3 Fatty Acid Receptor GPR120 Protects against Focal Cerebral Ischemic Injury by Preventing Inflammation and Apoptosis in Mice. J. Immunol. 2019, 202, 747–759. [Google Scholar] [CrossRef]
  147. Liu, G.J.; Tao, T.; Wang, H.; Zhou, Y.; Gao, X.; Gao, Y.Y.; Hang, C.H.; Li, W. Functions of resolvin D1-ALX/FPR2 receptor interaction in the hemoglobin-induced microglial inflammatory response and neuronal injury. J. Neuroinflammation 2020, 17, 239. [Google Scholar] [CrossRef]
  148. Mastromarino, M.; Favia, M.; Schepetkin, I.A.; Kirpotina, L.N.; Trojan, E.; Niso, M.; Carrieri, A.; Leśkiewicz, M.; Regulska, M.; Darida, M.; et al. Design, Synthesis, Biological Evaluation, and Computational Studies of Novel Ureidopropanamides as Formyl Peptide Receptor 2 (FPR2) Agonists to Target the Resolution of Inflammation in Central Nervous System Disorders. J. Med. Chem. 2022, 65, 5004–5028. [Google Scholar] [CrossRef]
  149. Konuskan, D.B.; Arslan, M.; Oksuz, A. Physicochemical properties of cold pressed sunflower, peanut, rapeseed, mustard and olive oils grown in the Eastern Mediterranean region. Saudi J. Biol. Sci. 2019, 26, 340–344. [Google Scholar] [CrossRef]
  150. Agaev, S.G.; Baida, A.A.; Georgiev, O.V.; Maiorova, O.O.; Mozyrev, A.G. Dielectric Spectroscopy of Vegetable Oils. Russ. J. Appl. Chem. 2020, 93, 748–756. [Google Scholar] [CrossRef]
  151. Alves, A.Q.; da Silva, V.A.; Goes, A.J.S.; Silva, M.S.; de Oliveira, G.G.; Bastos, I.; Neto, A.G.D.; Alves, A.J. The Fatty Acid Composition of Vegetable Oils and Their Potential Use in Wound Care. Adv. Ski. Wound Care 2019, 32, 1–8. [Google Scholar] [CrossRef]
  152. Muradoglu, F.; Oguz, H.I.; Yildiz, K.; Yilmaz, H. Some chemical composition of walnut (Juglans regia L.) selections from Eastern Turkey. Afr. J. Agric. Res. 2010, 5, 2379–2385. [Google Scholar]
  153. Ariel, A.; Li, P.L.; Wang, W.; Tang, W.X.; Fredman, G.; Hong, S.; Gotlinger, K.H.; Serhan, C.N. The docosatriene protectin D1 is produced by TH2 skewing and promotes human T cell apoptosis via lipid raft clustering. J. Biol. Chem. 2005, 280, 43079–43086. [Google Scholar] [CrossRef]
  154. Li, H.; Ruan, X.Z.; Powis, S.H.; Fernando, R.; Mon, W.Y.; Wheeler, D.C.; Moorhead, J.F.; Varghese, Z. EPA and DHA reduce LPS-induced inflammation responses in HK-2 cells: Evidence for a PPAR-gamma-dependent mechanism. Kidney Int. 2005, 67, 867–874. [Google Scholar] [CrossRef] [Green Version]
  155. Zúñiga, J.; Cancino, M.; Medina, F.; Varela, P.; Vargas, R.; Tapia, G.; Videla, L.A.; Fernández, V. N-3 PUFA supplementation triggers PPAR-α activation and PPAR-α/NF-κB interaction: Anti-inflammatory implications in liver ischemia-reperfusion injury. PLoS ONE 2011, 6, e28502. [Google Scholar] [CrossRef]
  156. Magee, P.; Pearson, S.; Whittingham-Dowd, J.; Allen, J. PPARγ as a molecular target of EPA anti-inflammatory activity during TNF-α-impaired skeletal muscle cell differentiation. J. Nutr. Biochem. 2012, 23, 1440–1448. [Google Scholar] [CrossRef]
  157. Kliewer, S.A.; Sundseth, S.S.; Jones, S.A.; Brown, P.J.; Wisely, G.B.; Koble, C.S.; Devchand, P.; Wahli, W.; Willson, T.M.; Lenhard, J.M.; et al. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proc. Natl. Acad. Sci. USA 1997, 94, 4318–4323. [Google Scholar] [CrossRef]
  158. Yang, L.; Yuan, J.; Liu, L.; Shi, C.; Wang, L.; Tian, F.; Liu, F.; Wang, H.; Shao, C.; Zhang, Q.; et al. α-linolenic acid inhibits human renal cell carcinoma cell proliferation through PPAR-γ activation and COX-2 inhibition. Oncol. Lett. 2013, 6, 197–202. [Google Scholar] [CrossRef]
  159. Smith, W.L.; Urade, Y.; Jakobsson, P.J. Enzymes of the cyclooxygenase pathways of prostanoid biosynthesis. Chem. Rev. 2011, 111, 5821–5865. [Google Scholar] [CrossRef]
  160. Moncada, S.; Higgs, E.A.; Vane, J.R. Human arterial and venous tissues generate prostacyclin (prostaglandin x), a potent inhibitor of platelet aggregation. Lancet 1977, 1, 18–20. [Google Scholar] [CrossRef]
  161. Bell, E.; Ponthan, F.; Whitworth, C.; Westermann, F.; Thomas, H.; Redfern, C.P. Cell survival signalling through PPARδ and arachidonic acid metabolites in neuroblastoma. PLoS ONE 2013, 8, e68859. [Google Scholar] [CrossRef]
  162. Moya-Camarena, S.Y.; Vanden Heuvel, J.P.; Blanchard, S.G.; Leesnitzer, L.A.; Belury, M.A. Conjugated linoleic acid is a potent naturally occurring ligand and activator of PPARalpha. J. Lipid Res. 1999, 40, 1426–1433. [Google Scholar] [CrossRef]
  163. Kandel, P.; Semerci, F.; Mishra, R.; Choi, W.; Bajic, A.; Baluya, D.; Ma, L.; Chen, K.; Cao, A.C.; Phongmekhin, T.; et al. Oleic acid is an endogenous ligand of TLX/NR2E1 that triggers hippocampal neurogenesis. Proc. Natl. Acad. Sci. USA 2022, 119, e2023784119. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Fatty acids important for human health.
Figure 1. Fatty acids important for human health.
Biology 12 00279 g001
Figure 2. An oversimplified scheme of plant fatty acid biosynthesis. Phopspholipids PA and PC are shown as examples. G3P—glycerol-3-phosphate, PA—phosphatidic acid, PG—phosphatidyl glycerol, PC—phosphatidylcholine, DAG—diacylglycerol.
Figure 2. An oversimplified scheme of plant fatty acid biosynthesis. Phopspholipids PA and PC are shown as examples. G3P—glycerol-3-phosphate, PA—phosphatidic acid, PG—phosphatidyl glycerol, PC—phosphatidylcholine, DAG—diacylglycerol.
Biology 12 00279 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Coniglio, S.; Shumskaya, M.; Vassiliou, E. Unsaturated Fatty Acids and Their Immunomodulatory Properties. Biology 2023, 12, 279. https://doi.org/10.3390/biology12020279

AMA Style

Coniglio S, Shumskaya M, Vassiliou E. Unsaturated Fatty Acids and Their Immunomodulatory Properties. Biology. 2023; 12(2):279. https://doi.org/10.3390/biology12020279

Chicago/Turabian Style

Coniglio, Salvatore, Maria Shumskaya, and Evros Vassiliou. 2023. "Unsaturated Fatty Acids and Their Immunomodulatory Properties" Biology 12, no. 2: 279. https://doi.org/10.3390/biology12020279

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