Next Article in Journal
The Diverse Binding Modes Explain the Nanomolar Levels of Inhibitory Activities Against 1-Deoxy-d-Xylulose 5-Phosphate Reductoisomerase from Plasmodium falciparum Exhibited by Reverse Hydroxamate Analogs of Fosmidomycin with Varying N-Substituents
Previous Article in Journal
Guava Leaf Extract Exhibits Antimicrobial Activity in Extensively Drug-Resistant (XDR) Acinetobacter baumannii
Previous Article in Special Issue
In Vitro Multi-Bioactive Potential of Enzymatic Hydrolysis of a Non-Toxic Jatropha curcas Cake Protein Isolate
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms Linking Omega-3 Fatty Acids and the Gut–Brain Axis

by
Anna Zinkow
,
Wojciech Grodzicki
,
Malwina Czerwińska
and
Katarzyna Dziendzikowska
*
Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences—SGGW, Nowoursynowska 159C, 02-776 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Submission received: 25 November 2024 / Revised: 20 December 2024 / Accepted: 26 December 2024 / Published: 28 December 2024
(This article belongs to the Special Issue Bioactive Compounds from Functional Foods, 2nd Edition)

Abstract

:
The gut–brain axis (GBA) is a complex communication network connecting the gastrointestinal tract (GIT) and the central nervous system (CNS) through neuronal, endocrine, metabolic, and immune pathways. Omega-3 (n-3) fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are crucial food components that may modulate the function of this axis through molecular mechanisms. Derived mainly from marine sources, these long-chain polyunsaturated fatty acids are integral to cell membrane structure, enhancing fluidity and influencing neurotransmitter function and signal transduction. Additionally, n-3 fatty acids modulate inflammation by altering eicosanoid production, reducing proinflammatory cytokines, and promoting anti-inflammatory mediators. These actions help preserve the integrity of cellular barriers like the intestinal and blood–brain barriers. In the CNS, EPA and DHA support neurogenesis, synaptic plasticity, and neurotransmission, improving cognitive functions. They also regulate the hypothalamic–pituitary–adrenal (HPA) axis by reducing excessive cortisol production, associated with stress responses and mental health disorders. Furthermore, n-3 fatty acids influence the composition and function of the gut microbiota, promoting beneficial bacterial populations abundance that contribute to gut health and improve systemic immunity. Their multifaceted roles within the GBA underscore their significance in maintaining homeostasis and supporting mental well-being.

1. Introduction

The gastrointestinal tract (GIT) plays a key physiological role in nutrient digestion and absorption but is also particularly vulnerable, since the alimentary canal is constantly exposed to external and potentially hazardous factors [1]. Given this dual character, GIT has to be closely monitored by the organism’s master controller—the brain [2]. The exchange of information between the intestines and the central nervous system (CNS) is mutual. It involves a plethora of neuronal, endocrine, metabolic, and immune factors, known collectively as the gut–brain axis (GBA). The GBA is crucial for maintaining homeostasis and mental health [3]. The pathways that form the axis include interactions with the nervous system as well as molecular signals, like microbial metabolites, tight junction protein expression, and cytokines released during inflammation [4]. Dietary factors, particularly omega-3 polyunsaturated fatty acids (PUFAs), significantly modulate the GBA by influencing gut microbiota composition, enhancing intestinal barrier integrity, and supporting neural function, showing the interplay between nutrition and gut–brain health. Based on the position of the first double bond relative to the terminal end of the carbon chain, PUFAs can be classified into two main groups: omega-3 (n-3) and omega-6 (n-6) fatty acids [5]. n-3 Fatty acids are crucial dietary fats derived from plant sources and marine organisms. α-linolenic acid (ALA, C18:3), an 18-carbon chain fatty acid, is the shortest in the n-3 family and is classified as a short-chain polyunsaturated fatty acid (SC-PUFA). It is also the most abundant n-3 fatty acid found in dietary sources, such as flaxseed, chia seeds, walnuts, and rapeseed oil [6]. ALA plays unique roles supporting cardiovascular health by reducing lipids, blood pressure, and inflammation. Additionally, ALA-derived oxylipins promote vascular health, and emerging evidence highlights its potential in improving cognitive function and supporting brain health [7]. Marine n-3 fatty acids, including eicosapentaenoic acid (EPA, C20:5) and docosahexaenoic acid (DHA, C22:6), are classified as a long-chain PUFAs (LC-PUFA) and exhibit greater biological activity than their plant-derived counterparts [8]. These essential fatty acids cannot be synthesized in the human body and must be supplied through the diet [9]. LCPUFAs, including EPA, DHA, and arachidonic acid (AA, C20:4, n-6 fatty acid), are major components of cellular membrane phospholipids (75–88%, depending on the cell type). Reduced dietary intake of n-3 fatty acids, such as ALA, EPA, and DHA, can lead to decreased content of those fatty acids in brain cells and organelles. DHA, in particular, is the most abundant n-3 fatty acid in the CNS, and is especially concentrated in the membrane lipids of gray matter [8,10]. Therefore, it is to be expected that n-3—and LCPUFAs in particular—would play a crucial role in the GBA, maintaining proper functioning of both the gut and the brain.

2. The Gut–Brain Axis: Definition and Overview

The fundamental part of the GBA is the enteric nervous system (ENS), which entails 100 million nerve cells located in two principal plexuses: the submucosal and the myenteric plexuses [11,12]. ENS forms part of the autonomic nervous system (ANS) and regulates processes related to digestion, nutrient absorption, the release of gastrointestinal hormones, and peristalsis. It serves as an intermediary between the GIT and the CNS by detecting diverse stimuli originating from the intestinal lumen and transferring them to the brain [11,12,13]. The bidirectional communication network between the CNS and the GIT involves the ENS and parasympathetic innervation, primarily through the vagus nerve fibers that innervate most of the GT. The vagus nerve, also known as the tenth cranial nerve, plays a central role in the complex communication network between the gut and the brain. This nerve is a branch of the ANS that connects the CNS to the ENS through both afferent and efferent nerve fibers [11,12,13]. Given their multifarious receptors, the constituents of the vagus nerve are sensitive to diverse stimuli, including mechanical tension, hormones, and other chemical incentives. The signals they provide are integrated by the solitary nucleus in the brainstem and elicit a wide array of effects in the brain, stimulating regions related to feeding behavior, anxiety, or emotions [11]. On the other hand, efferent vagal activity has an impact on the gut environment, influencing the immune system and metabolism [11,12].
The CNS also communicates with the GIT through the hypothalamic–pituitary–adrenal (HPA) axis, a key component of the gut–brain communication pathways [11]. The HPA axis activity is initiated by the release of corticotropin-releasing hormone (CRH) from the hypothalamus, which stimulates the anterior pituitary to secrete adrenocorticotropic hormone (ACTH). ACTH, in turn, stimulates the adrenal cortex to release cortisol, a steroid hormone, which plays an important role in regulating metabolism, immune response, and maintaining homeostasis under stress conditions [4,14]. The endocrine factors that influence and regulate the functioning of the GBA include CRH and cortisol. The latter can affect the functioning of immune cells, leading to the production of both proinflammatory cytokines (e.g., tumor necrosis factor-α (TNF-α), interferon-γ (INF-γ), and interleukin 6 (IL-6)) and anti-inflammatory cytokines (e.g., IL-10). Additionally, cortisol increases the permeability of the intestinal barrier, allowing bacterial antigens to pass from the intestinal lumen into the bloodstream, which contributes to the development of systemic inflammation [15].
Gut-associated lymphoid tissue (GALT), which consists of immune cells, is another component of the GBA, positioned at the interface between gut contents and the internal environment of the body [16]. It constitutes 70–80% of the human immune system, highlighting the crucial and sensitive role the intestines play in the body’s defense mechanisms [17]. M cells and dendritic cells interact with luminal antigens, activating T and B lymphocytes in Peyer’s patches [1]. Cytokine release from immune cells and enterocytes triggers an immune response that can extend beyond the GI tract, reaching the CNS via the bloodstream and affecting vagus nerve signaling. The immune cells’ defense is complemented by the protective function of the intestinal barrier [1,11]. The intestinal mucosa functions as a physical and immunological defense barrier, consisting of key components, such as the outer mucus layer with commensal microbiota, antimicrobial peptides, and secretory immunoglobulin A. It also includes a central single layer of epithelial cells and an inner lamina propria containing innate and adaptive immune cells like T cells, B cells, macrophages, and dendritic cells [18].
GALT is also a very important site of interaction with microbial agents and their metabolites [11,16]. Collectively known as the gut microbiota, these microorganisms significantly influence the host’s physiology [19]. Interestingly, the number of microbes within the digestive tract exceeds the number of human cells by a factor of 1.3, which highlights their essential role in both health and disease [20,21]. Intestinal microorganisms produce a number of neurotransmitters and neuromodulators—including serotonin, melatonin, γ-aminobutyric acid (GABA), catecholamines, and histamine—which contribute significantly to the proper functioning of brain areas involved in emotion processing, motor activity, and cognitive skills [21,22]. Intestinal microorganisms synthesize and metabolize tryptophan, producing about 95% of systemic serotonin in the gut, which also participates in gut–brain communication [23,24]. Current research demonstrates that the microbiota not only interacts with the CNS but also shapes its development by influencing growth and maturation of brain cells [11,16,25]. Additionally, alterations in microbiota composition have been associated with the onset of various CNS-related disorders, such as Alzheimer’s disease, Parkinson’s disease, autism, and depression [12].
GALT and gut microbiota collaboratively reinforce the intestinal barrier, contributing to immune protection and maintaining gut homeostasis. Epithelial cells are the primary physical components of the intestinal barrier [26]. Due to the impermeability of cell membranes to hydrophilic solutes without specific transporters, the passage of such molecules through intestinal epithelial cells (IECs) is highly restricted. Lipophilic or larger molecules are primarily absorbed via diffusion and endocytosis that is controlled by junctional complexes, with the most crucial being tight junctions (TJs), adherens junctions (AJs), and desmosomes. TJs, located at the apical region, seal the intercellular space and include proteins such as claudins, occludin, and zonula occludens (ZO)-1 and ZO-2. AJs are positioned below TJs and, together with desmosomes, maintain the epithelial integrity through strong adhesive bonds [27]. The proper functioning of the intestinal barrier is ensured by the tight adhesion of these cells, allowing for transcellular transport and enabling selective absorption.
The GBA also includes the physical barrier that protects the CNS. The blood–brain barrier (BBB) is formed by endothelial cells that are connected by protein junctions [28]. These specialized squamous epithelial cells create a single layer of polarized lining on the inner side of the capillary wall [29]. The intercellular spaces of the capillary endothelium are covered by a dense network of high-resistance junctions, with TJs being the most significant ones. The main components of these TJs are the proteins occludin and claudin [30]. Another set of proteins found in the barrier junctions of the brain are cell adhesion molecules— junctional adhesion molecules (JAM)-A, -B, -C, and -D—with JAM-A believed to influence TJs formation. Along with membrane proteins, the barrier connections also include cytosolic proteins such as ZO-1, -2, and -3 [31]. Additionally, astrocytes contribute to the neurovascular cell complex by forming an extra barrier that circulating blood compounds must cross to reach the brain, enhancing the selectivity of the BBB during periods of increased neuronal activity. Transport of substrates and metabolites is carefully regulated by membrane systems, including the sodium–potassium pump. The BBB serves as a protective barrier against neuroactive substances (e.g., catecholamines) and blood-borne toxins, while also supplying neurons with essential nutrients like glucose and amino acids. Moreover, this barrier protects also against the entry of immune system cells [30].
In summary, multiple communication pathways link the gut and the brain, encompassing systems like the ANS, HPA axis, ENS, intestinal barrier, GALT, microbiota, and the BBB. These systems engage in continuous interaction and information exchange within the GBA (Figure 1) [13].

3. Molecular Mechanisms Linking n-3 Fatty Acids, Microbiota, and Brain Function

3.1. Omega-3 Fatty Acids as Components of Cell Membranes

The multifaceted roles of n-3 fatty acids within biological systems include complex interactions with the GBA. They are integral components of phospholipids in the nerve cell membranes, which enhance their structure and are crucial for their optimal fluidity [32]. This fluidity influences neuronal information transfer by affecting neurotransmitter binding and the speed and integrity of cell signaling [33,34,35]. DHA represents approximately 15% of all fatty acids in the gray matter of the prefrontal cortex, while EPA and n-3 docosapentaenoic acid (DPA) account for only ~1% of brain fatty acids [36,37,38]. However, the hydroxy derivative of DHA (2-hydroxydocosahexaenoic acid, OHDHA) has demonstrated therapeutic potential in treating Alzheimer’s disease in a mouse model. Administration of this DHA derivative increased brain DHA levels and reduced Aβ (amyloid-β) levels as well as Aβ-induced tau phosphorylation—key factors in the progression of the disease. These effects supported neuronal cell membranes, preserving proper synaptic function, which is essential for signaling and membrane stability in the CNS [39]. The mechanism through which neuronal fluidity changes is based on the displacement of cholesterol from the membrane as well as the induction of non-lamellar structure formation in the membrane [40,41,42]. Different lipid concentrations in the cell membrane can alter its fluidity as well as the structure and functioning of embedded proteins, such as enzymes, receptors, and ion channels. It is believed that the incorporation of fatty acids into cell membranes also affects the inflammatory cellular responses [43,44,45]. EPA and DHA compete with dihomogammalinolenic acid and AA for incorporation into the phospholipid membrane, sharing enzymes involved in the eicosanoid production process [46,47]. Metabolic transformations of DHA and EPA carried out by cyclooxygenases (COX), lipoxygenases (LOX), and cytochrome P450 enzymes result in the production of numerous eicosanoids and docosanoids [48,49]. These metabolites influence the brain-derived neurotrophic factor (BDNF), increasing synaptic plasticity and enhancing neurotransmission, thus providing neuroprotective effects, as demonstrated in studies conducted on the multipotent human hippocampal progenitor cell line HPC0A07/03C and the human bone marrow neuroblastoma SH-SY5Y cell line (multipotent human hippocampal progenitor cell line) [48,49,50]. Both animal and human studies indicate that the incorporation of EPA and DHA into cell phospholipids during inflammatory processes is dose-dependent and occurs at the expense of AA content [47,51,52,53,54,55,56]. The role of n-3 fatty acids as components of CNS cellular membranes is summarized in Figure 2.

3.2. Impact of Omega-3 Fatty Acids on Inflammation

The modulation of inflammation by n-3 fatty acids, mainly through their effects on eicosanoid production, illustrates a critical mechanism by which dietary fats influence both neurological health and systemic inflammatory responses [57,58]. Prostaglandins, thromboxanes, leukotrienes, and hydroxyl and hydroxy fatty acids are enzymatic metabolic products of PUFA, known as eicosanoids. These compounds play a pivotal role in the inflammatory processes and neurological pathways within the GBA [59,60]. The structural differences in eicosanoids derived from EPA and AA affect the biological activity of EPA derivatives, as eicosanoid receptors have a lower affinity for the mediators derived from EPA than ARA [61]. An in vitro study on HEK293 cell lines demonstrated 50–80% lower activity of prostaglandin E3 (PGE3) compared to prostaglandin E2 (PGE2) on prostaglandin E2 receptors 1, 2, 3, and 4 (EP1, EP2, EP3, and EP4), indicating nuanced influence of dietary fats on neuroinflammatory responses [62]. Eicosanoids derived from EPA and DHA have anti-inflammatory properties [47,55,58,63]. EPA competes with AA for the cyclooxygenase enzyme system, effectively inhibiting the production of proinflammatory eicosanoids from AA. Both DHA and EPA reduce the release of proinflammatory cytokines, such as interleukin-1β (IL-1β), -2 (IL-2), and -6 (IL-6), along with IFN-γ and TNF-α [57,63,64,65]. These cytokines can affect the CNS both indirectly and directly by reducing the availability of neurotransmitter precursors, influencing their metabolism, transport, and regulation, as well as impacting the HPA axis and mRNA-encoding proteins involved in neurotransmitter metabolism [66,67,68,69]. Flaxseed oil inhibits the synthesis of proinflammatory cytokines, thereby regulating neurotransmitter production and the HPA axis [70]. Marine-derived n-3 fatty acids lead to the production of pro-resolving lipid mediators, such as EPA-derived resolvins (E series), DHA-derived resolvins (D series), and resolvins derived from DPA n-3 (RvDn-3 DPA), along with protectins (also known as neuroprotectins when produced in the nervous tissue) and maresins derived from DHA. Their synthesis occurs through COX and LOX pathways, acting intercellularly [57,71,72,73,74]. The anti-inflammatory actions of n-3 fatty acids involve binding to peroxisome proliferator-activated receptors (PPARs), G-protein–coupled receptor 40 (GPR40), and free fatty acid receptor 4 (FFA4), also known as GPR120, which promote the production of anti-inflammatory lipids—resolvins and protectins. These lipids play a crucial role in inhibiting the activation of key inflammatory regulators, including transcription factor nuclear factor kappa B (NF-kB), IL-1 β, and TNF-α release [75,76,77,78,79]. Supplementation with EPA and DHA in mice has been shown to increase the number of T lymphocytes [80]. Additionally, DHA and EPA are able to inhibit IL-6 and interleukin 8 (IL-8) production stimulated by lipopolysaccharide (LPS) in human endothelial cells. Moreover, EPA has been observed to inhibit TNF-α production by cultured monocytes [81,82]. LC-PUFAs also reduce LPS-induced proinflammatory cytokine production in human blood monocytes and in murine fetal liver-derived macrophages, leading to a decrease in TNF-α level in serum, NF-kB activation, and IL-1 β production by monocytes [82,83,84]. This reduction can trigger the release of substantial amounts of anti-inflammatory factors, such as interleukin-10 (IL-10), from resident macrophages. LC-PUFAs play a critical role in directly modulating cytokine production and immune cell regulation. They also exert broader anti-inflammatory effects through key intracellular signaling pathways, such as NFκB, that influence overall immune response and inflammatory status mainly due to the increased level of inflammatory cytokines, adhesion molecules, and cyclooxygenase-2 (COX-2) [82,85,86]. NFκB is activated by the signaling cascade triggered by external inflammatory stimuli, including endotoxin binding to toll-like receptor (TLR) 4. Upon activation, the NFκB dimer is translocated to the nucleus, where it upregulates gene expression [87,88,89,90].
The documented anti-inflammatory and regulatory capabilities of EPA and DHA on cytokine dynamics and cellular mediator pathways also help preserve the integrity of cellular barriers, such as the BBB, by reducing inflammation-induced disruption and enhancing barrier functions. Transmembrane proteins, including claudin-5 and occludin, along with scaffold proteins, like ZO-1 and ZO-2, participate in the tight junction complex of endothelial cells in the CNS [91,92]. LC-PUFAs indirectly enhance the integrity of the BBB and reduce its permeability by modulating levels of inflammatory cytokines [93,94]. Systemic administration of IL-1β has been demonstrated to cause prolonged BBB disruption, activate matrix metalloproteinase-9, and induce rearrangement of claudin-5 in cerebral vessels following transient middle cerebral artery occlusion in mice [95]. Therefore, it is suggested that n-3 fatty acids may strengthen the integrity of the BBB by downregulating the expression of proinflammatory cytokines. Interestingly, research indicates that certain anti-inflammatory cytokines, including IL-4, IL-10, and IL-13, can also contribute to damaging the BBB, but the precise mechanisms behind these effects are not yet well understood [96]. However, the disruption kinetics of the barrier via IL-4 seems to be similar to that of n-6 PUFAs, such as di-homo-gamma-linolenic acid (DGLA, C20:3 n-6) and AA. In comparison n-3 PUFAs, including EPA and DHA, support epithelial barrier integrity by enhancing trans-epithelial electrical resistance (TER) and significantly reducing IL-4-induced permeability. This suggests that LC-PUFAs play a crucial role in maintaining barrier function, as demonstrated by Willemsen Le et al. [97]. Tight junction complexes actively control paracellular permeability and are sensitive to soluble barrier-disrupting mediators [98,99]. Proteins such as occludin, ZO-1, and claudins play critical roles in regulating the intestinal barrier, controlled by the perijunctional actomyosin ring and myosin light chain kinase [100,101,102]. LC-PUFAs improve trans-epithelial resistance, but the exact mechanism of its impact on this process is not fully understood. Several mechanisms have been proposed to explain the effects of n-3 LC-PUFAs on BBB integrity, particularly their roles in modulating inflammatory pathways and enhancing antioxidant defense systems. Notably, n-3 PUFAs suppress IFN-γ-induced expression of TNF-α, IL-6, nitric oxide synthase (NOS), and COX-2, while promoting the upregulation of heme oxygenase-1 (HO-1) in BV-2 microglia cells. BBB disruption and “leaky gut” phenomenon increase the transfer of neurotoxins into the brain, leading to elevated production of proinflammatory molecules, reactive oxygen species, and increased bacterial adhesion through receptors, thereby disrupting endothelial connections [103]. n-3 Fatty acids, specifically EPA and DHA, have been shown to enhance basal trans-epithelial resistance (TER), thereby reducing the permeability of the gut barrier in human intestinal epithelial cells (T84) in vitro, indirectly through mechanisms involving IL-4. LCPUFAs, such as dihomo-γ-linolenic acid (DGLA), AA, EPA, and DHA, stimulate basal resistance and mitigate IL-4-induced permeability changes [97,104,105]. Supplementation with EPA in rats has been shown to increase the expression of occludin, a protein playing an important role in the intestinal barrier integrity [106,107,108]. These protective effects are associated with support and protection of the tight junction structure and function. Additionally, n-3 fatty acids have been shown to increase the number of goblet cells, promote the growth of beneficial bacteria, and elevate the expression of mucin 2, all of which contribute to improved intestinal barrier integrity [104,109]. Summarized anti-inflammatory effects of n-3 fatty acids in the gut and the brain are shown in Figure 3.

3.3. Impact of Omega-3 Fatty Acids on the Nervous System and Cognitive Functions

Omega-3 fatty acids, mainly DHA and EPA, are essential for maintaining neural integrity and improving cognitive functions, as they serve as integral components of neuron membrane structure and play a critical role in neurogenesis, neural signaling, and neuroprotection. DHA also promotes neuron growth by enhancing protein kinase B signaling [110]. This process requires the accumulation of lipids in newly formed membranes and DHA facilitates the incorporation of cholesterol into structures important for myelination and neurite extension, thereby assisting in the organization of the lipid raft domain of the membrane [111,112,113]. Summarized data presenting the effects of n-3 fatty acids on cognitive functions are presented in Table 1.
Neurogenesis is essential for cognitive function, as it supports brain plasticity and adaptability, fundamental for learning and memory retention. This process is most apparent during prenatal development, but it persists in certain parts of the adult brain, most notably in the hippocampus, a structure crucial for learning and memory formation [114,115]. BDNF is a protein that belongs to the neurotrophin family of growth factors, which are vital for the growth, survival, and differentiation of neuron cells. It plays a key role in neuroplasticity, and it is a critical mediator that links neurogenesis to broader brain functions, particularly those related to learning, memory, and overall cognitive health [116,117,118]. n-3 Fatty acids—especially DHA—increase BDNF levels, thereby influencing the growth, differentiation, and viability of neurons in the hippocampus, playing a significant role in the regulation of various neurotransmitter systems [119,120,121,122]. Direct administration of BDNF to the dorsal hippocampus in rats significantly increased the granule cell layer of the hilar region [123]. Deuterated polyunsaturated fatty acids (D-PUFAs) supplementation has been shown to alleviate cognitive decline through its antioxidant activity and reduction in lipid peroxidation in a Huntington’s disease mouse model [124]. In an animal model of spinal cord injury, DHA treatment following injury led to comprehensive functional improvements, evidenced by enhanced fore and hindlimb locomotion and better motor control in the grid exploration and staircase tests. This was accompanied by structural neural adaptations, including increased synaptophysin density around motor neurons, enhanced cortical synaptogenesis, amplified serotonergic innervation, and increased sprouting of corticospinal axons in both rostral and caudal regions of the injured area [125]. DHA also promotes hippocampal neuron development in vitro by supporting neurite growth and branching and also synaptogenesis. Moreover, in vivo DHA depletion in the fetal hippocampus resulted in inhibition of hippocampal neuron development in culture, which can be reversed with DHA supplementation [126].
n-3 Fatty acids increase synaptic plasticity in hippocampal neurons and improve its glutaminergic activity, which is crucial for cognitive function as it mediates synaptic plasticity and neurotransmission, fundamental for learning, memory formation, and overall brain cell connectivity [127,128,129]. A 6-month randomized controlled trial showed that supplementation at a dose of almost 2 g of EPA and DHA decreases depression symptoms in the Geriatric Depression Scale in geriatric patients and DHA improves cognitive function, i.e., verbal fluency tested with Initial Letter Fluency [130]. In a model of accelerated aging with prediabetic status, dietary enrichment with flaxseed and fish oils enhanced spatial learning and working memory performance in the Morris water maze test through reduction in inflammatory markers and toxic metabolites in the CNS of male rats. Elevated n-3 fatty acids levels in the frontal cortex following supplementation were associated with protective effects against cognitive impairment and reduced depressive-like behaviors linked to gray matter atrophy, suggesting a crucial role of EPA in preventing cognitive decline [131].
The cognitive enhancements provided by n-3 fatty acids primarily stem from their profound impact on cellular structures, which leads to the modulation of the physical aspects of brain health, particularly the stabilization of neuronal membranes and ion channels [132,133,134]. In a randomized, double-blind, controlled trial, healthy older adults who consumed 3.7 g per day of flaxseed oil containing 2.2 g of ALA over 12 weeks demonstrated improved verbal fluency. This improvement is believed to result from changes in neuronal cell membrane structure across broad anatomical regions, enhanced membrane fluidity, and improved intercellular connectivity, which typically decline with age [135]. LC-PUFAs influence the integrity of membrane proteins, including enzymes, receptors, and ion channels. They regulate membrane protein integrity, affecting enzymatic activity, receptor function, and ion channel conductance. Administration of DHA demonstrated neuromodulatory effects via ion channel regulation, subsequently reducing the amplitude of epileptiform discharges in experimental rodent models of seizure activity [134,136]. These changes occurred through sodium channel blockade and neuronal membrane stabilization by suppressing calcium-gated membrane tension, thereby blocking synaptic transmission [132,133,134,136,137,138]. Research indicates that n-3 LC-PUFAs play a crucial role in promoting brain health. They enhance neuroprotective capabilities, support neuroplasticity, and reduce neuroinflammation, all of which are essential for sustaining cognitive functions. Short-term exposure to EPA and DHA reversed spatial working memory deficits in older mice by reducing IL-1β expression in inflammation-associated hippocampi [139]. Similarly, a high-fat diet enriched with flaxseed oil improved spatial learning and working memory in male mice during the Morris water maze test by reducing levels of inflammatory markers and toxic metabolites in the CNS [140].
n-3 Fatty acids contribute to brain health by modulating membrane structures and reducing neuronal damage. Additionally, they directly impact cellular mechanisms that eliminate harmful proteins and facilitate cellular repair, highlighting their potential therapeutic roles in neurodegenerative diseases [141]. DHA and EPA stimulate and increase the expression of insulin-degrading enzyme (IDE) genes, which raises the levels of IDE, the main enzyme responsible for degrading amyloid-beta (Aβ) peptide secreted into the extracellular space of neuronal and microglial cells [142]. The accumulation of Aβ peptides is linked to the development of AD, and the effect of n-3 fatty acids may assist in removing these peptides from the brain and modulating inflammation, thereby supporting brain glial cells [143,144]. Additionally, DHA exhibits a protective effect on dopaminergic neurons in a Parkinson’s disease animal model induced by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [142]. Moreover, another study demonstrated that DHA administration significantly increased TH-positive neurons compared to the control group when exposed to MPTP, suggesting changes in dopaminergic activity [145]. It has been suggested that the DHA may delay or slow the progression of Parkinson disease development by blocking the conversion of MPTP to MPP+ (methylpyridinium ion) or by preventing the uptake of MPP+ into dopaminergic terminals. Furthermore, DHA has been shown to influence synaptic plasticity and cognitive functions by activating syntaxin 3, a plasma membrane protein that plays a significant role in membrane expansion [146,147].
Table 1. Effects of n-3 fatty acids on the CNS and cognitive functions.
Table 1. Effects of n-3 fatty acids on the CNS and cognitive functions.
ModelsType of StudySource and DoseExposureEffect Related to Nervous System and Cognitive FunctionsReference
In vitro model studies
Embryonic neurons from E18 mouse hippocampi pregnant C57/BL6 micein vitroDiet with 2.5 wt% of linolenic acid plus 0.9 wt% DHA 16 days- Increased neurite growth and synaptogenesis
- Enhance glutamatergic synaptic activity
[126]
Hippocampus of Sprague Dawley ratsin vitro50 μM of DHASingle dose- Attenuation of epileptic activity[136]
In vivo model studies
Q140 mouse model of Huntington’s diseasein vivoDeuterium-reinforced D2-Lin (KI D-PUFA) 5 months- Alleviation of cognitive decline[124]
Female Sprague Dawley ratsin vivo5 mL/kg of DHA (i.v. injection) post-injurySingle bolus- Improve locomotion and BBB score
- Enhanced synaptogenesis in cortical neurons
[125]
Male Sprague Dawley rats with prediabetic statusin vivo2% fish oil (EPA + DHA) or 2% flaxseed oil3 months- Improved spatial memory[131]
Adult male Sprague Dawley ratsin vivoDiet with n-6/n-3 PUFA ratio at 6:1 (1.25% DHA, 0.25% EPA)12 days- Enhanced synaptic function underlying learning and memory
- Improved spatial learning
[146]
C57Bl6/J mice 22 months old (aged) in vivoEPA and DHA from tuna oil2 months- Inhibition proinflammatory cytokine expression
- Prevention of morphological alterations in hippocampal tissue
- Amelioration of spatial memory impairments
[139]
Obese male C57BL/6 mice 8 weeks oldin vivon-3 PUFA from linseed oil16 weeks- Improved spatial memory
- Reduced inflammatory markers (TNF-α)
- Decreased toxic metabolite levels in the CNS
[140]
Male C57BL/6 mice model of Parkinson’s disease (3 months old)in vivo36 mg/kg/d of DHA (in corn oil)30 days- Decrease levels and activity of heme oxygenase (HO) in substantia nigra- Decrease in levels of Nuclear Factor E2-related factor 2, HO-1 and HO-2 in substantia nigra[148]
Male C57BL/6 mice model of Parkinson’s disease (10 months old)in vivo36 mg/kg/d of DHA (in corn oil)30 days- Protection against oxidative stress
- Significant increase in TH-positive neurons
[145]
Human subject studies
Healthy older adults 65–80 years oldhuman subjects3.7 g/day of flaxseed oil with 2.2 g of alpha-linolenic acid12 weeks- Improve lexical fluency[135]
Elderly people (over 65 years old)human subjects1.67 g EPA + 0.16 g DHA/d or 1.55 g DHA + 0.40 g EPA/d6 months- Decrease in depression symptoms [130]

3.4. Impact of Omega-3 Fatty Acids on the HPA Axis

As a neuroendocrine factor, cortisol is essential in the stress response and plays a critical role in regulating GBA functions. As a primary stress hormone released by the adrenal glands in response to stress, cortisol forms part of the body’s HPA axis, which is closely connected to the GBA. Dysregulation of this system, particularly HPA axis hyperactivity, has been implicated in the pathophysiology of anxiety, depression, and other stress-related conditions [149,150]. n-3 fatty acids, particularly EPA and DHA, have been shown to regulate the HPA axis by reducing excessive cortisol production. Research has shown a reduced cortisol response to acute mental stress in healthy men who were given 7.2 g/day of fish oil for 3 weeks [151]. Preclinical and clinical data indicate that low plasma levels of n-3 fatty acids are correlated with higher CRH [149] and plasma concentration of cortisol [152,153], while n-3 fatty acid supplementation may decrease CRH expression and corticosterone secretion [154,155]. CRH, crucial in regulating the HPA axis, is produced in neurons within the hypothalamic paraventricular nucleus (PVN), which receives input from the limbic system and the brainstem. This connectivity enables these neurons to respond to both psychological and physical stressors [156]. In vitro studies confirmed that n-3 fatty acid-deficient rats had exaggerated distress behaviours in comparison with rats with the appropriate n-3 fatty acid levels during administration of CRH and were normalized upon restoration of n-3 fatty acid levels [157]. Studies conducted on male Finnish psychiatric patients (with diagnosed depression, alcoholism, or both) and healthy patients without psychiatric disorders have also shown that excessive stress response and HPA hyperactivity may be related to concentrations of brain and plasma neuroactive steroids (NASs) [158]. NASs are produced in the CNS from cholesterol and have the ability to alter neuronal excitability rapidly. In a study carried out on male psychiatric patients from Finland, the authors observed that lower plasma levels of n-3 fatty acids were linked to higher plasma levels of neurosteroids. Specifically, reduced concentrations of DHA and EPA were correlated with increased levels of NASs in healthy control subjects [158]. DHA supplementation reduced stress-related increases in aggression and hostility among Japanese students [159]. The research conducted by Oravcova et al. [160] on patients aged 11–18 confirmed the hypothesis that long-term (12 weeks) supplementation of n-3 fatty acids from fish oil emulsion reduces morning cortisol levels in saliva. These findings are in line with the current understanding of the connection between HPA axis activity and fatty acid metabolism in adults with recurrent depressive disorder [155]. Adults with lower levels of n-3 fatty acids demonstrated HPA axis dysregulation, suggesting that supplementation with these fatty acids could potentially improve both physical and mental health [161]. Notably, dietary supplementation with LC-PUFAs from marine oils has been shown to lower corticosterone levels in rats and to reduce cortisol secretion in both healthy individuals and adults experiencing depression [151,162,163]. Results from a randomized, placebo-controlled trial in midlife adults demonstrated that EPA and DHA supplementation significantly reduced cortisol levels during stress in a dose–response manner. Patients receiving the higher doses of n-3 fatty acids had the lowest overall cortisol levels, while those in the placebo group exhibited the highest levels [164]. Additionally, supplementation with n-3 fatty acids (2400 mg of total omega-3 fatty acids; 1000 mg of EPA and 750 mg of DHA; EPA:DHA ratio of 1.33:1) over a 12-week period was shown to significantly reduce clinical symptoms of depression in older children and adolescents [165]. In adolescent patients, the positive treatment effects from n-3 fatty acid supplementation were also associated with a decrease in various oxidative stress markers, such as 8-isoprostane, advanced oxidation protein products, and nitrotyrosine blood levels, as well as increased Trolox equivalent antioxidant capacity and superoxide dismutase activity [166].
Chronic stress or inflammation can result in the overactivation of the HPA axis. Previous studies have demonstrated increased levels of inflammatory markers, including IL-6 and TNF-α, in numerous chronic diseases such as cardiovascular diseases, obesity, and depression, contributing to their development and progression [167,168,169]. Additionally, HPA axis hyperactivity has been observed in obesity, and increased morning cortisol levels have been reported in individuals with depression [170,171]. Given their anti-inflammatory and immunomodulatory effects, n-3 PUFAs may help mitigate these detrimental processes and thereby potentially alleviate conditions characterized by HPA axis dysregulation [172,173]. n-3 fatty acids can lower HPA axis hyperactivity by decreasing proinflammatory cytokines levels, such as IL-6 and TNF-α, which influence the stress response. By downregulating the proinflammatory cytokine pathways and modulating the stress response, n-3 fatty acids reduce the sensitivity of the HPA axis. This means that the axis is less likely to become overactive in response to minor stressors, potentially lowering the risk of stress-related disorders. In a randomized, double blind, placebo-controlled trial conduced on healthy man, n-3 supplementation in combination with phosphatidylserine significantly improved the functioning of the HPA axis by lowering chronic and acute stress [174]. The authors concluded that individuals experiencing high levels of chronic stress and/or having a dysfunctional HPA axis response could benefit from n-3 phosphatidylserine supplementation. The impact of n-3 fatty acids on regulating the activity of the HPA axis may be related to their anti-inflammatory properties, coupled with the increase in the HPA axis sensitivity to negative feedback [175,176].

3.5. Modulation of the Gut Microbiota by Omega-3 Fatty Acids

The beneficial physiological effects of LC-PUFAs encompass their influence on the composition and function of the gut microbiota, as demonstrated in various studies linking fatty acid intake to microbial diversity and health outcomes. n-3 fatty acids can influence the modulation and abundance of gut bacteria types. At the same time, gut microbiota can affect the absorption and metabolism of these fatty acids. Fish oil, for instance, reduces the growth of Enterobacteria, while increasing that of Bifidobacteria. EPA and DHA are partially metabolized by anaerobic bacteria, such as Bifidobacteria and Lactobacilli, in the distal gut [177]. In a study conducted on gnotobiotic piglets fed with PUFA from seal oil, a significant increase in the number of Lactobacillus paracasei adhering to the jejunal mucosa was shown [178]. Similarly, n-3 fatty acids administered to male transgenic fat-1 mice significantly increased the number and percentage of Bifidobacterium, Akkermansia muciniphila, Lactobacillus, Clostridium clusters IV and XIVa, and Enterococcus faecium in the intestines [179]. The modulatory functions of n-3 fatty acids are also attributed to increased levels of SCFAs. It has been shown that dietary supplementation with DHA and EPA in mice infected with Salmonella increased SCFA fecal content, enhancing resistance against the pathogen [180]. Consuming 3 g per day of DHA and EPA from sardines significantly altered the gut microbiota composition in patients with untreated type 2 diabetes. Specifically, the proportions of Bacteroides/Prevotella increased, while the Firmicutes/Bacteroidetes ratio decreased [181]. In a study of birds, a diet rich in omega-3 PUFA significantly increased the presence of Firmicutes (e.g., Faecalibacterium, Clostridium, and Ruminococcus, all of which are butyrate producers), in the gut microbiota [182]. Additionally, serum levels of omega-3 fatty acids, particularly DHA, were positively correlated with gut microbiome diversity and the abundance of specific bacterial taxa, such as Lachnospiraceae, in a cohort of 876 elderly women, suggesting a potential role for LC-PUFA intake in modulating microbiome composition [183]. A 6-week intervention study further demonstrated that omega-3 supplementation alters gut microbiome composition, increasing the abundance of butyrate-associated Coprococcus spp. and beneficial fermentation products, suggesting a potential prebiotic-like role for omega-3 fatty acids [184].
On the other hand, gut microbiota can both indirectly and directly modulate the absorption, bioavailability, and biotransformation of n-3 fatty acids [185,186,187]. Certain bacterial species, such as Bacillus proteus or Lactobacillus plantarum, convert ALA and linoleic acid (LA) into conjugated linoleic acid (CLA) and conjugated alpha-linolenic acid (CALA), which are then hydrogenated to stearic acid, changing the PUFA content in the brain and heart [188]. A study of mice demonstrated that high tissue levels of n-3 fatty acids were associated with variations in the amounts of Bifidobacterium and Lactobacillus [189]. Conversely, mice fed a diet low in n-3 fatty acids for two generations showed a significant decrease in lactic acid bacteria and an increase in Bifidobacteria in the oral cavity compared to those fed a diet adequate in n-3 fatty acids [190]. While studies suggest that Bifidobacterium can significantly modulate fatty acid metabolism and its absorption by the intestinal epithelium, specific mechanisms underlying this relationship remain unexplained [191,192,193].
n-3 Fatty acids can modulate gut microbiota by either inhibiting the production of proinflammatory mediators or promoting the production of anti-inflammatory mediators. For instance, DHA reduces its activation by lowering IkappaB kinase (IκB) phosphorylation in response to LPS in cultured macrophages and dendritic cells, thereby decreasing NFκB activation [194,195]. Similarly, EPA reduced NFκB activation induced by LPS in human monocytes due to reduced IκB phosphorylation [196]. Furthermore, peroxisome proliferator-activated receptor gamma (PPAR-γ), which functions by inhibiting NFκB translocation to the nucleus, is regulated by the binding of EPA and DHA [197]. This binding interaction significantly influences inflammatory processes [198]. Supplementation with these acids significantly influences changes in the gut microbiota by altering the content of immune cell membranes and influencing proinflammatory signaling pathways. n-3 Fatty acids increase the number of regulatory T lymphocytes (Tregs), thereby reducing inflammatory reactions [199]. n-3 Fatty acids are mainly absorbed in the intestine, where their metabolites can be directly utilized by certain microorganisms. Their protective action on the intestinal mucosa includes increasing its thickness and improving barrier functions [200]. Summarized data presenting the mechanistic effects of LC-PUFAs on gut microbiota, immune function, and intestinal barrier integrity are presented in Table 2.

4. Conclusions

n-3 Fatty acids, particularly EPA and DHA, play a pivotal role in the intricate communication network of the GBA. Their incorporation into cell membranes enhances membrane fluidity, which is essential for optimal neurotransmitter function and efficient signal transduction. By modulating inflammatory responses—reducing proinflammatory cytokines and promoting anti-inflammatory mediators—n-3 fatty acids help preserve the integrity of critical barriers, like the intestinal barrier and the BBB. This not only supports gut health but also protects the CNS from potential neurotoxins and inflammatory agents. In the CNS, EPA and DHA contribute to neurogenesis and synaptic plasticity, thereby enhancing cognitive functions such as learning and memory. They also regulate the HPA axis by mitigating excessive cortisol production, which is often associated with stress responses and mental health disorders, like depression and anxiety. Furthermore, n-3 fatty acids positively influence gut microbiota composition, promoting growth of beneficial bacterial populations that contribute to gut health and systemic immunity. The effects of n-3 fatty acids on the GBA are summarized in Figure 4.
Given the multifaceted benefits of n-3 fatty acids on the GBA, it is recommended to incorporate adequate amounts of EPA and DHA into the diet by consuming marine foods, like fatty fish, or through supplementation. Additionally, based on the significant benefits of n-3 fatty acids on the GBA and overall health, the following nutritional recommendations are proposed:
  • Incorporate fish rich in n-3 fatty acids, such as salmon, mackerel, sardines, trout, and herring, into the diet at least twice a week to boost EPA and DHA intake according to recommendation to achieve essentiality and cardiovascular benefits [201].
  • Consider n-3 fatty acid supplements based on fish oil or algae, especially for individuals with limited access to n-3-rich foods or with dietary restrictions [202].
  • Consume fiber-rich foods, like whole grains, fruits, and vegetables, along with fermented foods like yogurt, kefir, and sauerkraut, to support beneficial gut microbiota that synergizes with n-3 fatty acids [203].
  • Use cooking methods that preserve n-3 content, such as baking, steaming, or grilling, and avoid high-temperature frying, which can oxidize these delicate fats [204].
  • To ensure adequate intake of omega-3 fatty acids, individuals are encouraged to follow national or international dietary guidelines, such as those provided by the World Health Organization, the Institute of Medicine, or the European Food Safety Authority, which offer evidence-based recommendations for maintaining optimal health.

5. Limitations and Current Knowledge Gaps

Despite the existing research on the influence of LC-PUFAs on the GBA, critical gaps in our understanding of the precise mechanisms underlying this interaction persist. For instance, the exact processes by which n-3 PUFAs stabilize physiological barriers—such as the BBB and the intestinal barrier—and how they modulate the activity of both immune and neuronal cells, remain incompletely elucidated. Furthermore, the complex relationships between gut microbiota composition, n-3 fatty acid metabolism, and the regulation of inflammatory states require more investigations to fully clarify their interconnected roles. Despite substantial evidence demonstrating the beneficial effects of LC-PUFAs on the GBA, the major limitation is the difficulty of extrapolating doses and outcomes from in vitro and animal models to human populations. The concentrations of fatty acids used in cell culture studies and the dietary interventions used in animal experiments often exceed physiologically achievable levels in human.

Author Contributions

Conceptualization, K.D.; writing—original draft preparation, A.Z., W.G., M.C. and K.D.; writing—review and editing, W.G. and K.D.; visualization, A.Z. and W.G.; supervision, K.D. 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 conflicts of interest.

References

  1. Shen, L. Functional Morphology of the Gastrointestinal Tract. Mol. Mech. Bact. Infect. Gut 2009, 337, 1–35. [Google Scholar]
  2. Deisseroth, K. A Look Inside the Brain. Sci. Am. 2016, 315, 30. [Google Scholar] [CrossRef] [PubMed]
  3. Lu, S.; Zhao, Q.; Guan, Y.; Sun, Z.; Li, W.; Guo, S.; Zhang, A. The Communication Mechanism of the Gut-Brain Axis and Its Effect on Central Nervous System Diseases: A Systematic Review. Biomed. Pharmacother. 2024, 178, 117207. [Google Scholar] [CrossRef] [PubMed]
  4. Morys, J.; Małecki, A.; Nowacka-Chmielewska, M. Stress and the Gut-Brain Axis: An Inflammatory Perspective. Front. Mol. Neurosci. 2024, 17, 1415567. [Google Scholar] [CrossRef] [PubMed]
  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]
  6. Liput, K.P.; Lepczyński, A.; Ogłuszka, M.; Nawrocka, A.; Poławska, E.; Grzesiak, A.; Ślaska, B.; Pareek, C.S.; Czarnik, U.; Pierzchała, M. Effects of Dietary n–3 and n–6 Polyunsaturated Fatty Acids in Inflammation and Cancerogenesis. Int. J. Mol. Sci. 2021, 22, 6965. [Google Scholar] [CrossRef] [PubMed]
  7. Sala-Vila, A.; Fleming, J.; Kris-Etherton, P.; Ros, E. Impact of α-Linolenic Acid, the Vegetable ω-3 Fatty Acid, on Cardiovascular Disease and Cognition. Adv. Nutr. 2022, 13, 1584–1602. [Google Scholar] [CrossRef] [PubMed]
  8. Chandola, H.M.; Tanna, I. Role of Omega-3 Fatty Acids in Brain and Neurological Health with Special Reference to Clinical Depression. In Omega-3 Fatty Acids in Brain and Neurological Health; Academic Press: Cambridge, MA, USA, 2014; pp. 163–179. ISBN 9780124105270. [Google Scholar] [CrossRef]
  9. Drobner, T.; Braun, T.S.; Kiehntopf, M.; Schlattmann, P.; Lorkowski, S.; Dawczynski, C. Evaluation of Influencing Factors on Metabolism of Land-Based n-3 Poly Unsaturated Fatty Acids—The KoALA Study. Nutrients 2023, 15, 4461. [Google Scholar] [CrossRef] [PubMed]
  10. Yu, H.; Bi, Y.; Ma, W.; He, L.; Yuan, L.; Feng, J.; Xiao, R. Long-Term Effects of High Lipid and High Energy Diet on Serum Lipid, Brain Fatty Acid Composition, and Memory and Learning Ability in Mice. Int. J. Dev. Neurosci. 2010, 28, 271–276. [Google Scholar] [CrossRef]
  11. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.; Sandhu, K.V.; Bastiaanssen, T.F.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
  12. Morais, L.H.; Schreiber, H.L.; Mazmanian, S.K. The Gut Microbiota–Brain Axis in Behaviour and Brain Disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef]
  13. Carabotti, M.; Scirocco, A.; Antonietta Maselli, M.; Severi, C. The Gut-Brain Axis: Interactions between Enteric Microbiota, Central and Enteric Nervous Systems. Ann. Gastroenterol. 2015, 28, 203. [Google Scholar] [PubMed]
  14. Gjerstad, J.K.; Lightman, S.L.; Spiga, F. Role of Glucocorticoid Negative Feedback in the Regulation of HPA Axis Pulsatility. Stress 2018, 21, 403–416. [Google Scholar] [CrossRef] [PubMed]
  15. Gareau, M.G.; Jury, J.; MacQueen, G.; Sherman, P.M.; Perdue, M.H. Probiotic Treatment of Rat Pups Normalises Corticosterone Release and Ameliorates Colonic Dysfunction Induced by Maternal Separation. Gut 2007, 56, 1522–1528. [Google Scholar] [CrossRef] [PubMed]
  16. Liang, S.; Wu, X.; Jin, F. Gut-Brain Psychology: Rethinking Psychology from the Microbiota–Gut–Brain Axis. Front. Integr. Neurosci. 2018, 12, 33. [Google Scholar] [CrossRef] [PubMed]
  17. Kumar, S.; Kumar, A. Microbial Pathogenesis in Inflammatory Bowel Diseases. Microb. Pathog. 2022, 163, 105383. [Google Scholar] [CrossRef] [PubMed]
  18. Vancamelbeke, M.; Vermeire, S. The Intestinal Barrier: A Fundamental Role in Health and Disease. Expert Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef] [PubMed]
  19. Ma, Q.; Xing, C.; Long, W.; Wang, H.Y.; Liu, Q.; Wang, R.-F. Impact of Microbiota on Central Nervous System and Neurological Diseases: The Gut-Brain Axis. J. Neuroinflamm. 2019, 16, 53. [Google Scholar] [CrossRef]
  20. Sender, R.; Fuchs, S.; Milo, R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef]
  21. Bruce-Keller, A.J.; Salbaum, J.M.; Luo, M.; Blanchard, E.; Taylor, C.M.; Welsh, D.A.; Berthoud, H.R. Obese-Type Gut Microbiota Induce Neurobehavioral Changes in the Absence of Obesity. Biol. Psychiatry 2015, 77, 607–615. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef] [PubMed]
  23. Eisenstein, M. Microbiome: Bacterial Broadband. Nature 2016, 533, S104–S106. [Google Scholar] [CrossRef] [PubMed]
  24. Nichols, E.; Szoeke, C.E.I.; Vollset, S.E.; Abbasi, N.; Abd-Allah, F.; Abdela, J.; Aichour, M.T.E.; Akinyemi, R.O.; Alahdab, F.; Asgedom, S.W.; et al. Global, Regional, and National Burden of Alzheimer’s Disease and Other Dementias, 1990–2016: A Systematic Analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88–106. [Google Scholar] [CrossRef] [PubMed]
  25. Erny, D.; De Angelis, A.L.H.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host Microbiota Constantly Control Maturation and Function of Microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
  26. Di Tommaso, N.; Gasbarrini, A.; Ponziani, F.R. Intestinal Barrier in Human Health and Disease. Int. J. Environ. Res. Public Health 2021, 18, 12836. [Google Scholar] [CrossRef]
  27. Ghosh, S.; Whitley, C.S.; Haribabu, B.; Jala, V.R. Regulation of Intestinal Barrier Function by Microbial Metabolites. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 1463–1482. [Google Scholar] [CrossRef]
  28. Ballabh, P.; Braun, A.; Nedergaard, M. The Blood-Brain Barrier: An Overview: Structure, Regulation, and Clinical Implications. Neurobiol. Dis. 2004, 16, 1–13. [Google Scholar] [CrossRef]
  29. Adil, M.S.; Narayanan, S.P.; Somanath, P.R. Cell-Cell Junctions: Structure and Regulation in Physiology and Pathology. Tissue Barriers 2021, 9, 1848212. [Google Scholar] [CrossRef] [PubMed]
  30. Petty, M.A.; Lo, E.H. Junctional Complexes of the Blood-Brain Barrier: Permeability Changes in Neuroinflammation. Prog. Neurobiol. 2002, 68, 311–323. [Google Scholar] [CrossRef] [PubMed]
  31. Serlin, Y.; Shelef, I.; Knyazer, B.; Friedman, A. Anatomy and Physiology of the Blood-Brain Barrier. Semin. Cell Dev. Biol. 2015, 38, 2–6. [Google Scholar] [CrossRef]
  32. Guixà-González, R.; Javanainen, M.; Gómez-Soler, M.; Cordobilla, B.; Domingo, J.C.; Sanz, F.; Pastor, M.; Ciruela, F.; Martinez-Seara, H.; Selent, J. Membrane Omega-3 Fatty Acids Modulate the Oligomerisation Kinetics of Adenosine A2A and Dopamine D2 Receptors. Sci. Rep. 2016, 6, 19839. [Google Scholar] [CrossRef]
  33. Bazan, N.G.; Musto, A.E.; Knott, E.J. Endogenous Signaling by Omega-3 Docosahexaenoic Acid-Derived Mediators Sustains Homeostatic Synaptic and Circuitry Integrity. Mol. Neurobiol. 2011, 44, 216–222. [Google Scholar] [CrossRef] [PubMed]
  34. Heinrichs, S.C. Dietary ω-3 Fatty Acid Supplementation for Optimizing Neuronal Structure and Function. Mol. Nutr. Food Res. 2010, 54, 447–456. [Google Scholar] [CrossRef]
  35. Hennebelle, M.; Champeil-Potokar, G.; Lavialle, M.; Vancassel, S.; Denis, I. Omega-3 Polyunsaturated Fatty Acids and Chronic Stress-Induced Modulations of Glutamatergic Neurotransmission in the Hippocampus. Nutr. Rev. 2014, 72, 99–112. [Google Scholar] [CrossRef]
  36. McNamara, R.K.; Able, J.; Jandacek, R.; Rider, T.; Tso, P.; Eliassen, J.C.; Alfieri, D.; Weber, W.; Jarvis, K.; Delbello, M.P.; et al. Docosahexaenoic Acid Supplementation Increases Prefrontal Cortex Activation during Sustained Attention in Healthy Boys: A Placebo-Controlled, Dose-Ranging, Functional Magnetic Resonance Imaging Study. Am. J. Clin. Nutr. 2010, 91, 1060–1067. [Google Scholar] [CrossRef] [PubMed]
  37. 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]
  38. Carver, J.D.; Benford, V.J.; Han, B.; Cantor, A.B. The Relationship between Age and the Fatty Acid Composition of Cerebral Cortex and Erythrocytes in Human Subjects. Brain Res. Bull. 2001, 56, 79–85. [Google Scholar] [CrossRef] [PubMed]
  39. Torres, M.; Price, S.L.; Fiol-Deroque, M.A.; Marcilla-Etxenike, A.; Ahyayauch, H.; Barceló-Coblijn, G.; Terés, S.; Katsouri, L.; Ordinas, M.; López, D.J.; et al. Membrane Lipid Modifications and Therapeutic Effects Mediated by Hydroxydocosahexaenoic Acid on Alzheimer’s Disease. Biochim. Biophys. Acta Biomembr. 2014, 1838, 1680–1692. [Google Scholar] [CrossRef] [PubMed]
  40. de Santis, A.; Scoppola, E.; Ottaviani, M.F.; Koutsioubas, A.; Barnsley, L.C.; Paduano, L.; D’Errico, G.; Krauss, I.R. Order vs. Disorder: Cholesterol and Omega-3 Phospholipids Determine Biomembrane Organization. Int. J. Mol. Sci. 2022, 2, 5322. [Google Scholar] [CrossRef] [PubMed]
  41. De Santis, A.; Vitiello, G.; Appavou, M.S.; Scoppola, E.; Fragneto, G.; Barnsley, L.C.; Clifton, L.A.; Ottaviani, M.F.; Paduano, L.; Russo Krauss, I.; et al. Not Just a Fluidifying Effect: Omega-3 Phospholipids Induce Formation of Non-Lamellar Structures in Biomembranes. Soft Matter 2020, 16, 10425–10438. [Google Scholar] [CrossRef]
  42. Rubin, D.; Laposata, M. Cellular Interactions between N-6 and n-3 Fatty Acids: A Mass Analysis of Fatty Acid Elongation/Desaturation, Distribution among Complex Lipids, and Conversion to Eicosanoids. J. Lipid Res. 1992, 33, 1431–1440. [Google Scholar] [CrossRef]
  43. Sunshine, H.; Iruela-Arispe, M.L. Membrane Lipids and Cell Signaling. Curr. Opin. Lipidol. 2017, 28, 408–413. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, X.; Hurng, J.; Rateri, D.L.; Daugherty, A.; Schmid-Schönbein, G.W.; Shin, H.Y. Membrane Cholesterol Modulates the Fluid Shear Stress Response of Polymorphonuclear Leukocytes via Its Effects on Membrane Fluidity. Am. J. Physiol. Cell Physiol. 2011, 301, 451–460. [Google Scholar] [CrossRef] [PubMed]
  45. Cammarota, E.; Soriani, C.; Taub, R.; Morgan, F.; Sakai, J.; Veatch, S.L.; Bryant, C.E.; Cicuta, P. Criticality of Plasma Membrane Lipids Reflects Activation State of Macrophage Cells. J. R. Soc. Interface 2020, 17, 163. [Google Scholar] [CrossRef]
  46. Whelan, J. Antagonistic Effects of Dietary Arachidonic Acid and N-3 Polyunsaturated Fatty Acids. J. Nutr. 1996, 126, 1086S–1091S. [Google Scholar] [CrossRef] [PubMed]
  47. Calder, P.C. Polyunsaturated Fatty Acids and Inflammatory Processes: New Twists in an Old Tale. Biochimie 2009, 91, 791–795. [Google Scholar] [CrossRef] [PubMed]
  48. 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] [PubMed]
  49. Fischer, R.; Konkel, A.; Mehling, H.; Blossey, K.; Gapelyuk, A.; Wessel, N.; Von Schacky, C.; Dechend, R.; Muller, D.N.; Rothe, M.; et al. Dietary Omega-3 Fatty Acids Modulate the Eicosanoid Profile in Man Primarily via the CYP-Epoxygenase Pathway. J. Lipid Res. 2014, 55, 1150–1164. [Google Scholar] [CrossRef]
  50. Ceccarini, M.R.; Ceccarelli, V.; Codini, M.; Fettucciari, K.; Calvitti, M.; Cataldi, S.; Albi, E.; Vecchini, A.; Beccari, T. The Polyunsaturated Fatty Acid EPA, but Not DHA, Enhances Neurotrophic Factor Expression through Epigenetic Mechanisms and Protects against Parkinsonian Neuronal Cell Death. Int. J. Mol. Sci. 2022, 23, 16176. [Google Scholar] [CrossRef] [PubMed]
  51. Norris, P.C.; Dennis, E.A. Omega-3 Fatty Acids Cause Dramatic Changes in TLR4 and Purinergic Eicosanoid Signaling. Proc. Natl. Acad. Sci. USA 2012, 109, 8517–8522. [Google Scholar] [CrossRef] [PubMed]
  52. Palombo, J.D.; Demichele, S.J.; Lydon, E.E.; Gregory, T.J.; Banks, P.L.; Forse, R.A.; Bistrian, B.R. Rapid Modulation of Lung and Liver Macrophage Phospholipid Fatty Acids in Endotoxemic Rats by Continuous Enteral Feeding with N-3 and y-Linolenic Fatty Acids. Am. J. Clin. Nutr. 1996, 63, 208–219. [Google Scholar] [CrossRef] [PubMed]
  53. Hayashi, D.; Mouchlis, V.D.; Dennis, E.A. Omega-3 versus Omega-6 Fatty Acid Availability Is Controlled by Hydrophobic Site Geometries of Phospholipase A2s. J. Lipid Res. 2021, 62, 100113. [Google Scholar] [CrossRef]
  54. Nieves, D.; Moreno, J.J. Effect of Arachidonic and Eicosapentaenoic Acid Metabolism on RAW 264.7 Macrophage Proliferation. J. Cell Physiol. 2006, 208, 428–434. [Google Scholar] [CrossRef]
  55. So, J.; Wu, D.; Lichtenstein, A.H.; Tai, A.K.; Matthan, N.R.; Maddipati, K.R.; Lamon-Fava, S. EPA and DHA Differentially Modulate Monocyte Inflammatory Response in Subjects with Chronic Inflammation in Part via Plasma Specialized Pro-Resolving Lipid Mediators: A Randomized, Double-Blind, Crossover Study. Atherosclerosis 2021, 316, 90–98. [Google Scholar] [CrossRef]
  56. Rees, D.; Miles, E.A.; Banerjee, T.; Wells, S.J.; Roynette, C.E.; Wahle, K.W.; Calder, P.C. Dose-Related Effects of Eicosapentaenoic Acid on Innate Immune Function in Healthy Humans: A Comparison of Young and Older Men. Am. J. Clin. Nutr. 2006, 83, 331–342. [Google Scholar] [CrossRef]
  57. Calder, P.C. Omega-3 Fatty Acids and Inflammatory Processes. Nutrients 2010, 2, 355–374. [Google Scholar] [CrossRef]
  58. Wall, R.; Ross, R.P.; Fitzgerald, G.F.; Stanton, C. Fatty Acids from Fish: The Anti-Inflammatory Potential of Long-Chain Omega-3 Fatty Acids. Nutr. Rev. 2010, 68, 280–289. [Google Scholar] [CrossRef]
  59. Calder, P.C. Eicosapentaenoic and Docosahexaenoic Acid Derived Specialised Pro-Resolving Mediators: Concentrations in Humans and the Effects of Age, Sex, Disease and Increased Omega-3 Fatty Acid Intake. Biochimie 2020, 178, 105–123. [Google Scholar] [CrossRef] [PubMed]
  60. Calder, P.C. Dietary Fatty Acids, Lipid Mediators, Immunity, and Inflammation. In Functional Dietary Lipids: Food Formulation, Consumer Issues, and Innovation for Health, 2nd ed.; Woodhead Publishing: Sawston, UK, 2024; pp. 187–214. [Google Scholar] [CrossRef]
  61. Larsen, L.N.; Bremer, J.; Flock, S.; Skattebø, L. α- and β- Alkyl-Substituted Eicosapentaenoic Acids: Incorporation into Phospholipids and Effets on Prostaglandin H Synthase and 5-Lipoxygenase. Biochem. Pharmacol. 1998, 55, 405–411. [Google Scholar] [CrossRef]
  62. Wada, M.; DeLong, C.J.; Hong, Y.H.; Rieke, C.J.; Song, I.; Sidhu, R.S.; Yuan, C.; Warnock, M.; Schmaier, A.H.; Yokoyama, C.; et al. Enzymes and Receptors of Prostaglandin Pathways with Arachidonic Acid-Derived versus Eicosapentaenoic Acid-Derived Substrates and Products. J. Biol. Chem. 2007, 282, 22254–22266. [Google Scholar] [CrossRef]
  63. Allaire, J.; Couture, P.; Leclerc, M.; Charest, A.; Marin, J.; Lépine, M.C.; Talbot, D.; Tchernof, A.; Lamarche, B. A Randomized, Crossover, Head-to-Head Comparison of Eicosapentaenoic Acid and Docosahexaenoic Acid Supplementation to Reduce Inflammation Markers in Men and Women: The Comparing EPA to DHA (ComparED) Study. Am. J. Clin. Nutr. 2016, 104, 280–287. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, J.-Y. Editorial: Eicosanoids and Cytokines: Resolution of Inflammation. Front. Pharmacol. 2022, 13, 978331. [Google Scholar] [CrossRef] [PubMed]
  65. Sierra, S.; Lara-Villoslada, F.; Comalada, M.; Olivares, M.; Xaus, J. Dietary Eicosapentaenoic Acid and Docosahexaenoic Acid Equally Incorporate as Decosahexaenoic Acid but Differ in Inflammatory Effects. Nutrition 2008, 24, 245–254. [Google Scholar] [CrossRef] [PubMed]
  66. Skelly, D.T.; Hennessy, E.; Dansereau, M.A.; Cunningham, C. A Systematic Analysis of the Peripheral and CNS Effects of Systemic LPS, IL-1Β, TNF-α and IL-6 Challenges in C57BL/6 Mice. PLoS ONE 2013, 8, 10. [Google Scholar] [CrossRef]
  67. Vezzani, A.; Viviani, B. Neuromodulatory Properties of Inflammatory Cytokines and Their Impact on Neuronal Excitability. Neuropharmacology 2015, 96, 70–82. [Google Scholar] [CrossRef] [PubMed]
  68. Brebner, K.; Hayley, S.; Zacharko, R.; Merali, Z.; Anisman, H. Synergistic Effects of Interleukin-1, Interleukin-6, and Tumor Necrosis Factor-: Central Monoamine, Corticosterone, and Behavioral Variations. Neuropsychopharmacology 2000, 22, 566–580. [Google Scholar] [CrossRef]
  69. Wang, J.; Chen, Z.; Walston, J.D.; Gao, P.; Gao, M.; Leng, S.X. Interferon-γ Potentiates α-Synuclein-Induced Neurotoxicity Linked to Toll-like Receptors 2 and 3 and Tumor Necrosis Factor-α in Murine Astrocytes. Mol. Neurobiol. 2019, 56, 7664–7679. [Google Scholar] [CrossRef] [PubMed]
  70. Caroprese, M.; Ciliberti, M.G.; Annicchiarico, G.; Albenzio, M.; Muscio, A.; Sevi, A. Hypothalamic-Pituitary-Adrenal Axis Activation and Immune Regulation in Heat-Stressed Sheep after Supplementation with Polyunsaturated Fatty Acids. J. Dairy Sci. 2014, 97, 4247–4258. [Google Scholar] [CrossRef]
  71. Kwon, Y. Immuno-Resolving Ability of Resolvins, Protectins, and Maresins Derived from Omega-Fatty Acids in Metabolic Syndrome. Mol. Nutr. Food Res. 2020, 64, 1900824. [Google Scholar] [CrossRef] [PubMed]
  72. Pilkington, S.M.; Rhodes, L.E.; Al-Aasswad, N.M.I.; Massey, K.A.; Nicolaou, A. Impact of EPA Ingestion on COX- and LOX-Mediated Eicosanoid Synthesis in Skin with and without a pro-Inflammatory UVR Challenge—Report of a Randomised Controlled Study in Humans. Mol. Nutr. Food Res. 2014, 58, 580–590. [Google Scholar] [CrossRef]
  73. Ferreira, I.; Falcato, F.; Bandarra, N.; Rauter, A.P. Resolvins, Protectins, and Maresins: DHA-Derived Specialized Pro-Resolving Mediators, Biosynthetic Pathways, Synthetic Approaches, and Their Role in Inflammation. Molecules 2022, 27, 1677. [Google Scholar] [CrossRef] [PubMed]
  74. Singer, P.; Shapiro, H.; Theilla, M.; Anbar, R.; Singer, J.; Cohen, J. Anti-Inflammatory Properties of Omega-3 Fatty Acids in Critical Illness: Novel Mechanisms and an Integrative Perspective. Intensive Care Med. 2008, 34, 1580–1592. [Google Scholar] [CrossRef] [PubMed]
  75. 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] [PubMed]
  76. Tapia, G.; Valenzuela, R.; Espinosa, A.; Romanque, P.; Dossi, C.; Gonzalez-Mañán, D.; Videla, L.A.; D’Espessailles, A. N-3 Long-Chain PUFA Supplementation Prevents High Fat Diet Induced Mouse Liver Steatosis and Inflammation in Relation to PPAR-α Upregulation and NF-ΚB DNA Binding Abrogation. Mol. Nutr. Food Res. 2014, 58, 1333–1341. [Google Scholar] [CrossRef]
  77. Bosviel, R.; Joumard-Cubizolles, L.; Chinetti-Gbaguidi, G.; Bayle, D.; Copin, C.; Hennuyer, N.; Duplan, I.; Staels, B.; Zanoni, G.; Porta, A.; et al. DHA-Derived Oxylipins, Neuroprostanes and Protectins, Differentially and Dose-Dependently Modulate the Inflammatory Response in Human Macrophages: Putative Mechanisms through PPAR Activation. Free Radic. Biol. Med. 2017, 103, 146–154. [Google Scholar] [CrossRef] [PubMed]
  78. El-Ashmawy, N.E.; Khedr, N.F.; El-Bahrawy, H.A.; Helal, S.A. Upregulation of PPAR-γ Mediates the Renoprotective Effect of Omega-3 PUFA and Ferulic Acid in Gentamicin-Intoxicated Rats. Biomed. Pharmacother. 2018, 99, 504–510. [Google Scholar] [CrossRef] [PubMed]
  79. 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]
  80. Campanari, D.D.; Cipriano, U.G.; Fraga-Silva, T.F.D.C.; Ramalho, L.N.Z.; Ovidio, P.P.; Jordão Júnior, A.A.; Bonato, V.L.D.; Ferriolli, E. Effect of Dietary Supplementation with Omega-3 Fatty Acid on the Generation of Regulatory T Lymphocytes and on Antioxidant Parameters and Markers of Oxidative Stress in the Liver Tissue of IL−10 Knockout Mice. Nutrients 2024, 16, 634. [Google Scholar] [CrossRef]
  81. Khalfoun, B.; Thibault, F.; Watier, H.; Bardos, P.; Lebranchu, Y. Docosahexaenoic and Eicosapentaenoic Acids Inhibit In vitro Human Endothelial Cell Production of Interleukin-6. Adv. Exp. Med. Biol. 1997, 400, 589–597. [Google Scholar]
  82. Mullen, A.; Loscher, C.E.; Roche, H.M. Anti-Inflammatory Effects of EPA and DHA Are Dependent upon Time and Dose-Response Elements Associated with LPS Stimulation in THP-1-Derived Macrophages. J. Nutr. Biochem. 2010, 21, 444–450. [Google Scholar] [CrossRef] [PubMed]
  83. Wierenga, K.A.; Riemers, F.M.; Westendorp, B.; Harkema, J.R.; Pestka, J.J. Single Cell Analysis of Docosahexaenoic Acid Suppression of Sequential LPS-Induced Proinflammatory and Interferon-Regulated Gene Expression in the Macrophage. Front. Immunol. 2022, 13, 993614. [Google Scholar] [CrossRef] [PubMed]
  84. Weldon, S.M.; Mullen, A.C.; Loscher, C.E.; Hurley, L.A.; Roche, H.M. Docosahexaenoic Acid Induces an Anti-Inflammatory Profile in Lipopolysaccharide-Stimulated Human THP-1 Macrophages More Effectively than Eicosapentaenoic Acid. J. Nutr. Biochem. 2007, 18, 250–258. [Google Scholar] [CrossRef] [PubMed]
  85. Dennis, E.A.; Norris, P.C. Eicosanoid Storm in Infection and Inflammation. Nat. Rev. Immunol. 2015, 15, 511–523. [Google Scholar] [CrossRef] [PubMed]
  86. Draper, E.; Reynolds, C.M.; Canavan, M.; Mills, K.H.; Loscher, C.E.; Roche, H.M. Omega-3 Fatty Acids Attenuate Dendritic Cell Function via NF-ΚB Independent of PPARγ. J. Nutr. Biochem. 2011, 22, 784–790. [Google Scholar] [CrossRef]
  87. Perkins, N.D. Integrating Cell-Signalling Pathways with NF-ΚB and IKK Function. Nat. Rev. Mol. Cell Biol. 2007, 8, 49–62. [Google Scholar] [CrossRef]
  88. Downton, P.; Bagnall, J.S.; England, H.; Spiller, D.G.; Humphreys, N.E.; Jackson, D.A.; Paszek, P.; White, M.R.H.; Adamson, A.D. Overexpression of IκB⍺ Modulates NF-ΚB Activation of Inflammatory Target Gene Expression. Front. Mol. Biosci. 2023, 10, 1187187. [Google Scholar] [CrossRef]
  89. Adelaja, A.; Hoffmann, A. Signaling Crosstalk Mechanisms That May Fine-Tune Pathogen-Responsive NFκB. Front. Immunol. 2019, 10, 433. [Google Scholar] [CrossRef]
  90. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-ΚB Signaling in Inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed]
  91. Jiao, H.; Wang, Z.; Liu, Y.; Wang, P.; Xue, Y. Specific Role of Tight Junction Proteins Claudin-5, Occludin, and ZO-1 of the Blood-Brain Barrier in a Focal Cerebral Ischemic Insult. J. Mol. Neurosci. 2011, 44, 130–139. [Google Scholar] [CrossRef] [PubMed]
  92. Balbuena, P.; Li, W.; Ehrich, M. Assessments of Tight Junction Proteins Occludin, Claudin 5 and Scaffold Proteins ZO1 and ZO2 in Endothelial Cells of the Rat Blood-Brain Barrier: Cellular Responses to Neurotoxicants Malathion and Lead Acetate. Neurotoxicology 2011, 32, 58–67. [Google Scholar] [CrossRef]
  93. Wang, X.; Pan, L.; Lu, J.; Li, N.; Li, J. N-3 PUFAs Attenuate Ischemia/Reperfusion Induced Intestinal Barrier Injury by Activating I-FABP-PPARγ Pathway. Clin. Nutr. 2012, 31, 951–957. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, W.; Zhang, H.; Mu, H.; Zhu, W.; Jiang, X.; Hu, X.; Shi, Y.; Leak, R.K.; Dong, Q.; Chen, J.; et al. Omega-3 Polyunsaturated Fatty Acids Mitigate Blood-Brain Barrier Disruption after Hypoxic-Ischemic Brain Injury. Neurobiol. Dis. 2016, 91, 37–46. [Google Scholar] [CrossRef] [PubMed]
  95. McColl, B.W.; Rothwell, N.J.; Allan, S.M. Systemic Inflammation Alters the Kinetics of Cerebrovascular Tight Junction Disruption After Experimental Stroke in Mice. J. Neurosci. 2008, 28, 9451–9462. [Google Scholar] [CrossRef] [PubMed]
  96. Smyth, L.C.D.; Rustenhoven, J.; Park, T.I.H.; Schweder, P.; Jansson, D.; Heppner, P.A.; O’Carroll, S.J.; Mee, E.W.; Faull, R.L.M.; Curtis, M.; et al. Unique and Shared Inflammatory Profiles of Human Brain Endothelia and Pericytes. J. Neuroinflamm. 2018, 15, 138. [Google Scholar] [CrossRef]
  97. Willemsen, L.E.M.; Koetsier, M.A.; Balvers, M.; Beermann, C.; Stahl, B.; Van Tol, E.A.F. Polyunsaturated Fatty Acids Support Epithelial Barrier Integrity and Reduce IL-4 Mediated Permeability In Vitro. Eur. J. Nutr. 2008, 47, 183–191. [Google Scholar] [CrossRef] [PubMed]
  98. Naser, A.N.; Lu, Q.; Chen, Y.-H. Trans-Compartmental Regulation of Tight Junction Barrier Function. Tissue Barriers 2023, 11, 2133880. [Google Scholar] [CrossRef]
  99. Krug, S.M.; Schulzke, J.D.; Fromm, M. Tight Junction, Selective Permeability, and Related Diseases. Semin. Cell Dev. Biol. 2014, 36, 166–176. [Google Scholar] [CrossRef] [PubMed]
  100. Cunningham, K.E.; Turner, J.R. Myosin Light Chain Kinase: Pulling the Strings of Epithelial Tight Junction Function. Ann. N. Y. Acad. Sci. 2012, 1258, 34–42. [Google Scholar] [CrossRef]
  101. Musch, M.W.; Mary Walsh-Reitz, M.; Chang, E.B.; Chang Roles, E.B. Roles of ZO-1, Occludin, and Actin in Oxidant-Induced Barrier Disruption. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 290, 222–231. [Google Scholar] [CrossRef]
  102. Yu, D.; Marchiando, A.M.; Weber, C.R.; Raleigh, D.R.; Wang, Y.; Shen, L.; Turner, J.R. MLCK-Dependent Exchange and Actin Binding Region-Dependent Anchoring of ZO-1 Regulate Tight Junction Barrier Function. Proc. Natl. Acad. Sci. USA 2010, 107, 8237–8241. [Google Scholar] [CrossRef] [PubMed]
  103. Lukiw, W.J. Gastrointestinal (GI) Tract Microbiome-Derived Neurotoxins—Potent Neuro-Inflammatory Signals from the GI Tract via the Systemic Circulation into the Brain. Front. Cell Infect. Microbiol. 2020, 10, 22. [Google Scholar] [CrossRef] [PubMed]
  104. Xiao, G.; Tang, L.; Yuan, F.; Zhu, W.; Zhang, S.; Liu, Z.; Geng, Y.; Qiu, X.; Zhang, Y.; Su, L. Eicosapentaenoic Acid Enhances Heat Stress-Impaired Intestinal Epithelial Barrier Function in Caco-2 Cells. PLoS ONE 2013, 8, e73571. [Google Scholar] [CrossRef] [PubMed]
  105. Usami, M.; Muraki, K.; Iwamoto, M.; Ohata, A.; Matsushita, E.; Miki, A. Effect of Eicosapentaenoic Acid (EPA) on Tight Junction Permeability in Intestinal Monolayer Cells. Clin. Nutr. 2001, 20, 351–359. [Google Scholar] [CrossRef]
  106. Xiao, G.; Yuan, F.; Geng, Y.; Qiu, X.; Liu, Z.; Lu, J.; Tang, L.; Zhang, Y.; Su, L. Eicosapentaenoic Acid Enhances Heatstroke-Impaired Intestinal Epithelial Barrier Function in Rats. Shock 2015, 44, 348–356. [Google Scholar] [CrossRef] [PubMed]
  107. Yu, T.-X.; Wang, P.-Y.; Rao, J.N.; Zou, T.; Liu, L.; Xiao, L.; Gorospe, M.; Wang, J.-Y. Chk2-Dependent HuR Phosphorylation Regulates Occludin MRNA Translation and Epithelial Barrier Function. Nucleic Acids Res. 2011, 39, 8472–8487. [Google Scholar] [CrossRef] [PubMed]
  108. Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms Regulating Intestinal Barrier Integrity and Its Pathological Implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef] [PubMed]
  109. Tian, F.; Gao, X.; Zhang, L.; Wang, X.; Wan, X.; Jiang, T.; Wu, C.; Bi, J.; Lei, Q. Effects of N-3 PUFAs on Intestinal Mucosa Innate Immunity and Intestinal Microbiota in Mice after Hemorrhagic Shock Resuscitation. Nutrients 2016, 8, 609. [Google Scholar] [CrossRef] [PubMed]
  110. Kim, H.-Y.; Akbar, M.; Kim, Y.-S. Phosphatidylserine-Dependent Neuroprotective Signaling Promoted by Docosahexaenoic Acid. Prostaglandins Leukot. Essent. Fat. Acids 2010, 82, 165–172. [Google Scholar] [CrossRef] [PubMed]
  111. Xie, Q.; Zhang, Y.; Zhang, J.; Cui, D.; Zhou, Q.; Guo, M. Promotion Effect of the Blend Containing 2′-FL, OPN and DHA on Oligodendrocyte Progenitor Cells Myelination In Vitro. Front. Nutr. 2022, 9, 1054431. [Google Scholar] [CrossRef] [PubMed]
  112. Dagai, L.; Peri-Naor, R.; Birk, R.Z. Docosahexaenoic Acid Significantly Stimulates Immediate Early Response Genes and Neurite Outgrowth. Neurochem. Res. 2009, 34, 867–875. [Google Scholar] [CrossRef] [PubMed]
  113. 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]
  114. Götz, M.; Barde, Y.A. Radial Glial Cells: Defined and Major Intermediates between Embryonicstem Cells and CNS Neurons. Neuron 2005, 46, 369–372. [Google Scholar] [CrossRef] [PubMed]
  115. Ming, G.L.; Song, H. Adult Neurogenesis in the Mammalian Brain: Significant Answers and Significant Questions. Neuron 2011, 70, 687–702. [Google Scholar] [CrossRef]
  116. Kowiański, P.; Lietzau, G.; Czuba, E.; Waśkow, M.; Steliga, A.; Moryś, J. BDNF: A Key Factor with Multipotent Impact on Brain Signaling and Synaptic Plasticity. Cell. Mol. Neurobiol. 2018, 38, 579–593. [Google Scholar] [CrossRef] [PubMed]
  117. Miranda, M.; Morici, J.F.; Zanoni, M.B.; Bekinschtein, P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front. Cell. Neurosci. 2019, 13, 363. [Google Scholar] [CrossRef] [PubMed]
  118. Müller, P.; Duderstadt, Y.; Lessmann, V.; Müller, N.G. Lactate and BDNF: Key Mediators of Exercise Induced Neuroplasticity? J. Clin. Med. 2020, 9, 1136. [Google Scholar] [CrossRef]
  119. Cysneiros, R.M.; Ferrari, D.; Arida, R.M.; Terra, V.C.; de Almeida, A.C.G.; Cavalheiro, E.A.; Scorza, F.A. Qualitative Analysis of Hippocampal Plastic Changes in Rats with Epilepsy Supplemented with Oral Omega-3 Fatty Acids. Epilepsy Behav. 2010, 17, 33–38. [Google Scholar] [CrossRef]
  120. Kolarow, R.; Kuhlmann, C.R.W.; Munsch, T.; Zehendner, C.; Brigadski, T.; Luhmann, H.J.; Lessmann, V. BDNF-Induced Nitric Oxide Signals in Cultured Rat Hippocampal Neurons: Time Course, Mechanism of Generation, and Effect on Neurotrophin Secretion. Front. Cell. Neurosci. 2014, 8, 323. [Google Scholar] [CrossRef] [PubMed]
  121. Sugasini, D.; Yalagala, P.C.R.; Subbaiah, P.V. Plasma BDNF Is a More Reliable Biomarker than Erythrocyte Omega-3 Index for the Omega-3 Fatty Acid Enrichment of Brain. Sci. Rep. 2020, 10, 10809. [Google Scholar] [CrossRef] [PubMed]
  122. Mita, T.; Mayanagi, T.; Ichijo, H.; Fukumoto, K.; Otsuka, K.; Sakai, A.; Sobue, K. Docosahexaenoic Acid Promotes Axon Outgrowth by Translational Regulation of Tau and Collapsin Response Mediator Protein 2 Expression. J. Biol. Chem. 2016, 291, 4955–4965. [Google Scholar] [CrossRef] [PubMed]
  123. Scharfman, H.; Goodman, J.; Macleod, A.; Phani, S.; Antonelli, C.; Croll, S. Increased Neurogenesis and the Ectopic Granule Cells after Intrahippocampal BDNF Infusion in Adult Rats. Exp. Neurol. 2005, 192, 348–356. [Google Scholar] [CrossRef] [PubMed]
  124. Hatami, A.; Zhu, C.; Relaño-Gines, A.; Elias, C.; Galstyan, A.; Jun, M.; Milne, G.; Cantor, C.R.; Chesselet, M.F.; Shchepinov, M.S. Deuterium-Reinforced Linoleic Acid Lowers Lipid Peroxidation and Mitigates Cognitive Impairment in the Q140 Knock in Mouse Model of Huntington’s Disease. FEBS J. 2018, 285, 3002–3012. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, Z.-H.; Yip, P.K.; Adams, L.; Davies, M.; Lee, J.W.; Michael, G.J.; Priestley, J.V.; Michael-Titus, A.T. A Single Bolus of Docosahexaenoic Acid Promotes Neuroplastic Changes in the Innervation of Spinal Cord Interneurons and Motor Neurons and Improves Functional Recovery after Spinal Cord Injury. J. Neurosci. 2015, 35, 12733–12752. [Google Scholar] [CrossRef]
  126. Cao, D.; Kevala, K.; Kim, J.; Moon, H.S.; Jun, S.B.; Lovinger, D.; Kim, H.Y. Docosahexaenoic Acid Promotes Hippocampal Neuronal Development and Synaptic Function. J. Neurochem. 2009, 111, 510–521. [Google Scholar] [CrossRef] [PubMed]
  127. González, L.M.; Bourissai, A.; Lessard-Beaudoin, M.; Lebel, R.; Tremblay, L.; Lepage, M.; Graham, R.K. Amelioration of Cognitive and Olfactory System Deficits in APOE4 Transgenic Mice with DHA Treatment. Mol. Neurobiol. 2023, 60, 5624–5641. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, J.H.; Wang, Q.; You, Q.L.; Li, Z.L.; Hu, N.Y.; Wang, Y.; Jin, Z.L.; Li, S.J.; Li, X.W.; Yang, J.M.; et al. Acute EPA-Induced Learning and Memory Impairment in Mice Is Prevented by DHA. Nat. Commun. 2020, 11, 5465. [Google Scholar] [CrossRef] [PubMed]
  129. Xiao, M.; Xiang, W.; Chen, Y.; Peng, N.; Du, X.; Lu, S.; Zuo, Y.; Li, B.; Hu, Y.; Li, X. DHA Ameliorates Cognitive Ability, Reduces Amyloid Deposition, and Nerve Fiber Production in Alzheimer’s Disease. Front. Nutr. 2022, 9, 852433. [Google Scholar] [CrossRef]
  130. Sinn, N.; Milte, C.M.; Street, S.J.; Buckley, J.D.; Coates, A.M.; Petkov, J.; Howe, P.R.C. Effects of N-3 Fatty Acids, EPA v. DHA, on Depressive Symptoms, Quality of Life, Memory and Executive Function in Older Adults with Mild Cognitive Impairment: A 6-Month Randomised Controlled Trial. Br. J. Nutr. 2012, 107, 1682–1693. [Google Scholar] [CrossRef]
  131. Guo, Y.-R.; Lee, H.-C.; Lo, Y.-C.; Yu, S.-C.; Huang, S.-Y. N-3 Polyunsaturated Fatty Acids Prevent d-Galactose-Induced Cognitive Deficits in Prediabetic Rats. Food Funct. 2018, 9, 2228–2239. [Google Scholar] [CrossRef] [PubMed]
  132. Javanainen, M.; Enkavi, G.; Guixà-Gonzaléz, R.; Kulig, W.; Martinez-Seara, H.; Levental, I.; Vattulainen, I. Reduced Level of Docosahexaenoic Acid Shifts GPCR Neuroreceptors to Less Ordered Membrane Regions. PLoS Comput. Biol. 2019, 15, e1007033. [Google Scholar] [CrossRef] [PubMed]
  133. Moreno, C.; de la Cruz, A.; Valenzuela, C. In-Depth Study of the Interaction, Sensitivity, and Gating Modulation by PUFAs on K+ Channels; Interaction and New Targets. Front. Physiol. 2016, 7, 578. [Google Scholar] [CrossRef]
  134. Elinder, F.; Liin, S.I. Actions and Mechanisms of Polyunsaturated Fatty Acids on Voltage-Gated Ion Channels. Front. Physiol. 2017, 8, 43. [Google Scholar] [CrossRef]
  135. Ogawa, T.; Sawane, K.; Ookoshi, K.; Kawashima, R. Supplementation with Flaxseed Oil Rich in Alpha-Linolenic Acid Improves Verbal Fluency in Healthy Older Adults. Nutrients 2023, 15, 1499. [Google Scholar] [CrossRef]
  136. Young, C.; Gean, P.-W.; Chiou, L.-C.; Shen, Y.-Z. Docosahexaenoic Acid Inhibits Synaptic Transmission and Epileptiform Activity in the Rat Hippocampus. Synapse 2000, 37, 90–94. [Google Scholar] [CrossRef]
  137. Cordero-Morales, J.F.; Vásquez, V. How Lipids Contribute to Ion Channel Function, a Fat Perspective on Direct and Indirect Interactions. Curr. Opin. Struct. Biol. 2018, 51, 92–98. [Google Scholar] [CrossRef] [PubMed]
  138. Leaf, A.; Xiao, Y.-F.; Kang, J.X. Interactions of N-3 Fatty Acids with Ion Channels in Excitable Tissues. Prostaglandins Leukot. Essent. Fat. Acids 2002, 67, 113–120. [Google Scholar] [CrossRef] [PubMed]
  139. Labrousse, V.F.; Nadjar, A.; Joffre, C.; Costes, L.; Aubert, A.; Grégoire, S.; Bretillon, L.; Layé, S. Short-Term Long Chain Omega3 Diet Protects from Neuroinflammatory Processes and Memory Impairment in Aged Mice. PLoS ONE 2012, 7, e36861. [Google Scholar] [CrossRef] [PubMed]
  140. Fan, R.; Hua, Y.; Shen, J.; Xiao, R.; Ma, W. Dietary Fatty Acids Affect Learning and Memory Ability via Regulating Inflammatory Factors in Obese Mice. J. Nutr. Biochem. 2022, 103, 108959. [Google Scholar] [CrossRef] [PubMed]
  141. Luchtman, D.W.; Song, C. Cognitive Enhancement by Omega-3 Fatty Acids from Child-Hood to Old Age: Findings from Animal and Clinical Studies. Neuropharmacology 2013, 64, 550–565. [Google Scholar] [CrossRef]
  142. Grimm, M.O.W.; Mett, J.; Stahlmann, C.P.; Haupenthal, V.J.; Blümel, T.; Stötzel, H.; Grimm, H.S.; Hartmann, T. Eicosapentaenoic Acid and Docosahexaenoic Acid Increase the Degradation of Amyloid-β by Affecting Insulin-Degrading Enzyme1. Biochem. Cell Biol. 2016, 94, 534–542. [Google Scholar] [CrossRef] [PubMed]
  143. Yan, L.; Xie, Y.; Satyanarayanan, S.K.; Zeng, H.; Liu, Q.; Huang, M.; Ma, Y.; Wan, J.-B.; Yao, X.; Su, K.-P.; et al. Omega-3 Polyunsaturated Fatty Acids Promote Brain-to-Blood Clearance of β-Amyloid in a Mouse Model with Alzheimer’s Disease. Brain Behav. Immun. 2020, 85, 35–45. [Google Scholar] [CrossRef] [PubMed]
  144. Ren, H.; Luo, C.; Feng, Y.; Yao, X.; Shi, Z.; Liang, F.; Kang, J.X.; Wan, J.-B.; Pei, Z.; Su, H. Omega-3 Polyunsaturated Fatty Acids Promote Amyloid-b Clearance from the Brain through Mediating the Function of the Glymphatic System. FASEB J. 2017, 31, 282–293. [Google Scholar] [CrossRef]
  145. Ozsoy, O.; Seval-Celik, Y.; Hacioglu, G.; Yargicoglu, P.; Demir, R.; Agar, A.; Aslan, M. The Influence and the Mechanism of Docosahexaenoic Acid on a Mouse Model of Parkinson’s Disease. Neurochem. Int. 2011, 59, 664–670. [Google Scholar] [CrossRef]
  146. Chytrova, G.; Ying, Z.; Gomez-Pinilla, F. Exercise Contributes to the Effects of DHA Dietary Supplementation by Acting on Membrane-Related Synaptic Systems. Brain Res. 2010, 1341, 32–40. [Google Scholar] [CrossRef] [PubMed]
  147. Wu, A.; Ying, Z.; Gomez-Pinilla, F. The Salutary Effects of DHA Dietary Supplementation on Cognition, Neuroplasticity, and Membrane Homeostasis after Brain Trauma. J. Neurotrauma 2011, 28, 2113–2122. [Google Scholar] [CrossRef] [PubMed]
  148. Ozkan, A.; Parlak, H.; Tanriover, G.; Dilmac, S.; Ulker, S.N.; Birsen, L.; Agar, A. The Protective Mechanism of Docosahexaenoic Acid in Mouse Model of Parkinson: The Role of Heme Oxygenase. Neurochem. Int. 2016, 101, 110–119. [Google Scholar] [CrossRef]
  149. Jezova, D.; Hlavacova, N. Endocrine Factors in Stress and Psychiatric Disorders: Focus on Anxiety and Salivary Steroids. Ann. N. Y. Acad. Sci. 2008, 1148, 495–503. [Google Scholar] [CrossRef] [PubMed]
  150. Chrousos, G.P. Stress and Disorders of the Stress System. Nat. Rev. Endocrinol. 2009, 5, 374–381. [Google Scholar] [CrossRef]
  151. Delarue, J.; Matzinger, O.; Binnert, C.; Schneiter, P.; Chioléro, R.; Tappy, L. Fish Oil Prevents the Adrenal Activation Elicited by Mental Stress in Healthy Men. Diabetes Metab. 2003, 29, 289–295. [Google Scholar] [CrossRef]
  152. Song, C.; Xiang, Y.Z.; Manku, M. Increased Phospholipase A2 Activity and Inflammatory Response but Decreased Nerve Growth Factor Expression in the Olfactory Bulbectomized Rat Model of Depression: Effects of Chronic Ethyl-Eicosapentaenoate Treatment. J. Neurosci. 2009, 29, 14–22. [Google Scholar] [CrossRef] [PubMed]
  153. Levant, B.; Ozias, M.K.; Davis, P.F.; Winter, M.; Russell, K.L.; Carlson, S.E.; Reed, G.A.; McCarson, K.E. Decreased Brain Docosahexaenoic Acid Content Produces Neurobiological Effects Associated with Depression: Interactions with Reproductive Status in Female Rats. Psychoneuroendocrinology 2008, 33, 1279–1292. [Google Scholar] [CrossRef] [PubMed]
  154. Morgese, M.G.; Tucci, P.; Mhillaj, E.; Bove, M.; Schiavone, S.; Trabace, L.; Cuomo, V. Lifelong Nutritional Omega-3 Deficiency Evokes Depressive-Like State Through Soluble Beta Amyloid. Mol. Neurobiol. 2017, 54, 2079–2089. [Google Scholar] [CrossRef] [PubMed]
  155. Mocking, R.J.T.; Ruhé, H.G.; Assies, J.; Lok, A.; Koeter, M.W.J.; Visser, I.; Bockting, C.L.H.; Schene, A.H. Relationship between the Hypothalamic-Pituitary-Adrenal-Axis and Fatty Acid Metabolism in Recurrent Depression. Psychoneuroendocrinology 2013, 38, 1607–1617. [Google Scholar] [CrossRef]
  156. Lightman, S.L. The Neuroendocrinology of Stress: A Never Ending Story. J. Neuroendocrinol. 2008, 20, 880–884. [Google Scholar] [CrossRef] [PubMed]
  157. Takeuchi, T.; Iwanaga, M.; Harada, E. Possible Regulatory Mechanism of DHA-Induced Anti-Stress Reaction in Rats. Brain Res. 2003, 964, 136–143. [Google Scholar] [CrossRef]
  158. Nieminen, L.R.G.; Makino, K.K.; Mehta, N.; Virkkunen, M.; Kim, H.Y.; Hibbeln, J.R. Relationship between Omega-3 Fatty Acids and Plasma Neuroactive Steroids in Alcoholism, Depression and Controls. Prostaglandins Leukot. Essent. Fat. Acids 2006, 75, 309–314. [Google Scholar] [CrossRef] [PubMed]
  159. Hamazaki, T.; Sawazaki, S.; Itomura, M.; Asaoka, E.; Nagao, Y.; Nishimura, N.; Yazawa, K.; Kuwamori, T.; Kobayashi, M. Effect of Docosahexaenoic Acid on Aggression Rapid Publication The Effect of Docosahexaenoic Acid on Aggression in Young Adults A Placebo-Controlled Double-Blind Study. J. Clin. Investig. 1996, 97, 1129–1133. [Google Scholar] [CrossRef] [PubMed]
  160. Oravcova, H.; Katrencikova, B.; Garaiova, I.; Durackova, Z.; Trebaticka, J.; Jezova, D. Stress Hormones Cortisol and Aldosterone, and Selected Markers of Oxidative Stress in Response to Long-Term Supplementation with Omega-3 Fatty Acids in Adolescent Children with Depression. Antioxidants 2022, 11, 1546. [Google Scholar] [CrossRef]
  161. Thesing, C.S.; Bot, M.; Milaneschi, Y.; Giltay, E.J.; Penninx, B.W.J.H. Omega-3 Polyunsaturated Fatty Acid Levels and Dysregulations in Biological Stress Systems. Psychoneuroendocrinology 2018, 97, 206–215. [Google Scholar] [CrossRef] [PubMed]
  162. Jazayeri, S.; Keshavarz, S.A.; Tehrani-Doost, M.; Djalali, M.; Hosseini, M.; Amini, H.; Chamari, M.; Djazayery, A. Effects of Eicosapentaenoic Acid and Fluoxetine on Plasma Cortisol, Serum Interleukin-1beta and Interleukin-6 Concentrations in Patients with Major Depressive Disorder. Psychiatry Res. 2010, 178, 112–115. [Google Scholar] [CrossRef]
  163. Song, C.; Phillips, A.G.; Leonard, B.E.; Horrobin, D.F. Ethyl-Eicosapentaenoic Acid Ingestion Prevents Corticosterone-Mediated Memory Impairment Induced by Central Administration of Interleukin-1β in Rats. Mol. Psychiatry 2004, 9, 630–638. [Google Scholar] [CrossRef]
  164. Madison, A.A.; Belury, M.A.; Andridge, R.; Renna, M.E.; Rosie Shrout, M.; Malarkey, W.B.; Lin, J.; Epel, E.S.; Kiecolt-Glaser, J.K. Omega-3 Supplementation and Stress Reactivity of Cellular Aging Biomarkers: An Ancillary Substudy of a Randomized, Controlled Trial in Midlife Adults. Mol. Psychiatry 2021, 26, 3034–3042. [Google Scholar] [CrossRef]
  165. Trebatická, J.; Hradečná, Z.; Surovcová, A.; Katrenčíková, B.; Gushina, I.; Waczulíková, I.; Sušienková, K.; Garaiova, I.; Šuba, J.; Ďuračková, Z. Omega-3 Fatty-Acids Modulate Symptoms of Depressive Disorder, Serum Levels of Omega-3 Fatty Acids and Omega-6/Omega-3 Ratio in Children. A Randomized, Double-Blind and Controlled Trial. Psychiatry Res. 2020, 287, 112911. [Google Scholar] [CrossRef]
  166. Katrenčíková, B.; Vaváková, M.; Paduchová, Z.; Nagyová, Z.; Garaiova, I.; Muchová, J.; Ďuračková, Z.; Trebatická, J. Oxidative Stress Markers and Antioxidant Enzymes in Children and Adolescents with Depressive Disorder and Impact of Omega-3 Fatty Acids in Randomised Clinical Trial. Antioxidants 2021, 10, 1256. [Google Scholar] [CrossRef] [PubMed]
  167. Lasselin, J.; Magne, E.; Beau, C.; Ledaguenel, P.; Dexpert, S.; Aubert, A.; Layé, S.; Capuron, L. Adipose Inflammation in Obesity: Relationship with Circulating Levels of Inflammatory Markers and Association with Surgery-Induced Weight Loss. J. Clin. Endocrinol. Metab. 2014, 99, E53–E61. [Google Scholar] [CrossRef] [PubMed]
  168. Kiecolt-Glaser, J.K.; Derry, H.M.; Fagundes, C.P. Inflammation: Depression Fans the Flames and Feasts on the Heat. Am. J. Psychiatry 2015, 172, 1075–1091. [Google Scholar] [CrossRef]
  169. Gupta, S.; Gupta, I.; Gupta, R.; Gupta, P. Role of C-Reactive Protein in Periodontal Disease—A Review. Int. J. Contemp. Med. Res. 2017, 4, 980–985. [Google Scholar]
  170. Incollingo Rodriguez, A.C.; Epel, E.S.; White, M.L.; Standen, E.C.; Seckl, J.R.; Tomiyama, A.J. Hypothalamic-Pituitary-Adrenal Axis Dysregulation and Cortisol Activity in Obesity: A Systematic Review. Psychoneuroendocrinology 2015, 62, 301–318. [Google Scholar] [CrossRef] [PubMed]
  171. Vreeburg, S.A.; Hoogendijk, W.J.; van Pelt, J.; DeRijk, R.H.; Verhagen, J.C.; van Dyck, R.; Smit, J.H.; Zitman, F.G.; Penninx, B.W. Major Depressive Disorder and Hypothalamic-Pituitary-Adrenal Axis Activity Results from a Large Cohort Study. Arch. Gen. Psychiatry 2009, 66, 617–626. [Google Scholar] [CrossRef] [PubMed]
  172. Bowden, R.G.; Wilson, R.L.; Deike, E.; Gentile, M. Fish Oil Supplementation Lowers C-Reactive Protein Levels Independent of Triglyceride Reduction in Patients with End-Stage Renal Disease. Nutr. Clin. Pract. 2009, 24, 508–512. [Google Scholar] [CrossRef] [PubMed]
  173. Zhao, Y.T.; Shao, L.; Teng, L.L.; Hu, B.; Luo, Y.; Yu, X.; Zhang, D.F.; Zhang, H. Effects of N-3 Polyunsaturated Fatty Acid Therapy on Plasma Inflammatory Markers and N-Terminal Pro-Brain Natriuretic Peptide in Elderly Patients with Chronic Heart Failure. J. Int. Med. Res. 2009, 37, 1831–1841. [Google Scholar] [CrossRef] [PubMed]
  174. Hellhammer, J.; Hero, T.; Franz, N.; Contreras, C.; Schubert, M. Omega-3 Fatty Acids Administered in Phosphatidylserine Improved Certain Aspects of High Chronic Stress in Men. Nutr. Res. 2012, 32, 241–250. [Google Scholar] [CrossRef] [PubMed]
  175. Pace, T.W.; Hu, F.; Miller, A.H. Cytokine-Effects on Glucocorticoid Receptor Function: Relevance to Glucocorticoid Resistance and the Pathophysiology and Treatment of Major Depression. Brain Behav. Immun. 2007, 21, 9–19. [Google Scholar] [CrossRef] [PubMed]
  176. Escoll, P.; Ranz, I.; Muñoz-Antón, N.; Van-Den-Rym, A.; Alvarez-Mon, M.; Martínez-Alonso, C.; Sanz, E.; De-La-Hera, A. Sustained Interleukin-1β Exposure Modulates Multiple Steps in Glucocorticoid Receptor Signaling, Promoting Split-Resistance to the Transactivation of Prominent Anti-Inflammatory Genes by Glucocorticoids. Mediat. Inflamm. 2015, 2015, 347965. [Google Scholar] [CrossRef] [PubMed]
  177. Cao, W.; Wang, C.; Chin, Y.; Chen, X.; Gao, Y.; Yuan, S.; Xue, C.; Wang, Y.; Tang, Q. DHA-Phospholipids (DHA-PL) and EPA-Phospholipids (EPA-PL) Prevent Intestinal Dysfunction Induced by Chronic Stress. Food Funct. 2019, 10, 277–288. [Google Scholar] [CrossRef]
  178. Bomba, A.; Nemcová, R.; Gancarcíková, S.; Herich, R.; Guba, P.; Mudronová, D. Improvement of the Probiotic Effect of Micro-Organisms by Their Combination with Maltodextrins, Fructo-Oligosaccharides and Polyunsaturated Fatty Acids. Br. J. Nutr. 2002, 88, S95–S99. [Google Scholar] [CrossRef] [PubMed]
  179. Kaliannan, K.; Wang, B.; Li, X.-Y.; Kim, K.-J.; Kang, J.X. A Host-Microbiome Interaction Mediates the Opposing Effects of Omega-6 and Omega-3 Fatty Acids on Metabolic Endotoxemia. Sci. Rep. 2015, 5, 11276. [Google Scholar] [CrossRef]
  180. Liu, J.; Huang, H.; Yang, Q.; Zhao, J.; Zhang, H.; Chen, W.; Peng, X.; Gu, Z. Dietary Supplementation of N-3 LCPUFAs Prevents Salmonellosis in a Murine Model. J. Agric. Food Chem. 2020, 68, 128–137. [Google Scholar] [CrossRef] [PubMed]
  181. Balfegò, M.; Canivell, S.; Hanzu, F.A.; Sala-Vila, A.; Martínez-Medina, M.; Murillo, S.; Mur, T.; Ruano, E.G.; Linares, F.; Porras, N.; et al. Effects of Sardine-Enriched Diet on Metabolic Control, Inflammation and Gut Microbiota in Drug-Naïve Patients with Type 2 Diabetes: A Pilot Randomized Trial. Lipids Health Dis. 2016, 15, 78. [Google Scholar] [CrossRef] [PubMed]
  182. Neijat, M.; Habtewold, J.; Li, S.; Jing, M.; House, J.D. Effect of Dietary N-3 Polyunsaturated Fatty Acids on the Composition of Cecal Microbiome of Lohmann Hens. Prostaglandins Leukot. Essent. Fat. Acids 2020, 162, 102182. [Google Scholar] [CrossRef]
  183. Menni, C.; Zierer, J.; Pallister, T.; Jackson, M.A.; Long, T.; Mohney, R.P.; Steves, C.J.; Spector, T.D.; Valdes, A.M. Omega-3 Fatty Acids Correlate with Gut Microbiome Diversity and Production of N-Carbamylglutamate in Middle Aged and Elderly Women. Sci. Rep. 2017, 7, 11079. [Google Scholar] [CrossRef] [PubMed]
  184. Vijay, A.; Astbury, S.; Le Roy, C.; Spector, T.D.; Valdes, A.M. The Prebiotic Effects of Omega-3 Fatty Acid Supplementation: A Six-Week Randomised Intervention Trial. Gut Microbes 2021, 13, 1863133. [Google Scholar] [CrossRef]
  185. Jayapala, H.P.S.; Lim, S.Y. N-3 Polyunsaturated Fatty Acids and Gut Microbiota. Comb. Chem. High Throughput Screen. 2022, 26, 892–905. [Google Scholar] [CrossRef]
  186. Tao, F.; Xing, X.; Wu, J.; Jiang, R. Enteral Nutrition Modulation with N-3 PUFAs Directs Microbiome and Lipid Metabolism in Mice. PLoS ONE 2021, 16, e0248482. [Google Scholar] [CrossRef]
  187. Robertson, R.C.; Oriach, C.S.; Murphy, K.; Moloney, G.M.; Cryan, J.F.; Dinan, T.G.; Ross, R.P.; Stanton, C. Deficiency of Essential Dietary N-3 PUFA Disrupts the Caecal Microbiome and Metabolome in Mice. Br. J. Nutr. 2017, 118, 959–970. [Google Scholar] [CrossRef]
  188. Blanchard, H.; Pédrono, F.; Boulier-Monthéan, N.; Catheline, D.; Rioux, V.; Legrand, P. Comparative Effects of Well-Balanced Diets Enriched in α-Linolenic or Linoleic Acids on LC-PUFA Metabolism in Rat Tissues. Prostaglandins Leukot. Essent. Fat. Acids 2013, 88, 383–389. [Google Scholar] [CrossRef] [PubMed]
  189. Robertson, R.C.; Seira Oriach, C.; Murphy, K.; Moloney, G.M.; Cryan, J.F.; Dinan, T.G.; Paul Ross, R.; Stanton, C. Omega-3 Polyunsaturated Fatty Acids Critically Regulate Behaviour and Gut Microbiota Development in Adolescence and Adulthood. Brain Behav. Immun. 2017, 59, 21–37. [Google Scholar] [CrossRef]
  190. Pachikian, B.D.; Neyrinck, A.M.; Portois, L.; De Backer, F.C.; Sohet, F.M.; Hacquebard, M.; Carpentier, Y.A.; Cani, P.D.; Delzenne, N.M. Involvement of Gut Microbial Fermentation in the Metabolic Alterations Occurring in N-3 Polyunsaturated Fatty Acids-Depleted Mice. Nutr. Metab. 2011, 8, 44. [Google Scholar] [CrossRef]
  191. Horiuchi, H.; Kamikado, K.; Aoki, R.; Suganuma, N.; Nishijima, T.; Nakatani, A.; Kimura, I. Bifidobacterium Animalis Subsp. Lactis GCL2505 Modulates Host Energy Metabolism via the Short-Chain Fatty Acid Receptor GPR43. Sci. Rep. 2020, 10, 4158. [Google Scholar] [CrossRef]
  192. Patterson, E.; Wall, R.; Lisai, S.; Ross, R.P.; Dinan, T.G.; Cryan, J.F.; Fitzgerald, G.F.; Banni, S.; Quigley, E.M.; Shanahan, F.; et al. Bifidobacterium Breve with α-Linolenic Acid Alters the Composition, Distribution and Transcription Factor Activity Associated with Metabolism and Absorption of Fat. Sci. Rep. 2017, 7, 43300. [Google Scholar] [CrossRef]
  193. Lu, J.; Shataer, D.; Yan, H.; Dong, X.; Zhang, M.; Qin, Y.; Cui, J.; Wang, L. Probiotics and Non-Alcoholic Fatty Liver Disease: Unveiling the Mechanisms of Lactobacillus Plantarum and Bifidobacterium Bifidum in Modulating Lipid Metabolism, Inflammation, and Intestinal Barrier Integrity. Foods 2024, 13, 2992. [Google Scholar] [CrossRef] [PubMed]
  194. Honda, K.L.; Lamon-Fava, S.; Matthan, N.R.; Wu, D.; Lichtenstein, A.H. Docosahexaenoic Acid Differentially Affects TNFα and IL-6 Expression in LPS-Stimulated RAW 264.7 Murine Macrophages. Prostaglandins Leukot. Essent. Fat. Acids 2015, 97, 27–34. [Google Scholar] [CrossRef] [PubMed]
  195. 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] [PubMed]
  196. Zhao, Y.; Joshi-Barve, S.; Barve, S.; Chen, L.H. Eicosapentaenoic Acid Prevents LPS-Induced TNF-α Expression by Preventing NF-ΚB Activation. J. Am. Coll. Nutr. 2004, 23, 71–78. [Google Scholar] [CrossRef] [PubMed]
  197. Vanden Berghe, W.; Vermeulen, L.; Delerive, P.; De Bosscher, K.; Staels, B.; Haegeman, G. A Paradigm for Gene Regulation: Inflammation, NF-KB and PPAR. In Peroxisomal Disorders and Regulation of Genes; Springer: Boston, MA, USA, 2003. [Google Scholar]
  198. Novak, T.E.; Babcock, T.A.; Jho, D.H.; Helton, W.S.; Espat, N.J.; Joseph, N. NF-B Inhibition by-3 Fatty Acids Modulates LPS-Stimulated Macrophage TNF-Transcription. Am. J. Physiol. Lung Cell. Mol. Physiol. 2003, 284, 84–89. [Google Scholar] [CrossRef] [PubMed]
  199. Zeyda, M.; Staffler, G.; Hořejší, V.; Waldhäusl, W.; Stulnig, T.M. LAT Displacement from Lipid Rafts as a Molecular Mechanism for the Inhibition of T Cell Signaling by Polyunsaturated Fatty Acids. J. Biol. Chem. 2002, 277, 28418–28423. [Google Scholar] [CrossRef] [PubMed]
  200. Koppelmann, T.; Pollak, Y.; Mogilner, J.; Bejar, J.; Coran, A.G.; Sukhotnik, I. Reversal of Severe Methotrexate-Induced Intestinal Damage Using Enteral n-3 Fatty Acids. Br. J. Nutr. 2013, 109, 89–98. [Google Scholar] [CrossRef] [PubMed]
  201. Gebauer, S.K.; Psota, T.L.; Harris, W.S.; Kris-Etherton, P.M. N−3 Fatty Acid Dietary Recommendations and Food Sources to Achieve Essentiality and Cardiovascular Benefits. Am. J. Clin. Nutr. 2006, 83, 1526S–1535S. [Google Scholar] [CrossRef] [PubMed]
  202. Van Dael, P. Role of N-3 Long-Chain Polyunsaturated Fatty Acids in Human Nutrition and Health: Review of Recent Studies and Recommendations. Nutr. Res. Pract. 2021, 15, 137–159. [Google Scholar] [CrossRef]
  203. Schneider, E.; Balasubramanian, R.; Ferri, A.; Cotter, P.D.; Clarke, G.; Cryan, J.F. Fibre & fermented foods: Differential effects on the microbiota-gut-brain axis. Proc Nutr Soc. [CrossRef]
  204. de Oliveira, V.S.; Viana, D.S.B.; Keller, L.M.; de Melo, M.T.T.; Mulandeza, O.F.; Barbosa, M.I.M.J.; Barbosa Júnior, J.L.; Saldanha, T. Impact of Air Frying on Food Lipids: Oxidative Evidence, Current Research, and Insights into Domestic Mitigation by Natural Antioxidants. Trends Food Sci. Technol. 2024, 147, 104465. [Google Scholar] [CrossRef]
Figure 1. Interaction Between the gut and the brain through the gut–brain axis. HPA axis, hypothalamic-pituitary-adrenal axis; ENS, enteric nervous system; SCFA, short-chain fatty acids. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Figure 1. Interaction Between the gut and the brain through the gut–brain axis. HPA axis, hypothalamic-pituitary-adrenal axis; ENS, enteric nervous system; SCFA, short-chain fatty acids. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Molecules 30 00071 g001
Figure 2. The role of omega-3 fatty acids as components of cell membranes. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Figure 2. The role of omega-3 fatty acids as components of cell membranes. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Molecules 30 00071 g002
Figure 3. Anti-inflammatory role of n-3 fatty acids. NF-kB—nuclear factor kappa B; COX-2—cyclooxygenase-2, prostaglandin-endoperoxide synthase 2; LOX—lipoxygenase; NOS—nitric oxide synthase; PPAR—peroxisome proliferator-activated receptors; GPR40—G-protein–coupled receptor 40; GPR120—G-protein–coupled receptor 120; RvDn DPA—resolvins derived from DPA n-3; Il-1β—interleukin 1β; IL-2—interleukin 2; IL-6—interleukin 6; INF-γ—interferon-γ; TNF-α—tumor necrosis factor α; TER—trans-epithelial electrical resistance. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Figure 3. Anti-inflammatory role of n-3 fatty acids. NF-kB—nuclear factor kappa B; COX-2—cyclooxygenase-2, prostaglandin-endoperoxide synthase 2; LOX—lipoxygenase; NOS—nitric oxide synthase; PPAR—peroxisome proliferator-activated receptors; GPR40—G-protein–coupled receptor 40; GPR120—G-protein–coupled receptor 120; RvDn DPA—resolvins derived from DPA n-3; Il-1β—interleukin 1β; IL-2—interleukin 2; IL-6—interleukin 6; INF-γ—interferon-γ; TNF-α—tumor necrosis factor α; TER—trans-epithelial electrical resistance. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Molecules 30 00071 g003
Figure 4. Summary of n-3 PUFAs’ mechanisms of action: a schematic representation of the possible mechanisms through which n-3 PUFAs influence the gut–brain axis. DHA—docosahexaenoic acid; EPA—eicosapentaenoic acid; DPA—n-3 docosapentaenoic acid; HPA—hypothalamic-pituitary-adrenal; BBB—blood–brain barrier; SCFAs—short-chain fatty acids. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Figure 4. Summary of n-3 PUFAs’ mechanisms of action: a schematic representation of the possible mechanisms through which n-3 PUFAs influence the gut–brain axis. DHA—docosahexaenoic acid; EPA—eicosapentaenoic acid; DPA—n-3 docosapentaenoic acid; HPA—hypothalamic-pituitary-adrenal; BBB—blood–brain barrier; SCFAs—short-chain fatty acids. ↑ indicates an increase, ↓ indicates a decrease. Designed using elements by ©Canva, sparklestroke, Pixeden, iconsy, OpenClipart-Vectors via Canva.com (access date: 18 November 2024).
Molecules 30 00071 g004
Table 2. Summarized effects of LC-PUFAs on the gut microbiota, immune function, and intestinal barrier integrity.
Table 2. Summarized effects of LC-PUFAs on the gut microbiota, immune function, and intestinal barrier integrity.
ModelsType of StudySource and Dose of n-3 PUFAExposureEffectReference
In vitro model studies
RAW 264.7 murine macrophage-like cell line in vitro100 μM DHA 24 h- DHA reduced NFκB-DNA binding activity
- Reduced inflammation
[194]
Murine bone marrow-derived DC in vitro100 μM DHA 24 h- Reduction NFκB translocation mediated by inhibition of IκB degradation
- Reduced inflammation
[195]
RAW 264.7 murine MØ cell line in vitro12 mg%, ω-3 FA emulsion 4 h- Reduction in endotoxin-induced NFκB activation through decreased IκB phosphorylation
- Reduced inflammation
[198]
Human Jurkat T cell lines E6-1 in vitro50 μm EPA 48 h - Promotion of regulatory T lymphocyte (Treg) induction and prevention of excessive development of T helper 17 (Th17) cells increase the number of regulatory T lymphocytes (Tregs)
- Reduced inflammation
[199]
In vivo model and human subject studies
Male BALB/c mice with chronic stress in vivoSquid egg and sea cucumber (9% EPA and 38.9% DHA
or 36% EPA and 5% DHA or
79% EPA and 10% DHA)
21 days- Increase Lactobacillus, Prevotella spp., Bacteroides fragilis, and Roseburia spp.
- Decrease Enterobacteriaceae and Enterococcus spp.
- Protection against intestinal dysfunction
- Attenuation of proinflammatory processes
- Amelioration of LPS increase
[177]
Poultry in vivo0.2% and 0.6% of total n-3 PUFA in the diet (marine algal biomass or flaxseed oil)8 weeks- Increased population of Firmicutes (e.g., Faecalibacterium, Clostridium and Ruminococcus [182]
C57BL/6J female mice were and their male offspringin vivo∼1 g EPADHA/100 g of the diet 12 weeks- Increased fecal Bifidobacterium and Lactobacillus abundance in offspring[189]
Male Sprague Dawley rats with intestinal damage in vivo 300 μg/kg per day (EPA 180 μg + DHA 120 μg) Once per day 48 h before and 72 h after MTX injection- Increased mass of the colon and ileum
- Greater mass of the ileal mucosa—increased villus height and crypt depth in the ileum
[200]
Drug-naïve patients with type 2 diabeteshuman subjects3.0 ± 0.2 g EPA + DHA/d (sardines) 5 days a week for 6 months- Increased ratio of Bacteroides/Prevotella
- Decreased ratio of Firmicutes/Bacteroidetes
[181]
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

Zinkow, A.; Grodzicki, W.; Czerwińska, M.; Dziendzikowska, K. Molecular Mechanisms Linking Omega-3 Fatty Acids and the Gut–Brain Axis. Molecules 2025, 30, 71. https://doi.org/10.3390/molecules30010071

AMA Style

Zinkow A, Grodzicki W, Czerwińska M, Dziendzikowska K. Molecular Mechanisms Linking Omega-3 Fatty Acids and the Gut–Brain Axis. Molecules. 2025; 30(1):71. https://doi.org/10.3390/molecules30010071

Chicago/Turabian Style

Zinkow, Anna, Wojciech Grodzicki, Malwina Czerwińska, and Katarzyna Dziendzikowska. 2025. "Molecular Mechanisms Linking Omega-3 Fatty Acids and the Gut–Brain Axis" Molecules 30, no. 1: 71. https://doi.org/10.3390/molecules30010071

APA Style

Zinkow, A., Grodzicki, W., Czerwińska, M., & Dziendzikowska, K. (2025). Molecular Mechanisms Linking Omega-3 Fatty Acids and the Gut–Brain Axis. Molecules, 30(1), 71. https://doi.org/10.3390/molecules30010071

Article Metrics

Back to TopTop