Next Article in Journal
Micronutrient Antioxidants for Men (Menevit®) Improve Sperm Function by Reducing Oxidative Stress, Resulting in Improved Assisted Reproductive Technology Outcomes
Next Article in Special Issue
Isobolographic Analysis of the Cytoprotective Effect of Dapsone and Cannabidiol Alone or Combination upon Oxygen–Glucose Deprivation/Reoxygenation Model in SH-SY5Y Cells
Previous Article in Journal
Photobiomodulation Inhibits Ischemia-Induced Brain Endothelial Senescence via Endothelial Nitric Oxide Synthase
Previous Article in Special Issue
Modulating Nitric Oxide: Implications for Cytotoxicity and Cytoprotection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ischemic Brain Injury: Involvement of Lipids in the Pathophysiology of Stroke and Therapeutic Strategies

by
Nathalie Bernoud-Hubac
*,
Amanda Lo Van
,
Adina-Nicoleta Lazar
and
Michel Lagarde
Univ Lyon, INSA Lyon, CNRS, LAMCOS, UMR5259, 69621 Villeurbanne, France
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(6), 634; https://doi.org/10.3390/antiox13060634
Submission received: 6 April 2024 / Revised: 14 May 2024 / Accepted: 21 May 2024 / Published: 23 May 2024

Abstract

:
Stroke is a devastating neurological disorder that is characterized by the sudden disruption of blood flow to the brain. Lipids are essential components of brain structure and function and play pivotal roles in stroke pathophysiology. Dysregulation of lipid signaling pathways modulates key cellular processes such as apoptosis, inflammation, and oxidative stress, exacerbating ischemic brain injury. In the present review, we summarize the roles of lipids in stroke pathology in different models (cell cultures, animal, and human studies). Additionally, the potential of lipids, especially polyunsaturated fatty acids, to promote neuroprotection and their use as biomarkers in stroke are discussed.

1. Introduction

Hypoxic–ischemic brain damage (HIBD) results in partial or complete hypoxia of brain tissue due to decreased or suspended cerebral blood flow. Among HIBD, ischemic stroke is a leading cause of mortality and disability worldwide [1,2]. Strokes mainly affect the brain. Still, in less than 1.5% of cases, the occlusion of the blood flow to the spine may also occur, leading to spinal strokes. Cerebral stroke can be classified into hemorrhagic stroke and ischemic stroke, the latter accounting for 80 to 85% of cases. Lipids play critical roles in maintaining the structural integrity, energy metabolism, and signaling pathways in the brain. Dysregulation of lipid metabolism has been implicated in the pathogenesis of stroke [3,4,5,6]. Ischemia–reperfusion injury leads to the excessive production of reactive oxygen species (ROS), resulting in lipid, protein, and DNA oxidation. Lipid-derived oxidative stress contributes to neuronal injury, blood–brain barrier disruption, and neuroinflammation in stroke. Targeting lipid-mediated mechanisms represents a promising therapeutic approach for stroke prevention and treatment. Understanding the relationship between ischemic stroke and lipids is essential for elucidating potential preventive and therapeutic strategies.
This review aims to describe the mechanisms involved in the pathophysiology of hypoxic–ischemic brain injury, especially cerebral stroke, with a focus on the importance of reactive oxygen species production and lipid peroxidation that can lead to the clinical condition of hypoxic–ischemic. Highlighting the roles of lipids in these pathologies could help to define lipid-based strategies for the prevention of stroke or treatment.

2. Stroke

Stroke, also known as cerebrovascular accident (CVA), is the second leading cause of death and disability worldwide [1,2] with 16 million incidents, an annual mortality rate of about 6.6 million, and 50% of survivors being chronically disabled [7,8,9]. Both in Europe and the USA, stroke has been the leading neurological disease in terms of disability-adjusted life years (DALYs) [10,11,12]. Alarming studies showed a less favorable trend in the incidence of stroke in younger vs. older individuals [13,14,15,16]. Globally, between 1990 and 2013, there was a significant increase in the absolute number and prevalence rate of stroke for younger adults [17]. Studies predict that stroke mortality worldwide will increase by 50%, from 6.6 million in 2020 to 9.7 million in 2050, and that DALYs will rise over the same period, from 144.8 million in 2020 to 189.3 million in 2050 [1]. Age is the most important demographic risk factor of stroke, and the lifetime risk of stroke has increased with aging [18]. Ethnic differences also exist in the prevalence of stroke risk factors [19,20]. The major risk factors for stroke are previous stroke or transient ischemic attack, hypertension (being the most significant risk factor for stroke), diabetes mellitus, obesity, hyperlipidemia, smoking, and drug abuse [21,22,23,24].
A stroke occurs when the blood supply to some part of the brain is interrupted or reduced, depriving brain tissue of oxygen and nutrients. Strokes can be classified into two major categories: hemorrhagic stroke and ischemic stroke. Both types of strokes impair the correct functioning of the brain. Hemorrhagic strokes occur when a weakened blood vessel ruptures and bleeds into the surrounding brain tissue [25,26,27]. Hemorrhagic stroke may be further subdivided into two main types including intracerebral hemorrhage (ICH), when blood spills into the surrounding tissue following the rupture of a blood vessel, causing damage to nearby cells, and subarachnoid hemorrhage (SAH), when bleeding occurs into the space between the brain and the surrounding membrane (the arachnoid membrane). Hemorrhagic stroke is associated with severe morbidity and high mortality [28]. The symptoms of a hemorrhagic stroke may include severe increases in blood pressure, sudden severe headache, vomiting, agitation, and mydriatic pupil [29]. The treatment for hemorrhagic stroke may involve surgery to repair blood vessels, medications to reduce blood pressure and prevent further bleeding, and supportive care to manage symptoms and prevent complications [30].
Ischemic stroke, also known as cerebral ischemia, generally accounts for about 80% to 85% of all strokes and is the third leading cause of death worldwide, with nearly 15 million people affected yearly [29,31]. Ischemic strokes occur when a blood clot blocks or narrows an artery leading to the brain, reducing blood flow to a part of the brain. There are two main types of ischemic strokes including thrombotic stroke, when a blood clot (thrombus) develops within a blood vessel supplying blood to the brain, and embolic stroke, when a blood clot or other debris forms elsewhere in the body (often in the heart, cardioembolic stroke, or large arteries) and travels through the bloodstream to the brain. Ischemic stroke may have various clinical manifestations including vomiting, paresis, paralysis, ataxia, acute headache, and agitation [29].
The main therapeutic approaches in patients with acute cerebral ischemia are reperfusion treatments, either via mechanical thrombectomy or more commonly via intravenous thrombolysis. Intravenous thrombolysis or intravenous alteplase, with tissue-type plasminogen activator (tPA), which is aimed at restoring the cerebral blood flow, is restricted to few patients because of a narrow therapeutic window of 4.5 h and a high hemorrhagic risk [32,33]. Understanding the mechanisms involved in the pathophysiology of stroke is crucial for developing effective interventions to prevent and treat ischemic stroke. The interruption of blood flow deprives the affected brain tissue of oxygen and nutrients, leading to cellular dysfunction and death. A cascade of pathophysiological and interconnected changes occurs, including pathological permeability of the blood–brain barrier (BBB), neuroinflammation, neuronal apoptosis, autophagy, excitotoxicity, and ionic imbalance, as well as oxidative stress.

3. Oxidative Stress and Ischemic Stroke

During an ischemic stroke, the lack of oxygen and nutrients supplied to the brain cells can lead to the production of free radicals, resulting in oxidative stress. Free radicals are highly reactive molecules that contain oxygen and can cause damage to cells, proteins, lipids, and DNA. This oxidative stress can exacerbate the damage caused by the stroke and contribute to further injury to brain tissue. Oxidative stress, characterized by the overproduction of reactive oxygen species (ROS) and impaired antioxidant defenses, represents a hallmark feature of ischemic stroke pathology [34,35,36]. Reactive nitrogen species (RNS) are also predominantly involved in the pathogenesis of stroke. The brain is particularly vulnerable to ROS and RNS compared with other organs because of high oxygen consumption, low neuronal antioxidant activity, high levels of peroxidizable lipids, and high concentrations of iron [37,38]. Ischemia–reperfusion injury triggers the excessive production of ROS, including superoxide anions (O2), hydroxyl radicals (OH), and hydrogen peroxide (H2O2), primarily through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, xanthine oxidase, and mitochondrial dysfunction [39]. In physiological conditions, antioxidant defenses (enzymatic or non-enzymatic) protect brain tissues against oxidative stress cytotoxicity [40,41]. Enzymatic antioxidant defenses include superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). SOD catalyzes the conversion of superoxide radicals into hydrogen peroxide, which is less toxic than superoxide radicals. CAT catalyzes the decomposition of hydrogen peroxide into water and oxygen, thereby preventing the formation of hydroxyl radicals [42]. CAT is particularly abundant in peroxisomes and helps to protect neurons from oxidative damage during stroke. GPx utilizes glutathione as a co-factor to reduce hydrogen peroxide and lipid hydroperoxides into their corresponding alcohols [43]. GPx is crucial for maintaining cellular redox balance and protecting neurons from oxidative stress-induced damage. Non-enzymatic antioxidants include vitamins E (α-tocopherol) and C (ascorbic acid), glutathione (GSH), carotenoids, and flavonoids [44,45]. The Nf2 (nuclear factor erythroid 2-related factor 2) pathway is also considered one of the most critical antioxidant transcription factors in cells [45,46]. Mitochondrial dysfunction plays a significant role in the pathophysiology of ischemic stroke. Oxidative stress disrupts mitochondrial homeostasis, impairing electron transport chain function, causing electron leakage, reducing ATP production, and promoting cytochrome c release, thereby initiating apoptotic cascades and exacerbating neuronal death. So, upon reperfusion, the sudden reintroduction of oxygen amplifies ROS generation by dysfunctional mitochondria, exacerbating tissue injury.

4. Lipids and Ischemic Stroke

4.1. Lipids and the Brain

Lipids, as essential components of brain structure and function, play pivotal roles in stroke pathophysiology. The brain is highly enriched in lipids and contains high concentrations of polyunsaturated fatty acids (PUFAs), especially docosahexaenoic acid [47,48]. Docosahexaenoic acid (22:6n-3, DHA), the most abundant PUFA in neuronal membranes, is a bioactive nutrient essential for brain development, learning ability, and memory [49,50]. Lipids play critical roles in maintaining the structural integrity of neuronal membranes, influencing membrane fluidity, permeability, and normal functioning of receptor proteins. PUFAs can be oxygenated into oxylipins, either via a non-enzymatic free radical-catalyzed pathway or an enzymatic pathway, which regulate several biological processes within the brain.

4.1.1. Enzymatic Metabolism of PUFAs

Three main enzymes are involved in the production of oxylipins including cyclooxygenases (COX), lipoxygenases (LOX), and cytochrome P450 monooxygenases (CYP450) [51,52,53] (Figure 1). The biosynthesis of oxilipins is initiated by the initial release of PUFAs from membrane phospholipids, mainly by the action of phospholipase A2 (PLA2) [54,55]. COX, also named prostaglandin endoperoxide H synthases, possesses both oxygenase and peroxidase activity. COX catalyzes the formation of PGG2, a 15-hydroperoxide converted into its 15-hydroxyl derivative PGH2 by hydroperoxidase [56,57]. PGH2 is then further metabolized to other prostaglandins (D, E, F, I, or prostacyclin), thromboxanes, levuglandins, and hydroxy fatty acids [52,58]. There are two major human isoforms of cyclooxygenase, COX-1, which is constitutively expressed in most tissues, and COX-2, which is induced by inflammatory and proliferative stimuli [59,60]. The main PUFA metabolized by COX is arachidonic acid (AA, 20:4n-6), but COX also oxygenates omega-3 PUFAs like eicopapentaenoic acid (EPA, 20:5n-3). Another prostanoid precursor is dihomo-gamma-linolenic acid (DGLA, 20:3n-6). Lipoxygenase enzymes are a family of dioxygenases, including 5-lipoxygenase (5-LOX), 12-LOX, 15-LOX, and 12/15-LOX isoforms, that catalyze the oxidation of AA, linoleic acid (LA, 18:2n-6), EPA, and DHA, to produce leukotrienes (LTs from AA and EPA), hydroxyperoxyeicosatetraenoic acids (HpETEs) than can be reduced to hydroxyeicosatetraenoic acid (HETEs), lipoxins (from AA), and SPMs (including resolvins Rvs from EPA and DHA; protectins PDs from DHA; and maresins MaRs from DHA) [61,62]. A further class of metabolites generated from omega-3 PUFAs by LOX are the electrophilic fatty acid oxo-derivatives (EFOX), with 7-oxo-DHA, 7-oxo-DPAn-3, and 5-oxo-EPA produced from DHA, DPAn-3, and EPA, respectively [63]. CYP450 oxidases can produce different epoxyeicosatrienoic acids (EETs) and hydroxy-eicosatetraenes (HETEs) from AA, epoxyeicosatetraenoic acids (EpETEs) and hydroxyeicosapentaenoic acids (HEPEs) from EPA, and epoxydocosapentaenoic acids (EDPs) and hydroxydocosahexaenoic acids (HDoHEs) from DHA [64,65].

4.1.2. Peroxidation of Lipids

Lipid peroxidation is a complex process involving the oxidative degradation of PUFAs, leading to the formation of various reactive lipid products and playing a pivotal role in various physiological and pathological conditions. Lipid peroxidation occurs in three consecutive phases [66] as follows: initiation, propagation, and termination. Initiation involves the abstraction of hydrogen atoms from PUFA carbon atoms by ROS or RNS, at the origin of a PUFA radical. Subsequent propagation reactions involve the interaction of lipid radicals with molecular oxygen, forming lipid peroxyl radicals and lipid hydroperoxides. These reactive intermediates can propagate lipid peroxidation through autocatalytic cycles, causing extensive damage to cellular membranes and organelles. The termination of lipid peroxidation occurs through various mechanisms, including radical scavenging by antioxidants, enzymatic degradation of lipid hydroperoxides, and reaction of radical species with each other to give non-radical or non-propagating species.
Isoprostanes (IsoPs) and neuroprostanes (NPs) are prostaglandin (PG)-like compounds that are produced by free radical non-enzymatic peroxidation of AA and DHA, respectively [67,68]. F2-isoprostanes and F4-NPs are primarily formed in situ from AA esterified in phospholipids and subsequently released [69]. Their formation proceeds via PGH2-like bicyclic endoperoxide intermediates, which are reduced to form F-ring IsoPs (F2-IsoPs) [67] or undergo rearrangement to form E-ring and D-ring IsoPs [70] and isothromboxanes [71]. Analogous to the formation of IsoPs, the formation of NPs also proceeds through bicyclic endoperoxide intermediates that not only are reduced to F4-ring compounds but also undergo rearrangement in vivo to form D4- and E4-ring NPs [68,72]. Furthermore, a series of highly reactive γ-ketoaldehydes termed isoketals (IsoKs) and neuroketals (NKs) are formed via the isoprostane and neuroprostane pathways, respectively [73,74,75]. IsoKs and NKs have a remarkable proclivity to form covalent adducts in vivo with proteins by binding the ε-amino group of lysine residues, forming lactam and Schiff base adducts, and to crosslink proteins. These γ-ketoaldehydes also covalently modify aminophospholipids, forming pyrrole and Schiff base adducts with phosphatidylethanolamine [76]. All these molecules are detected in the human brain [74]. Malondialdehyde (MDA) and hydroxyalkenals are also aldehydes that are formed during n-3 and n-6 fatty acid peroxidation. 4-hydroxynonenal (4-HNE) is an end-product of the peroxidation of LA, AA, and 15-hydroperoxy arachidonic acid [77]. 4-hydroxy-2E-hexenal (4-HHE) is described as a major degradation product of n-3 PUFA peroxidation, such as DHA [78]. All these reactive aldehydes derived from lipid peroxidation have been suggested to be key mediators of oxidant injury because of their capacity to covalently modify proteins, lipids, and DNA [79,80,81,82,83,84].

4.2. Dysregulation of Lipid Metabolism in the Pathogenesis of Stroke

Emerging evidence suggests that dysregulation of lipid metabolism plays a pivotal role in stroke pathogenesis and progression. The implication of potent bioactive eicosanoids derived from the arachidonate cascade in ischemic stroke has been attracting more attention.
The elevation of intracellular calcium and oxidative stress produced by ischemia/reperfusion in rats activates enzymes including calcium-dependent cytosolic PLA2 (cPLA2), leading to a rapid release of AA [85] (Figure 2). The release of brain PUFAs from phospholipids by PLA2 is exacerbated by the deprivation of oxygen in brain tissue. Plasma PUFAs, including AA, EPA, and DHA, are also increased under ischemic stroke in mice models [86]. Secretory PLA2 (sPLA2-IIA) is induced in reactive astrocytes in response to transient focal cerebral ischemia by occlusion of the middle cerebral artery (MCA) in the rat brain. sPLA2-IIA gene expression is up-regulated in rat brains after transient global ischemia [87]. The infarct size is reduced after the administration of quercitine, a PLA2 inhibitor, to MCA-occulted rats [88].
COX-2 is also upregulated in the infracted human brain [89] and in vivo models of stroke by occlusion of the MCA [90]. The selective COX-2 inhibitor NS-398 [91,92] reduces the accumulation of PGE2 in the post-ischemic brain and ameliorates cerebral ischemic damage [90]. PGD2, PGE2, PGF, and TXA2 are elevated in stroke [3,6,93]. The effects of PGs on the brain are dependent on their respective receptors. PGE2-EP1 receptors contribute to the neurotoxicity mediated by PGE2 by increasing the Ca2+ dysregulation underlying excitotoxic neuronal death [94]. ONO-AE-248, a selective EP3 agonist, significantly increased infarct size in the MCA occlusion model and aggravated the lesion caused by N-methyl-D-aspartic acid-induced excitotoxicity. Conversely, genetic deletion of EP3 receptor provided protection from acute excitotoxicity [95]. FP receptor of PGF has been shown to significantly contribute to excitotoxic brain injury and cerebral ischemia [96].
Lipoxygenase products were also detected in the brain after ischemia [4,97,98]. Concentrations of LTC4 and LTD4, which are potent inflammatory mediators [99], increased in the brain after ischemia [97]. The amounts of 5-, 9-, 11-, and 15-HETEs were significantly increased in ischemic rat brains after MCA occlusion [4]. The amelioration of stroke damage, by decreasing mortality-adjusted infarct size, was observed in FLAP knockout mice that do not produce LTs [100].
Markers of oxidative damage are increased immediately after ischemic stroke and remain elevated for several days after stroke onset. IsoPs are increased in different ischemic stroke models. The level of 8-epi-PGF2α is more elevated in primary rat cortical neurons exposed to hypoxia followed by reoxygenation as well as in in vivo model (rats submitted MCA occlusion followed by a reperfusion period) [5]. F2-IsoP levels are increased in the plasma of acute ischemic stroke patients [101]. 15-A2t-IsoP is abundantly produced in infarcted human cortical tissue, and 15-A2t-IsoP led to a rapid induction of the mitochondrial permeability transition pore and induced the release of cytochrome c from mitochondria in primary neuronal cultures [102]. 15-A2t-IsoP also induces neuronal apoptosis and potentiates oxidative glutamate toxicity in vitro [103]. The levels of F4-NPs and HETEs were significantly higher in the plasma of stroke patients [104]. These increases were highest from 6 to 12 h after stroke onset. Elevated levels of urinary 8-iso- and 2,3-dinor-F2-IsoPs were observed in stroke subjects, whereas urinary levels of 2,3-dinor-5,6-dihydro-F2-isoPs were decreased [104]. 4-HNE concentrations were higher in the plasma of experimental stroke rats with occlusion of the MCA [105]. Moreover, intravenous injection of 4-HNE increased both brain oxidative stress and infarct area induced by ischemia. Guo et al. (2013) [106] showed that mitochondrial aldehyde dehydrogenase 2 (ALDH2), an enzyme detoxifying aldehydes such as 4-HNE, protects against ischemic stroke. Regarding MDA, plasma and serum levels are positively correlated with stroke outcome [107,108,109] and negatively correlated with Mini-Mental State Examination (MMSE) scores [110].

4.3. Lipids as Neuroprotective Agents for Ischemic Stroke

4.3.1. Omega 3 PUFAs

The high level of lipids in the brain delineates their critical role in preserving neuronal function and synaptic plasticity. Among them, PUFAs are known to optimize synaptic membrane organization and function, rendering neurons more resistant to neurodegenerative processes. PUFA brain content varies between 25 and 30% and is composed mainly of DHA (12–14% of total fatty acids) and AA (8–10% of total fatty acids) [111,112], both being critical for brain function and contributing to the prevention of age-related neurodegeneration and cognitive deficits [113,114,115]. Omega-3 PUFAs play a key role in stroke neuroprotection. A high EPA/AA ratio was associated with good outcomes in ischemic stroke, suggesting that pre-stroke nutrition habits influence the severity of ischemic stroke in patients [116]. Low DHA level was also shown to be a potential factor affecting the risk of depression in stroke patients [117]. The protective effects of omega-3 PUFAs against damage in stroke patients have been shown to involve multiple pathways. The administration of DHA conferred high-grade neuroprotection in an MCA occlusion model [118,119]. The protective effects of omega-3 PUFAs against damage in stroke patients have been shown to involve multiple pathways. DHA reduces oxidative stress (F4-NPs and 8-iso-PGF2) following hypoxia–ischemia in the urine of piglets [120]. Post-stroke DHA injection efficiently reduced brain infarct and ameliorated neurological deficits 3 days after transient MCA occlusion, inhibiting the infiltration of immune cells and promoting the polarization of macrophages toward an anti-inflammatory M2 phenotype, therefore reducing central and peripheral inflammation after stroke [121]. DHA can reduce oxidative stress and cerebral ischemia–reperfusion injury by reducing COX-2 expression [122]. In rat brain microvascular endothelial cells under an oxygen- and glucose-deprivation environment (OGD), that mimics ischemic stroke in vitro, DHA was shown to decrease apoptosis, COX-2 protein expression, and the secretion of PGE2, PGI2, vascular endothelial growth factor (VEGF), and angiopoietin-2 (Ang-2, which has been shown to cause blood–brain barrier damage and to increase endothelial cell apoptosis) [123]. DHA can also decrease hypoxia/reoxygenation injury by activating Src-suppressed C kinase (SSeCKS), a substrate of protein kinase C that plays a key role in maintaining cell morphology and tight junctions of the blood–brain barrier and regulating cell permeability [124,125].
Other PUFAs have been shown to protect against ischemia. EPA inhibits oxidative damage and the inflammatory response after ischemic brain injury [126]. The deuterated form of linoleic PUFA (D4-Lnn) enhances its protective properties against oxidative stress caused by ischemia-like conditions [127]. ALA, the precursor of DHA, was also shown to reduce the consequence of stroke [128]. ALA, delivered either by intravenous injection (iv as a pre-treatment or a post-treatment therapy) or by dietary supplementation, has protective effects against cerebral ischemia [129,130,131,132].

4.3.2. PUFA Derivatives from Enzymatic Pathways

The roles of PUFA mediators in stroke neuroprotection are emerging [133]. Neuroprotectin D1 (NPD1), a DHA metabolite formed via a 15-LOX-initiated mechanism, has been shown to be neuroprotective in the ischemic brain [118,119].
NPD1 inhibits leukocyte infiltration mediated by brain ischemia–reperfusion, decreases proinflammatory gene expression, and increases neurogenesis [118,134]. The combination of resolvin D1 (RvD1) with NPD1 improves neuroprotection after focal cerebral ischemia in rats [135]. The exogenous supply of RvD2 via intraperitoneal injection, another derivative of ω-3 PUFAs, reversed cerebral ischemia–reperfusion injury caused by MCA occlusion by decreasing inflammation, brain edema, and neurological scores [136,137]. The neuroprotective actions of lipoxins (LXs) have been well established. Intracerebroventricular administration of LXA4 decreases infarct volume and neurological deficit after MCA occlusion, partly via PPARg agonistic actions and by decreasing inflammation and neutrophil infiltration [138,139,140]. Maresin 1 (MaR1) also protects against ischemia–reperfusion injury by inhibiting pro-inflammatory cytokines and NF-kB p65 activation [141].

4.3.3. Delivery of Omega-3 PUFAs to the Brain

The brain requires a constant supply of DHA from the blood to maintain DHA levels within the brain. Several plasma pools have been proposed to supply the brain with DHA, including plasma lipoproteins, lysophosphatidylcholine (LysoPC), and unesterified fatty acids. LysoPC-DHA is a privileged physiological carrier of DHA across the blood–brain barrier into the brain [142,143,144,145]. Studies showed that dietary DHA as LysoPC, but not as free acid, enriches the brain and improves memory in adult mice [146] and that LysoPC is more efficient than triacylglycerol or phosphatidylcholine in enriching the brain with DHA [147]. A specific protein expressed in brain endothelium called Mfsd2a has been reported to bind LysoPC-DHA, but not free DHA, for its uptake into the brain [148,149,150]. AceDoPC (1-acetyl,2-docosahexaenoyl-glycerophosphocholine), a stabilized form of LysoPC-DHA, targets efficiently and specifically DHA to the brain [151,152,153,154]. It was shown to prevent post-ischemic stroke consequences and to decrease oxidative stress in a stroke animal model of ischemia-reperfusion [155], to decrease the neuroinflammation induced by lipopolysaccharides in mice and microglia cells [156], and to stimulate neurogenesis in an in vitro OGD model of ischemia [157]. AceDoPC decreased the level of AA metabolites involved in oxidative stress and inflammation, including 8-epi-PGF2α, PGD2, PGF2, LTB4, and 15-HETE, and activated the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway [157], a critical antioxidant transcription factor in cells upregulating enzymes such as heme oxygenase 1 [158]. DHA carried in triacylglycerol emulsion exhibits neuroprotectin effects in mice subjected to hypoxic–ischemic brain injury [159]. Omega3-rich diets, like fish oil supplementations, combined with intraperitoneal DHA injections, exerted neuroprotective actions on ischemic brain injury [160].
The bioavailability and the delivery of omega-3 PUFAs or other drugs to the brain can be improved by nanoparticles. Positive outcomes highlight the efficiency of lipid nanoparticles and nanostructured lipid carriers to target the brain and protect against ischemic stroke [161,162,163]. The intranasal route of PUFAs or other drug delivery via lipid nanoparticles is also very promising, allowing the drug to directly reach the brain by bypassing the blood–brain barrier, a structure that restricts the passage of therapeutic agents and limits the efficacity of the delivery [164,165,166,167].

4.4. Lipid Biomarkers in Ischemic Stroke

Lipid-derived biomarkers in stroke research have gained significant attention because of their potential roles as diagnosis, prognosis, and therapeutic targets in stroke management. Different key lipid-derived biomarkers are studied in relation to stroke. This includes lipid peroxidation products (MDA, 4-HNE, IsoPs, and NPs), PLA2, and oxylipins, as previously described. Free FAs, as discussed above, also represent a significant predictive factor for the prognosis of ischemic stroke patients. For example, the DHA/EPA ratio has a positive correlation, while the EPA/AA ratio negatively correlates with all prognostic parameters (clinical, paraclinical, and outcome parameters) [168]. Saturated FAs play a negative role in long-term cognitive outcomes in stroke patients [169]. Increased low-density lipoprotein cholesterol (LDL-C) is also considered a relevant marker for ischemic stroke and was significantly associated with an increased risk of ischemic stroke [170,171,172]. However, some studies found no association between high levels of LDL-C and increased risk of ischemic stroke [173]. Non-high-density lipoprotein (HDL) cholesterol (meaning cholesterol of all atherogenic lipoproteins) was shown to be a better marker of risk of ischemic stroke than LDL-C [174]. The ratio of triglycerides (TGs)/HDL-C was significantly higher in young patients with ischemic stroke compared with older cases or healthy adults [175]. The TG/HDL cholesterol ratio was positively correlated and the total cholesterol/TG ratio was negatively correlated with silent brain infarct lesion burden [176], and the total cholesterol to HDL-C ratio was associated with an increased risk of ischemic stroke [169]. Another lipid class considered a biomarker of stroke is LysoPC. Low plasma levels of LysoPC 16:0 are a potential predictor of stroke recurrence [177]. LysoPC was closely associated with stroke recovery [178]. All these studies show an interest in lipidomic profile analyses. Indeed, comprehensive lipidomic analyses have revealed alterations in the overall lipid profiles of stroke patients. These lipidomic signatures may serve as diagnostic and prognostic biomarkers for stroke subtypes and severity. Multi-omics approaches are essential for lipid biomarker research. Opto-lipidomics also opens new opportunities to study lipid biomarkers [179]. Research into lipid-derived biomarkers in stroke is ongoing, and further elucidation of their roles in stroke pathophysiology may lead to the development of novel therapeutic strategies and personalized treatment approaches.

5. Conclusions

In conclusion, this review sheds light on the roles of oxidative stress and lipids in ischemic stroke. Lipids play multifaceted roles in stroke pathophysiology, contributing to neuronal injury, oxidative stress, inflammation, and blood–brain barrier dysfunction. Elucidating the specific lipid pathways involved in stroke pathogenesis and evaluating the efficacy of lipid-targeted interventions hold promise for improved outcomes following ischemic events. The identification of reliable lipid-derived biomarkers remains crucial for early diagnosis, prognostication, and therapeutic targeting. Different therapeutic options targeting lipid-derived biomarkers exist such as drugs that target more specifically LDL-C (statins; ezetimide; evinacumab; PCSK9 inhibitors…), the TG/HDL-C ratio (niacin or omega-3 fatty acids...), and oxylipins (acetylsalicylic acid, aspirin, preventing the production of thromboxane by the inhibition of COX and then platelet aggregation; omega-3 fatty acids; selective COX-2 inhibitors…). Edaravone and Dl-3-n-Butylphthalide are two neuroprotective antioxidants that are approved for clinical use. While many promising novel drug therapies are in various stages of preclinical and clinical development, translating these findings into effective treatments for ischemic stroke remains a significant challenge. Rigorous clinical trials are essential to evaluate the safety and efficacy of these therapies in human patients and to bring them to widespread clinical use. Ongoing research is exploring novel therapeutic approaches for ischemic stroke, including stem cell therapy, neurorecovery agents, anti-inflammatory drugs, and neurovascular protective agents. Lipid nanoparticles also hold great promise as drug delivery vehicles in the treatment of stroke, offering targeted delivery, controlled release, and the potential for combination therapy and diagnostic applications. Further research and development in this field are essential to validate the clinical utility of lipid biomarkers, explore their potential as therapeutic targets in ischemic stroke management, and optimize the strategies of administration for stroke treatments.

Author Contributions

N.B.-H.: contributed to the conceptualization and supervision of this study and the writing of this manuscript; M.L., A.-N.L., and A.L.V.: contributed to the review of this manuscript and figure design. All authors have read and agreed to the published version of the manuscript.

Funding

The authors are thankful for the support of the National Institute of Applied Sciences Lyon and the French National Centre for Scientific Research.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. GBD 2019 Stroke Collaborators; Feigin, V.L.; Stark, B.A.; Johnson, C.O.; Roth, G.A.; Bisignano, C.; Abady, G.G.; Abbasifard, M.; Abbasi-Kangevari, M.; Abd-Allah, F.; et al. Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021, 20, 795–820. [Google Scholar] [CrossRef] [PubMed]
  2. Ge, R.; You, S.; Zheng, D.; Zhang, Z.; Cao, Y.; Chang, J. Global, regional, and national temporal trends of diet-related ischemic stroke mortality and disability from 1990 to 2019. Int. J. Stroke 2024, 17474930241237932. [Google Scholar] [CrossRef] [PubMed]
  3. Ogawa, H.; Sasaki, T.; Kassell, N.F.; Nakagomi, T.; Lehman, R.M.; Hongo, K. Immunohistochemical demonstration of increase in prostaglandin F2-alpha after recirculation in global ischemic rat brains. Acta Neuropathol. 1987, 75, 62–68. [Google Scholar] [CrossRef] [PubMed]
  4. Usui, M.; Asano, T.; Takakura, K. Identification and quantitative analysis of hydroxy-eicosatetraenoic acids in rat brains exposed to regional ischemia. Stroke 1987, 18, 490–494. [Google Scholar] [CrossRef] [PubMed]
  5. Marin, J.G.; Cornet, S.; Spinnewyn, B.; Demerlé-Pallardy, C.; Auguet, M.; Chabrier, P.E. BN 80933 inhibits F2-isoprostane elevation in focal cerebral ischaemia and hypoxic neuronal cultures. Neuroreport 2000, 11, 1357–1360. [Google Scholar] [CrossRef] [PubMed]
  6. Yokota, C.; Kaji, T.; Kuge, Y.; Inoue, H.; Tamaki, N.; Minematsu, K. Temporal and topographic profiles of cyclooxygenase-2 expression during 24 h of focal brain ishemia in rats. Neurosci. Lett. 2004, 357, 219–222. [Google Scholar] [CrossRef] [PubMed]
  7. Lopez, A.D.; Mathers, C.D.; Ezzati, M.; Jamison, D.T.; Murray, C.J. Global and regional burden of disease and risk factors, 2001: Systematic analysis of population health data. Lancet 2006, 367, 1747–1757. [Google Scholar] [CrossRef] [PubMed]
  8. Donkor, E.S. Stroke in the 21st Century: A Snapshot of the Burden, Epidemiology, and Quality of Life. Stroke Res. Treat. 2018, 2018, 3238165. [Google Scholar] [CrossRef] [PubMed]
  9. Katan, M.; Luft, A. Global Burden of Stroke. Semin. Neurol. 2018, 38, 208–211. [Google Scholar] [CrossRef]
  10. GBD 2017 US Neurological Disorders Collaborators; Feigin, V.L.; Vos, T.; Alahdab, F.; Amit, A.M.L.; Bärnighausen, T.W.; Beghi, E.; Beheshti, M.; Chavan, P.P.; Criqui, M.H.; et al. Burden of Neurological Disorders Across the US From 1990-2017: A Global Burden of Disease Study. JAMA Neurol. 2021, 78, 165–176. [Google Scholar] [CrossRef]
  11. GBD 2021 Nervous System Disorders Collaborators; Steinmetz, J.D.; Seeher, K.M.; Schiess, N.; Nichols, E.; Cao, B.; Servili, C.; Cavallera, V.; Cousin, E.; Hagins, H.; et al. Global, regional, and national burden of disorders affecting the nervous system, 1990–2021: A systematic analysis for the Global Burden of Disease Study 2021. Lancet Neurol. 2024, 23, 344–381. [Google Scholar] [CrossRef]
  12. Deuschl, G.; Beghi, E.; Fazekas, F.; Varga, T.; Christoforidi, K.A.; Sipido, E.; Bassetti, C.L.; Vos, T.; Feigin, V.L. The burden of neurological diseases in Europe: An analysis for the Global Burden of Disease Study 2017. Lancet Public Health 2020, 5, e551–e567. [Google Scholar] [CrossRef] [PubMed]
  13. Kissela, B.M.; Khoury, J.C.; Alwell, K.; Moomaw, C.J.; Woo, D.; Adeoye, O.; Flaherty, M.L.; Khatri, P.; Ferioli, S.; De Los Rios La Rosa, F.; et al. Age at stroke: Temporal trends in stroke incidence in a large, biracial population. Neurology 2012, 79, 1781–1787. [Google Scholar] [CrossRef]
  14. Béjot, Y.; Daubail, B.; Jacquin, A.; Durier, J.; Osseby, G.V.; Rouaud, O.; Giroud, M. Trends in the incidence of ischaemic stroke in young adults between 1985 and 2011: The Dijon Stroke Registry. J. Neurol. Neurosurg. Psychiatry 2014, 85, 509–513. [Google Scholar] [CrossRef]
  15. Burke, J.F.; Skolarus, L.E. Are More Young People Having Strokes?—A Simple Question with an Uncertain Answer. JAMA Neurol. 2017, 74, 639–641. [Google Scholar] [CrossRef]
  16. Scott, C.A.; Li, L.; Rothwell, P.M. Diverging Temporal Trends in Stroke Incidence in Younger vs Older People: A Systematic Review and Meta-analysis. JAMA Neurol. 2022, 79, 1036–1048. [Google Scholar] [CrossRef]
  17. Feigin, V.L.; Norrving, B.; Mensah, G.A. Global Burden of Stroke. Circ. Res. 2017, 120, 439–448. [Google Scholar] [CrossRef] [PubMed]
  18. Johnston, S.C.; Mendis, S.; Mathers, C.D. Global variation in stroke burden and mortality: Estimates from monitoring, surveillance, and modelling. Lancet Neurol. 2009, 8, 345–354. [Google Scholar] [CrossRef]
  19. Owolabi, M.O.; Ugoya, S.; Platz, T. Racial disparity in stroke risk factors: The Berlin-Ibadan experience; a retrospective study. Acta Neurol. Scand. 2009, 119, 81–87. [Google Scholar] [CrossRef] [PubMed]
  20. Bravata, D.M.; Wells, C.K.; Gulanski, B.; Kernan, W.N.; Brass, L.M.; Long, J.; Concato, J. Racial disparities in stroke risk factors: The impact of socioeconomic status. Stroke 2005, 36, 1507–1511. [Google Scholar] [CrossRef]
  21. González, H.M.; Tarraf, W.; Rodríguez, C.J.; Gallo, L.C.; Sacco, R.L.; Talavera, G.A.; Heiss, G.; Kizer, J.R.; Hernandez, R.; Davis, S.; et al. Cardiovascular health among diverse Hispanics/Latinos: Hispanic Community Health Study/Study of Latinos (HCHS/SOL) results. Am. Heart J. 2016, 176, 134–144. [Google Scholar] [CrossRef]
  22. Silva, G.S.; Koroshetz, W.J.; González, R.G.; Schwamm, L.H. Causes of Ischemic Stroke. In Acute Ischemic Stroke; González, R., Hirsch, J., Lev, M., Schaefer, P., Schwamm, L., Eds.; Springer: Berlin/Heidelberg, Germany, 2011. [Google Scholar] [CrossRef]
  23. Goldstein, L.B.; Adams, R.; Alberts, M.J.; Appel, L.J.; Brass, L.M.; Bushnell, C.D.; Culebras, A.; Degraba, T.J.; Gorelick, P.B.; Guyton, J.R.; et al. Primary prevention of ischemic stroke: A guideline from the American Heart Association/American Stroke Association Stroke Council: Cosponsored by the Atherosclerotic Peripheral Vascular Disease Interdisciplinary Working Group; Cardiovascular Nursing Council; Clinical Cardiology Council; Nutrition, Physical Activity, and Metabolism Council; and the Quality of Care and Outcomes Research Interdisciplinary Working Group: The American Academy of Neurology affirms the value of this guideline. Stroke 2006, 37, 1583–1633. [Google Scholar] [CrossRef]
  24. Sacco, R.L.; Adams, R.; Albers, G.; Alberts, M.J.; Benavente, O.; Furie, K.; Goldstein, L.B.; Gorelick, P.; Halperin, J.; Harbaugh, R.; et al. Guidelines for prevention of stroke in patients with ischemic stroke or transient ischemic attack: A statement for healthcare professionals from the American Heart Association/American Stroke Association Council on Stroke: Co-sponsored by the Council on Cardiovascular Radiology and Intervention: The American Academy of Neurology affirms the value of this guideline. Stroke 2006, 37, 577–617. [Google Scholar] [CrossRef]
  25. Donnan, G.A. International Journal of Stroke. Editorial. Int. J. Stroke 2008, 3, 157. [Google Scholar] [CrossRef]
  26. Amarenco, P.; Bogousslavsky, J.; Caplan, L.R.; Donnan, G.A.; Hennerici, M.G. Classification of stroke subtypes. Cerebrovasc. Dis. 2009, 27, 493–501. [Google Scholar] [CrossRef]
  27. Unnithan, A.K.A.; Das, J.M.; Mehta, P. Hemorrhagic Stroke—Abstract—Europe PMC. 2022. Available online: https://europepmc.org/article/nbk/nbk559173?utm_medium=email&utm_source=transaction&client=bot&client=bot (accessed on 17 April 2023).
  28. Chen, S.; Zeng, L.; Hu, Z. Progressing haemorrhagic stroke: Categories, causes, mechanisms and managements. J. Neurol. 2014, 261, 2061–2078. [Google Scholar] [CrossRef]
  29. Ojaghihaghighi, S.; Vahdati, S.S.; Mikaeilpour, A.; Ramouz, A. Comparison of neurological clinical manifestation in patients with hemorrhagic and ischemic stroke. World J Emerg Med. 2017, 8, 34–38. [Google Scholar] [CrossRef]
  30. Montaño, A.; Hanley, D.F.; Hemphill, J.C., 3rd. Hemorrhagic stroke. Handb. Clin. Neurol. 2021, 176, 229–248. [Google Scholar] [CrossRef]
  31. Paul, S.; Candelario-Jalil, E. Emerging neuroprotective strategies for the treatment of ischemic stroke: An overview of clinical and preclinical studies. Exp. Neurol. 2021, 335, 113518. [Google Scholar] [CrossRef]
  32. Emberson, J.; Lees, K.R.; Lyden, P.; Blackwell, L.; Albers, G.; Bluhmki, E.; Brott, T.; Cohen, G.; Davis, S.; Donnan, G.; et al. Effect of treatment delay, age, and stroke severity on the effects of intravenous thrombolysis with alteplase for acute ischaemic stroke: A meta-analysis of individual patient data from randomised trials. Lancet 2014, 384, 1929–1935. [Google Scholar] [CrossRef]
  33. Hacke, W.; Kaste, M.; Bluhmki, E.; Brozman, M.; Dávalos, A.; Guidetti, D.; Larrue, V.; Lees, K.R.; Medeghri, Z.; Machnig, T.; et al. Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N. Engl. J. Med. 2008, 359, 1317–1329. [Google Scholar] [CrossRef] [PubMed]
  34. Narne, P.; Pandey, V.; Phanithi, P.B. The interplay between mitochondrial metabolism and oxidative stress in ischemic stroke: An epigenetic connection. Mol. Cell. Neurosci. 2017, 82, 176–194. [Google Scholar] [CrossRef]
  35. Ren, J.X.; Li, C.; Yan, X.L.; Qu, Y.; Yang, Y.; Guo, Z.N. Crosstalk between oxidative stress and ferroptosis/oxytosis in ischemic stroke: Possible targets and molecular mechanisms. Oxid. Med. Cell. Longev. 2021, 2021, 6643382. [Google Scholar] [CrossRef] [PubMed]
  36. Chavda, V.; Chaurasia, B.; Garg, K.; Deora, H.; Umana, G.E.; Palmisciano, P.; Scalia, G.; Lu, B. Molecular mechanisms of oxidative stress in stroke and cancer. Brain Disord. 2022, 5, 100029. [Google Scholar] [CrossRef]
  37. Dringen, R. Metabolism and functions of glutathione in brain. Prog. Neurobiol. 2000, 62, 649–671. [Google Scholar] [CrossRef]
  38. Saeed, S.A.; Shad, K.F.; Saleem, T.; Javed, F.; Khan, M.U. Some new prospects in the understanding of the molecular basis of the pathogenesis of stroke. Exp. Brain Res. 2007, 182, 1–10. [Google Scholar] [CrossRef] [PubMed]
  39. Briones-Valdivieso, C.; Briones, F.; Orellana-Urzúa, S.; Chichiarelli, S.; Saso, L.; Rodrigo, R. Novel Multi-Antioxidant Approach for Ischemic Stroke Therapy Targeting the Role of Oxidative Stress. Biomedicines 2024, 12, 501. [Google Scholar] [CrossRef]
  40. Cadenas, E. Basic mechanisms of antioxidant activity. Biofactors 1997, 6, 391–397. [Google Scholar] [CrossRef] [PubMed]
  41. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M. Telser Free radicals and antioxidants in normal physiological functions and human disease. J. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  42. Khan, R.A.; Khan, M.R.; Sahreen, S. Brain antioxidant markers, cognitive performance and acetylcholinesterase activity of rats: Efficiency of Sonchus asper. Behav. Brain Funct. 2012, 8, 21. [Google Scholar] [CrossRef]
  43. Lubos, E.; Loscalzo, J.; Handy, D.E. Glutathione peroxidase-1 in health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2011, 15, 1957–1997. [Google Scholar] [CrossRef] [PubMed]
  44. Yoshioka, M.; Tanaka Ki Miyazaki, I.; Fujita, N.; Higashi, Y.; Asanuma, M.; Ogawa, N. The dopamine agonist cabergoline provides neuroprotection by activation of the glutathione system and scavenging free radicals. Neurosci. Res. 2002, 43, 259–267. [Google Scholar] [CrossRef] [PubMed]
  45. Sivandzade, F.; Prasad, S.; Bhalerao, A.; Cucullo, L. NRF2 and NF-κB interplay in cerebrovascular and neurodegenerative disorders: Molecular mechanisms and possible therapeutic approaches. Redox Biol. 2019, 21, 101059. [Google Scholar] [CrossRef]
  46. Wang, L.; Zhang, X.; Xiong, X.; Zhu, H.; Chen, R.; Zhang, S.; Chen, G.; Jian, Z. Nrf2 Regulates Oxidative Stress and Its Role in Cerebral Ischemic Stroke. Antioxidants 2022, 11, 2377. [Google Scholar] [CrossRef] [PubMed]
  47. O’Brien, J.S.; Sampson, E.L. Fatty acid and fatty aldehyde composition of the major brain lipids in normal human gray matter, white matter, and myelin. J. Lipid Res. 1965, 6, 545–551. [Google Scholar] [CrossRef] [PubMed]
  48. Bourre, J.M.; Bonneil, M.; Clément, M.; Dumont, O.; Durand, G.; Lafont, H.; Nalbone, G.; Piciotti, M. Function of dietary polyunsaturated fatty acids in the nervous system. Prostaglandins Leukot. Essent. Fat. Acids 1993, 48, 5–15. [Google Scholar] [CrossRef] [PubMed]
  49. Belkouch, M.; Hachem, M.; Elgot, A.; Lo Van, A.; Picq, M.; Guichardant, M.; Lagarde, M.; Bernoud-Hubac, N. The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer’s disease. J. Nutr. Biochem. 2016, 38, 1–11. [Google Scholar] [CrossRef] [PubMed]
  50. Bazinet, R.P.; Bernoud-Hubac, N.; Lagarde, M. How the plasma lysophospholipid and unesterified fatty acid pools supply the brain with docosahexaenoic acid. Prostaglandins Leukot. Essent. Fat. Acids 2019, 142, 1–3. [Google Scholar] [CrossRef] [PubMed]
  51. Phillis, J.W.; Horrocks, L.A.; Farooqui, A.A. Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: Their role and involvement in neurological disorders. Brain Res. Rev. 2006, 52, 201–243. [Google Scholar] [CrossRef]
  52. Smith, M.L.; Murphy, R.C. The eicosanoids: Cyclooxygenase, lipoxygenase and epoxygenase pathways. In Biochemistry of Lipids, Lipoproteins and Membranes, 6th ed.; Ridgeway, N.D., McLeod, R.S., Eds.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 260–296. [Google Scholar]
  53. Christie, W.W.; Harwood, J.L. Oxidation of polyunsaturated fatty acids to produce lipid mediators. Essays Biochem. 2020, 64, 401–421. [Google Scholar] [CrossRef]
  54. Dennis, E.A.; Cao, J.; Hsu, Y.H.; Magrioti, V.; Kokotos, G. Phospholipase A2 enzymes: Physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem. Rev. 2011, 111, 6130–6185. [Google Scholar] [CrossRef]
  55. Murakami, M.; Sato, H.; Taketomi, Y. Updating Phospholipase A2 Biology. Biomolecules 2020, 10, 1457. [Google Scholar] [CrossRef] [PubMed]
  56. Lands, W.E.; Samuelsson, B. Phospholipid precursors of prostaglandins. Biochim. Biophys. Acta. 1968, 164, 426–429. [Google Scholar] [CrossRef] [PubMed]
  57. Hamberg, M.; Svensson, J.; Samuelsson, B. Prostaglandin endoperoxides. A new concept concerning the mode of action and release of prostaglandins. Proc. Natl. Acad. Sci. USA 1974, 71, 3824–3828. [Google Scholar] [CrossRef]
  58. Salomon, R.G.; Miller, D.B. Levuglandins: Isolation, characterization, and total synthesis of new secoprostanoid products from prostaglandin endoperoxides. Adv. Prostaglandin Thromboxane Leukot. Res. 1985, 15, 323–326. [Google Scholar]
  59. Kulmacz, R.J.; van der Donk, W.A.; Tsai, A.L. Comparison of the properties of prostaglandin H synthase-1 and -2. Prog. Lipid Res. 2003, 42, 377–404. [Google Scholar] [CrossRef]
  60. Rouzer, C.A.; Marnett, L.J. Cyclooxygenases: Structural and functional insights. J. Lipid Res. 2009, 50, S29–S34. [Google Scholar] [CrossRef] [PubMed]
  61. Dyall, S.C.; Balas, L.; Bazan, N.G.; Brenna, J.T.; Chiang, N.; da Costa Souza, F.; Dalli, J.; Durand, T.; Galano, J.M.; Lein, P.J.; et al. Polyunsaturated fatty acids and fatty acid-derived lipid mediators: Recent advances in the understanding of their biosynthesis, structures, and functions. Prog. Lipid Res. 2022, 86, 101165. [Google Scholar] [CrossRef]
  62. Guichardant, M.; Chen, P.; Liu, M.; Lo Van, A.; Jouvène, C.; Bernoud-Hubac, N.; Véricel, E.; Lagarde, M. Double lipoxygenation of polyunsaturated fatty acids of nutritional interest. Prostaglandins Leukot. Essent. Fat. Acids 2020, 162, 102185. [Google Scholar] [CrossRef]
  63. Cipollina, C.; Salvatore, S.R.; Muldoon, M.F.; Freeman, B.A.; Schopfer, F.J. Generation and dietary modulation of anti-inflammatory electrophilic omega-3 fatty acid derivatives. PLoS ONE. 2014, 9, e94836. [Google Scholar] [CrossRef]
  64. Capdevila, J.H.; Falck, J.R. The arachidonic acid monooxygenase: From biochemical curiosity to physiological/pathophysiological significance. J. Lipid Res. 2018, 59, 2047–2062. [Google Scholar] [CrossRef] [PubMed]
  65. Arnold, C.; Konkel, A.; Fischer, R.; Schunck, W.H. Cytochrome P450-dependent metabolism of omega-6 and omega-3 long-chain polyunsaturated fatty acids. Pharmacol. Rep. 2010, 62, 536–547. [Google Scholar] [CrossRef] [PubMed]
  66. Porter, N.A.; Caldwell, S.E.; Mills, K.A. Mechanisms of free radical oxidation of unsaturated lipids. Lipids 1995, 30, 277–290. [Google Scholar] [CrossRef] [PubMed]
  67. Morrow, J.D.; Hill, K.E.; Burk, R.F.; Nammour, T.M.; Badr, K.F.; Roberts, L.J., 2nd. A series of prostaglandin F2-like compounds are produced in vivo in humans by a non-cyclooxygenase, free radical-catalyzed mechanism. Proc. Natl. Acad. Sci. USA 1990, 87, 9383–9387. [Google Scholar] [CrossRef] [PubMed]
  68. Reich, E.E.; Zackert, W.E.; Brame, C.J.; Chen, Y.; Roberts, L.J., 2nd; Hachey, D.L.; Montine, T.J.; Morrow, J.D. Formation of novel D-ring and E-ring isoprostane-like compounds (D4/E4-neuroprostanes) in vivo from docosahexaenoic acid. Biochemistry 2000, 39, 2376–2383. [Google Scholar] [CrossRef] [PubMed]
  69. Morrow, J.D.; Awad, J.A.; Boss, H.J.; Blair, I.A.; Roberts, L.J., 2nd. Non-cyclooxygenase-derived prostanoids (F2-isoprostanes) are formed in situ on phospholipids. Proc. Natl. Acad. Sci. USA 1992, 89, 10721–10725. [Google Scholar] [CrossRef] [PubMed]
  70. Morrow, J.D.; Minton, T.A.; Mukundan, C.R.; Campbell, M.D.; Zackert, W.E.; Daniel, V.C.; Badr, K.F.; Blair, I.A.; Roberts, L.J., 2nd. Free radical-induced generation of isoprostanes in vivo. Evidence for the formation of D-ring and E-ring isoprostanes. J. Biol. Chem. 1994, 269, 4317–4326. [Google Scholar] [CrossRef] [PubMed]
  71. Morrow, J.D.; Awad, J.A.; Wu, A.; Zackert, W.E.; Daniel, V.C.; Roberts, L.J., 2nd. Nonenzymatic free radical-catalyzed generation of thromboxane-like compounds (isothromboxanes) in vivo. J. Biol. Chem. 1996, 271, 23185–23190. [Google Scholar] [CrossRef] [PubMed]
  72. Roberts, L.J., 2nd; Montine, T.J.; Markesbery, W.R.; Tapper, A.R.; Hardy, P.; Chemtob, S.; Dettbarn, W.D.; Morrow, J.D. Formation of isoprostane-like compounds (neuroprostanes) in vivo from docosahexaenoic acid. J. Biol. Chem. 1998, 273, 13605–13612. [Google Scholar] [CrossRef]
  73. Brame, C.J.; Salomon, R.G.; Morrow, J.D.; Roberts, L.J., 2nd. Identification of extremely reactive gamma-ketoaldehydes (isolevuglandins) as products of the isoprostane pathway and characterization of their lysyl protein adducts. J. Biol. Chem. 1999, 274, 13139–13146. [Google Scholar] [CrossRef]
  74. Bernoud-Hubac, N.; Davies, S.S.; Boutaud, O.; Montine, T.J.; Roberts, L.J., 2nd. Formation of highly reactive gamma-ketoaldehydes (neuroketals) as products of the neuroprostane pathway. J. Biol. Chem. 2001, 276, 30964–30970. [Google Scholar] [CrossRef]
  75. Bernoud-Hubac, N.; Roberts, L.J., 2nd. Identification of oxidized derivatives of neuroketals. Biochemistry 2002, 41, 11466–11471. [Google Scholar] [CrossRef]
  76. Bernoud-Hubac, N.; Fay, L.B.; Armarnath, V.; Guichardant, M.; Bacot, S.; Davies, S.S.; Roberts, L.J., 2nd; Lagarde, M. Covalent binding of isoketals to ethanolamine phospholipids. Free Radic. Biol Med. 2004, 37, 1604–1611. [Google Scholar] [CrossRef]
  77. Esterbauer, H.; Benedetti, A.; Lang, J.; Fulceri, R.; Fauler, G.; Comporti, M. Studies on the mechanism of formation of 4-hydroxynonenal during microsomal lipid peroxidation. Biochim. Biophys. Acta 1986, 876, 154–166. [Google Scholar] [CrossRef]
  78. Van Kuijk, F.J.; Holte, L.L.; Dratz, E.A. 4-Hydroxyhexenal: A lipid peroxidation product derived from oxidized docosahexaenoic acid. Biochim. Biophys. Acta 1990, 1043, 116–118. [Google Scholar] [CrossRef]
  79. Murthi, K.K.; Friedman, L.R.; Oleinick, N.L.; Salomon, R.G. Formation of DNA-protein cross-links in mammalian cells by levuglandin E2. Biochemistry 1993, 32, 4090–4097. [Google Scholar] [CrossRef]
  80. Davies, S.S.; Amarnath, V.; Montine, K.S.; Bernoud-Hubac, N.; Boutaud, O.; Montine, T.J.; Roberts, L.J., 2nd. Effects of reactive gamma-ketoaldehydes formed by the isoprostane pathway (isoketals) and cyclooxygenase pathway (levuglandins) on proteasome function. FASEB J. 2002, 16, 715–717. [Google Scholar] [CrossRef]
  81. Bacot, S.; Bernoud-Hubac, N.; Baddas, N.; Chantegrel, B.; Deshayes, C.; Doutheau, A.; Lagarde, M.; Guichardant, M. Covalent binding of hydroxy-alkenals 4-HDDE, 4-HHE, and 4-HNE to ethanolamine phospholipid subclasses. J. Lipid Res. 2003, 44, 917–926. [Google Scholar] [CrossRef]
  82. Brame, C.J.; Boutaud, O.; Davies, S.S.; Yang, T.; Oates, J.A.; Roden, D.; Roberts, L.J., 2nd. Modification of proteins by isoketal-containing oxidized phospholipids. J. Biol. Chem. 2004, 279, 13447–13451. [Google Scholar] [CrossRef]
  83. Bernoud-Hubac, N.; Alam, D.A.; Lefils, J.; Davies, S.S.; Amarnath, V.; Guichardant, M.; Roberts, L.J., 2nd; Lagarde, M. Low concentrations of reactive gamma-ketoaldehydes prime thromboxane-dependent human platelet aggregation via p38-MAPK activation. Biochim. Biophys. Acta 2009, 1791, 307–313. [Google Scholar] [CrossRef]
  84. Davies, S.S.; Bodine, C.; Matafonova, E.; Pantazides, B.G.; Bernoud-Hubac, N.; Harrison, F.E.; Olson, S.J.; Montine, T.J.; Amarnath, V.; Roberts, L.J., 2nd. Treatment with a γ-ketoaldehyde scavenger prevents working memory deficits in hApoE4 mice. J. Alzheimers Dis. 2011, 27, 49–59. [Google Scholar] [CrossRef]
  85. Rodriguez de Turco, E.B.; Belayev, L.; Liu, Y.; Busto, R.; Parkins, N.; Bazan, N.G.; Ginsberg, M.D. Systemic fatty acid responses to transient focal cerebral ischemia: Influence of neuroprotectant therapy with human albumin. J. Neurochem. 2002, 83, 515–524. [Google Scholar] [CrossRef]
  86. Golovko, S.A.; Golovko, M.Y. Plasma Unesterified Fatty-Acid Profile Is Dramatically and Acutely Changed under Ischemic Stroke in the Mouse Model. Lipids 2018, 53, 641–645. [Google Scholar] [CrossRef]
  87. Lauritzen, I.; Heurteaux, C.; Lazdunski, M. Expression of group II phospholipase A2 in rat brain after severe forebrain ischemia and in endotoxic shock. Brain Res. 1994, 651, 353–356. [Google Scholar] [CrossRef]
  88. Estevez, A.Y.; Phillis, J.W. The phospholipase A2 inhibitor, quinacrine, reduces infarct size in rats after transient middle cerebral artery occlusion. Brain Res. 1997, 752, 203–208. [Google Scholar] [CrossRef]
  89. Sairanen, T.; Ristimäki, A.; Karjalainen-Lindsberg, M.L.; Paetau, A.; Kaste, M.; Lindsberg, P.J. Cyclooxygenase-2 is induced globally in infarcted human brain. Ann. Neurol. 1998, 43, 738–747. [Google Scholar] [CrossRef]
  90. Nogawa, S.; Zhang, F.; Ross, M.E.; Iadecola, C. Cyclo-oxygenase-2 gene expression in neurons contributes to ischemic brain damage. J. Neurosci. 1997, 17, 2746–2755. [Google Scholar] [CrossRef]
  91. Futaki, N.; Yoshikawa, K.; Hamasaka, Y.; Arai, I.; Higuchi, S.; Iizuka, H.; Otomo, S. NS-398, a novel non-steroidal anti-inflammatory drug with potent analgesic and antipyretic effects, which causes minimal stomach lesions. Gen. Pharmacol. 1993, 24, 105–110. [Google Scholar] [CrossRef]
  92. Masferrer, J.L.; Zweifel, B.S.; Manning, P.T.; Hauser, S.D.; Leahy, K.M.; Smith, W.G.; Isakson, P.C.; Seibert, K. Selective inhibition of inducible cyclooxygenase 2 in vivo is antiinflammatory and nonulcerogenic. Proc. Natl. Acad. Sci. USA 1994, 91, 3228–3232. [Google Scholar] [CrossRef]
  93. Fagan, S.C.; Castellani, D.; Gengo, F.M. Prostanoid concentrations in human CSF following acute ischaemic brain infarction. Clin. Exp. Pharmacol. Physiol. 1986, 13, 629–632. [Google Scholar] [CrossRef]
  94. Kawano, T.; Anrather, J.; Zhou, P.; Park, L.; Wang, G.; Frys, K.A.; Kunz, A.; Cho, S.; Orio, M.; Iadecola, C. Prostaglandin E2 EP1 receptors: Downstream effectors of COX-2 neurotoxicity. Nat. Med. 2006, 12, 225–229. [Google Scholar] [CrossRef]
  95. Ahmad, M.; Ahmad, A.S.; Zhuang, H.; Maruyama, T.; Narumiya, S.; Doré, S. Stimulation of prostaglandin E2-EP3 receptors exacerbates stroke and excitotoxic injury. J. Neuroimmunol. 2007, 184, 172–179. [Google Scholar] [CrossRef]
  96. Saleem, S.; Ahmad, A.S.; Maruyama, T.; Narumiya, S.; Doré, S. PGF FP receptor contributes to brain damage following transient focal brain ischemia. Neurotox. Res. 2009, 15, 62–70. [Google Scholar] [CrossRef]
  97. Moskowitz, M.A.; Kiwak, K.J.; Hekimian, K.; Levine, L. Synthesis of compounds with properties of leukotrienes C4 and D4 in gerbil brains after ischemia and reperfusion. Science 1984, 224, 886–889. [Google Scholar] [CrossRef]
  98. Kiwak, K.J.; Moskowitz, M.A.; Levine, L. Leukotriene production in gerbil brain after ischemic insult, subarachnoid hemorrhage, and concussive injury. J. Neurosurg. 1985, 62, 865–869. [Google Scholar] [CrossRef]
  99. Samuelsson, B.; Hammarström, S. Leukotrienes: A novel group of biologically active compounds. Vitam. Horm. 1982, 39, 1–30. [Google Scholar] [CrossRef]
  100. Ström, J.O.; Strid, T.; Hammarström, S. Disruption of the alox5ap gene ameliorates focal ischemic stroke: Possible consequence of impaired leukotriene biosynthesis. BMC Neurosci. 2012, 13, 146. [Google Scholar] [CrossRef]
  101. Kelly, P.J.; Morrow, J.D.; Ning, M.; Koroshetz, W.; Lo, E.H.; Terry, E.; Milne, G.L.; Hubbard, J.; Lee, H.; Stevenson, E.; et al. Oxidative stress and matrix metalloproteinase-9 in acute ischemic stroke: The Biomarker Evaluation for Antioxidant Therapies in Stroke (BEAT-Stroke) study. Stroke 2008, 39, 100–104. [Google Scholar] [CrossRef]
  102. Zeiger, S.L.; Musiek, E.S.; Zanoni, G.; Vidari, G.; Morrow, J.D.; Milne, G.J.; McLaughlin, B. Neurotoxic lipid peroxidation species formed by ischemic stroke increase injury. Free Radic. Biol. Med. 2009, 47, 1422–1431. [Google Scholar] [CrossRef]
  103. Musiek, E.S.; Breeding, R.S.; Milne, G.L.; Zanoni, G.; Morrow, J.D.; McLaughlin, B. Cyclopentenone isoprostanes are novel bioactive products of lipid oxidation which enhance neurodegeneration. J. Neurochem. 2006, 97, 1301–1313. [Google Scholar] [CrossRef]
  104. Seet, R.C.; Lee, C.Y.; Chan, B.P.; Sharma, V.K.; Teoh, H.L.; Venketasubramanian, N.; Lim, E.C.; Chong, W.L.; Looi, W.F.; Huang, S.H.; et al. Oxidative damage in ischemic stroke revealed using multiple biomarkers. Stroke 2011, 42, 2326–2329. [Google Scholar] [CrossRef]
  105. Lee, W.C.; Wong, H.Y.; Chai, Y.Y.; Shi, C.W.; Amino, N.; Kikuchi, S.; Huang, S.H. Lipid peroxidation dysregulation in ischemic stroke: Plasma 4-HNE as a potential biomarker? Biochem. Biophys. Res. Commun. 2012, 425, 842–847. [Google Scholar] [CrossRef]
  106. Guo, J.M.; Liu, A.J.; Zang, P.; Dong, W.Z.; Ying, L.; Wang, W.; Xu, P.; Song, X.R.; Cai, J.; Zhang, S.Q.; et al. ALDH2 protects against stroke by clearing 4-HNE. Cell Res. 2013, 23, 915–930. [Google Scholar] [CrossRef]
  107. Cojocaru, I.M.; Cojocaru, M.; Sapira, V.; Ionescu, A. Evaluation of oxidative stress in patients with acute ischemic stroke. Rom. J. Intern. Med. 2013, 51, 97–106. [Google Scholar]
  108. Elsayed, W.M.; Abdel-Gawad, E.H.A.; Mesallam, D.I.; El-Serafy, T.S. The relationship between oxidative stress and acute ischemic stroke severity and functional outcome. Egypt. J. Neurol. Psychiatry Neurosurg. 2020, 56, 74. [Google Scholar] [CrossRef]
  109. Menon, B.; Ramalingam, K.; Kumar, R. Evaluating the Role of Oxidative Stress in Acute Ischemic Stroke. J. Neurosci. Rural Pract. 2020, 11, 156–159. [Google Scholar] [CrossRef]
  110. Liu, Z.; Liu, Y.; Tu, X.; Shen, H.; Qiu, H.; Chen, H.; He, J. High Serum Levels of Malondialdehyde and 8-OHdG are both Associated with Early Cognitive Impairment in Patients with Acute Ischaemic Stroke. Sci. Rep. 2017, 7, 9493. [Google Scholar] [CrossRef]
  111. Carrié, I.; Clément, M.; de Javel, D.; Francès, H.; Bourre, J.M. Specific phospholipid fatty acid composition of brain regions in mice. Effects of n-3 polyunsaturated fatty acid deficiency and phospholipid supplementation. J. Lipid Res. 2000, 41, 465–472. [Google Scholar] [CrossRef]
  112. Joffre, C.; Grégoire, S.; De Smedt, V.; Acar, N.; Bretillon, L.; Nadjar, A.; Layé, S. Modulation of brain PUFA content in different experimental models of mice. Prostaglandins Leukot. Essent. Fat. Acids 2016, 114, 1–10. [Google Scholar] [CrossRef]
  113. Peet, M.; Stokes, C. Omega-3 fatty acids in the treatment of psychiatric disorders. Drugs 2005, 65, 1051–1059. [Google Scholar] [CrossRef]
  114. Gao, X.; Chen, H.; Fung, T.T.; Logroscino, G.; Schwarzschild, M.A.; Hu, F.B.; Ascherio, A. Prospective study of dietary pattern and risk of Parkinson disease. Am. J. Clin. Nutr. 2007, 86, 1486–1494. [Google Scholar] [CrossRef] [PubMed]
  115. Wysoczański, T.; Sokoła-Wysoczańska, E.; Pękala, J.; Lochyński, S.; Czyż, K.; Bodkowski, R.; Herbinger, G.; Patkowska-Sokoła, B.; Librowski, T. Omega-3 Fatty Acids and their Role in Central Nervous System—A Review. Curr. Med. Chem. 2016, 23, 816–831. [Google Scholar] [CrossRef] [PubMed]
  116. Mishina, M.; Kim, K.; Kominami, S.; Mizunari, T.; Kobayashi, S.; Katayama, Y. Impact of polyunsaturated fatty acid consumption prior to ischemic stroke. Acta Neurol. Scand. 2013, 127, 181–185. [Google Scholar] [CrossRef] [PubMed]
  117. Kotlega, D.; Zembron-Lacny, A.; Golab-Janowska, M.; Nowacki, P.; Szczuko, M. The Association of Free Fatty Acids and Eicosanoids with the Severity of Depressive Symptoms in Stroke Patients. Int. J. Mol. Sci. 2020, 21, 5220. [Google Scholar] [CrossRef] [PubMed]
  118. Belayev, L.; Marcheselli, V.L.; Khoutorova, L.; Rodriguez de Turco, E.B.; Busto, R.; Ginsberg, M.D.; Bazan, N.G. Docosahexaenoic acid complexed to albumin elicits high-grade ischemic neuroprotection. Stroke 2005, 36, 118–123. [Google Scholar] [CrossRef] [PubMed]
  119. Belayev, L.; Khoutorova, L.; Atkins, K.D.; Bazan, N.G. Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient, focal cerebral ischemia. Stroke 2009, 40, 3121–3126. [Google Scholar] [CrossRef] [PubMed]
  120. Huun, M.U.; Garberg, H.T.; Escobar, J.; Chafer, C.; Vento, M.; Holme, I.M.; Saugstad, O.D.; Solberg, R. DHA reduces oxidative stress following hypoxia-ischemia in newborn piglets: A study of lipid peroxidation products in urine and plasma. J. Perinat. Med. 2018, 46, 209–217. [Google Scholar] [CrossRef] [PubMed]
  121. Cai, W.; Liu, S.; Hu, M.; Sun, X.; Qiu, W.; Zheng, S.; Hu, X.; Lu, Z. Post-stroke DHA Treatment Protects Against Acute Ischemic Brain Injury by Skewing Macrophage Polarity Toward the M2 Phenotype. Transl. Stroke Res. 2018, 9, 669–680. [Google Scholar] [CrossRef]
  122. Quartu, M.; Serra, M.P.; Boi, M.; Pillolla, G.; Melis, T.; Poddighe, L.; Del Fiacco, M.; Falconieri, D.; Carta, G.; Murru, E.; et al. Effect of acute administration of Pistacia lentiscus L. essential oil on rat cerebral cortex following transient bilateral common carotid artery occlusion. Lipids Health Dis. 2012, 11, 8. [Google Scholar] [CrossRef]
  123. Chen, X.; Wang, Q.; Zhan, L.; Shu, A. Effects and mechanisms of docosahexaenoic acid on the generation of angiopoietin-2 by rat brain microvascular endothelial cells under an oxygen- and glucose-deprivation environment. Springerplus 2016, 5, 1518. [Google Scholar] [CrossRef]
  124. Lee, S.W.; Kim, W.J.; Choi, Y.K.; Song, H.S.; Son, M.J.; Gelman, I.H.; Kim, Y.J.; Kim, K.W. SSeCKS regulates angiogenesis and tight junction formation in blood-brain barrier. Nat. Med. 2003, 9, 900–906. [Google Scholar] [CrossRef]
  125. Yu, Y.; Fang, H.; Qiu, Z.; Xia, Z.; Zhou, B. DHA Attenuates Hypoxia/Reoxygenation Injury by Activating SSeCKS in Human Cerebrovascular Pericytes. Neurochem. Res. 2020, 45, 310–321. [Google Scholar] [CrossRef]
  126. Okabe, N.; Nakamura, T.; Toyoshima, T.; Miyamoto, O.; Lu, F.; Itano, T. Eicosapentaenoic acid prevents memory impairment after ischemia by inhibiting inflammatory response and oxidative damage. J. Stroke Cerebrovasc. Dis. 2011, 20, 188–195. [Google Scholar] [CrossRef]
  127. Turovsky, E.A.; Varlamova, E.G.; Gudkov, S.V.; Plotnikov, E.Y. The Protective Mechanism of Deuterated Linoleic Acid Involves the Activation of the Ca2+ Signaling System of Astrocytes in Ischemia In Vitro. Int. J. Mol. Sci. 2021, 22, 13216. [Google Scholar] [CrossRef] [PubMed]
  128. Blondeau, N. The nutraceutical potential of omega-3 alpha-linolenic acid in reducing the consequences of stroke. Biochimie 2016, 120, 49–55. [Google Scholar] [CrossRef]
  129. Blondeau, N.; Nguemeni, C.; Debruyne, D.N.; Piens, M.; Wu, X.; Pan, H.; Hu, X.; Gandin, C.; Lipsky, R.H.; Plumier, J.C.; et al. Subchronic alpha-linolenic acid treatment enhances brain plasticity and exerts an antidepressant effect: A versatile potential therapy for stroke. Neuropsychopharmacology 2009, 34, 2548–2559. [Google Scholar] [CrossRef]
  130. Heurteaux, C.; Guy, N.; Laigle, C.; Blondeau, N.; Duprat, F.; Mazzuca, M.; Lang-Lazdunski, L.; Widmann, C.; Zanzouri, M.; Romey, G.; et al. TREK-1, a K+ channel involved in neuroprotection and general anesthesia. EMBO J. 2004, 23, 2684–2695. [Google Scholar] [CrossRef]
  131. Nguemeni, C.; Delplanque, B.; Rovère, C.; Simon-Rousseau, N.; Gandin, C.; Agnani, G.; Nahon, J.L.; Heurteaux, C.; Blondeau, N. Dietary supplementation of alpha-linolenic acid in an enriched rapeseed oil diet protects from stroke. Pharmacol. Res. 2010, 61, 226–233. [Google Scholar] [CrossRef]
  132. Bourourou, M.; Heurteaux, C.; Blondeau, N. Alpha-linolenic acid given as enteral or parenteral nutritional intervention against sensorimotor and cognitive deficits in a mouse model of ischemic stroke. Neuropharmacology 2016, 108, 60–72. [Google Scholar] [CrossRef]
  133. Miao, Z.; Schultzberg, M.; Wang, X.; Zhao, Y. Role of polyunsaturated fatty acids in ischemic stroke—A perspective of specialized pro-resolving mediators. Clin. Nutr. 2021, 40, 2974–2987. [Google Scholar] [CrossRef]
  134. Marcheselli, V.L.; Hong, S.; Lukiw, W.J.; Tian, X.H.; Gronert, K.; Musto, A.; Hardy, M.; Gimenez, J.M.; Chiang, N.; Serhan, C.N.; et al. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. J. Biol. Chem. 2003, 278, 43807–43817. [Google Scholar] [CrossRef] [PubMed]
  135. Reid, M.M.; Kautzmann, M.I.; Andrew, G.; Obenaus, A.; Mukherjee, P.K.; Khoutorova, L.; Ji, J.X.; Roque, C.R.; Oria, R.B.; Habeb, B.F.; et al. NPD1 Plus RvD1 Mediated Ischemic Stroke Penumbra Protection Increases Expression of Pro-homeostatic Microglial and Astrocyte Genes. Cell. Mol. Neurobiol. 2023, 43, 3555–3573. [Google Scholar] [CrossRef]
  136. Zuo, G.; Zhang, D.; Mu, R.; Shen, H.; Li, X.; Wang, Z.; Li, H.; Chen, G. Resolvin D2 protects against cerebral ischemia/reperfusion injury in rats. Mol. Brain 2018, 11, 9. [Google Scholar] [CrossRef] [PubMed]
  137. Tułowiecka, N.; Kotlęga, D.; Prowans, P.; Szczuko, M. The Role of Resolvins: EPA and DHA Derivatives Can Be Useful in the Prevention and Treatment of Ischemic Stroke. Int. J. Mol. Sci. 2020, 21, 7628. [Google Scholar] [CrossRef] [PubMed]
  138. Sobrado, M.; Pereira, M.P.; Ballesteros, I.; Hurtado, O.; Fernández-López, D.; Pradillo, J.M.; Caso, J.R.; Vivancos, J.; Nombela, F.; Serena, J.; et al. Synthesis of lipoxin A4 by 5-lipoxygenase mediates PPARgamma-dependent, neuroprotective effects of rosiglitazone in experimental stroke. J. Neurosci. 2009, 29, 3875–3884. [Google Scholar] [CrossRef] [PubMed]
  139. Tułowiecka, N.; Kotlęga, D.; Bohatyrewicz, A.; Szczuko, M. Could Lipoxins Represent a New Standard in Ischemic Stroke Treatment? Int. J. Mol. Sci. 2021, 22, 4207. [Google Scholar] [CrossRef] [PubMed]
  140. Zhang, J.; Li, Z.; Fan, M.; Jin, W. Lipoxins in the Nervous System: Brighter Prospects for Neuroprotection. Front. Pharmacol. 2022, 13, 781889. [Google Scholar] [CrossRef] [PubMed]
  141. Xian, W.; Wu, Y.; Xiong, W.; Li, L.; Li, T.; Pan, S.; Song, L.; Hu, L.; Pei, L.; Yao, S.; et al. The pro-resolving lipid mediator Maresin 1 protects against cerebral ischemia/reperfusion injury by attenuating the pro-inflammatory response. Biochem. Biophys. Res. Commun. 2016, 472, 175–181. [Google Scholar] [CrossRef] [PubMed]
  142. Thies, F.; Pillon, C.; Moliere, P.; Lagarde, M.; Lecerf, J. Preferential incorporation of sn-2 lysoPC DHA over unesterified DHA in the young rat brain. Am. J. Physiol. 1994, 267 Pt 2, R1273–R1279. [Google Scholar] [CrossRef]
  143. Bernoud, N.; Fenart, L.; Molière, P.; Dehouck, M.P.; Lagarde, M.; Cecchelli, R.; Lecerf, J. Preferential transfer of 2-docosahexaenoyl-1-lysophosphatidylcholine through an in vitro blood-brain barrier over unesterified docosahexaenoic acid. J. Neurochem. 1999, 72, 338–345. [Google Scholar] [CrossRef]
  144. Lagarde, M.; Bernoud, N.; Brossard, N.; Lemaitre-Delaunay, D.; Thiès, F.; Croset, M.; Lecerf, J. Lysophosphatidylcholine as a preferred carrier form of docosahexaenoic acid to the brain. J. Mol. Neurosci. 2001, 16, 201–204; discussion 215–221. [Google Scholar] [CrossRef]
  145. Lo Van, A.; Sakayori, N.; Hachem, M.; Belkouch, M.; Picq, M.; Lagarde, M.; Osumi, N.; Bernoud-Hubac, N. Mechanisms of DHA transport to the brain and potential therapy to neurodegenerative diseases. Biochimie 2016, 130, 163–167. [Google Scholar] [CrossRef]
  146. Sugasini, D.; Thomas, R.; Yalagala, P.C.R.; Tai, L.M.; Subbaiah, P.V. Dietary docosahexaenoic acid (DHA) as lysophosphatidylcholine, but not as free acid, enriches brain DHA and improves memory in adult mice. Sci. Rep. 2017, 7, 11263. [Google Scholar] [CrossRef]
  147. Sugasini, D.; Yalagala, P.C.R.; Goggin, A.; Tai, L.M.; Subbaiah, P.V. Enrichment of brain docosahexaenoic acid (DHA) is highly dependent upon the molecular carrier of dietary DHA: Lysophosphatidylcholine is more efficient than either phosphatidylcholine or triacylglycerol. J. Nutr. Biochem. 2019, 74, 108231. [Google Scholar] [CrossRef]
  148. Nguyen, L.N.; Ma, D.; Shui, G.; Wong, P.; Cazenave-Gassiot, A.; Zhang, X.; Wenk, M.R.; Goh, E.L.; Silver, D.L. Mfsd2a is a transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature 2014, 509, 503–506. [Google Scholar] [CrossRef]
  149. Wong, B.H.; Silver, D.L. Mfsd2a: A Physiologically Important Lysolipid Transporter in the Brain and Eye. Adv. Exp. Med. Biol. 2020, 1276, 223–234. [Google Scholar] [CrossRef]
  150. Cater, R.J.; Chua, G.L.; Erramilli, S.K.; Keener, J.E.; Choy, B.C.; Tokarz, P.; Chin, C.F.; Quek, D.Q.Y.; Kloss, B.; Pepe, J.G.; et al. Structural basis of omega-3 fatty acid transport across the blood-brain barrier. Nature 2021, 595, 315–319. [Google Scholar] [CrossRef]
  151. Lagarde, M.; Hachem, M.; Bernoud-Hubac, N.; Picq, M.; Véricel, E.; Guichardant, M. Biological properties of a DHA-containing structured phospholipid (AceDoPC) to target the brain. Prostaglandins Leukot. Essent. Fat. Acids. 2015, 92, 63–65. [Google Scholar] [CrossRef]
  152. Hachem, M.; Géloën, A.; Van, A.L.; Foumaux, B.; Fenart, L.; Gosselet, F.; Da Silva, P.; Breton, G.; Lagarde, M.; Picq, M.; et al. Efficient Docosahexaenoic Acid Uptake by the Brain from a Structured Phospholipid. Mol. Neurobiol. 2016, 53, 3205–3215. [Google Scholar] [CrossRef] [PubMed]
  153. Hachem, M.; Nacir, H.; Picq, M.; Belkouch, M.; Bernoud-Hubac, N.; Windust, A.; Meiller, L.; Sauvinet, V.; Feugier, N.; Lambert-Porcheron, S.; et al. Docosahexaenoic Acid (DHA) Bioavailability in Humans after Oral Intake of DHA-Containing Triacylglycerol or the Structured Phospholipid AceDoPC. Nutrients 2020, 12, 251. [Google Scholar] [CrossRef] [PubMed]
  154. Lo Van, A.; Bernoud-Hubac, N.; Lagarde, M. Esterification of Docosahexaenoic Acid Enhances Its Transport to the Brain and Its Potential Therapeutic Use in Brain Diseases. Nutrients 2022, 14, 4550. [Google Scholar] [CrossRef] [PubMed]
  155. Chauveau, F.; Cho, T.H.; Perez, M.; Guichardant, M.; Riou, A.; Aguettaz, P.; Picq, M.; Lagarde, M.; Berthezène, Y.; Nighoghossian, N.; et al. Brain-targeting form of docosahexaenoic acid for experimental stroke treatment: MRI evaluation and anti-oxidant impact. Curr. Neurovasc. Res. 2011, 8, 95–102. [Google Scholar] [CrossRef] [PubMed]
  156. Fourrier, C.; Remus-Borel, J.; Greenhalgh, A.D.; Guichardant, M.; Bernoud-Hubac, N.; Lagarde, M.; Joffre, C.; Layé, S. Docosahexaenoic acid-containing choline phospholipid modulates LPS-induced neuroinflammation in vivo and in microglia in vitro. J. Neuroinflamm. 2017, 14, 170. [Google Scholar] [CrossRef] [PubMed]
  157. Lo Van, A.; Sakayori, N.; Hachem, M.; Belkouch, M.; Picq, M.; Fourmaux, B.; Lagarde, M.; Osumi, N.; Bernoud-Hubac, N. Targeting the Brain with a Neuroprotective Omega-3 Fatty Acid to Enhance Neurogenesis in Hypoxic Condition in Culture. Mol. Neurobiol. 2019, 56, 986–999. [Google Scholar] [CrossRef]
  158. Zhang, M.; Wang, S.; Mao, L.; Leak, R.K.; Shi, Y.; Zhang, W.; Hu, X.; Sun, B.; Cao, G.; Gao, Y.; et al. Omega-3 fatty acids protect the brain against ischemic injury by activating Nrf2 and upregulating heme oxygenase 1. J. Neurosci. 2014, 34, 1903–1915. [Google Scholar] [CrossRef]
  159. Williams, J.J.; Mayurasakorn, K.; Vannucci, S.J.; Mastropietro, C.; Bazan, N.G.; Ten, V.S.; Deckelbaum, R.J. N-3 fatty acid rich triglyceride emulsions are neuroprotective after cerebral hypoxic-ischemic injury in neonatal mice. PLoS ONE 2013, 8, e56233. [Google Scholar] [CrossRef] [PubMed]
  160. Pu, H.; Jiang, X.; Hu, X.; Xia, J.; Hong, D.; Zhang, W.; Gao, Y.; Chen, J.; Shi, Y. Delayed Docosahexaenoic Acid Treatment Combined with Dietary Supplementation of Omega-3 Fatty Acids Promotes Long-Term Neurovascular Restoration After Ischemic Stroke. Transl. Stroke Res. 2016, 7, 521–534. [Google Scholar] [CrossRef] [PubMed]
  161. Satapathy, M.K.; Yen, T.L.; Jan, J.S.; Tang, R.D.; Wang, J.Y.; Taliyan, R.; Yang, C.H. Solid Lipid Nanoparticles (SLNs): An Advanced Drug Delivery System Targeting Brain through BBB. Pharmaceutics 2021, 13, 1183. [Google Scholar] [CrossRef] [PubMed]
  162. Sabry, S.A.; Abd El Razek, A.M.; Nabil, M.; Khedr, S.M.; El-Nahas, H.M.; Eissa, N.G. Brain-targeted delivery of Valsartan using solid lipid nanoparticles labeled with Rhodamine B; a promising technique for mitigating the negative effects of stroke. Drug Deliv. 2023, 30, 2179127. [Google Scholar] [CrossRef]
  163. Nong, J.; Glassman, P.M.; Shuvaev, V.V.; Reyes-Esteves, S.; Descamps, H.C.; Kiseleva, R.Y.; Papp, T.E.; Alameh, M.G.; Tam, Y.K.; Mui, B.L.; et al. Targeting lipid nanoparticles to the blood-brain barrier to ameliorate acute ischemic stroke. Mol. Ther. 2024, 32, 1344–1358. [Google Scholar] [CrossRef]
  164. Erdő, F.; Bors, L.A.; Farkas, D.; Bajza, Á.; Gizurarson, S. Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res. Bull. 2018, 143, 155–170. [Google Scholar] [CrossRef]
  165. Battaglia, L.; Panciani, P.P.; Muntoni, E.; Capucchio, M.T.; Biasibetti, E.; De Bonis, P.; Mioletti, S.; Fontanella, M.; Swaminathan, S. Lipid nanoparticles for intranasal administration: Application to nose-to-brain delivery. Expert Opin. Drug Deliv. 2018, 15, 369–378. [Google Scholar] [CrossRef]
  166. Passeri, E.; Elkhoury, K.; Jiménez Garavito, M.C.; Desor, F.; Huguet, M.; Soligot-Hognon, C.; Linder, M.; Malaplate, C.; Yen, F.T.; Arab-Tehrany, E. Use of Active Salmon-Lecithin Nanoliposomes to Increase Polyunsaturated Fatty Acid Bioavailability in Cortical Neurons and Mice. Int. J. Mol. Sci. 2021, 22, 11859. [Google Scholar] [CrossRef]
  167. Passeri, E.; Bun, P.; Elkhoury, K.; Linder, M.; Malaplate, C.; Yen, F.T.; Arab-Tehrany, E. Transfer Phenomena of Nanoliposomes by Live Imaging of Primary Cultures of Cortical Neurons. Pharmaceutics 2022, 14, 2172. [Google Scholar] [CrossRef]
  168. Andone, S.; Farczádi, L.; Imre, S.; Bălașa, R. Fatty Acids and Lipid Paradox-Neuroprotective Biomarkers in Ischemic Stroke. Int. J. Mol. Sci. 2022, 23, 10810. [Google Scholar] [CrossRef]
  169. Kotlęga, D.; Peda, B.; Palma, J.; Zembroń-Łacny, A.; Gołąb-Janowska, M.; Masztalewicz, M.; Nowacki, P.; Szczuko, M. Free Fatty Acids Are Associated with the Cognitive Functions in Stroke Survivors. Int. J. Environ. Res. Public Health 2021, 18, 6500. [Google Scholar] [CrossRef]
  170. Kurth, T.; Everett, B.M.; Buring, J.E.; Kase, C.S.; Ridker, P.M.; Gaziano, J.M. Lipid levels and the risk of ischemic stroke in women. Neurology 2007, 68, 556–562. [Google Scholar] [CrossRef]
  171. Glasser, S.P.; Mosher, A.; Howard, G.; Banach, M. What is the association of lipid levels and incident stroke? Int. J. Cardiol. 2016, 220, 890–894. [Google Scholar] [CrossRef]
  172. Gu, X.; Li, Y.; Chen, S.; Yang, X.; Liu, F.; Li, Y.; Li, J.; Cao, J.; Liu, X.; Chen, J.; et al. Association of Lipids with Ischemic and Hemorrhagic Stroke: A Prospective Cohort Study Among 267 500 Chinese. Stroke 2019, 50, 3376–3384. [Google Scholar] [CrossRef]
  173. Shahar, E.; Chambless, L.E.; Rosamond, W.D.; Boland, L.L.; Ballantyne, C.M.; McGovern, P.G.; Sharrett, A.R. Plasma lipid profile and incident ischemic stroke: The atherosclerosis risk in communities (ARIC) study. Stroke 2003, 34, 623–631. [Google Scholar] [CrossRef]
  174. Johannesen, C.D.L.; Mortensen, M.B.; Langsted, A.; Nordestgaard, B.G. ApoB and Non-HDL Cholesterol Versus LDL Cholesterol for Ischemic Stroke Risk. Ann. Neurol. 2022, 92, 379–389. [Google Scholar] [CrossRef] [PubMed]
  175. Çoban, E.K. Can TG/HDL Ratio be an Accurate Predictor in the Determination of the Risk of Cerebrovascular Events in Youngsters? Sisli Etfal Hastan Tip Bul. 2018, 52, 201–205. [Google Scholar] [CrossRef] [PubMed]
  176. Nam, K.W.; Kwon, H.M.; Jeong, H.Y.; Park, J.H.; Kwon, H.; Jeong, S.M. High triglyceride/HDL cholesterol ratio is associated with silent brain infarcts in a healthy population. BMC Neurol. 2019, 19, 147. [Google Scholar] [CrossRef]
  177. Jové, M.; Mauri-Capdevila, G.; Suárez, I.; Cambray, S.; Sanahuja, J.; Quílez, A.; Farré, J.; Benabdelhak, I.; Pamplona, R.; Portero-Otín, M.; et al. Metabolomics predicts stroke recurrence after transient ischemic attack. Neurology 2015, 84, 36–45. [Google Scholar] [CrossRef] [PubMed]
  178. Huang, M.; Xu, S.; Zhou, M.; Luo, J.; Zha, F.; Shan, L.; Yang, Q.; Zhou, B.; Wang, Y. Lysophosphatidylcholines and phosphatidylcholines as biomarkers for stroke recovery. Front. Neurol. 2022, 13, 1047101. [Google Scholar] [CrossRef]
  179. Jensen, M.; Liu, S.; Stepula, E.; Martella, D.; Birjandi, A.A.; Farrell-Dillon, K.; Chan, K.L.A.; Parsons, M.; Chiappini, C.; Chapple, S.J.; et al. Opto-Lipidomics of Tissues. Adv. Sci. 2024, 11, e2302962. [Google Scholar] [CrossRef]
Figure 1. Metabolism of omega-3 and omega-6 PUFAs.
Figure 1. Metabolism of omega-3 and omega-6 PUFAs.
Antioxidants 13 00634 g001
Figure 2. Schematic illustration of the major oxidative mechanisms in ischemic stroke leading to lipid mediators formed by enzymatic or non-enzymatic pathways and playing important roles in various biological processes.
Figure 2. Schematic illustration of the major oxidative mechanisms in ischemic stroke leading to lipid mediators formed by enzymatic or non-enzymatic pathways and playing important roles in various biological processes.
Antioxidants 13 00634 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bernoud-Hubac, N.; Lo Van, A.; Lazar, A.-N.; Lagarde, M. Ischemic Brain Injury: Involvement of Lipids in the Pathophysiology of Stroke and Therapeutic Strategies. Antioxidants 2024, 13, 634. https://doi.org/10.3390/antiox13060634

AMA Style

Bernoud-Hubac N, Lo Van A, Lazar A-N, Lagarde M. Ischemic Brain Injury: Involvement of Lipids in the Pathophysiology of Stroke and Therapeutic Strategies. Antioxidants. 2024; 13(6):634. https://doi.org/10.3390/antiox13060634

Chicago/Turabian Style

Bernoud-Hubac, Nathalie, Amanda Lo Van, Adina-Nicoleta Lazar, and Michel Lagarde. 2024. "Ischemic Brain Injury: Involvement of Lipids in the Pathophysiology of Stroke and Therapeutic Strategies" Antioxidants 13, no. 6: 634. https://doi.org/10.3390/antiox13060634

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

Article Metrics

Back to TopTop