Next Article in Journal
Nicking Activity of M13 Bacteriophage Protein 2
Previous Article in Journal
Analysis of the Transcriptome Provides Insights into the Photosynthate of Maize Response to Salt Stress by 5-Aminolevulinic Acid
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Protection Strategies Against Palmitic Acid-Induced Lipotoxicity in Metabolic Syndrome and Related Diseases

by
Zeltzin Alejandra Ceja-Galicia
1,
Carlos Leonardo Armando Cespedes-Acuña
2 and
Mohammed El-Hafidi
1,*
1
Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
2
Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Avenida Andres Bello 720, Chillan 3780708, Chile
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(2), 788; https://doi.org/10.3390/ijms26020788
Submission received: 26 November 2024 / Revised: 26 December 2024 / Accepted: 3 January 2025 / Published: 18 January 2025
(This article belongs to the Section Molecular Endocrinology and Metabolism)

Abstract

:
Diets rich in carbohydrate and saturated fat contents, when combined with a sedentary lifestyle, contribute to the development of obesity and metabolic syndrome (MetS), which subsequently increase palmitic acid (PA) levels. At high concentrations, PA induces lipotoxicity through several mechanisms involving endoplasmic reticulum (ER) stress, mitochondrial dysfunction, inflammation and cell death. Nevertheless, there are endogenous strategies to mitigate PA-induced lipotoxicity through its unsaturation and elongation and its channeling and storage in lipid droplets (LDs), which plays a crucial role in sequestering oxidized lipids, thereby reducing oxidative damage to lipid membranes. While extended exposure to PA promotes mitochondrial reactive oxygen species (ROS) generation leading to cell damage, acute exposure of ß-cells to PA increases glucose-stimulated insulin secretion (GSIS), through the activation of free fatty acid receptors (FFARs). Subsequently, the activation of FFARs by exogenous agonists has been suggested as a potential therapeutic strategy to prevent PA-induced lipotoxicity in ß cells. Moreover, some saturated fatty acids, including oleic acid, can counteract the negative impact of PA on cellular health, suggesting a complex interaction between different dietary fats and cellular outcomes. Therefore, the challenge is to prevent the lipid peroxidation of dietary unsaturated fatty acids through the utilization of natural antioxidants. This complexity indicates the necessity for further research into the function of palmitic acid in diverse pathological conditions and to find the main therapeutic target against its lipotoxicity. The aim of this review is, therefore, to examine recent data regarding the mechanism underlying PA-induced lipotoxicity in order to identify strategies that can promote protection mechanisms against lipotoxicity, dysfunction and apoptosis in MetS and obesity.

1. Introduction

Dietary habits characterized by a high intake of carbohydrates and saturated fats, in combination with a sedentary lifestyle, related to the incidence of metabolic syndrome (MetS) and obesity, are risk factors for cardiovascular diseases and type II diabetes. The high-carbohydrate diet exhibits a stronger association with the accumulation of adipose tissue compared with a high-fat diet; furthermore, the restriction of dietary carbohydrates is related to a significant reduction in body weight compared to a low-fat diet in overweight and MetS patients [1]. Moreover, the reduction in carbohydrate uptake decreases blood pressure and triglyceride (TG) concentrations and increases high-density lipoprotein (HDL) levels in overweight individuals with MetS [2,3]. Thus, a low-carbohydrate diet reduces the prevalence of MetS and offers several potential benefits for the treatment of obesity and type II diabetes in many people, even if they do not lose weight [4,5,6]. The high-carbohydrate diet is well known to enhance de novo lipogenesis and increases the biosynthesis of palmitic acid (16:0, PA), the major saturated fatty acid, corresponding to 20 to 30% of total fatty acids in the human body. PA metabolism is tightly regulated with the endogenous synthesis via de novo lipogenesis (DNL) and dietary intake [7]. Palmitic acid serves as a major substrate for myocardial metabolism, influencing energy production. However, depending on the concentration and exposure duration, PA can potentially affect energy production and cardiovascular tissue health. Therefore, the balance between low and excessive exposure to palmitic acid plays a critical role in cell health and death due to lipotoxicity [8,9]. Moderate PA levels support cardiac health, whereas excessive concentrations can trigger detrimental metabolic pathways, highlighting the need for a balanced dietary intake to maintain cardiovascular lipid homeostasis. PA at high concentrations promotes lipotoxicity that results in cell dysfunction and death in many cell types, including skeletal muscle cells, liver cells, β-cells and cardiomyocytes [10]. PA induces lipotoxicity through several mechanisms involving mitochondria dysfunction, endoplasmic reticulum (ER) stress, oxidative stress, inflammation and cell death through apoptosis [11]. In pathophysiological conditions of obesity, insulin resistance and MetS to store PA in lipid droplets (LDs) as triglycerides in adipose and non-adipose tissues such as the liver, skeletal muscle and heart is considered as an endogenous protection strategy to protect cells from excessive PA. The utility of LDs in providing energy for metabolic processes and membrane biosynthesis has been extensively characterized in adipocytes, where triglyceride hydrolysis enzymes such as adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) activities depend on feeding, starvation and hormonal status [12,13]. In obesity and MetS, a chronic increase in triglyceride biosynthesis and accumulation in LDs within both adipose and non-adipose tissues results in enhanced FFA release that promotes oxidative stress and endoplasmic reticulum stress, thereby increasing inflammation, insulin resistance, diabetes and cardiovascular diseases [14].
To attenuate the lipotoxic effect of PA, some strategies have been addressed. As saturated and monounsaturated fatty acids differ significantly in their contributions to lipotoxicity, it appears that monounsaturated, such as palmitoleic and oleic, acids have been found to protect against PA-induced lipotoxicity in cell culture and rodent models by enhancing whole body insulin sensitivity, stimulating insulin secretion by β-cells, increasing hepatic fatty acid oxidation and improving the blood lipid profile [15,16]. Furthermore, oleic acid has been found to protect against PA-induced lipotoxicity, by channeling the PA and oxidized PUFA into LDs as a TG within the cytosol of hepatocytes, skeletal muscle and pancreatic β-cells in an obesity rat model [17,18]. Nevertheless, the oxidative stress linked to PA-induced lipotoxicity and the exposure of food lipids to high temperatures and oxygen that harm human health suggest the need for antioxidant protection strategies against PA-induced lipotoxicity. Therefore, this review aims to discuss the recent findings concerning the mechanisms involved in PA-induced lipotoxicity, as well as to examine various endogenous and exogenous interventions aimed to protect cells from mitochondrial dysfunction, oxidative stress, ER stress and cell death and to protect lipid-rich foods from oxidation and subsequent health degradation.

2. Palmitic Acid Biosynthesis in Metabolic Syndrome and Obesity

PA accounts for approximately 20 to 30% of total fatty acids in human and animal bodies [7,19]. PA is synthetized endogenously from the end-products of glycolysis (pyruvate from amino acids and from other fatty acids). As illustrated in Figure 1, PA biosynthesis starts at the glycolysis end-product via the transformation of pyruvate to citrate, subsequently undergoing a conversion into acetyl-CoA. This acetyl-CoA is converted by acetyl-CoA carboxylase (ACC) to malonyl-CoA, which serves as a substrate for fatty acid synthase (FAS) that catalyzes the biosynthesis of PA. Therefore, a high-carbohydrate diet not only induces hyperinsulinemia but also increases de novo lipogenesis, contributing to increased levels of PA and triglycerides in adipose and non-adipose tissues [20]. Furthermore, PA as a free fatty acid is also enhanced by high lipolytic activity in adipose tissue due to insulin resistance.
Adipose triglycerides lipases, hormone-sensitive lipases, are among the key enzymes in the hydrolysis of triacylglycerol, diacylglycerol and monoacylglycerol, producing FFAs and glycerol. FFA release and mobilization from adipose tissue to peripheral tissue are regulated by insulin levels, fasting states, feeding conditions and hormonal status [21]. Nevertheless, when the release of PA via lipolysis activity exceeds its subsequent beta-oxidation, its accumulation leads to several metabolic alterations, as well as cell dysfunction and death through apoptosis and necrosis [22]. The disruption of the homeostatic balance related to the uncontrolled endogenous biosynthesis of PA involves different physiopathological conditions such as MetS, obesity and related metabolic diseases. In the liver, the stimulation of de novo lipogenesis through hyperinsulinemia involves the transcription factor sterol regulatory element-binding proteins-1c (SREBP-1c), which up-regulates the enzymes that catalyze lipogenesis [23]. Lipogenesis is also stimulated by glucose through the activation of the transcription factor of carbohydrate regulatory element-binding proteins (ChREBP) [24]. Both SREBP-1c and ChREBP up-regulate different genes involved in fatty acid biosynthesis, including desaturation and elongation enzymes, as shown in Figure 1 [25,26]. However, under the conditions of a high sucrose intake, the accumulation of PA is partially prevented by its enhanced activation to palmitoyl-CoA by acyl-CoA synthetase and its unsaturation to palmitoleyl-CoA (16:1n−7) by Δ-9 desaturase. Palmitoyl-CoA can also be elongated to stearoyl-CoA (C18:0) by fatty acid-like family member 6 (ELOVL6) elongase, and it is further unsaturated to oleyl-CoA (18:1n−9) by stearoyl-CoA desaturase and, finally, incorporation into triglycerides by Acyl-CoA:diacylglycerol O-acyltransferase (DGAT), which catalyzes the final step in TG synthesis, converting diacylglycerol into triglycerides, which, in turn, are stored in lipid droplets [11,27]. During the development of MetS and obesity due to sucrose feeding, the stearoyl-CoA desaturase activity positively correlates with palmitic and palmitoleic acid and plasma TG concentrations [28,29,30]. The desaturation of PA to palmitoleic acid is considered as endogenous protection against PA-induced lipotoxicity. Therefore, some studies have proposed that the substitution of palmitic acid by monounsaturated acid should be beneficial for diabetic and related cardiovascular diseases [31,32]. In addition, losing weight decreases the hepatic TG content by decreasing de novo lipogenesis in patients with non-alcoholic fatty liver disease (NAFLD) and increasing circulating glucose and insulin [33].
In addition to de novo lipogenesis, PA comes from the diet, where it is found in very large quantities compared to other fatty acids. In physiological conditions, the postprandial PA from food is quickly metabolized, incorporated in the cellular membranes and/or used for energy expenditure, making it an important nutrient that must be consumed [7]. The esterified PA in food triglycerides and phospholipids is absorbed in the small intestine, packing in chylomicrons and, subsequently, in lipoproteins from the liver, then being distributed to the organs through the blood stream [34]. In the capillary, the lipoprotein lipase (LPL) hydrolyzes the fatty acids from the glycerol to be absorbed from the cell and normally esterified in adipose tissue or oxidized in other organs. Moderated levels of PA in the cell promote beta-oxidation and ATP production, principally in the heart, skeletal muscle and kidney, maintaining mitochondrial function and cellular homeostasis [35]. On the other hand, a high-fat diet rich in saturated fatty acids such as PA is associated with insulin resistance in adipose tissue and enhanced lipolytic activity due to insulin resistance and an increase in non-esterified fatty acids, including PA.

3. Palmitic Acid-Induced Lipotoxicity

As previously elucidated, de novo PA is typically esterified in phospholipids or triglycerides to avoid its deleterious effect. However, when the PA availability exceeds its esterification capacity, the free form of PA can trigger mitochondrial dysfunction, endoplasmic reticulum (ER) stress, oxidative stress and inflammation, which are involved in the lipotoxicity induced by PA and contribute to cell death through apoptosis. However, mitochondria ROS generation and ER stress are the major participating mechanisms and can be attenuated by several antioxidant treatments using cell culture and experimental animals of obesity and MetS.

3.1. Palmitic Acid-Induced Endoplasmic Reticulum (ER) Stress

Palmitate-induced cardiomyocyte dysfunction and β-cell death in type II diabetes involve the participation of ER stress as the primary site of oxidative protein folding catalysis mediated by oxidoreductin-1α and ERO-1β isoenzymes through an over-production of hydrogen peroxide (H2O2) [36]. ER stress induced by PA is associated with the unfolded protein response (UPR) as a compensatory mechanism mediated through the up-regulation of activating transcription factor 4 (ATF4), inositol-requiring enzyme 1 (IRE1) and C/EBP homologous protein (CHOP), involved in the proapoptotic signaling pathway to mitigate dysfunctional cells [37]. In the myocardium of patients with type II diabetes, the levels of ER stress markers such as ATF4 and CHOP are found to be elevated due to metabolic disturbances of PA overload [38]. Among the mechanisms proposed to explain the induction of ER stress and loss of β-cell viability in response to PA treatment, the protein palmitoylation is a post-translational modification that regulates protein localization, stability and activity. Indeed, PA-induced ER stress in β-cells has been described to be mediated by excessive protein palmitoylation [39]. In MetS and obesity, the high availability of PA and its activated form palmitoyl-CoA serves as the S-palmitoylation protein agent. The S-palmitoylation of protein is a reversible reaction catalyzed by palmitoyl protein acyl transferase (PPT) and acyl protein thioesterase (APT), which depend on PA availability. Some proteins involved in the lipid metabolism such as CD36, a fatty acid transporter, are activated via palmitoylation to enhance fatty acid accumulation within cells (for a review, see [40]). The palmitate-induced protein palmitoylation has been described for the serine palmitoyl transferase 2 (SPTLC2), the rate-limiting enzyme of de novo ceramide synthesis. SPTLC2 catalyzes the condensation of serine and palmitoyl-CoA, forming 3-ketosphinganine, which is, thereafter, transformed into ceramide in hepatocytes exposed to palmitic acid [41]. Furthermore, ER stress is related to the increased total ceramide content and, specifically, due to the increased ceramide C14:0 and ceramide C16:0 [41]. In muscles from mice fed a high-fat diet, ceramide accumulation is linked to insulin resistance [42], impacts liver homeostasis and induces metabolic disorders [43]. Additionally, inhibiting desaturase SCD or SREBF1/2 leads to increased dihydroceramide levels, indicating their involvement in ceramide biosynthesis [44]. In primary mouse hepatocytes exposed to PA, cell death via apoptosis can be prevented using myriocin as an inhibitor of ceramide synthesis or by taurourso-deoxy-cholic to prevent ER stress in hepatocytes [44]. The inhibition of ceramide synthase effectively mitigates PA-induced β-cell ferroptosis [45]. Similarly, PA contributes to apoptosis through de novo ceramide synthesis in various cell types via oxidative stress mechanisms induced by the activation of the TLR4-ROS-p53 pathway [46] and the ERK1/2 signaling pathway, leading to increased levels of Bax, as well as a decrease in Bcl-2, which contributes to the apoptosis of H9c2 cells [47].
In addition, ceramides play a significant role in inducing apoptosis through other mechanisms, involving mitochondrial pathways and redox signaling. Ceramides bind to voltage-dependent anion channels (VDACs) 1 and 2 in mitochondria, which is crucial for triggering apoptotic pathways. The loss of VDAC2 significantly reduces ceramide-induced apoptosis [48]. In relation to redox signaling, ceramides enhance the expression of thioredoxin-interacting protein (Txnip), concomitantly reducing thioredoxin 1 (Trx1) activity, which subsequently leads to increased apoptosis [49]. The altered redox signaling induced by ceramide is also linked to a shift in the balance of superoxide dismutases (SOD) 1 and 2 [50]. Furthermore, the activation of ER stress by PA induces the release of calcium from the ER and results in elevated cytosolic calcium concentrations, thereby perturbing calcium homeostasis and facilitating the accumulation of reactive oxygen species (ROS), which, in turn, promotes the occurrence of ferroptotic cell death [51]. Thus, the disruption of calcium homeostasis through excessive Ca2+ release in the ER and the resulting mitochondrial Ca2+ overload increase oxidative stress and lipid droplet accumulation, which are essential in the progression of insulin resistance and other diabetic complications [52].

3.2. Palmitic Acid-Induced Mitochondrial Dysfunction

Palmitic acid-induced mitochondria dysfunction is mediated by several mechanisms, involving altered fatty acid beta-oxidation, ROS generation and the collapse of the mitochondrial membrane potential (MMP). All these mechanisms involve cytochrome c release, with caspase-3 activation being a key enzyme in the cell death process via apoptosis [53].
In cardiomyocytes, PA at high concentrations has been described to impair mitochondria energy metabolism by inhibiting fatty acid beta-oxidation, resulting in lipid accumulation and decreased ATP biosynthesis [54]. In addition, PA-increased ROS generation and -decreased ATP biosynthesis induce alterations in the expression of peroxisome proliferator-activated receptor (PPAR) α, δ and γ, PGC1α and UCP2 involved in fatty acid metabolism [54]. Thus, PPARs play a significant regulatory role in the mitochondrial biogenesis signaling pathway and fatty acid metabolism in diabetic cardiomyopathy. In response to PA treatment, the expression levels of PPARα and PPARγ were found to be decreased, while PGC-1α and UCP2 levels were found to be increased, suggesting complex interaction between lipid accumulation, ROS production and PPAR signaling [54].
Palmitic acid also down-regulates the expression of fusion genes (MFN1) and up-regulates the fission genes (FIS1), disrupting the balance between fusion and fission, leading to apoptosis and impaired oxidative metabolism [55]. Mitofusin2 (Mfn2), the inducer of the mitochondria fusion, directly interacts with carnitine palmitoyl transferase (Cpt1α), a fatty acid transporter, into mitochondria, via its GTPase domains, which seems to be essential for maintaining the activity of Cpt1α and promoting mitochondrial fatty acid beta-oxidation [56].
In macrophages, palmitic acid impairs the mitochondrial function by decreasing respiratory complex IV and succinate dehydrogenase activities and up-regulating the adipocyte fatty acid-binding protein (FABP4), which compromise fatty acid oxidation and increase ROS generation [57]. In addition, palmitic acid negatively impacts aconitase and isocitrate dehydrogenase (IDH) activities [58]. In summary, PA and its biologically active derivative, palmitoyl-CA, engage multiple biochemical pathways that result in elevated ROS production via the inhibition of mitochondrial complexes I and III and the stimulation of peroxisomal activity and protein kinase C (PKC) signaling pathways. Collectively, these mechanisms lead to increased ROS production in hepatic mitochondria and macrophages, which is associated with increased expression of proinflammatory cytokines, thereby intensifying oxidative stress [59].
In an H9C2 cell model of lipotoxicity induced by high levels of PA exposure, the externalization of mtDNA in the cytosol is a result of the PA-induced over-production of mitochondrial ROS, promoting diabetic cardiomyopathy and cell death via apoptosis [60,61]. Indeed, PA also enhances caspase-3, leading to programed cell death and DNA fragmentation, and alters cell signaling in HepG2 cells under glucolipotoxic conditions [53,62]. PA-induced apoptosis is also accompanied by autophagy that, when blocked, exacerbates cell damage [63].
In a model of metabolic syndrome and obesity induced by high-sucrose consumption, a change in membrane phospholipids, especially cardiolipin (CL), exclusive of the mitochondrial inner membrane (MIM), was observed [64]. Its four-chain polyunsaturated fatty acid and conical structure enable it to interact with several proteins, enhancing the communication between them and regulating mitochondrial dynamics and homeostasis. For example, CL concentrates near the membrane contact sites of proteins such as OPA1, which are essential for membrane fusion and remodeling [65]. The high availability of PA [64] promotes its incorporation into CL molecules, and the enrichment of CL with PA modifies its tridimensional structure and, therefore, mitochondrial function. This modification contributes to the pathogenesis of metabolic and cardiovascular disorders via various molecular mechanisms, mainly those associated with lipid metabolism, mitochondrial architecture and respiratory efficiency [66]. These may also be related to decreased mitochondrial membrane fluidity and the stability of respiratory complexes [67].
Under conditions of lipid saturation stress, CL is crucial for maintaining the ultrastructure of the MIM, as shown in yeast models [68]. It has been reported that PA, when incorporated into CL molecules, alters the composition and potentially affects mitochondrial function and cell proliferation through the activation of the NOD-like receptor (NLR) signaling pathway [69,70]. Prolonged exposure of mitochondria to PA increases the production of ROS by inducing oxidative stress and damaging cardiomyocytes [8,71].
On the other hand, high levels of PA-induced oxidative stress within cardiomyocytes modify lipid metabolism, adversely influencing CL composition [8,9] and impacting mitochondrial biogenesis and dynamics [9]. Indeed, unsaturated fatty acids in cardiolipin are particularly susceptible to oxidative damage, leading to altered fatty acyl profiles [72]. Altered cardiolipin composition due to oxidative stress is linked to increased apoptosis in cardiomyocytes [71]. However, oxidative stress triggers remodeling processes that replace cardiolipin oxidized fatty acids with non-oxidized polyunsaturated fatty acids, further compromising mitochondrial function [72].
The loss of CL remodeling enzymes results in altered structural integrity of the MIM, such as the accumulation of monolyso-cardiolipin, interacting with membrane-associated proteins and altering the activity of these proteins, thus affecting mitochondrial dynamics [65]. CL remodeling is essential for adapting to different lipid environments and is crucial for mitochondrial health [68]. Unlike palmitic acid, which exerts adverse effects on cardiolipin composition and overall cellular vitality, specific fatty acids, including oleic acid, have the ability to prevent these negative consequences. This finding suggests a complex interplay between different dietary lipids and their cellular implications [69]. Thus, CL remodeling could be induced by the presence of a high PA concentration or because of an oxidative process to protect MIM against ROS. Such complexity points to the need for further research into the role of cardiolipin in various physiological and pathological contexts.

3.3. Palmitic Acid-Induced Inflammation

PA is well known to induce inflammatory responses, with disturbances in glucose metabolism and insulin resistance [73]. PA induces ROS inflammation via two main pathways: the production of superoxide anion during its metabolism and its direct accumulation in cells as diacylglycerols (DAG). This leads to the activation of protein kinase C, which phosphorylates and activates IKK, subsequently enhancing NF-kB activation [74]. This is the most approved theory; PA induces an indirect activation of NF-κB through the increased ER stress and mitochondria ROS production. Nevertheless, PA could directly participate in the inflammatory process. It has been hypothesized that, in macrophages, PA could induce this process though direct interaction with Troll-like receptors (TRL), specifically 4, and the differentiation cluster 36 (CD36) complex or with the interaction of the free fatty acid receptor (FFAR1/GPR40), enhancing the NF-κB inflammatory response through the PI3K/PKB/AKT or PLC/PKC/MAPK signaling pathways [73].
Also, it has been described that, in addition to TRL4, TLR6 and 2 could participate in the PA activation of the inflammasome NLPR3 and interleukin 1ß via NF-κB and through the activation of the PI3K/AKT/mTOR signaling pathway [75,76]. On the other hand, the PA activation of TRL4 also induces resistin/insulin resistance in human neuroblastoma through the formation of the TRL4/MYD38/TRIAP complex [77]. The interaction between PA, the TRL family and the direct relationship of PA-induced lipotoxicity with inflammation and insulin resistance have been demonstrated. In addition, it has been described that PA decreases the anti-inhibitor of nuclear factor κB (IκBα), interrupting the interaction and cytoplasmic stabilization of NF-κB, and, thus, allowing for its translocation to the nucleus in primary human myotubes [78]. Thus, a high concentration of PA in obesity and metabolic syndrome could activate, directly and indirectly, the inflammatory process through different signaling pathways, exacerbating lipotoxicity and breaking cell homeostasis to develop insulin resistance.
On the contrary, it has been mentioned that PA could play a role in the anti-inflammatory process and glucose metabolism disorders through free fatty acid receptors FFAR1 or FFAR4/F-κB/KLF7 signaling pathway in epididymal white adipose tissue and livers from mice fed a high-fat diet [19]. This approximation is related to PA-induced long noncoding RNAs (PARAIL)/ELAV like RNA-binding protein 1 (ELAV1) interaction in macrophages, where the PARAIL down-regulation of cytosolic ELVA1 decreases the activity on NF-κB and the synthesis of proinflammatory cytokines [79]. However, PARAIL could be a protective cell mechanism in response to high concentrations of PA. These results could support the idea that PA-induced inflammation is not only related to its concentration but that the long duration of exposure to a high concentration plays a crucial role in this respect.

4. PA-Induced Lipotoxicity and Free Fatty Acid Receptor Intervention

Free fatty acid receptors (FFARs) are G protein-coupled receptors (GPRs), such as GPR40, GPR41, GPR43, GPR119 and GPR120, are expressed in almost all tissues and are activated by a wide variety of ligands, such as hormones, neurotransmitters, peptides, proteins, steroids and FFAs, mediating their effects through the intracellular signaling cascade [80]. In particular, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain saturated and unsaturated fatty acids, whereas FFAR3 (GPR41) and FFAR2 (GPR43) are activated by short-chain fatty acids (SCFAs), mainly acetate, butyrate, and propionate [81]. Among the five members of the FFAR family, FFAR1, also known as GPR40, has been the most extensively studied in the β-cell. FFAR1, predominantly expressed in pancreatic β-cells, structurally possesses seven transmembrane domains and is activated by various medium- and long-chain FFAs (C12–C22), triggering a signaling cascade, which leads to increased intracellular calcium levels and potentiates glucose-stimulated insulin secretion (GSIS), managing blood glucose in diabetic patients [82]. The mechanism by which FFAs acutely potentiate GSIS is also through FFAR1 and Gαq signaling, activating phospholipase C (PLC) that, in turn, hydrolyzes phosphatidylinositol-4,5-bisphosphate (PIP2) into inositol trisphosphate (IP3) [83]. The inositol trisphosphate targets its receptors at the ER membrane, promoting calcium release from ER stores, which results in the further elevation of cytosolic calcium, primarily initiated by glucose stimulation [83]. The DAG resulting from PIP2 hydrolysis directly participates in the potentiation of GSIS by targeting PKC and modulating insulin granule exocytosis [84,85]. Indeed, PLC inhibition and FFAR1 RNA interference block intracellular calcium mobilization and insulin secretion in islets from rosiglitazone-treated OLETF rats with an over-expression of FFAR1 [86]. In physiological conditions, PA-treated cells enhance mitochondrial respiration via the combined action of the intracellular metabolism of the fatty acid and Gαq-coupled FFAR1 signaling, suggesting a synergic action of the two pathways on insulin secretion [87].
On the other hand, the prolonged exposure of cells to PA disrupts this intracellular signaling, impairing beta-cell function and GSIS [88]. In type II diabetes, increased levels of PA induce the loss of the β-cell function due to the involvement of FFAR1 in crosstalk with mTOR-Akt and IRS-1, altering insulin signaling [89,90,91]. In hepatocytes, mTOR signaling is implicated in the cellular response to lipotoxicity by enhancing triglyceride secretion and reducing cell survival [92]. The selective inhibition of mTOR promotes fatty acid catabolism and suppresses lipogenesis, thereby protecting against non-alcoholic fatty liver in mice [93] and managing metabolic disorder related to obesity [94]. In addition, an excess of PA as an FFA may increase ROS generation related to lipotoxicity and alters the pathways of GSIS through FFAR1 [95]. Indeed, palmitic acid has been found to promote the generation of superoxide anion via the activation of NADPH oxidase and mitochondria, which correlates with a reduction in FFAR1-mediated signal transduction pathways, inhibiting insulin secretion, even when glucose concentrations are elevated [96]. Thus, these works were addressed to elucidate the signaling pathways by which FFARs mediate the effects of fatty acids on insulin secretion and lipotoxicity, which may lead to new therapeutic strategies for managing insulin resistance and related metabolic disorders.
Several strategies have been considered to improve FFAR signal transduction via the up-regulation of their expression and the stimulation of their activities. FFARs can be activated by natural agonists, such as oleic acid, linoleic acid and do-cosahexaenoic acid, which can be considered safe. In the case of synthetic agonists, which are considered much more effective than natural ones, only TAK-875, and not GW9508 or NCG75, which have been used in research involving cells in culture and experimental animals, has shown efficacy with no side effects [97]. TAK-875, a selective FFAR1 agonist, has been found to increase insulin secretion and improve glucose tolerance without causing hypoglycemia and other serious side effects in patients with type II diabetes for the first time, making it a potential new treatment for type II diabetes, as reported by Leifke et al. and Araki et al. [97,98].
In a recent study by Dragano et al. [99], it was described that FFAR1 activation increases energy expenditure and promotes the browning of subcutaneous white adipose tissue, thus contributing to weight loss. However, significant differences in the effects of FFAR1 activation can be found between obese and non-obese individuals, particularly in terms of energy balance and metabolic responses. In obese individuals, FFAR1 activation leads to increased energy expenditure and enhanced thermogenesis in brown adipose tissue, with a reduction in inflammation and endoplasmic reticulum stress in the hypothalamus [99]. In contrast, non-obese individuals may not benefit from these pronounced metabolic advantages, as their basic energy regulation mechanisms are different. In rat proximal tubular cells (NRK52E), the stimulation of FFAR1 by specific agonists mitigates the superoxide production and cytotoxic effects induced by PA in a dependent manner on Nrf2/HO-1 [100,101]. In β-cells, the up-regulation of the expression of FFAR1 at the mRNA through pioglitazone treatment, an insulin sensitizer, enhances the antioxidative stress protein levels and reduces PA-induced lipoapoptosis [102]. Unlike saturated FFAs, long-chain unsaturated FFA-induced FFAR1 over-expression protects rat pancreatic β-cells (INS-1) against lipotoxicity [103]. The FFAR1 overexpression induced by unsaturated FFAs is more related to the acute protection of FFA-activated FFAR1 than that of FFAs, mediating chronic β-cell lipotoxicity [104]. Furthermore, FFAR4, which is also stimulated by long-chain fatty acids, plays a crucial role in mediating a protective response against lipotoxicity; this effect is completely diminished in cells where FFAR4 is inactivated and in islets isolated from FFAR4 knockout mice that exhibit glucose intolerance and insulin resistance [105]. This observation implies that FFAR4 activation could potentially serve as a preventive and therapeutic target for the management of obesity and diabetes [105].
Hence, future research should address and elucidate the signaling pathways by which FFARs mediate the effects of fatty acids on insulin secretion and lipotoxicity, which could lead to new therapeutic strategies for managing insulin resistance and related metabolic disorders.

5. Palmitic Acid Reduced Insulin Sensitivity and Increased Vascular Dysfunction

In obesity and MetS, the excessive accumulation of PA enhances metabolic complications, leading to insulin resistance (IR) and type II diabetes [106,107]. Among the mechanisms of PA, reducing insulin sensitivity in several cell types, the influence of PA on the plasma membrane lipid organization may influence the amount and affinity of insulin receptors to insulin and the modulation of its phosphorylation [108,109,110]. In skeletal muscle cells, PA reduces glucose uptake by enhancing the serine phosphorylation of insulin receptor substrate-1 (IRS-1) and decreasing insulin-mediated Akt activation and glucose transporter 4 (GLUT4) translocation [111]. In pancreatic cells, PA affects GSIS through the phosphorylation of both IRS-1 in the serine residue S636/639 and Akt in S473 [89]. Moreover, PA decreases the expression of the insulin receptor 1ß (IR1ß) [89]. The high content of PA in the membrane of erythrocyte and hepatocytes makes it more rigid, with a decrease in the affinity of insulin to its receptor that results in decreased insulin sensitivity and hyperinsulinemia [108,112]. The insulin and glucose impairment induces type II diabetes development. According to this, reviews have shown that high-carbohydrate and high-fat diets contribute to lipotoxicity in pancreatic ß-cells, ultimately leading to disfunction attributable to oxidative stress and ER stress [113]. Also, the deficiency of depalmitoylation enzymes or increase in the palmitoylation process of diverse proteins activates insulin hypersecretion that causes stress to ß-cells, progressing to insulin deficiency [114]. The lipotoxicity process in diabetes can lead to diabetic cardiomyopathy through PA-induced ROS production and mitochondrial DNA release that activates the expression of proinflammatory factors [57].
PA is implicated in the development of cardiovascular diseases through several molecular mechanisms. These mechanisms primarily involve inflammatory responses, endothelial dysfunction and atherosclerotic plaque instability. In normal cardiomyocytes, palmitate alters calcium handling, reducing cytosolic Ca2+ transience and cell contraction, whereas in insulin-resistant cells, palmitate potentiates these parameters [115]. This dual effect is related to the increased mitochondrial production of ROS [115] and to the activation of the NADPH oxidase (NOX)/ROS signaling pathway, which decreases the phosphorylation of endothelial nitric oxide synthase (eNOS) at critical sites, impairing nitric oxide production in endothelial cells. These alterations are linked to increased oxidative stress and reduced vascular function. In addition, PA activates long-chain acyl-CoA synthetase-1, further promoting inflammation in endothelial cells via TLR pathways [116]. It binds to the TLR4 accessory protein MD2, leading to myocardial inflammatory injuries [117]. In type II diabetes, PA enhances atherosclerotic plaque vulnerability through macrophage delta-like ligand 4 (Dll4) signaling, which induces vascular smooth muscle cell senescence and reduces collagen synthesis and plaque stability [118]. In addition, high levels of PA impair the bioavailability of endothelial progenitor cells, crucial for vascular repair, via a PPARγ-mediated mechanism, which may exacerbate cardiovascular disease [119]. Therefore, PA at high concentrations influences the structural integrity of phospholipid membranes, thereby modulating insulin sensitivity and inducing cellular starvation. This produces changes in the cell homeostasis, making them vulnerable to oxidative stress and the inflammatory process, resulting in tissue injury and the development of diabetes or cardiovascular disease.
Although the lipotoxic effects of a high concentration of PA are well documented, short-term exposure to low-dose palmitic acid has been shown to improve mitochondrial function and oxidative status in cardiomyoblasts, suggesting that controlled exposure may enhance cellular resistance to mitochondrial dysfunction, oxidative stress, inflammation and insulin resistance [8]. Thus, the possibility that low doses of PA confer protective benefits suggests the need to explore dose–response relationships further in clinical settings. This duality could inspire dietetic recommendations and personalized therapeutic strategies.

6. Palmitic Acid-Induced Lipotoxicity Is Attenuated by Natural Antioxidants

The impact of mitochondria-targeted antioxidant MitoQ on the cardiac damage induced by obesity suggests crosstalk between PA-induced lipotoxicity and mitochondrial ROS generation [120]. In an in vitro model of liver steatosis induced by palmitic/oleic, chlorogenic and lipoic acid, the prosthetic groups of the pyruvate dehydrogenase complex and a potent antioxidant bioactive, respectively, reduced PA-induced lipotoxicity, improved mitochondrial membrane potential, and decreased ROS production and Bax expression, thereby reducing mitochondria-mediated caspase-dependent apoptosis in hepatocytes [121,122]. Between polyphenolic compounds, chlorogenic acid also attenuates PA-induced lipotoxicity through the activation of SIRT1 (silent information regulator1) in hepatocytes and suppresses the activation of the c-Jun NH2-teminal kinase (JNK) signaling pathway, inhibiting ER stress [123]. PA-induced ROS production, ER stress and death in liver cells were also prevented by luteolin and melatonin treatment, a known antioxidant, and anti-inflammatory compounds, through the up-regulation of the expression of antioxidant enzymes, i.e., heme oxygenase-1 and glutathione peroxidase [100,124].
Between coumarins, scopoletin, esculetin and umbelliferone are molecules with antioxidant and anti-inflammatory properties, which significantly reduce oxidative stress in PA-treated hepatocytes by decreasing mitochondrial superoxide production and increasing the expression of antioxidant genes, including Nrf2 and Gpx1, thus reducing ER stress by inhibiting the phosphorylation of JNK, a cell death signaling intermediate [125]. In the same way, highland barley tea extract, rich in polyphenols and flavonoids, represses PA-induced apoptosis and mitochondrial dysfunction in C2C12 myocytes by promoting the expression of Sirt3 through the phosphorylation of adenosine 5‘-monophosphate (AMP)-activated protein kinase (AMPK), which also activated SIRT3/FoxO3a to scavenge ROS in excess [126]. In summary, various antioxidants have been developed and tested to mitigate PA-induced lipotoxicity. However, further investigations into the efficacy and specificity of natural antioxidants in reducing oxidative stress and improving cellular health in models of metabolic MetS and obesity are needed.
PA-induced IR also involves intracellular mechanisms sensitive to oxidative stress and ROS generation. Several natural extracts from higher plants, such as phenolic and flavonoid compounds with antioxidant properties, have been tested and shown to be protective against PA-induced IR in cell cultures [127,128]. For example, Epigallocatechin gallate and resveratrol stimulate glucose uptake mediated by the PI3K-independent mechanism that involves SIRT1 in skeletal muscle cells [129] and improve PA-induced IR in C2C12 cells through the DNA damage-inducible transcript 4 (DDIT4)/mTOR/IRS-1/PI3K/AKT/GLUT4 signaling pathway [130]. Epigallocatechin gallate also improves PA-induced IR and reduces inflammatory and oxidative stress levels through NF-κB, tumor necrosis factor-α, interleukin-6, p53, ROS and by enhancing SOD and glutathione peroxidase [129]. In HepG2 cells, theaflavins, the bioactive polyphenols in black tea, protect against PA-induced insulin resistance by up-regulating total and membrane-bound GLUT4, increasing the phosphorylation of Akt at Ser473 residue, and decreasing the phosphorylation of IRS-1 at Ser307 [131]. Other natural polyphenols, such as salvianolic acid A, extracted from Salvia miltiorrhiza Bunge protect against PA-induced lipotoxicity in livers from high-fat–high-carbohydrate (HFCD)-fed mice and in hepatocytes through a TLR4/MAPKs-mediated mechanism [123,124]. Other natural phenolic compounds, such as curcumin, quercetin, resveratrol and sulforaphane, widely known as natural Keap1-Nrf2 activators, react with critical thiol groups of Keap1, leading to the release and activation of Nrf2, enhancing cellular protection against toxicity, which could mitigate PA-induced oxidative stress through the expression of antioxidant enzymes [132]. In summary, various antioxidants have been developed and tested to mitigate PA-induced lipotoxicity. Notably, the main mechanism is related to the activation of sirtuins, Akt and AMPK, with polyphenols being the most effective. Also, it appears that PA-induced lipotoxicity is mainly related to oxidative stress and that antioxidants diminish this process. However, lipotoxicity is not totally attenuated, suggesting that PA induces lipotoxicity through other signaling pathways. Further investigations on the efficacy and specificity of natural antioxidants in reducing oxidative stress and improving cellular health in models of metabolic MetS and obesity are needed to provide an appropriate therapeutic target that could help improve antioxidant therapy.

7. PA-Induced Lipotoxicity Is Attenuated by Lipid Droplet (LD) Functionality

The biogenesis of LDs from ER is induced in cells exposed to excessive amounts of lipids, nutrients and oxidative stress [133]. However, several other conditions, such as energy and redox imbalances, limit LDs and accelerate lipophagy, a part of autophagy that maintains cellular homeostasis, by removing damaged proteins and lipids [134,135]. In normal beta-cells, triacylglycerol accumulation in LDs is considered a protective mechanism to prevent the cellular rise of toxic free fatty acyl moieties that serve as inducers of cell death [136]. Thus, the biosynthesis and turnover of LDs within cells are precisely regulated to maintain the balanced lipid distribution and allow for cellular adaptation during saturated fatty acid stress [137].
In non-adipose cells, cytosolic LDs store neutral lipid triacylglycerols, and, when energy is required, they are hydrolyzed to FFA and glycerol as an oxidative substrate for ATP biosynthesis. Lipid droplets, however, have been described to form dynamic contact sites with mitochondria and peroxisomes, facilitating the transfer of fatty acids for beta-oxidation and energy production [138]. These interactions will depend on the metabolic state of the cell and will vary according to cell and tissue type. While lipid droplets are essential for efficient energy utilization, their dysfunction can also lead to metabolic disorders, such as obesity and diabetes, underscoring the importance of new research that will enable us to understand their biogenesis and interactions with other organelles in order to develop therapeutic strategies for lipid-associated diseases.
In primary cultures of cardiomyocytes from adult rats, the protective mechanism against PA-induced apoptosis involves channeling PA into triglyceride pools, which accumulate in LDs, decreasing DNA fragmentation and cell death through apoptosis [139,140]. The storage of harmful PA into inert triglyceride (TG) in LDs within skeletal muscle myotubes has been evidenced by the rapid incorporation of the fluorescent PA analogue into arachidonic acid-driven TG droplets [17]. Adipocyte LDs have a great capacity to store the exogenous PUFA delivered from the diet as TGs to avoid their accumulation within the phospholipid membrane that render cells more susceptible to oxidative stress [141]. In addition, LDs protect against lipid peroxidation, control the availability of energy for muscle function, modulate mitochondrial activity and enable the transfer of fatty acids between organelles, but they influence lipotoxicity in metabolic diseases [142]. In cardiomyocytes, lipids that exceed the cell capacity for LD storage induce non-ischemic and non-hypertensive cardiomyopathy, known as lipotoxic cardiomyopathy [143].
Lipid droplets have also been reported to control PUFA storage in triglycerides in order to reduce membrane lipid peroxidation and, in turn, may act as antioxidant organelles to preserve the cell function and prevent cell death due to ferroptosis [135,144]. Consequently, LDs play a crucial role in regulating PUFA transport and facilitating the management of membrane lipid peroxidation, ferroptosis and lipid mediator signaling [145]. The presence of LDs within renal cells suggests significant potential for the diagnosis and enhanced comprehension of chronic kidney disease, due to the concurrent presence of oxidized lipid species, such as triglycerides, phosphatidylcholines and phosphatidylethanolamine hydroperoxides, in addition to the dysregulation of lipid metabolic pathways [146]. The protective effect of LDs is also associated with the replacement of PUFAs with MUFAs that are more resistant to lipid peroxidation. Indeed, the treatment of cells with exogenous MUFAs reduces the sensitivity of plasma membrane lipids to oxidation and activates acyl-coenzyme A synthetase long-chain family member 3 (ACSL3) involved in LD formation and the insertion of MUFA in membrane phospholipids [147]. Additionally, PUFAs, including linoleic acid obtained from nutritional intake, are transferred from cellular membranes to the nucleus of LDs, where they are more protected from peroxidation, thereby highlighting the antioxidant functions of LDs [145]. Thus, the endogenous formation of LDs diminishes PA lipotoxicity, reducing the presence of FFAs, avoiding interaction with lipolysis enzymes and, therefore, the excessive production of oxidative phosphorylation substrates that can produce ROS. Also, it could prevent increased phospholipids enriched with saturated fatty acids in the membrane and the palmytoylation of several proteins, preserving cellular homeostasis.

8. Natural Antioxidants and PA-Induced Lipid Droplets

The intracellular accumulation of TG and its oxidized species (TG hydroperoxides) in LDs triggered by PA in hepatocytes can also be regulated by polyphenols and flavonoids with antioxidant activities, decreasing the release of lactate dehydrogenase, an important indicator of cytotoxicity [148,149]. In HepG2 cells with oleic acid and PA-induced lipid accumulation, baicalein, a flavonoid derived from Scutellaria-baicalensis, reduces the production of TG, total cholesterol and lipid droplets by decreasing the expression of SREBP1, as well as the lipogenic enzymes fatty acid synthase (FAS) and stearoyl-CoA desaturase-1 (SCD1) [150]. The addition of N-acetylcysteine (NAC) as an antioxidant to the retinal pigment epithelium reduces oxidative stress mediated by lipid metabolism, which plays an important role in lipid droplet (LD) accumulation and cellular senescence induced by high glucose level [151]. On the other hand, exercise training elevates the markers of lipid droplet dynamics such as the perilipin proteins and triglyceride lipases ATGL and HSL, as well as mitochondrial efficiency, improving lipid turnover and reducing lipotoxicity [152].
Thus, LDs play an important role in the regulation of cellular stress, suggesting their application as safe and effective biomaterials to reduce cellular stress and lipotoxicity. Therefore, the generation of stable and pure artificial LDs coated with perilipins, lipase regulatory proteins, has reduced hydrogen peroxide-induced oxidized lipid species and alleviated cellular lipotoxicity caused by excess FFAs [142,153].
In summary, LDs are usually protective in many diseases; however, the long-term accumulation of and uncontrolled increase in LDs in chronic diseases such as obesity and MetS can exacerbate disease progression [154]. It is, therefore, necessary to understand the lipid–protein interactions of physiological and pathological LDs in order to better understand their transition from beneficial to detrimental in different pathological contexts. In this case, investigating the sites of contact of lipid droplets with other organelles in healthy and diseased states will contribute to elucidating the mechanisms of disease progression and, thus, the development of therapeutic strategies based on the biogenesis and function of lipid droplets. Therefore, more studies are needed to clarify how lipid droplet dynamics can be manipulated to enhance their protective effects against lipotoxicity and their potential as therapeutic targets in cellular stress regulation.

9. Antioxidant Strategies to Prevent Lipid Peroxidation

Mitochondrial PA beta-oxidation has been described as a source of superoxide anion (O2)(O2), which, in turn, is protonated to generate the perhydroxy radical (HOO.) that promotes the lipid peroxidation of PUFA [155,156]. In order to attenuate lipid oxidation in the human body and food, several effective strategies using natural antioxidants, including tocopherols, ascorbic acid, rosemary extracts, lycopene and some flavonoids, should be considered as inhibitors of lipid hydroperoxide formation and subsequent degradation into aldehydic by-products [157]. These natural antioxidants are also added to foods to protect nutrients and the human body from oxidation and the generation of volatile organic compounds, especially aldehydes [158]. However, once phenolic antioxidants trap or neutralize free radical-initiated lipid peroxidation, they are also converted into reactive species, such as phenoxy radicals, which may enhance lipid peroxidation over time [159]. To prevent the accumulation of these phenoxy radicals, they are reduced back to their original compound by other phenolic compounds existing in the total plant extract as an electron donor, ensuring a continuous supply of antioxidants. The underlying mechanisms of synergistic interactions between several antioxidants are poorly understood and rarely studied [160]. Nevertheless, the most natural antioxidants used have several phenolic rings in their molecular structure and are effective in inhibiting lipid peroxidation by blocking the formation of free radicals. In addition, the increased number of hydroxyl groups, the presence of allyl carboxylic acid and the substitution of hydroxy by methoxy groups in the molecule strongly increase their reducing and antioxidant capacities [161,162]. In regard to lipid food protection strategies against oxidation, the reduction of O2, through airtightness and vacuum, encapsulation and light protection have been used [163,164]. Further, the strategies of antioxidant supplementation in the food industry have been successfully used to improve food shelf life. Synthetic antioxidants, such as tert-butyl-4-hydroxyanisole (BHA), 2,6-di-tert-butyl-4-methylphenol (BHT), tert-butylhydroquinone (TBHQ) and propyl gallate (PG), have been commonly added to processed foods to avoid rancidity and malodor resulting from nutrient oxidation, but their use has become limited due to growing concerns about their potential mutagenic and carcinogenic effects [165]. Therefore, the use of the total polyphenolic extract from plants allows the antioxidants to continuously quench free radicals, prevent the oxidation of biomolecules and the accumulation of oxidized phenols and the formation of secondary products, preserving the flavors, colors and texture of foods during storage [165,166]. The antioxidant capacity depends on the individual (poly)phenol content of mangosteen peel extract. The reducing capacities of their phenolic compounds can be predicted on the basis of their structural characteristics. For example, mangosteen peel extract, rich in α, β and γ-mangostin as major polyphenols with several hydroxyl and methoxy groups, is used as a potential source of natural antioxidants for prolonged application in the food industry to suppress lipid peroxidation (for structure, see Figure 2) [167]. These polyphenols can prevent some foods enriched with n-3 PUFA from quality deteriorations and lipid oxidation [168]. A study indicated that n-3 PUFA-enriched eggs were relatively stable during storage and home cooking in the presence of antioxidants [169]. N-3 PUFA ingestion without antioxidant protection results in increased plasma and urinary concentrations of hydroxyl alkanals due to peroxidized oil absorption or in vivo oxidation [170,171]. Indeed, 4-HNE and lipoxygenase (LOX)-catalyzed linoleic acid (LA) oxidation products affect the digestibility and gel properties in myofibrillar proteins from bighead carp, resulting in decreased free amino acid content in myofibrillar proteins [172]. Aldehydes can be found in many foods, including red meat, chicken, fish, dairy, grains, fruits, vegetables and alcoholic beverages. However, fried foods are particularly high in oxidized fat and can contain high concentrations of aldehydes [173].
As these lipid aldehyde compounds have adverse effects on nutrient quality and human health, it is a challenge to reduce their formation. In tissues, lipid aldehydes are metabolically removed through oxidation and reduction. They are catalyzed via the enzymes aldehyde dehydrogenase (ALDH2) and aldose reductase (AKR1B1) and are also removed by endogenous glutathione and histidyl dipeptides, such as carnosine (β-alanine-histidine). When both the enzymatic and non-enzymatic removal of lipid aldehydes is diminished, aldehyde-modified proteins accumulate in atrophic skeletal muscle during heart failure. Therefore, reducing or blocking their formation during PUFA oxidation in foods will allow for their preservation and extend the shelf life of nutrients [174].

10. Conclusions

Oxidative stress caused by PA exposure is the main mechanism elucidated because it can be reversed through antioxidant treatments. Other processes, such as the activation of both lipid droplets and FFARs, are gaining importance, and, therefore, several works are oriented to determine how lipid droplets are formed and which contact sites between different organelles and lipid droplets are involved in the protection against PA-induced lipotoxicity. Therefore, it is essential to use formulations that are able to effectively preserve the bioactivity of n-3 PUFA in food by adding natural antioxidants from total plant extracts to block the propagation of lipid peroxidation and to reinforce the activity of the endogenous antioxidant systems, providing protection against PUFA peroxidation and preserving foods, leading to better health conditions.

Author Contributions

Conceptualization, M.E.-H. and Z.A.C.-G.; methodology, M.E.-H. and Z.A.C.-G.; investigation, M.E.-H. and Z.A.C.-G.; writing—original draft preparation, M.E.-H. and Z.A.C.-G.; writing—review and editing, M.E.-H., C.L.A.C.-A. and Z.A.C.-G.; supervision, M.E.-H.; project administration, M.E.-H.; funding acquisition, M.E.-H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank the Instituto Nacional de Cardiología Ignacio Chavez for the financial support that sustained the publication of this article.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Apekey, T.A.; Maynard, M.J.; Kittana, M.; Kunutsor, S.K. Comparison of the Effectiveness of Low Carbohydrate Versus Low Fat Diets, in Type 2 Diabetes: Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2022, 14, 4391. [Google Scholar] [CrossRef] [PubMed]
  2. Pour Abbasi, M.S.; Shojaei, N.; Farhangi, M.A. Low-carbohydrate diet score is associated with improved blood pressure and cardio-metabolic risk factors among obese adults. Physiol. Rep. 2022, 10, e15375. [Google Scholar] [CrossRef] [PubMed]
  3. Samaha, F.F.; Iqbal, N.; Seshadri, P.; Chicano, K.L.; Daily, D.A.; McGrory, J.; Williams, T.; Williams, M.; Gracely, E.J.; Stern, L. A Low-Carbohydrate as Compared with a Low-Fat Diet in Severe Obesity. N. Engl. J. Med. 2003, 348, 2074–2081. [Google Scholar] [CrossRef] [PubMed]
  4. Hyde, P.N.; Sapper, T.N.; Crabtree, C.D.; LaFountain, R.A.; Bowling, M.L.; Buga, A.; Fell, B.; McSwiney, F.T.; Dickerson, R.M.; Miller, V.J.; et al. Dietary carbohydrate restriction improves metabolic syndrome independent of weight loss. JCI Insight 2019, 4, e128308. [Google Scholar] [CrossRef]
  5. Hall, K.D.; Chung, S.T. Low-carbohydrate diets for the treatment of obesity and type 2 diabetes. Curr. Opin. Clin. Nutr. Metab. Care 2018, 21, 308–312. [Google Scholar] [CrossRef]
  6. Ismael, S.A. Effects of low carbohydrate diet compared to low fat diet on reversing the metabolic syndrome, using NCEP ATP III criteria: A randomized clinical trial. BMC Nutr. 2021, 7, 62. [Google Scholar] [CrossRef]
  7. Murru, E.; Manca, C.; Carta, G.; Banni, S. Impact of Dietary Palmitic Acid on Lipid Metabolism. Front. Nutr. 2022, 9, 861664. [Google Scholar] [CrossRef]
  8. Mthembu, S.X.H.; Mazibuko-Mbeje, S.E.; Silvestri, S.; Orlando, P.; Marcheggiani, F.; Cirilli, I.; Nkambule, B.B.; Muller, C.J.F.; Tiano, L.; Dludla, P.V. Low levels and partial exposure to palmitic acid improves mitochondrial function and the oxidative status of cultured cardiomyoblasts. Toxicol. Rep. 2024, 12, 234–243. [Google Scholar] [CrossRef]
  9. Rocca, C.; De Bartolo, A.; Guzzi, R.; Crocco, M.C.; Rago, V.; Romeo, N.; Perrotta, I.; De Francesco, E.M.; Muoio, M.G.; Granieri, M.C.; et al. Palmitate-Induced Cardiac Lipotoxicity Is Relieved by the Redox-Active Motif of SELENOT through Improving Mitochondrial Function and Regulating Metabolic State. Cells 2023, 12, 1042. [Google Scholar] [CrossRef]
  10. Engin, A.B. What Is Lipotoxicity? In Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2017; pp. 197–220. [Google Scholar]
  11. Chong, M.F.-F.; Hodson, L.; Bickerton, A.S.; Roberts, R.; Neville, M.; Karpe, F.; Frayn, K.N.; Fielding, B.A. Parallel activation of de novo lipogenesis and stearoyl-CoA desaturase activity after 3 d of high-carbohydrate feeding. Am. J. Clin. Nutr. 2008, 87, 817–823. [Google Scholar] [CrossRef]
  12. Cerk, I.K.; Wechselberger, L.; Oberer, M. Adipose Triglyceride Lipase Regulation: An Overview. Curr. Protein Pept. Sci. 2017, 19, 221–233. [Google Scholar] [CrossRef] [PubMed]
  13. Li, Y.; Li, Z.; Ngandiri, D.A.; Llerins Perez, M.; Wolf, A.; Wang, Y. The Molecular Brakes of Adipose Tissue Lipolysis. Front. Physiol. 2022, 13, 826314. [Google Scholar] [CrossRef] [PubMed]
  14. Alvheim, A.R.; Malde, M.K.; Osei-Hyiaman, D.; Hong, Y.H.; Pawlosky, R.J.; Madsen, L.; Kristiansen, K.; Frøyland, L.; Hibbeln, J.R. Dietary Linoleic Acid Elevates Endogenous 2-AG and Anandamide and Induces Obesity. Obesity 2012, 20, 1984–1994. [Google Scholar] [CrossRef]
  15. Miklankova, D.; Markova, I.; Hüttl, M.; Stankova, B.; Malinska, H. The Different Insulin-Sensitising and Anti-Inflammatory Effects of Palmitoleic Acid and Oleic Acid in a Prediabetes Model. J. Diabetes Res. 2022, 2022, 4587907. [Google Scholar] [CrossRef]
  16. Chen, X.; Li, L.; Liu, X.; Luo, R.; Liao, G.; Li, L.; Liu, J.; Cheng, J.; Lu, Y.; Chen, Y. Oleic acid protects saturated fatty acid mediated lipotoxicity in hepatocytes and rat of non-alcoholic steatohepatitis. Life Sci. 2018, 203, 291–304. [Google Scholar] [CrossRef] [PubMed]
  17. Cheon, H.; Cho, Y. Protection of palmitic acid-mediated lipotoxicity by arachidonic acid via channeling of palmitic acid into triglycerides in C2C12. J. Biomed. Sci. 2014, 21, 13. [Google Scholar] [CrossRef]
  18. Cho, Y.S.; Kim, C.H.; Kim, K.Y.; Cheon, H.G. Protective effects of arachidonic acid against palmitic acid-mediated lipotoxicity in HIT-T15 cells. Mol. Cell. Biochem. 2012, 364, 19–28. [Google Scholar] [CrossRef]
  19. Qiu, T.; Yang, X.; Wang, J.; Pan, C.; Chu, X.; Xiong, J.; Xie, J.; Chang, Y.; Wang, C.; Zhang, J. Obesity-induced elevated palmitic acid promotes inflammation and glucose metabolism disorders through GPRs/NF-κB/KLF7 pathway. Nutr. Diabetes 2022, 12, 23. [Google Scholar] [CrossRef]
  20. Schwarz, J.-M.; Linfoot, P.; Dare, D.; Aghajanian, K. Hepatic de novo lipogenesis in normoinsulinemic and hyperinsulinemic subjects consuming high-fat, low-carbohydrate and low-fat, high-carbohydrate isoenergetic diets. Am. J. Clin. Nutr. 2003, 77, 43–50. [Google Scholar] [CrossRef]
  21. Schweiger, M.; Schreiber, R.; Haemmerle, G.; Lass, A.; Fledelius, C.; Jacobsen, P.; Tornqvist, H.; Zechner, R.; Zimmermann, R. Adipose Triglyceride Lipase and Hormone-sensitive Lipase Are the Major Enzymes in Adipose Tissue Triacylglycerol Catabolism. J. Biol. Chem. 2006, 281, 40236–40241. [Google Scholar] [CrossRef]
  22. Kennedy, A.; Martinez, K.; Chuang, C.-C.; LaPoint, K.; McIntosh, M. Saturated Fatty Acid-Mediated Inflammation and Insulin Resistance in Adipose Tissue: Mechanisms of Action and Implications. J. Nutr. 2009, 139, 1–4. [Google Scholar] [CrossRef] [PubMed]
  23. Ferré, P.; Phan, F.; Foufelle, F. SREBP-1c and lipogenesis in the liver: An update. Biochem. J. 2021, 478, 3723–3739. [Google Scholar] [CrossRef] [PubMed]
  24. Régnier, M.; Carbinatti, T.; Parlati, L.; Benhamed, F.; Postic, C. The role of ChREBP in carbohydrate sensing and NAFLD development. Nat. Rev. Endocrinol. 2023, 19, 336–349. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, Y.; Botolin, D.; Xu, J.; Christian, B.; Mitchell, E.; Jayaprakasam, B.; Nair, M.; Peters, J.M.; Busik, J.; Olson, L.K.; et al. Regulation of hepatic fatty acid elongase and desaturase expression in diabetes and obesity. J. Lipid Res. 2006, 47, 2028–2041. [Google Scholar] [CrossRef]
  26. Benhamed, F.; Denechaud, P.-D.; Lemoine, M.; Robichon, C.; Moldes, M.; Bertrand-Michel, J.; Ratziu, V.; Serfaty, L.; Housset, C.; Capeau, J.; et al. The lipogenic transcription factor ChREBP dissociates hepatic steatosis from insulin resistance in mice and humans. J. Clin. Investig. 2012, 122, 2176–2194. [Google Scholar] [CrossRef]
  27. Moon, Y.A.; Ochoa, C.R.; Mitsche, M.A.; Hammer, R.E.; Horton, J.D. Deletion of ELOVL6 blocks the synthesis of oleic acid but does not prevent the development of fatty liver or insulin resistance. J. Lipid Res. 2015, 55, 2597–2605. [Google Scholar] [CrossRef]
  28. Poudyal, H.; Brown, L. Stearoyl-CoA Desaturase: A Vital Checkpoint in the Development and Progression of Obesity. Endocr. Metab. Immune Disord.-Drug Targets 2011, 11, 217–231. [Google Scholar] [CrossRef]
  29. El Hafidi, M.; Cuéllar, A.; Ramırez, J.; Baños, G. Effect of sucrose addition to drinking water, that induces hypertension in the rats, on liver microsomal Δ9 and Δ5-desaturase activities. J. Nutr. Biochem. 2001, 12, 396–403. [Google Scholar] [CrossRef]
  30. Ruiz-Ramírez, A.; Chávez-Salgado, M.; Peñeda-Flores, J.A.; Zapata, E.; Masso, F.; El-Hafidi, M. High-sucrose diet increases ROS generation, FFA accumulation, UCP2 level, and proton leak in liver mitochondria. Am. J. Physiol.-Endocrinol. Metab. 2011, 301, E1198–E1207. [Google Scholar] [CrossRef]
  31. Ravaut, G.; Légiot, A.; Bergeron, K.-F.; Mounier, C. Monounsaturated Fatty Acids in Obesity-Related Inflammation. Int. J. Mol. Sci. 2020, 22, 330. [Google Scholar] [CrossRef]
  32. Lenighan, Y.M.; McNulty, B.A.; Roche, H.M. Dietary fat composition: Replacement of saturated fatty acids with PUFA as a public health strategy, with an emphasis on α-linolenic acid. Proc. Nutr. Soc. 2019, 78, 234–245. [Google Scholar] [CrossRef] [PubMed]
  33. Smith, G.I.; Shankaran, M.; Yoshino, M.; Schweitzer, G.G.; Chondronikola, M.; Beals, J.W.; Okunade, A.L.; Patterson, B.W.; Nyangau, E.; Field, T.; et al. Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J. Clin. Investig. 2020, 130, 1453–1460. [Google Scholar] [CrossRef] [PubMed]
  34. Feingold, K.R. Lipid and Lipoprotein Metabolism. Endocrinol. Metab. Clin. N. Am. 2022, 51, 437–458. [Google Scholar] [CrossRef] [PubMed]
  35. Dikalov, S.; Panov, A.; Dikalova, A. Critical Role of Mitochondrial Fatty Acid Metabolism in Normal Cell Function and Pathological Conditions. Int. J. Mol. Sci. 2024, 25, 6498. [Google Scholar] [CrossRef]
  36. Sharifi, S.; Yamamoto, T.; Zeug, A.; Elsner, M.; Avezov, E.; Mehmeti, I. Non-esterified fatty acid palmitate facilitates oxidative endoplasmic reticulum stress and apoptosis of β-cells by upregulating ERO-1α expression. Redox Biol. 2024, 73, 103170. [Google Scholar] [CrossRef]
  37. Yang, H.-Y.; Chen, J.-Y.; Huo, Y.-N.; Yu, P.-L.; Lin, P.-Z.; Hsu, S.-C.; Huang, S.-M.; Tsai, C.-S.; Lin, C.-Y. The Role of Sirtuin 1 in Palmitic Acid-Induced Endoplasmic Reticulum Stress in Cardiac Myoblasts. Life 2022, 12, 182. [Google Scholar] [CrossRef]
  38. Ljubkovic, M.; Gressette, M.; Bulat, C.; Cavar, M.; Bakovic, D.; Fabijanic, D.; Grkovic, I.; Lemaire, C.; Marinovic, J. Disturbed Fatty Acid Oxidation, Endoplasmic Reticulum Stress, and Apoptosis in Left Ventricle of Patients With Type 2 Diabetes. Diabetes 2019, 68, 1924–1933. [Google Scholar] [CrossRef]
  39. Baldwin, A.C.; Green, C.D.; Olson, L.K.; Moxley, M.A.; Corbett, J.A. A role for aberrant protein palmitoylation in FFA-induced ER stress and β-cell death. Am. J. Physiol. Metab. 2012, 302, E1390–E1398. [Google Scholar] [CrossRef]
  40. Yuan, Y.; Li, P.; Li, J.; Zhao, Q.; Chang, Y.; He, X. Protein lipidation in health and disease: Molecular basis, physiological function and pathological implication. Signal Transduct. Target. Ther. 2024, 9, 60. [Google Scholar] [CrossRef]
  41. Martínez, L.; Torres, S.; Baulies, A.; Alarcón-Vila, C.; Elena, M.; Fabriàs, G.; Casas, J.; Caballeria, J.; Fernandez-Checa, J.C.; García-Ruiz, C. Myristic acid potentiates palmitic acid-induced lipotoxicity and steatohepatitis associated with lipodystrophy by sustaning de novo ceramide synthesis. Oncotarget 2015, 6, 41479–41496. [Google Scholar] [CrossRef]
  42. Imierska, M.; Zabielski, P.; Roszczyc-Owsiejczuk, K.; Sokołowska, E.; Pogodzińska, K.; Kojta, I.; Błachnio-Zabielska, A. Serine Palmitoyltransferase Gene Silencing Prevents Ceramide Accumulation and Insulin Resistance in Muscles in Mice Fed a High-Fat Diet. Cells 2022, 11, 1123. [Google Scholar] [CrossRef]
  43. Lallement, J.; Raho, I.; Merlen, G.; Rainteau, D.; Croyal, M.; Schiffano, M.; Kassis, N.; Doignon, I.; Soty, M.; Lachkar, F.; et al. Deletion of serine palmitoyl transferase 2 in hepatocytes impairs ceramide/sphingomyelin balance, prevents obesity and leads to liver damage in mice. bioRxiv 2021. [Google Scholar] [CrossRef]
  44. Ruiz, M.; Henricsson, M.; Borén, J.; Pilon, M. Palmitic acid causes increased dihydroceramide levels when desaturase expression is directly silenced or indirectly lowered by silencing AdipoR2. Lipids Health Dis. 2021, 20, 173. [Google Scholar] [CrossRef] [PubMed]
  45. Guo, M.; Huang, X.; Zhang, J.; Huang, Y.; Tang, Y.; Wen, H.; Xu, Y.; Zhang, S.; Wei, X.; Sun, S.; et al. Palmitic acid induces β-cell ferroptosis by activating ceramide signaling pathway. Exp. Cell Res. 2024, 440, 114134. [Google Scholar] [CrossRef] [PubMed]
  46. Yu, G.; Luo, H.; Zhang, N.; Wang, Y.; Li, Y.; Huang, H.; Liu, Y.; Hu, Y.; Liu, H.; Zhang, J.; et al. Loss of p53 Sensitizes Cells to Palmitic Acid-Induced Apoptosis by Reactive Oxygen Species Accumulation. Int. J. Mol. Sci. 2019, 20, 6268. [Google Scholar] [CrossRef]
  47. Wei, C.-D.; Li, Y.; Zheng, H.-Y.; Tong, Y.-Q.; Dai, W. Palmitate induces H9c2 cell apoptosis by increasing reactive oxygen species generation and activation of the ERK1/2 signaling pathway. Mol. Med. Rep. 2013, 7, 855–861. [Google Scholar] [CrossRef]
  48. Dadsena, S.; Bockelmann, S.; Mina, J.G.M.; Hassan, D.G.; Korneev, S.; Razzera, G.; Jahn, H.; Niekamp, P.; Müller, D.; Schneider, M.; et al. Ceramides bind VDAC2 to trigger mitochondrial apoptosis. Nat. Commun. 2019, 10, 1832. [Google Scholar] [CrossRef]
  49. Alizadeh, J.; da Silva Rosa, S.C.; Weng, X.; Jacobs, J.; Lorzadeh, S.; Ravandi, A.; Vitorino, R.; Pecic, S.; Zivkovic, A.; Stark, H.; et al. Ceramides and ceramide synthases in cancer: Focus on apoptosis and autophagy. Eur. J. Cell Biol. 2023, 102, 151337. [Google Scholar] [CrossRef]
  50. Chang, Y.C.; Fong, Y.; Tsai, E.-M.; Chang, Y.-G.; Chou, H.L.; Wu, C.-Y.; Teng, Y.-N.; Liu, T.-C.; Yuan, S.-S.; Chiu, C.-C. Exogenous C8-Ceramide Induces Apoptosis by Overproduction of ROS and the Switch of Superoxide Dismutases SOD1 to SOD2 in Human Lung Cancer Cells. Int. J. Mol. Sci. 2018, 19, 3010. [Google Scholar] [CrossRef]
  51. Kuang, H.; Sun, X.; Liu, Y.; Tang, M.; Wei, Y.; Shi, Y.; Li, R.; Xiao, G.; Kang, J.; Wang, F.; et al. Palmitic acid-induced ferroptosis via CD36 activates ER stress to break calcium-iron balance in colon cancer cells. FEBS J. 2023, 290, 3664–3687. [Google Scholar] [CrossRef]
  52. Chen, X.; Chen, K.; Hu, J.; Dong, Y.; Zheng, M.; Jiang, J.; Hu, Q.; Zhang, W. Palmitic acid induces lipid droplet accumulation and senescence in nucleus pulposus cells via ER-stress pathway. Commun. Biol. 2024, 7, 539. [Google Scholar] [CrossRef]
  53. Alnahdi, A.; John, A.; Raza, H. Augmentation of Glucotoxicity, Oxidative Stress, Apoptosis and Mitochondrial Dysfunction in HepG2 Cells by Palmitic Acid. Nutrients 2019, 11, 1979. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, X.; Mao, M.; Zuo, Z. Palmitate Induces Mitochondrial Energy Metabolism Disorder and Cellular Damage via the PPAR Signaling Pathway in Diabetic Cardiomyopathy. Diabetes Metab. Syndr. Obes. Targets Ther. 2022, 15, 2287–2299. [Google Scholar] [CrossRef] [PubMed]
  55. Raja, A.A.; Dandare, A.; Khan, M.J.; Khan, M.J. Free Fatty Acid Overload Targets Mitochondria: Gene Expression Analysis of Palmitic Acid-Treated Endothelial Cells. Genes 2022, 13, 1704. [Google Scholar] [CrossRef]
  56. Song, Y.; Wang, L.; Luo, Z.; Hogstrand, C.; Lai, X.-H.; Zheng, F. Moderate replacement of fish oil with palmitic acid-stimulated mitochondrial fusion promotes β-oxidation by Mfn2 interacting with Cpt1α via its GTPase-domain. J. Nutr. Biochem. 2024, 126, 109559. [Google Scholar] [CrossRef]
  57. Li, H.; Xiao, Y.; Tang, L.; Zhong, F.; Huang, G.; Xu, J.-M.; Xu, A.-M.; Dai, R.-P.; Zhou, Z.-G. Adipocyte Fatty Acid-Binding Protein Promotes Palmitate-Induced Mitochondrial Dysfunction and Apoptosis in Macrophages. Front. Immunol. 2018, 9, 81. [Google Scholar] [CrossRef]
  58. Haffar, T.; Akoumi, A.; Bousette, N. Lipotoxic Palmitate Impairs the Rate of β-Oxidation and Citric Acid Cycle Flux in Rat Neonatal Cardiomyocytes. Cell. Physiol. Biochem. 2016, 40, 969–981. [Google Scholar] [CrossRef]
  59. Barrios-Maya, M.-A.; Ruiz-Ramírez, A.; Quezada, H.; Céspedes Acuña, C.L.; El-Hafidi, M. Palmitoyl-CoA effect on cytochrome c release, a key process of apoptosis, from liver mitochondria of rat with sucrose diet-induced obesity. Food Chem. Toxicol. 2021, 154, 112351. [Google Scholar] [CrossRef]
  60. Ma, X.M.; Geng, K.; Law, B.Y.-K.; Wang, P.; Pu, Y.L.; Chen, Q.; Xu, H.W.; Tan, X.Z.; Jiang, Z.Z.; Xu, Y. Lipotoxicity-induced mtDNA release promotes diabetic cardiomyopathy by activating the cGAS-STING pathway in obesity-related diabetes. Cell Biol. Toxicol. 2023, 39, 277–299. [Google Scholar] [CrossRef]
  61. Heilig, R.; Lee, J.; Tait, S.W.G. Mitochondrial DNA in cell death and inflammation. Biochem. Soc. Trans. 2023, 51, 457–472. [Google Scholar] [CrossRef]
  62. Mansuri, M.L.; Sharma, G.; Parihar, P.; Dube, K.T.; Sharma, T.; Parihar, A.; Parihar, M.S. Increased oxidative stress and mitochondrial impairments associated with increased expression of TNF-α and caspase-3 in palmitic acid-induced lipotoxicity in myoblasts. J. Biochem. Mol. Toxicol. 2021, 35, e22744. [Google Scholar] [CrossRef] [PubMed]
  63. Park, E.-J.; Lee, A.Y.; Park, S.; Kim, J.-H.; Cho, M.-H. Multiple pathways are involved in palmitic acid-induced toxicity. Food Chem. Toxicol. 2014, 67, 26–34. [Google Scholar] [CrossRef] [PubMed]
  64. Ruiz-Ramírez, A.; Barrios-Maya, M.-A.; López-Acosta, O.; Molina-Ortiz, D.; El-Hafidi, M. Cytochrome c release from rat liver mitochondria is compromised by increased saturated cardiolipin species induced by sucrose feeding. Am. J. Physiol.-Endocrinol. Metab. 2015, 309, E777–E786. [Google Scholar] [CrossRef] [PubMed]
  65. Zuccaro, K.E.; Abriata, L.A.; Meireles, F.T.P.; Moss, F.R.; Frost, A.; Dal Peraro, M.; Aydin, H. Cardiolipin clustering promotes mitochondrial membrane dynamics. bioRxiv 2024. [Google Scholar] [CrossRef]
  66. Oemer, G.; Edenhofer, M.-L.; Lackner, K.; Leman, G.; Koch, J.; Lindner, H.H.; Dubrac, S.; Zschocke, J.; Keller, M.A. Fatty Acyl Availability Modulates Cardiolipin Composition and Alters Mitochondrial Function in HeLa Cells. J. Lipid Res. 2020, 62, 100111. [Google Scholar] [CrossRef]
  67. El-Hafidi, M.; Correa, F.; Zazueta, C. Mitochondrial dysfunction in metabolic and cardiovascular diseases associated with cardiolipin remodeling. Biochim. Biophys. Acta-Mol. Basis Dis. 2020, 1866, 165744. [Google Scholar] [CrossRef]
  68. Venkatraman, K.; Budin, I. Cardiolipin remodeling maintains the inner mitochondrial membrane in cells with saturated lipidomes. J. Lipid Res. 2024, 65, 100601. [Google Scholar] [CrossRef]
  69. Sapandowski, A.; Stope, M.; Evert, K.; Evert, M.; Zimmermann, U.; Peter, D.; Päge, I.; Burchardt, M.; Schild, L. Cardiolipin composition correlates with prostate cancer cell proliferation. Mol. Cell. Biochem. 2015, 410, 175–185. [Google Scholar] [CrossRef]
  70. Bi, C.; Zhang, T.; Li, Y.; Zhao, H.; Zhang, P.; Wang, Y.; Xu, Y.; Gu, K.; Liu, Y.; Yu, J.; et al. A Proteomics- and Metabolomics-Based Study Revealed That Disorder of Palmitic Acid Metabolism by Aconitine Induces Cardiac Injury. Chem. Res. Toxicol. 2020, 33, 3031–3040. [Google Scholar] [CrossRef]
  71. Yang, L.; Guan, G.; Lei, L.; Liu, J.; Cao, L.; Wang, X. Oxidative and endoplasmic reticulum stresses are involved in palmitic acid-induced H9c2 cell apoptosis. Biosci. Rep. 2019, 39, BSR20190225. [Google Scholar] [CrossRef]
  72. Shilovsky, G.A.; Putyatina, T.S.; Ashapkin, V.V.; Yamskova, O.V.; Lyubetsky, V.A.; Sorokina, E.V.; Shram, S.I.; Markov, A.V.; Vyssokikh, M.Y. Biological Diversity and Remodeling of Cardiolipin in Oxidative Stress and Age-Related Pathologies. Biochem. 2019, 84, 1469–1483. [Google Scholar] [CrossRef] [PubMed]
  73. Korbecki, J.; Bajdak-Rusinek, K. The effect of palmitic acid on inflammatory response in macrophages: An overview of molecular mechanisms. Inflamm. Res. 2019, 68, 915–932. [Google Scholar] [CrossRef] [PubMed]
  74. Lambertucci, R.H.; Leandro, C.G.; Vinolo, M.A.; Nachbar, R.T.; dos Reis Silveira, L.; Hirabara, S.M.; Curi, R.; Pithon-Curi, T.C. The Effects of Palmitic Acid on Nitric Oxide Production by Rat Skeletal Muscle: Mechanism via Superoxide and iNOS Activation. Cell. Physiol. Biochem. 2012, 30, 1169–1180. [Google Scholar] [CrossRef] [PubMed]
  75. Sano, M.; Shimazaki, S.; Kaneko, Y.; Karasawa, T.; Takahashi, M.; Ohkuchi, A.; Takahashi, H.; Kurosawa, A.; Torii, Y.; Iwata, H.; et al. Palmitic acid activates NLRP3 inflammasome and induces placental inflammation during pregnancy in mice. J. Reprod. Dev. 2020, 66, 241–248. [Google Scholar] [CrossRef]
  76. Liu, B.; Deng, X.; Jiang, Q.; Li, G.; Zhang, J.; Zhang, N.; Xin, S.; Xu, K. Scoparone improves hepatic inflammation and autophagy in mice with nonalcoholic steatohepatitis by regulating the ROS/P38/Nrf2 axis and PI3K/AKT/mTOR pathway in macrophages. Biomed. Pharmacother. 2020, 125, 109895. [Google Scholar] [CrossRef]
  77. Amine, H.; Benomar, Y.; Taouis, M. Palmitic acid promotes resistin-induced insulin resistance and inflammation in SH-SY5Y human neuroblastoma. Sci. Rep. 2021, 11, 5427. [Google Scholar] [CrossRef]
  78. Sergi, D.; Luscombe-Marsh, N.; Naumovski, N.; Abeywardena, M.; O’Callaghan, N. Palmitic Acid, but Not Lauric Acid, Induces Metabolic Inflammation, Mitochondrial Fragmentation, and a Drop in Mitochondrial Membrane Potential in Human Primary Myotubes. Front. Nutr. 2021, 8, 663838. [Google Scholar] [CrossRef]
  79. Tanwar, V.S.; Reddy, M.A.; Das, S.; Samara, V.A.; Abdollahi, M.; Dey, S.; Malek, V.; Ganguly, R.; Stapleton, K.; Lanting, L.; et al. Palmitic Acid–Induced Long Noncoding RNA PARAIL Regulates Inflammation via Interaction With RNA-Binding Protein ELAVL1 in Monocytes and Macrophages. Arterioscler. Thromb. Vasc. Biol. 2023, 43, 1157–1175. [Google Scholar] [CrossRef]
  80. Al Mahri, S.; Malik, S.S.; Al Ibrahim, M.; Haji, E.; Dairi, G.; Mohammad, S. Free Fatty Acid Receptors (FFARs) in Adipose: Physiological Role and Therapeutic Outlook. Cells 2022, 11, 750. [Google Scholar] [CrossRef]
  81. Kimura, I.; Ichimura, A.; Ohue-Kitano, R.; Igarashi, M. Free Fatty Acid Receptors in Health and Disease. Physiol. Rev. 2020, 100, 171–210. [Google Scholar] [CrossRef]
  82. Latour, M.G.; Alquier, T.; Oseid, E.; Tremblay, C.; Jetton, T.L.; Luo, J.; Lin, D.C.-H.; Poitout, V. GPR40 Is Necessary but Not Sufficient for Fatty Acid Stimulation of Insulin Secretion In Vivo. Diabetes 2007, 56, 1087–1094. [Google Scholar] [CrossRef] [PubMed]
  83. Shapiro, H.; Shachar, S.; Sekler, I.; Hershfinkel, M.; Walker, M.D. Role of GPR40 in fatty acid action on the β cell line INS-1E. Biochem. Biophys. Res. Commun. 2005, 335, 97–104. [Google Scholar] [CrossRef] [PubMed]
  84. Sakuma, K.; Yabuki, C.; Maruyama, M.; Abiru, A.; Komatsu, H.; Negoro, N.; Tsujihata, Y.; Takeuchi, K.; Habata, Y.; Mori, M. Fasiglifam (TAK-875) has dual potentiating mechanisms via G α q-GPR40/FFAR1 signaling branches on glucose-dependent insulin secretion. Pharmacol. Res. Perspect. 2016, 4, e00237. [Google Scholar] [CrossRef]
  85. Trexler, A.J.; Taraska, J.W. Regulation of insulin exocytosis by calcium-dependent protein kinase C in beta cells. Cell Calcium 2017, 67, 1–10. [Google Scholar] [CrossRef]
  86. Kim, H.-S.; Hwang, Y.-C.; Koo, S.-H.; Park, K.S.; Lee, M.-S.; Kim, K.-W.; Lee, M.-K. PPAR-γ Activation Increases Insulin Secretion through the Up-regulation of the Free Fatty Acid Receptor GPR40 in Pancreatic β-Cells. PLoS ONE 2013, 8, e50128. [Google Scholar] [CrossRef] [PubMed]
  87. Kristinsson, H.; Bergsten, P.; Sargsyan, E. Free fatty acid receptor 1 (FFAR1/GPR40) signaling affects insulin secretion by enhancing mitochondrial respiration during palmitate exposure. Biochim. Biophys. Acta-Mol. Cell Res. 2015, 1853, 3248–3257. [Google Scholar] [CrossRef] [PubMed]
  88. Kristinsson, H.; Smith, D.M.; Bergsten, P.; Sargsyan, E. FFAR1 Is Involved in Both the Acute and Chronic Effects of Palmitate on Insulin Secretion. Endocrinology 2013, 154, 4078–4088. [Google Scholar] [CrossRef]
  89. Marafie, S.K.; Al-Shawaf, E.M.; Abubaker, J.; Arefanian, H. Palmitic acid-induced lipotoxicity promotes a novel interplay between Akt-mTOR, IRS-1, and FFAR1 signaling in pancreatic β-cells. Biol. Res. 2019, 52, 44. [Google Scholar] [CrossRef]
  90. Briscoe, C.P.; Tadayyon, M.; Andrews, J.L.; Benson, W.G.; Chambers, J.K.; Eilert, M.M.; Ellis, C.; Elshourbagy, N.A.; Goetz, A.S.; Minnick, D.T.; et al. The Orphan G Protein-coupled Receptor GPR40 Is Activated by Medium and Long Chain Fatty Acids. J. Biol. Chem. 2003, 278, 11303–11311. [Google Scholar] [CrossRef]
  91. Itoh, Y.; Kawamata, Y.; Harada, M.; Kobayashi, M.; Fujii, R.; Fukusumi, S.; Ogi, K.; Hosoya, M.; Tanaka, Y.; Uejima, H.; et al. Free fatty acids regulate insulin secretion from pancreatic β cells through GPR40. Nature 2003, 422, 173–176. [Google Scholar] [CrossRef]
  92. Wang, J.; Chen, Y.; Song, Q.; Griffiths, A.; Song, Z. mTORC1-IRE1α pathway activation contributes to palmitate-elicited triglyceride secretion and cell death in hepatocytes. Exp. Biol. Med. 2020, 245, 1268–1279. [Google Scholar] [CrossRef] [PubMed]
  93. Gosis, B.S.; Wada, S.; Thorsheim, C.; Li, K.; Jung, S.; Rhoades, J.H.; Yang, Y.; Brandimarto, J.; Li, L.; Uehara, K.; et al. Inhibition of nonalcoholic fatty liver disease in mice by selective inhibition of mTORC1. Science 2022, 376, eabf8271. [Google Scholar] [CrossRef] [PubMed]
  94. Arora, M.; Pavlíková, Z.; Kučera, T.; Kozlík, P.; Šopin, T.; Vacík, T.; Ľupták, M.; Duda, M.; Slanař, O.; Kutinová Canová, N. Pharmacological effects of mTORC1/C2 inhibitor in a preclinical model of NASH progression. Biomed. Pharmacother. 2023, 167, 115447. [Google Scholar] [CrossRef]
  95. Gehrmann, W.; Elsner, M.; Lenzen, S. Role of metabolically generated reactive oxygen species for lipotoxicity in pancreatic β -cells. Diabetes Obes. Metab. 2010, 12, 149–158. [Google Scholar] [CrossRef]
  96. Graciano, M.F.; Valle, M.M.; Curi, R.; Carpinelli, A.R. Evidence for the involvement of GPR40 and NADPH oxidase in palmitic acid-induced superoxide production and insulin secretion. Islets 2013, 5, 139–148. [Google Scholar] [CrossRef]
  97. Leifke, E.; Naik, H.; Wu, J.; Viswanathan, P.; DeManno, D.; Kipnes, M.; Vakilynejad, M. A multiple-ascending-dose study to evaluate safety, pharmacokinetics, and pharmacodynamics of a novel GPR40 agonist, TAK-875, in subjects with type 2 diabetes. Clin. Pharmacol. Ther. 2012, 92, 29–39. [Google Scholar] [CrossRef]
  98. Araki, T.; Hirayama, M.; Hiroi, S.; Kaku, K. GPR40-induced insulin secretion by the novel agonist TAK-875: First clinical findings in patients with type 2 diabetes. Diabetes Obes. Metab. 2012, 14, 271–278. [Google Scholar] [CrossRef]
  99. Dragano, N.R.V.; Milbank, E.; Haddad-Tóvolli, R.; Garrido-Gil, P.; Nóvoa, E.; Fondevilla, M.F.; Capelli, V.; Zanesco, A.M.; Solon, C.; Morari, J.; et al. Hypothalamic free fatty acid receptor-1 regulates whole-body energy balance. Mol. Metab. 2024, 79, 101840. [Google Scholar] [CrossRef]
  100. Huang, C.-Y.; Chen, H.-W.; Lo, C.-W.; Wang, Y.-R.; Li, C.-C.; Liu, K.-L.; Lii, C.-K. Luteolin ameliorates palmitate-induced lipotoxicity in hepatocytes by mediating endoplasmic reticulum stress and autophagy. Food Chem. Toxicol. 2023, 171, 113554. [Google Scholar] [CrossRef]
  101. Kim, C.; Joo, S.; Kim, I.; Choi, H.-I.; Bae, E.; Kim, S.; Ma, S. Anti-Apoptotic Effect of G-Protein-Coupled Receptor 40 Activation on Tumor Necrosis Factor-α-Induced Injury of Rat Proximal Tubular Cells. Int. J. Mol. Sci. 2019, 20, 3386. [Google Scholar] [CrossRef]
  102. Shen, X.; Yang, L.; Yan, S.; Wei, W.; Liang, L.; Zheng, H.; Cai, X. The effect of FFAR1 on pioglitazone-mediated attenuation of palmitic acid-induced oxidative stress and apoptosis in βTC6 cells. Metabolism 2014, 63, 335–351. [Google Scholar] [CrossRef] [PubMed]
  103. Tuo, Y.; Feng, D.D.; Wang, D.; Sun, J.; Li, S.; Chen, C. Long-term in vitro treatment of INS -1 rat pancreatic β-cells by unsaturated free fatty acids protects cells against gluco- and lipotoxicities via activation of GPR 40 receptors. Clin. Exp. Pharmacol. Physiol. 2012, 39, 423–428. [Google Scholar] [CrossRef] [PubMed]
  104. Wu, P.; Yang, L.; Shen, X. The relationship between GPR40 and lipotoxicity of the pancreatic β-cells as well as the effect of pioglitazone. Biochem. Biophys. Res. Commun. 2010, 403, 36–39. [Google Scholar] [CrossRef]
  105. Wang, Y.; Xie, T.; Zhang, D.; Leung, P.S. GPR120 protects lipotoxicity-induced pancreatic β-cell dysfunction through regulation of PDX1 expression and inhibition of islet inflammation. Clin. Sci. 2019, 133, 101–116. [Google Scholar] [CrossRef]
  106. Martínez-García, C.; Izquierdo-Lahuerta, A.; Vivas, Y.; Velasco, I.; Yeo, T.-K.; Chen, S.; Medina-Gomez, G. Renal Lipotoxicity-Associated Inflammation and Insulin Resistance Affects Actin Cytoskeleton Organization in Podocytes. PLoS ONE 2015, 10, e0142291. [Google Scholar] [CrossRef]
  107. Pan, Y.; Hui, X.; Hoo, R.L.C.; Ye, D.; Chan, C.Y.C.; Feng, T.; Wang, Y.; Lam, K.S.L.; Xu, A. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J. Clin. Investig. 2019, 129, 834–849. [Google Scholar] [CrossRef]
  108. Garcia-Martinez, I.; Alen, R.; Pereira, L.; Povo-Retana, A.; Astudillo, A.M.; Hitos, A.B.; Gomez-Hurtado, I.; Lopez-Collazo, E.; Boscá, L.; Francés, R.; et al. Saturated fatty acid-enriched small extracellular vesicles mediate a crosstalk inducing liver inflammation and hepatocyte insulin resistance. JHEP Rep. 2023, 5, 100756. [Google Scholar] [CrossRef]
  109. Ferrara, P.J.; Rong, X.; Maschek, J.A.; Verkerke, A.R.P.; Siripoksup, P.; Song, H.; Green, T.D.; Krishnan, K.C.; Johnson, J.M.; Turk, J.; et al. Lysophospholipid acylation modulates plasma membrane lipid organization and insulin sensitivity in skeletal muscle. J. Clin. Investig. 2021, 131, e135963. [Google Scholar] [CrossRef]
  110. Tokarz, V.L.; Mylvaganam, S.; Klip, A. Palmitate-induced insulin resistance causes actin filament stiffness and GLUT4 mis-sorting without altered Akt signalling. J. Cell Sci. 2023, 136, jcs261300. [Google Scholar] [CrossRef]
  111. Den Hartogh, D.J.; Vlavcheski, F.; Tsiani, E. Muscle Cell Insulin Resistance is Attenuated by Rosmarinic Acid: Elucidating the Mechanisms Involved. Int. J. Mol. Sci. 2023, 24, 5094. [Google Scholar] [CrossRef]
  112. Tang, C.; Wang, Y.; Xu, Z.; Chen, D.; Xu, J.; Yang, D.; Zhang, L.; Liu, J.; Kan, J. The relationships between high-fat diet and metabolic syndrome: Potential mechanisms. Food Biosci. 2024, 59, 104261. [Google Scholar] [CrossRef]
  113. Vilas-Boas, E.A.; Almeida, D.C.; Roma, L.P.; Ortis, F.; Carpinelli, A.R. Lipotoxicity and β-cell failure in type 2 diabetes: Oxidative stress linked to NADPH oxidase and ER stress. Cells 2021, 10, 3328. [Google Scholar] [CrossRef] [PubMed]
  114. Dong, G.; Adak, S.; Spyropoulos, G.; Zhang, Q.; Feng, C.; Yin, L.; Speck, S.L.; Shyr, Z.; Morikawa, S.; Kitamura, R.A.; et al. Palmitoylation couples insulin hypersecretion with β cell failure in diabetes. Cell Metab. 2023, 35, 332–344. [Google Scholar] [CrossRef] [PubMed]
  115. Fauconnier, J.; Andersson, D.C.; Zhang, S.-J.; Lanner, J.T.; Wibom, R.; Katz, A.; Bruton, J.D.; Westerblad, H. Effects of Palmitate on Ca2+ Handling in Adult Control and ob/ob Cardiomyocytes. Diabetes 2007, 56, 1136–1142. [Google Scholar] [CrossRef]
  116. Ren, G.; Bhatnagar, S.; Hahn, D.J.; Kim, J. Long-chain acyl-CoA synthetase-1 mediates the palmitic acid-induced inflammatory response in human aortic endothelial cells. Am. J. Physiol. Metab. 2020, 319, E893–E903. [Google Scholar] [CrossRef]
  117. Wang, Y.; Qian, Y.; Fang, Q.; Zhong, P.; Li, W.; Wang, L.; Fu, W.; Zhang, Y.; Xu, Z.; Li, X.; et al. Saturated palmitic acid induces myocardial inflammatory injuries through direct binding to TLR4 accessory protein MD2. Nat. Commun. 2017, 8, 13997. [Google Scholar] [CrossRef]
  118. Wang, X.; Zhu, L.; Liu, J.; Ma, Y.; Qiu, C.; Liu, C.; Gong, Y.; Yuwen, Y.; Guan, G.; Zhang, Y.; et al. Palmitic acid in type 2 diabetes mellitus promotes atherosclerotic plaque vulnerability via macrophage Dll4 signaling. Nat. Commun. 2024, 15, 1281. [Google Scholar] [CrossRef]
  119. Trombetta, A.; Togliatto, G.; Rosso, A.; Dentelli, P.; Olgasi, C.; Cotogni, P.; Brizzi, M.F. Increase of Palmitic Acid Concentration Impairs Endothelial Progenitor Cell and Bone Marrow–Derived Progenitor Cell Bioavailability. Diabetes 2013, 62, 1245–1257. [Google Scholar] [CrossRef]
  120. Jiménez-González, S.; Marín-Royo, G.; Jurado-López, R.; Bartolomé, M.V.; Romero-Miranda, A.; Luaces, M.; Islas, F.; Nieto, M.L.; Martínez-Martínez, E.; Cachofeiro, V. The Crosstalk between Cardiac Lipotoxicity and Mitochondrial Oxidative Stress in the Cardiac Alterations in Diet-Induced Obesity in Rats. Cells 2020, 9, 451. [Google Scholar] [CrossRef]
  121. Longhitano, L.; Distefano, A.; Amorini, A.M.; Orlando, L.; Giallongo, S.; Tibullo, D.; Lazzarino, G.; Nicolosi, A.; Alanazi, A.M.; Saoca, C.; et al. (+)-Lipoic Acid Reduces Lipotoxicity and Regulates Mitochondrial Homeostasis and Energy Balance in an In Vitro Model of Liver Steatosis. Int. J. Mol. Sci. 2023, 24, 14491. [Google Scholar] [CrossRef]
  122. Wong, K.-L.; Wu, Y.-R.; Cheng, K.-S.; Chan, P.; Cheung, C.-W.; Lu, D.-Y.; Su, T.-H.; Liu, Z.-M.; Leung, Y.-M. Palmitic acid-induced lipotoxicity and protection by (+)-catechin in rat cortical astrocytes. Pharmacol. Rep. 2014, 66, 1106–1113. [Google Scholar] [CrossRef] [PubMed]
  123. Yang, L.; Wei, J.; Sheng, F.; Li, P. Attenuation of Palmitic Acid—Induced Lipotoxicity by Chlorogenic Acid through Activation of SIRT1 in Hepatocytes. Mol. Nutr. Food Res. 2019, 63, 1801432. [Google Scholar] [CrossRef] [PubMed]
  124. Xu, D.; Liu, L.; Zhao, Y.; Yang, L.; Cheng, J.; Hua, R.; Zhang, Z.; Li, Q. Melatonin protects mouse testes from palmitic acid-induced lipotoxicity by attenuating oxidative stress and DNA damage in a SIRT1-dependent manner. J. Pineal Res. 2020, 69, e12690. [Google Scholar] [CrossRef]
  125. Wu, Z.; Geng, Y.; Buist-Homan, M.; Moshage, H. Scopoletin and umbelliferone protect hepatocytes against palmitate- and bile acid-induced cell death by reducing endoplasmic reticulum stress and oxidative stress. Toxicol. Appl. Pharmacol. 2022, 436, 115858. [Google Scholar] [CrossRef] [PubMed]
  126. Nie, C.; Li, Y.; Guan, Y.; Zhang, K.; Liu, J.; Fan, M.; Qian, H.; Wang, L. Highland barley tea represses palmitic acid-induced apoptosis and mitochondrial dysfunction via regulating AMPK/SIRT3/FoxO3a in myocytes. Food Biosci. 2021, 40, 100893. [Google Scholar] [CrossRef]
  127. Yoon, H.J.; Bang, M.-H.; Kim, H.; Imm, J.-Y. Improvement of palmitate-induced insulin resistance in C2C12 skeletal muscle cells using Platycodon grandiflorum seed extracts. Food Biosci. 2018, 25, 61–67. [Google Scholar] [CrossRef]
  128. Xia, M.; Wu, Z.; Wang, J.; Buist-Homan, M.; Moshage, H. The Coumarin-Derivative Esculetin Protects against Lipotoxicity in Primary Rat Hepatocytes via Attenuating JNK-Mediated Oxidative Stress and Attenuates Free Fatty Acid-Induced Lipid Accumulation. Antioxidants 2023, 12, 1922. [Google Scholar] [CrossRef]
  129. Kang, B.-B.; Chiang, B.-H. EGCG regulation of non-insulin-responsive endosomal compartments in insulin-resistant skeletal muscle. Food Biosci. 2019, 28, 689. [Google Scholar] [CrossRef]
  130. Pan, X.; Liu, C.; Wang, X.; Zhao, M.; Zhang, Z.; Zhang, X.; Wang, C.; Song, G. Resveratrol improves palmitic acid-induced insulin resistance via the DDIT4/mTOR pathway in C2C12 cells. Mol. Med. Rep. 2023, 28, 181. [Google Scholar] [CrossRef]
  131. Tong, T.; Ren, N.; Soomi, P.; Wu, J.; Guo, N.; Kang, H.; Kim, E.; Wu, Y.; He, P.; Tu, Y.; et al. Theaflavins Improve Insulin Sensitivity through Regulating Mitochondrial Biosynthesis in Palmitic Acid-Induced HepG2 Cells. Molecules 2018, 23, 3382. [Google Scholar] [CrossRef]
  132. Turpaev, K.T. Keap1-Nrf2 signaling pathway: Mechanisms of regulation and role in protection of cells against toxicity caused by xenobiotics and electrophiles. Biochemistry 2013, 78, 111–126. [Google Scholar] [CrossRef] [PubMed]
  133. Petan, T.; Jarc, E.; Jusović, M. Lipid Droplets in Cancer: Guardians of Fat in a Stressful World. Molecules 2018, 23, 1941. [Google Scholar] [CrossRef] [PubMed]
  134. Tang, Y.; Majewska, M.; Leß, B.; Mehmeti, I.; Wollnitzke, P.; Semleit, N.; Levkau, B.; Saba, J.D.; van Echten-Deckert, G.; Gurgul-Convey, E. The fate of intracellular S1P regulates lipid droplet turnover and lipotoxicity in pancreatic beta-cells. J. Lipid Res. 2024, 65, 100587. [Google Scholar] [CrossRef] [PubMed]
  135. Jarc, E.; Petan, T. Lipid Droplets and the Management of Cellular Stress. Yale J. Biol. Med. 2019, 92, 435–452. [Google Scholar] [PubMed]
  136. Cnop, M.; Hannaert, J.C.; Hoorens, A.; Eizirik, D.L.; Pipeleers, D.G. Inverse Relationship Between Cytotoxicity of Free Fatty Acids in Pancreatic Islet Cells and Cellular Triglyceride Accumulation. Diabetes 2001, 50, 1771–1777. [Google Scholar] [CrossRef]
  137. Obaseki, E.; Adebayo, D.; Bandyopadhyay, S.; Hariri, H. Lipid droplets and fatty acid-induced lipotoxicity: In a nutshell. FEBS Lett. 2024, 598, 1207–1214. [Google Scholar] [CrossRef]
  138. Lin, P.; Zhou, D.H. Subcellular organelles: Lipid droplets and the multifunctional roles. In The Molecular Nutrition of Fats; Academic Press: Cambridge, MA, USA, 2018. [Google Scholar]
  139. de Souza, C.O.; Vannice, G.K.; Rosa Neto, J.C.; Calder, P.C. Is Palmitoleic Acid a Plausible Nonpharmacological Strategy to Prevent or Control Chronic Metabolic and Inflammatory Disorders? Mol. Nutr. Food Res. 2018, 62, 1700504. [Google Scholar] [CrossRef]
  140. Listenberger, L.L.; Han, X.; Lewis, S.E.; Cases, S.; Farese, R.V.; Ory, D.S.; Schaffer, J.E. Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc. Natl. Acad. Sci. USA 2003, 100, 3077–3082. [Google Scholar] [CrossRef]
  141. Li, N.; Sancak, Y.; Frasor, J.; Atilla-Gokcumen, G.E. A Protective Role for Triacylglycerols during Apoptosis. Biochemistry 2018, 57, 72–80. [Google Scholar] [CrossRef]
  142. Cabodevilla, A.G.; Son, N.; Goldberg, I.J. Intracellular lipase and regulation of the lipid droplet. Curr. Opin. Lipidol. 2024, 35, 85–92. [Google Scholar] [CrossRef]
  143. Nakamura, M.; Sadoshima, J. Cardiomyopathy in obesity, insulin resistance and diabetes. J. Physiol. 2020, 598, 2977–2993. [Google Scholar] [CrossRef] [PubMed]
  144. Drizyte-Miller, K.; Schott, M.B.; McNiven, M.A. Lipid Droplet Contacts With Autophagosomes, Lysosomes, and Other Degradative Vesicles. Contact 2020, 3, 1–13. [Google Scholar] [CrossRef]
  145. Danielli, M.; Perne, L.; Jarc Jovičić, E.; Petan, T. Lipid droplets and polyunsaturated fatty acid trafficking: Balancing life and death. Front. Cell Dev. Biol. 2023, 11, 1104725. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, Z.; Shrestha, R.; Yang, X.; Wu, X.; Jia, J.; Chiba, H.; Hui, S.-P. Oxidative Stress and Lipid Dysregulation in Lipid Droplets: A Connection to Chronic Kidney Disease Revealed in Human Kidney Cells. Antioxidants 2022, 11, 1387. [Google Scholar] [CrossRef] [PubMed]
  147. Magtanong, L.; Ko, P.-J.; To, M.; Cao, J.Y.; Forcina, G.C.; Tarangelo, A.; Ward, C.C.; Cho, K.; Patti, G.J.; Nomura, D.K.; et al. Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State. Cell Chem. Biol. 2019, 26, 420–432.e9. [Google Scholar] [CrossRef]
  148. Dibwe, D.F.; Kitayama, E.; Oba, S.; Takeishi, N.; Chiba, H.; Hui, S.-P. Inhibition of Lipid Accumulation and Oxidation in Hepatocytes by Bioactive Bean Extracts. Antioxidants 2024, 13, 513. [Google Scholar] [CrossRef]
  149. Granieri, M.C.; Rocca, C.; De Bartolo, A.; Nettore, I.C.; Rago, V.; Romeo, N.; Ceramella, J.; Mariconda, A.; Macchia, P.E.; Ungaro, P.; et al. Quercetin and Its Derivative Counteract Palmitate-Dependent Lipotoxicity by Inhibiting Oxidative Stress and Inflammation in Cardiomyocytes. Int. J. Environ. Res. Public Health 2023, 20, 3492. [Google Scholar] [CrossRef]
  150. Jiang, Q.; Zhang, T.; Liang, Y.; Zhang, J. Baicalein ameliorates lipid accumulation in HepG2 cells via the pregnane X receptor signaling pathway. Food Biosci. 2024, 60, 104482. [Google Scholar] [CrossRef]
  151. Chen, Q.; Tang, L.; Xin, G.; Li, S.; Ma, L.; Xu, Y.; Zhuang, M.; Xiong, Q.; Wei, Z.; Xing, Z.; et al. Oxidative stress mediated by lipid metabolism contributes to high glucose-induced senescence in retinal pigment epithelium. Free Radic. Biol. Med. 2019, 130, 48–58. [Google Scholar] [CrossRef]
  152. Zacharewicz, E.; Hesselink, M.K.C.; Schrauwen, P. Exercise counteracts lipotoxicity by improving lipid turnover and lipid droplet quality. J. Intern. Med. 2018, 284, 505–518. [Google Scholar] [CrossRef]
  153. Zhao, P.; Jin, Y.; Wu, X.; Huang, J.; Chen, L.; Tan, Y.; Yuan, H.; Wu, J.; Ren, Z. Artificial Lipid Droplets: Novel Effective Biomaterials to Protect Cells against Oxidative Stress and Lipotoxicity. Nanomaterials 2022, 12, 672. [Google Scholar] [CrossRef] [PubMed]
  154. Zadoorian, A.; Du, X.; Yang, H. Lipid droplet biogenesis and functions in health and disease. Nat. Rev. Endocrinol. 2023, 19, 443–459. [Google Scholar] [CrossRef] [PubMed]
  155. Panov, A. Perhydroxyl Radical (HO2•) as Inducer of the Isoprostane Lipid Peroxidation in Mitochondria. Mol. Biol. 2018, 52, 295–305. [Google Scholar] [CrossRef]
  156. Panov, A.; Mayorov, V.I.; Dikalov, S. Metabolic Syndrome and β-Oxidation of Long-Chain Fatty Acids in the Brain, Heart, and Kidney Mitochondria. Int. J. Mol. Sci. 2022, 23, 4047. [Google Scholar] [CrossRef] [PubMed]
  157. Rahmani-Manglano, N.E.; García-Moreno, P.J.; Pérez-Gálvez, R.; Guadix, E.M. Antioxidants location affects the oxidative stability of spray-dried microcapsules loaded with fish oil. Food Biosci. 2023, 56, 103074. [Google Scholar] [CrossRef]
  158. Xi, B.-N.; Zhang, J.-J.; Li, C.; Xu, X.; Zeng, Q.; Zhang, Y.; Chen, B.; Shen, Y. Effects of natural and synthetic antioxidants addition on the characteristic flavor and metabolites of walnut oil during oxidation. Food Biosci. 2024, 61, 104788. [Google Scholar] [CrossRef]
  159. Nyanhongo, G.S.; Sygmund, C.; Ludwig, R.; Prasetyo, E.N.; Guebitz, G.M. An antioxidant regenerating system for continuous quenching of free radicals in chronic wounds. Eur. J. Pharm. Biopharm. 2013, 83, 396–404. [Google Scholar] [CrossRef]
  160. Bayram, I.; Decker, E.A. Underlying mechanisms of synergistic antioxidant interactions during lipid oxidation. Trends Food Sci. Technol. 2023, 133, 219–230. [Google Scholar] [CrossRef]
  161. Pérez, M.; Dominguez-López, I.; Lamuela-Raventós, R.M. The Chemistry Behind the Folin—Ciocalteu Method for the Estimation of (Poly)phenol Content in Food: Total Phenolic Intake in a Mediterranean Dietary Pattern. J. Agric. Food Chem. 2023, 71, 17543–17553. [Google Scholar] [CrossRef]
  162. Platzer, M.; Kiese, S.; Tybussek, T.; Herfellner, T.; Schneider, F.; Schweiggert-Weisz, U.; Eisner, P. Radical Scavenging Mechanisms of Phenolic Compounds: A Quantitative Structure-Property Relationship (QSPR) Study. Front. Nutr. 2022, 9, 882458. [Google Scholar] [CrossRef]
  163. Fernández Sosa, E.I.; Ehman, N.; Felissia, F.E.; Chaves, M.G.; Area, M.C. Progress and potentialities in wood extractives-based materials for active food packaging applications. Food Biosci. 2024, 60, 104489. [Google Scholar] [CrossRef]
  164. da Costa Monção, É.; Grisi, C.V.B.; de Moura Fernandes, J.; Souza, P.S.; de Souza, A.L. Active packaging for lipid foods and development challenges for marketing. Food Biosci. 2022, 45, 101370. [Google Scholar] [CrossRef]
  165. Liu, R.; Mabury, S.A. Synthetic Phenolic Antioxidants: A Review of Environmental Occurrence, Fate, Human Exposure, and Toxicity. Environ. Sci. Technol. 2020, 54, 11706–11719. [Google Scholar] [CrossRef] [PubMed]
  166. Xu, X.; Liu, A.; Hu, S.; Ares, I.; Martínez-Larrañaga, M.-R.; Wang, X.; Martínez, M.; Anadón, A.; Martínez, M.-A. Synthetic phenolic antioxidants: Metabolism, hazards and mechanism of action. Food Chem. 2021, 353, 129488. [Google Scholar] [CrossRef]
  167. Chong, Y.M.; Chang, S.K.; Sia, W.C.M.; Yim, H.S. Antioxidant efficacy of mangosteen (Garcinia mangostana Linn.) peel extracts in sunflower oil during accelerated storage. Food Biosci. 2015, 12, 18–25. [Google Scholar] [CrossRef]
  168. Shahid, M.S.; Zhou, S.; Nie, W.; Wang, L.; Lv, H.; Yuan, J. Phytogenic Antioxidants Prolong n-3 Fatty Acid-Enriched Eggs’ Shelf Life by Activating the Nrf-2 Pathway through Phosphorylation of MAPK. Foods 2022, 11, 3158. [Google Scholar] [CrossRef] [PubMed]
  169. Ren, Y.; Perez, T.I.; Zuidhof, M.J.; Renema, R.A.; Wu, J. Oxidative Stability of Omega-3 Polyunsaturated Fatty Acids Enriched Eggs. J. Agric. Food Chem. 2013, 61, 11595–11602. [Google Scholar] [CrossRef] [PubMed]
  170. Larsson, K.; Harrysson, H.; Havenaar, R.; Alminger, M.; Undeland, I. Formation of malondialdehyde (MDA), 4-hydroxy-2-hexenal (HHE) and 4-hydroxy-2-nonenal (HNE) in fish and fish oil during dynamic gastrointestinal in vitro digestion. Food Funct. 2016, 7, 1176–1187. [Google Scholar] [CrossRef]
  171. Guichardant, M.; Bacot, S.; Molière, P.; Lagarde, M. Hydroxy-alkenals from the peroxidation of n-3 and n-6 fatty acids and urinary metabolites. Prostaglandins Leukot. Essent. Fat. Acids 2006, 75, 179–182. [Google Scholar] [CrossRef]
  172. Feng, R.; Zhang, H.; Ding, N.; Ma, H.; Luo, Y.; Tan, Y.; Wangtueai, S.; Hong, H. Effect of lipoxygenase-catalyzed linoleic acid oxidation and 4-hydroxy-2-nonenal on digestibility and gel properties of myofibrillar protein. Food Biosci. 2024, 61, 104882. [Google Scholar] [CrossRef]
  173. Moumtaz, S.; Percival, B.C.; Parmar, D.; Grootveld, K.L.; Jansson, P.; Grootveld, M. Toxic aldehyde generation in and food uptake from culinary oils during frying practices: Peroxidative resistance of a monounsaturate-rich algae oil. Sci. Rep. 2019, 9, 4125. [Google Scholar] [CrossRef] [PubMed]
  174. Wu, H.; Richards, M.P.; Undeland, I. Lipid oxidation and antioxidant delivery systems in muscle food. Compr. Rev. Food Sci. Food Saf. 2022, 21, 1275–1299. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PA-induced lipotoxicity and the possible mechanism involved. Excess-carbohydrate diet increases glycolysis products and the activation of SREBP and ChREBP and, therefore, transcription of lipogenesis-associated proteins. Lipogenesis produces a high concentration of palmitic acid (PA), which will be elongated and/or unsaturated to be esterified in phospholipids, altering the composition of the membrane, or in triglycerides that will accumulate as lipid droplets. On the other hand, an excess of PA involves the inhibition of ß-oxidation as well as malonyl-CoA, leading to an increase in ROS production or in the synthesis of ceramides that produce ER stress. Alterations in the lipid composition of the membrane, the excessive production of ROS and ER stress inhibit the signaling of insulin receptors and, consequently, the translocation of GLUT receptors, promoting insulin resistance and apoptosis. These processes can be inhibited with the support of the consumption of ceramide synthesis inhibitors or natural antioxidants that prevent oxidative stress. Glucose transporter (GLUT), insulin receptor (IR), acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), acetyl-CoA synthase (ACS), sterol regulatory element-binding protein (SREBP), carbohydrate response element-binding protein (ChREBP), monounsaturated fatty acids (MUFAs), palmitic acid (PA), reactive oxygen species (ROS), endoplasmic reticulum (ER), phosphorylation (P).
Figure 1. PA-induced lipotoxicity and the possible mechanism involved. Excess-carbohydrate diet increases glycolysis products and the activation of SREBP and ChREBP and, therefore, transcription of lipogenesis-associated proteins. Lipogenesis produces a high concentration of palmitic acid (PA), which will be elongated and/or unsaturated to be esterified in phospholipids, altering the composition of the membrane, or in triglycerides that will accumulate as lipid droplets. On the other hand, an excess of PA involves the inhibition of ß-oxidation as well as malonyl-CoA, leading to an increase in ROS production or in the synthesis of ceramides that produce ER stress. Alterations in the lipid composition of the membrane, the excessive production of ROS and ER stress inhibit the signaling of insulin receptors and, consequently, the translocation of GLUT receptors, promoting insulin resistance and apoptosis. These processes can be inhibited with the support of the consumption of ceramide synthesis inhibitors or natural antioxidants that prevent oxidative stress. Glucose transporter (GLUT), insulin receptor (IR), acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), acetyl-CoA synthase (ACS), sterol regulatory element-binding protein (SREBP), carbohydrate response element-binding protein (ChREBP), monounsaturated fatty acids (MUFAs), palmitic acid (PA), reactive oxygen species (ROS), endoplasmic reticulum (ER), phosphorylation (P).
Ijms 26 00788 g001
Figure 2. Chemical structure of synthetic and natural antioxidants used as lipid protectors. The increased number of hydroxyl groups, the substitution of hydroxy by methoxy groups and the presence of allyl carboxylic acid in the molecule strongly increase their reductions and antioxidants.
Figure 2. Chemical structure of synthetic and natural antioxidants used as lipid protectors. The increased number of hydroxyl groups, the substitution of hydroxy by methoxy groups and the presence of allyl carboxylic acid in the molecule strongly increase their reductions and antioxidants.
Ijms 26 00788 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

Ceja-Galicia, Z.A.; Cespedes-Acuña, C.L.A.; El-Hafidi, M. Protection Strategies Against Palmitic Acid-Induced Lipotoxicity in Metabolic Syndrome and Related Diseases. Int. J. Mol. Sci. 2025, 26, 788. https://doi.org/10.3390/ijms26020788

AMA Style

Ceja-Galicia ZA, Cespedes-Acuña CLA, El-Hafidi M. Protection Strategies Against Palmitic Acid-Induced Lipotoxicity in Metabolic Syndrome and Related Diseases. International Journal of Molecular Sciences. 2025; 26(2):788. https://doi.org/10.3390/ijms26020788

Chicago/Turabian Style

Ceja-Galicia, Zeltzin Alejandra, Carlos Leonardo Armando Cespedes-Acuña, and Mohammed El-Hafidi. 2025. "Protection Strategies Against Palmitic Acid-Induced Lipotoxicity in Metabolic Syndrome and Related Diseases" International Journal of Molecular Sciences 26, no. 2: 788. https://doi.org/10.3390/ijms26020788

APA Style

Ceja-Galicia, Z. A., Cespedes-Acuña, C. L. A., & El-Hafidi, M. (2025). Protection Strategies Against Palmitic Acid-Induced Lipotoxicity in Metabolic Syndrome and Related Diseases. International Journal of Molecular Sciences, 26(2), 788. https://doi.org/10.3390/ijms26020788

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