Open access peer-reviewed chapter

Lipid Peroxidation as a Link between Unhealthy Diets and the Metabolic Syndrome

Written By

Arnold N. Onyango

Submitted: 14 April 2021 Reviewed: 28 April 2021 Published: 20 May 2021

DOI: 10.5772/intechopen.98183

From the Edited Volume

Accenting Lipid Peroxidation

Edited by Pınar Atukeren

Chapter metrics overview

393 Chapter Downloads

View Full Metrics

Abstract

Unhealthy diets, such as those high in saturated fat and sugar accelerate the development of non-communicable diseases. The metabolic syndrome is a conglomeration of disorders such as abdominal obesity, hypertension, impaired glucose regulation and dyslipidemia, which increases the risk for diabetes and cardiovascular disease. The prevalence of the metabolic syndrome is increasing globally, and dietary interventions may help to reverse this trend. A good understanding of its pathophysiological mechanisms is needed for the proper design of such interventions. This chapter discusses how lipid peroxidation is associated with the development of this syndrome, mainly through the formation of bioactive aldehydes, such as 4-hydroxy-2-nonenal, malondialdehyde, acrolein and glyoxal, which modify biomolecules to induce cellular dysfunction, including the enhancement of oxidative stress and inflammatory signaling. It gives a current understanding of the mechanisms of formation of these aldehydes and how dietary components such as saturated fatty acids promote oxidative stress, leading to lipid oxidation. It also outlines mechanisms, apart from free radical scavenging and singlet oxygen quenching, by which various dietary constituents prevent oxidative stress and lipid oxidation in vivo.

Keywords

  • Oxidative stress
  • lipid peroxidation
  • insulin resistance; metabolic syndrome

1. Introduction

The metabolic syndrome (MS) refers to the occurrence in an individual of multiple physiological disorders related to obesity, hypertension, dysregulated blood glucose and dysregulated blood lipids, and is a risk factor for diabetes and cardiovascular disease [1]. It has been defined more specifically, and in slightly different ways by the National Cholesterol Education Program (NCEP) Adult Treatment Panel III, and by the World Health Organization (WHO). According to the former, MS is characterized by at least three of the following five clinical or biochemical abnormalities: abdominal obesity, arterial hypertension, elevated fasting blood glucose, high plasma triglycerides, and reduced high density lipoprotein cholesterol (HDL-c) [2]. On the other hand, WHO defined it as the occurrence of impaired glucose tolerance or impaired fasting glucose or diabetes and any two of the following: hypertension; elevated trigycerides or low HDL-c; abdominal obesity or obesity as determined by BMI; or microalbuminaria [1].

A proper understanding of the etiology of MS is necessary for its prevention and treatment. This chapter focuses on the role of lipid peroxidation in this pathophysiological process. It begins with a current understanding of the mechanisms of lipid oxidation, with emphasis on the formation of highly reactive lipid oxidation products such as 4-hydroxy-2-nonenal, malondialdehyde, acrolein and glyoxal. This is followed by a discussion of how these aldehydes and other lipid oxidation products contribute to the different MS components. The role of major dietary components in the initiation of oxidative stress and lipid oxidation, as well as mechanisms by which specific dietary components inhibit such undesirable events are also discussed.

Advertisement

2. Mechanisms of lipid peroxidation (LPO) and the formation of bioactive lipid oxidation products

In cells, extensive lipid oxidation and the accumulation of lipid oxidation products occurs under conditions of oxidative stress, when the concentrations of reactive oxygen species (ROS) such as superoxide anions, hydrogen peroxide and singlet oxygen increase, and are not matched by an increase in the cellular antioxidant capacity [3]. Electron leakage from the mitochondrial electron transport chain, or the actions of enzymes such as NADPH oxidases and xanthine oxidase generate superoxide anions (·- O2), which are converted by superoxide dismutase to hydrogen peroxide (H2O2), which may be converted by ferrous ions (Fe 2+) to hydroxyl radicals (·OH) according to the Fenton reaction Eq. (1). Superoxide anion also reacts with nitric oxide (NO), formed by nitric oxide synthases, to form the highly reactive peroxynitrite anion (-OONO), which reacts with H2O2 to form singlet oxygen according to Eq. (2), and this is only one of many possible mechanisms of formation of singlet oxygen in biological systems [4, 5, 6].

HOOH+Fe2+Fe3++OH+HO·E1
ONOO+HOOHO21+ONO+H2OE2

Lipid peroxidation involves a reaction between unsaturated lipids and oxygen. This may be enzyme-catalysed or non-enzymatic. Non-enzymatic lipid oxidation is either mediated by singlet oxygen, or it may involve free radical oxidation [7]. Singlet oxygen reacts by electrophilic addition to any of the double bonds in an unsaturated fatty acid such as linoleic acid (LA) to form hydroperoxide isomers such as the 10-, 12- and 13-LA hydroperoxides (10-LA-OOH, 12-LA-OOH and 13-LAOOH) as shown in Figure 1.

Figure 1.

Formation of different hydroperoxide isomers by the singlet oxygen-mediated oxidation of linoleic acid.

On the other hand, free radical oxidation begins by the abstraction of a hydrogen atom from a fatty acid, for example by the hydroxyl radical, to form a carbon centred radical, which rearranges to form a relatively stable conjugated radical (Figure 2). The latter reacts with oxygen to form a peroxyl radical, which abstracts a hydrogen from another fatty acid molecule to form a hydroperoxide and a new carbon centred radical, hence establishing a free radical chain reaction (Figure 2).

Figure 2.

Free radical peroxidation of linoleic acid showing the formation of one of the two most readily formed hydroperoxides, the 9-hydroperoxide. The other easily formed hydroperoxide is the 13-hydroperoxide (shown in Figure 1).

A fatty acid hydroperoxide can be converted to an alkoxyl radical by Fe2+ (Figure 3), in analogy to the conversion of H2O2 to the hydroxyl radical according to Eq. (1). The alkoxyl radical can be converted to a number of non-aldehydic products, including a hydroxy acid and a keto-acid, or it can cyclize to form an epoxy-allylic radical whose further oxygenation affords a hydroperoxy-epoxide (not shown) that can further be converted to various products including epoxy-keto-acids, such as 12,13-epoxy-9-keto-10E-octadecenoic acid (Figure 3), which contributes to hypertension as discussed in Section 3.2.

Figure 3.

Conversion of the 13-hydroperoxide of linoleic acid (13-LA-OOH) via the corresponding alkoxyl radical to different types of non-aldehydic products.

An alkoxyl radical can also undergo beta scission (C-C cleavage) to form an aldehyde and a carbon centred radical, and this is only facile if the latter is a resonance stabilized allylic radical, such as would be formed from the 10-LA-OOH (Figure 4) or 12-LA-OOH but not 13-LA-OOH [8]. Beta scission is also facile if the carbon bearing the alkoxyl radical occurs next to another oxygen-bearing carbon [9]. Various pathways fulfilling these conditions have been proposed for the formation of the major bioactive lipid-derived aldehydes such as MDA, HNE, acrolein and glyoxal [9, 10].

Figure 4.

b-Scission of an alkoxy radical to form an aldehyde (2-heptenal) and an allylic radical. Scission on the other side of the alkoxyl radical to form a vinyl radical and 12-oxo-9-dodecenoic acid is energetically unfavourable.

Acrolein is mainly formed from PUFAS with more than two double bonds, such as arachidonic acid, eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA) [9]. Figure 5 shows examples of how MDA, HNE and glyoxal can all be formed from linoleic acid, the most abundant PUFA in most human tissues [3, 4, 5, 6]. It starts with the 13-LA-OOH, formed by singlet oxygen -mediated or free radical oxidation, which further reacts with singlet oxygen to form a hydroperoxy-dioxetane (addition of singlet oxygen to a conjugated double bond system forms dioxetanes rather than hydroperoxides). The dioxetane is unstable, and decomposes to form two aldehydes, namely 9-oxononanoic acid and 4-hydroperoxy-2-nonenal (4-HPNE). The former is one of the predominant products of linoleic acid oxidation, and contributes to hypertension through activating phospholipase A2 (Section 3.2). A primary amine (RNH2) such as lysine or phosphatidylethanolamine may catalyse the conversion of 4-HPNE via a dioxetanyl anion to a dioxetane whose cleavage affords MDA and hexanal (Figure 5). While it has long been known that linoleic acid is a precursor of MDA, albeit not as readily as from more highly unsaturated PUFAS, its mechanism of formation from linoleic acid remained elusive [11]. 4-HPNE can alternatively react with another singlet oxygen molecule to form a hydroperoxy-dioxetanyl aldehyde whose decomposition affords glyoxal and 2-hydroperoxy-heptanal (not shown). 4-HPNE can also be converted to an alkoxyl radical, which can abstract a hydrogen to form 4-HNE, or to an epoxy-alkyl radical which can rearrange to an ether radical whose further reaction with oxygen leads to formation of a per acetal that can decompose to MDA and hexane. The epoxy-alkyl radical can also directly react with oxygen to form a hydroperoxy-epoxide whose decomposition affords glyoxal. In the cell, glutathione peroxidase may contribute to the conversion of 4-HPNE to 4-HNE.

Figure 5.

Mechanisms of the conversion of the 13-hydroperoxide of linoleic acid (LA-OOH) to 4-HNE, MDA, glyoxal, hexanal and 9-oxo-nonanoic acid. Other pathways to these products exist but are not shown.

Advertisement

3. Lipid peroxidation products contribute to the development of the metabolic syndrome

Lipid oxidation products influence the pathogenesis of metabolic syndrome components such as obesity, hypertension, impaired fasting glucose/diabetes, and dyslipidemia, in various ways [12].

3.1 Obesity

Obesity occurs when adipocytes increase in number and/or size, coupled with increased fat storage and reduced fat oxidation. Adipose tissue (AT) is functionally classified as brown or white (BAT and WAT, respectively). BAT consists of adipocytes specialized for thermogenesis, and hence contribute to reduction of obesity; while WAT, the major type of adipose tissue in humans, has less capacity for fat oxidation, and may contribute to obesity [13]. White adipocytes can exist in or acquire a brown-like (beige or brite) phenotype with higher fat oxidation than ordinary white adipocytes, and a higher number of beige adipocytes reduces an individual’s susceptibility to obesity [13]. Expansion of WAT by differentiation of preadipocytes (hyperplasia) into mature adipocytes with adequate lipid filling and fat oxidation capacity is beneficial for safe storage of fat; but mere expansion of mature adipocytes because of excessive lipid filling and reduced fat oxidation (hypertrophy) is associated with adverse health outcomes.

A certain amount of ROS is required for proper preadipocyte and mature adipocyte physiology. However, oxidative stress and excessive autophagy may inhibit preadipocyte differentiation and promote hypertrophy of mature adipocytes (Figure 6) [14]. Likewise, brown or beige adipocytes have many mitochondria for the enhanced fat oxidation, but mitochondrial oxidative stress causes loss of the mitochondria through mitophagy, thus leading to whitening, increased lipid storage and hypertrophy (Figure 6) [15]. Adenosine 5-monophosphate kinase (AMPK), sirtuins 1 and 3, protein kinase B (akt), peroxisome proliferator activated receptor gamma and alpha (PPARγ and PPARα, respectively), and PPARγ coactivator-1α (PGC-1α) are among the proteins that reduce oxidative stress and/or promote mitochondrial biogenesis in adipocytes [16, 17]. Both protein kinase A (PKA) and akt are required for PPARγ expression [18], which is required for differentiation of both brown and white adipocytes [19]. PPARγ promotes thermogenesis in mature brown adipocytes through activation of uncoupled protein 1 (UCP-1), and by upregulating glycerol kinase which catalyses glycerol-3-phosphate synthesis, which is required for TG synthesis [20]. While this looks paradoxical, TG synthesis may help reduce the lipotoxicity and oxidative stress induction by free fatty acids (discussed in Section 4), and allow targeted, β-adrenergic signaling-associated release of fatty acids for mitochondrial oxidation. AMPK activates autophagy and induces the transcription factor nrf2; and the latter upregulates antioxidant enzymes such as catalase, glutathione peroxidase, superoxide dismutase and heme oxygenase 1 [21]. Sirt1, which is mainly localized in the nucleus, increases the expression catalase and SOD as reviewed by Iside et al. [22]. In addition, it upregulates autophagy genes, and autophagy defect associated with its inhibition promotes release of exosomes which induce toll-like receptor 4 (TLR4) signaling, downstream activation of nuclear factor kappa B (NF-kB), and NF-kB-mediated upregulation of oxidative stress and inflammation-promoting genes [23].

Figure 6.

Role of oxidative stress and lipid oxidation-induced carbonyl stress in the pathogenesis of hypertrophic obesity.

Conditions that promote adipose tissue oxidative stress, including inappropriate diets (Section 4), induce lipid oxidation, and the latter generates carbonyl stress due to formation of various aldehydes as explained in Section 2. These aldehydes, including HNE and acrolein modify and inhibit AMPK and sirt1, thus amplifying oxidative stress and their own formation (Figure 6) [24, 25, 26]. HNE also carbonylates insulin receptor substrate1/2 (IRS1), leading to degradation and inhibition of the latter, thus inducing insulin resistance and downstream akt inhibition [27]. Thus, insulin resistant obese individuals have lower akt, AMPK ad sirt1 activity, but higher reactive carbonyls and carbonylated proteins [28]. Acrolein and HNE additionally aggravate oxidative stress through readily reacting with, and depleting the antioxidant glutathione [29, 30]. They modify the endoplasmic reticulum calcium pump SERCA, leading to its inhibition and ER stress [3], which aggravates oxidative stress, insulin resistance, sirt1 inhibition, expression of the pro-inflammatory cytokines, TNFα and IL6, and adipocyte whitening [31, 32]. Glutathionylated HNE and other aldehydes released from adipocytes under conditions of oxidative stress promote macrophage infiltration into WAT, and their acquisition of a proinflammatory M1 phenotype [33]. Malondialdehyde reacts with albumin, and the MDA-albumin conjugates promote a proinflammatory phenotype in macrophages and T cells [34]. Cytokines such as interleukin1-β, released from inflammatory macrophages, in turn promote adipocyte oxidative stress and whitening [35].

3.2 Hypertension

Arterial hypertension occurs because of (i) increased renal retention of sodium and water (ii) dysregulation of vasodilators and vasoconstrictors and (iii) arterial stiffness. Obesity is a major risk factor for hypertension [36]. For example, adiponectin inhibits adrenal production of aldosterone, a potent inducer of hypertension [37], but obesity reduces adiponectin secretion and increases circulating aldosterone [38]. Thus, by promoting obesity, lipid peroxidation products indirectly promote hypertension. However, they also induce hypertension independently of obesity. For example, the non-aldehydic linoleic acid oxidation product, 12,13-epoxy-9-keto-10-Ε-octadecenoic acid (Shown in Figure 3) also promotes adrenal production of aldosterone to induce hypertension [39].

Aldosterone binds to the renal tubular epithelial cell mineralocorticoid receptor, which, as a transcription factor, upregulates the expression of the epithelial sodium channel, which promotes sodium retention [40]. Independently of gene transcription, aldosterone activates the non-receptor tyrosine kinase cSrc in these cells, probably through the angiotensin receptor type 1 (AT1R), and cSrc activates epidermal growth factor receptor (EGFR) signaling, leading to activation of the mitogen activated protein kinase Erk1/2 [40]. Erk1/2 activates Na+/K+ ATPase, which promotes sodium and water retention [41]. Aldosterone-cSrc signaling also induces oxidative stress [40], which induces formation of lipid oxidation products. 4-HNE, inhibits AMPK and sirt1, thus inhibiting eNOS, leading to reduced NO bioavailability, and this causes increased transactivation of EGFR and downstream Erk1/2 [42, 43, 44]. Thus, blood HNE levels are increased in hypertension [45], and the latter can be ameliorated by carbonyl quenching [46]. Oxidized low density lipoprotein, which contains oxidatively modified lipids such as HNE, induces oxidative stress in renal tubular endothelial cells, which activates the renal renin-angiotensin system (RAS); whose component, angiotensin 2, overstimulates sodium transporters and thus induces hypertension [47, 48]. Hypertension in turn promotes oxidative stress and LDL oxidation, thereby creating a vicious circle [49].

Inhibition of endothelial cell sirt1, sirt3 and AMPK, which can be mediated by LPO products, causes inhibition of endothelial nitric oxide synthase (eNOS) and decreases production of NO, the main arterial vasodilator [50, 51, 52]. HNE induces endothelial cell insulin resistance, and the associated akt inhibition both inhibits eNOS and upregulates the vasoconstrictor, endothelin [3, 53]. The dysfunctional endothelial cells further produce pro-inflammatory factors such as TNFα, IL-1β, IL-8 and MCP-1 which recruit circulating neutrophils, platelets and monocytes, and the latter differentiate into macrophages [53, 54]. Neutrophils, monocytes and macrophages secrete myeloperoxidase [55]. Myeloperoxidase oxidizes LDL [56]. It also promotes the activation of endothelial cyp4a12a, which catalyzes the formation of 20-hydroxy-eicosatetraenoic acid (20-HETE) from arachidonic acid [57]. 20-HETE upregulates endothelial RAS components including angiotensin 2, a potent vasoconstrictor, which also induces aldosterone secretion [58]. Both angiotensin 2 and aldosterone aggravate endothelial oxidative stress and dysfunction. Androgens promote 20-HETE synthesis, and this may explain the higher occurrence of hypertension in men than women [58].

Stiffness of the coronary artery and other major arteries inhibits their systolic dilatation, and thus promotes systolic hypertension [59]. Degradation of the elastic fiber, elastin, in the walls of the major arteries, and its replacement with collagen fibres is a hallmark of the pathogenesis of arterial stiffness [59]. The myeloperoxidase product, 20-HETE, activates matrix metalloproteinase 12 (MMP-12, macrophage elastase), which degrades elastin [60]. Myeloperoxidase additionally inhibits the elastase inhibitor, α1, and this is antagonized by sulfur compounds such as glutathione [61]. Acrolein and HNE, on the other hand, deplete glutathione [62]. 20-HETE additionally sensitizes vascular smooth muscle cells to stimuli that promote their dedifferentiation and proliferation [58], which contributes to arterial stiffening especially of the muscular arteries [63]. One of the most readily formed aldehydic linoleic acid oxidation products, 9-oxononanoic acid (Figure 3) activates phospholipase A2 (PLA2) leading to generation of eicosanoids and thromboxane A2 in blood [64]. Thromboxane A2 causes vasoconstriction and the proliferation of smooth muscle cells [65].

Malondialdehyde forms collagen crosslinks that prevent collagen degradation, thus promoting arterial stiffness [66]. Thus, MDA-modified LDL independently predicts arterial stiffness [67]. Glyoxal contributes to arterial stiffness by reacting with collagen to form advanced glycation end products such as GOLA, GOLD, GODIC and carboxymethyl lysine (CML) [68]. CML induces endothelial oxidative stress through the RAGE receptor, which activates components of NF-kB signaling that promote expression of collagen 1 and 2 [69, 70].

3.3 Dyslipidemia

Dyslipidemia in metabolic syndrome is defined by elevated circulating triglycerides (hypertriglyceridemia) or low levels of high-density lipoprotein cholesterol (low HDLc); and hepatic steatosis, a component of non-alcoholic fatty liver disease (NAFLD) is its main risk factor [71]. This is because, in hepatic steatosis, there occurs greater production and secretion of triglyceride-rich very low-density lipoproteins (VLDL), leading to hypertriglyceridemia; as well as higher hepatic lipase activity, which increases the hepatic uptake and degradation of HDL [71]. Hepatocyte oxidative stress, ER stress and associated lipid peroxidation are involved in the development of hepatic steatosis [72, 73], and this makes lipid peroxidation an important factor in the development of dyslipidemia [74].

Low HDLc also occurs in obesity independently of elevated triglycerides, indicating that it occurs even independently of NAFLD [75]. Hypoadiponectinemia, which depends on obesity rather than NAFLD [76], may cause reduced HDLc through increased hepatic lipase activity; reduced hepatic expression of the HDL protein apo A; reduced expression of the cholesterol export protein ABCA1 which transfers cholesterol to HDL; and upregulated synthesis of LCAT which transfers cholesterol from HDLc to chylomicrons [77]. Obesity is also associated with increased plasma TNFα [78] which suppresses hepatocyte apo AI gene expression via ERK and JNK [79]. HNE contributes to JNK over-activation in hepatocytes [80].

3.4 Prediabetes and diabetes

Diabetes is a state of elevated postprandial and/or fasting blood glucose that, if not controlled, leads to the damage of various organs; while prediabetes refers to an intermediate level of fasting and/or postprandial blood glucose, higher than normal but less than diabetic blood glucose levels [1]. It is an earlier stage toward the development of diabetes, but which can revert to normoglycemia. The major causes of (pre)diabetes are pancreatic alpha and beta cell dysfunctions leading to glucagon over-secretion and insulin under-secretion, respectively; coupled with skeletal muscle, adipose and/or hepatic insulin resistance [81].

Both obesity and hypertension contribute to the pathogenesis of prediabetes, hence the lipid oxidation products that induce obesity and hypertension indirectly promote diabetes thereby. Nevertheless, lipid oxidation products also directly promote (pre)diabetes. For example, malondialdehyde was found to dose-dependently reduce the insulin content in the pancreas and to contribute to beta cell death [82]. HDL prevents beta cell apoptosis and diabetes by promoting cholesterol efflux from these cells, but acrolein- or MDA-modified HDL loses this protective property [83, 84, 85]. oxLDL impairs insulin gene expression and causes death of pancreatic beta cells, through induction of oxidative stress and ER stress [86]. As already discussed, lipid oxidation products induce endothelial dysfunction. Pancreatic endothelial cell dysfunction contributes to diabetes, being associated with leukocyte recruitment and increased production of proinflammatory cytokines [87]. Cytokines such as IL-1β and TNFα induce alpha and beta cell oxidative stress [88] and associated lipid peroxidation. Insulin resistance, which can be induced by HNE and acrolein, is part of the alpha cell and beta cell dysfunctions leading to diabetes [81].

Advertisement

4. Role of dietary constituents in inducing tissue oxidative stress, lipid peroxidation and the metabolic syndrome

Diets high in saturated fatty acids, cholesterol, sugar, salt, and red meat, contribute to higher lipid oxidation in the tissues and organs that have a central role in the metabolic syndrome, such as adipose tissue, endothelial tissue, muscle, liver and pancreas.

Although saturated fatty acids do not undergo peroxidation, they contribute to the induction of oxidative stress in cells, which then leads to peroxidation of unsaturated fatty acids. For example, the most abundant saturated fatty acid in the diet, palmitic acid, is a key substrate for the first reaction in ceramide biosynthesis [89]. Ceramides induce oxidative stress, for example by inhibiting components of the electron transport chain [90].

Palmitate also induces oxidative stress and ER stress independently of ceramide. For example, it increases diacylglycerol levels, which is associated with activation of protein kinase C (PKC), which inhibits the Kreb’s cycle enzymes aconitase and isocitrate dehydrogenase [91]. Thus, the acetyl COA generated from peroxisomal and mitochondrial fatty acid beta oxidation accumulates in the cell, promoting acetylation of mammalian target of rapamycin complex (MTORC-1) and high mobility group box-1 (HMGB-1), as has been demonstrated in hepatocytes [92]. Acetylation activates MTORC-1, which inhibits akt and further promotes oxidative stress by upregulating the expression of TLR4, thus upregulating the NF-kB-NADPH oxidase/iNOS axis [92]. Acetylation of HMGB-1 causes its translocation out of the cell, enabling it to induce oxidative stress by interacting with the receptor for advanced glycation end products (RAGE) as well as TLR4, which both induce NF-kB activation [93]. Obesity is associated with increased circulating HMGB-1, which accelerates the pathogenesis of obesity, hypertension and diabetes [72, 73, 94, 95]. TLR2/4 signaling also activates RAS components including angiotensin 2, whose signaling via its receptor AT1R induces NFkB and oxidative stress [3, 96].

PUFAS undergo peroxidation during cooking as well as in the digestive tract [97]. This is more pronounced when they are part of a meal containing meat, especially red meat, which has higher myoglobin content; since iron from the latter promotes lipid oxidation [98]. This leads to a postprandial increase in circulating carbonyls such as malondialdehyde and HNE, which promote oxidative stress, HDL modification and postprandial inflammation [98, 99]. On the other hand, absorbed, unoxidized unsaturated fatty acids including MUFAs and PUFAs reduce palmitate-induced oxidative stress and lipotoxicity in many cell types by promoting the incorporation of palmitate into TGs for safe storage [100, 101, 102]. Nevertheless, high concentrations of arachidonic acid also induce deleterious effects. Thus, supplementation of arachidonic acid to a high fat diet led to enhanced obesity in mice [103], which is attributable to the fact that this n-6 fatty acid promotes adipogenesis from preadipocytes, but its cyclooxygenase-mediated oxidation products, prostaglandins E2 and F2a (PGE2 and PGF2a) inhibit browning via ERK activation and associated decrease in UCP-1 expression [104, 105]. These prostaglandins activate NF-kB, diminish adiponectin production, upregulate pro-inflammatory mediators such as TNFα and MCP-1, and thus promote macrophage activation [106]. They promote oxidative stress and lipid oxidation, and the lipid oxidation product HNE in turn induces cyclooxygenase 2 [107]. Adipose inflammation has systemic effects, hence adipose tissue arachidonic acid was found to be independently associated with abdominal obesity, dyslipidemia, hypertension and fasting glucose [108]. Its myeloperoxidase products, 20-HETE is associated with insulin resistance and hyperglycemia [109]. Since humans synthesize arachidonic acid from linoleic acid, the arachidonic acid content in human adipose tissues does not necessarily reflect its dietary intake [110].

Although linoleic acid is a precursor of arachidonic acid, studies of its effects on the metabolic syndrome have given mixed results, with both harmful and protective roles reported [111, 112, 113]. The differences are partly due to genetic factors. For example, there are individual and ethnic differences in the expression of fatty acid desaturase 1 and 2 (FADS 1/2); with genotypes favouring greater FADS1/2 activity and arachidonic acid synthesis being associated with greater susceptibility to metabolic dysregulation [114, 115]. Black people and Indians significantly generate arachidonic acid from dietary linoleic acid, unlike people of European origin [114, 116]. A high adipose tissue linoleic: arachidonic acid is inversely associated with cardiovascular mortality and hypertension [112]. Likewise, a low linoleic: arachidonic acid ratio in plasma phospholipids is associated with hypertension [117]. Polymorphisms in the receptor for oxLDL, Lox-1, might also determine differences in the response to increased dietary linoleic acid; since this PUFA increases Lox-1 expression in aortic endothelial cells [118]. The effects of linoleic acid may also be dependent on the overall diet. If the diet is high in other factors that induce oxidative stress such as dietary sugar and salt, the pro-oxidative environment thus created may abrogate potential linoleic acid benefits through its increased oxidation. This is in analogy to the fact that high glycemic index foods abrogate the anti-obesity effects of fish oil [119]. A high linoleic acid diet may also be unfavourable for people who have already developed some component of the metabolic syndrome and thus have a more pro-inflammatory status.

Oleic acid is the major dietary fatty acid in the Mediterranean diet, which is generally associated with health benefits. This fatty acid is relatively resistant to peroxidation. Besides promoting the safe storage of palmitate in TGs, it induces thermogenesis by upregulating adipose triglyceride lipase and hormone sensitive lipase, which induce lipolysis coupled with fatty acid oxidation [120]. It promotes M2 macrophage phenotype in visceral adipose [121].

The dietary n-3 fatty acids are generally highly susceptible to oxidation because they all contain at least 3 double bonds. Although they are not 4-HNE precursors, decomposition of their hydroperoxides very readily produces acrolein, MDA, glyoxal and methylglyoxal. Despite this, they are largely beneficial, suppressing development of the metabolic syndrome [122]. In adipocytes, their binding to the GPR120/Ffar4 receptor inhibits TLR2 and TLR4 signaling and associated NFkB activation, oxidative stress and inflammation [123]. This receptor also upregulates miR-30b and 378, and induces FGF21 secretion, whose signaling activates AMPK, promotes browning and induces adiponectin [123, 124, 125, 126]. The n-3 PUFAs are metabolized by cyclooxygenase to resolvins, protectins, maresins and isoprostanes which help in resolving inflammation [127].

A high dietary n-3: n-6 PUFA ratio has been found to be protective against the metabolic syndrome in some studies but not others [128, 129]. This might be partly due to inter-individual differences in the metabolism of n-6 fatty acids.

High carbohydrate diets promote obesity because excess sugars are stored as lipids. High sucrose or high fructose diets are particularly obesogenic [130]. Fructose metabolism robustly increases palmitate synthesis in adipocytes [131]. Moreover, fructose metabolism is associated with decreased cellular ATP, purine degradation and activation of xanthine oxidase which generates reactive oxygen species and associated lipid peroxidation [132], which is involved in adipocyte whitening and less thermogenesis. Uric acid, a product of purine degradation also induces oxidative stress through increased NADPH oxidase activity and RAS activation [131, 132, 133, 134].

High salt (sodium chloride) diets promote obesity, by salt-induced activation of adipocyte Na/K+ ATPase, which is coupled to activation of src, which generates ROS, and transactivates PI3-K-Akt–MTOR and EGFR-ERK/MAPK pathways [135, 136]. This is associated with increased expression of proinflammatory mediators such as TNFα, MCP-1, COX-2, IL-17A, IL-6, leptin, and leptin [136, 137]. Sodium chloride also activates Na+/K+ ATPase and induces oxidative stress in endothelial cells and renal tubular epithelial cells, thereby promoting hypertension [132], and this is also subject to genetic susceptibility [138].

High dietary cholesterol is associated with a high risk of dyslipidemia [139]. Cholesterol-rich chylomicron remnants mainly deliver their cholesterol to the liver, and cholesterol accumulation in hepatocytes strongly induces oxidative stress, by modification of the mitochondrial membrane and limiting import of glutathione into the mitochondria, as well by inducing ER stress and proinflammatory cytokines [140].

The lipopolysaccharide (LPS) component of the walls of gram-negative bacteria is a pro-inflammatory molecule that contributes to metabolic low-grade inflammation (endotoxemia), by signaling through TLR2 and 4 in various cell types, leading to NFkB activation and release of pro-inflammatory cytokines. High sucrose and high saturated fat diets promote the growth of gram-negative bacteria, and thus increase the entry of LPS into the circulation [141].

Advertisement

5. Mechanisms of the antioxidant and metabolic syndrome-suppressing effects of dietary factors

While some food components promote a pro-oxidative and pro-inflammatory state as discussed in the previous section, other dietary factors inhibit oxidative stress and inflammation. They do this through various mechanisms, but the most widely considered mechanisms are those associated with lipid oxidation, including scavenging of free radicals such as peroxyl radicals and alkoxyl radicals, chelation of metal ions that participate in formation of such radicals, and singlet oxygen quenching.

5.1 Free radical scavenging and singlet oxygen quenching

Carotenoids, phenolic substances, tocopherols and ascorbic acid are well known for their antioxidant activities targeting the neutralization of reactive radicals and/or singlet oxygen quenching. Thus, carotenoids reduce oxidative stress and lipid oxidation, resulting in adipocyte beiging and obesity prevention [142]. There is decreased adipose beta carotene in obese subjects, and this was suggested to at least partly be due to their depletion under the high ROS environment [142]. Likewise, tocopherols and tocotrienols have been shown to be protective against all components of the metabolic syndrome [143]. Thus, the high tocotrienol content of palm oil may reduce its potential harm from the high palmitate content [144]. Unfortunately, radical scavenging antioxidants also exhibit pro-oxidant activity, depending on their concentrations and the level of prooxidative factors [145]. Hence, there is need to consider a broad range of dietary factors that prevent oxidative by alternative mechanisms, such as those outlined hereafter. A single molecule can act by multiple mechanisms, and the more mechanisms involved, the greater might be the benefit.

5.2 Insulin-mimicking

Insulin signaling activates akt, which reduces oxidative stress by promoting mitophagy and by activating nrf2 to induce antioxidant enzymes [81, 146]. Moreover, nrf2, via heme oxygenase 1 (HO-1), inhibits NFkB and associated upregulation of NADPH oxidase and iNOS [147]. Quercetin and ferulic acid are examples of molecules that have demonstrated oxidative stress and metabolic syndrome amelioration at least partly through PI3K-akt signaling in various cell types [148, 149, 150]. Resveratrol and ferulic acid inhibit LPS- and oxidative stress-induced intestinal barrier injury through this signaling pathway [151, 152].

5.3 AMPK and SIRT1 activation

AMPK and/or sirt1 reduce mitochondrial oxidative stress in adipocytes, pancreatic beta cells, hepatocytes, endothelial cells, and thus are useful in preventing all aspects of the metabolic syndrome. In addition to insulin mimicking, quercetin and ferulic acid, also activate these proteins [153, 154, 155].

5.4 Adiponectin and adiponectin receptor enhancement

Compounds that activate AMPK, sirt1 and/or PI3K-akt in adipose tissues limit adipocyte hypertrophy and inflammation, and enhance adiponectin production. Adiponectin has systemic effects in reducing insulin resistance and oxidative stress, because it activates both PI3K-akt and AMPK in insulin target tissues, and also promotes anti-inflammatory polarization of macrophages [156]. Dietary compounds than ameliorate metabolic syndrome through enhanced adiponectin secretion and/or upregulating adiponectin receptor include n3-fatty acids, sesamin, the citrus derived polymethoxyflavonoids nobiletin and tangeretin, quercetin and resveratrol [157, 158, 159, 160].

5.5 Ceramide reduction

Adiponectin signaling increases ceramidase activity, thus reducing ceramide levels [161]. Hence, the adiponectin and adiponectin receptor enhancers should contribute to reducing ceramide-induced oxidative stress. Not much research has been done along this line, but it has been reported that DHA inhibits ceramide biosynthesis [162]. In mice, dietary inulin reduces ceramide synthesis by suppressing neutral sphingomyelinase expression and activity [163].

5.6 Vasodilation

Vasodilation reduces blood pressure, and thus reduces pressure-dependent oxidative stress as well as LDL oxidation and Lox-1 dependent oxidative stress [164]. Thus, for people with prehypertension or hypertension, vasodilation may be a major strategy for reducing oxidative stress and lipid oxidation. Cinnamaldehyde has vasodilatory and antihypertensive activity through effects on smooth muscle contractility [165]. Dietary nitrate achieves vasodilation through NO release, and this is associated not only with pressure regulation, but also other components of the metabolic syndrome including blood glucose and lipid improvement [166]. Adiponectin induces AMPK dependent eNOS activation in endothelial cells, hence adiponectin enhancers such as imperatorin also promote NO synthesis and vasodilatation [167].

5.7 Reactive carbonyl, ALEs and AGEs scavenging

Scavengers of reactive carbonyls such as HNE, acrolein and MDA have been demonstrated to ameliorate oxidative stress, lipid peroxidation and the metabolic syndrome. Examples of compounds with such effects include carnosine, carnosinol, epigallocatechin-3-gallate and the mulberry anthocyanins cyanidin 3-glucoside (C3G) and cyanidin 3-rutinoside (C3R) [46, 168, 169, 170]. Aminoguanidine attenuates hypertension by scavenging AGES [171].

5.8 Gut microbiota modulation

Probiotic microorganisms suppress the growth of pathogenic microorganisms. They also produce metabolites such as short chain fatty acids with beneficial effects on the metabolic syndrome. For example, butyrate promotes PI3K-akt signaling to prevent oxidative stress and maintain intestinal barrier integrity [172, 173]. Quercetin, resveratrol and n-3 fatty acids have been demonstrated to positively influence gut microbiota and decrease intestinal barrier permeability in animal studies [153, 174].

Advertisement

6. Conclusions

Lipid peroxidation is a major contributor to the pathogenesis of the metabolic syndrome, especially through highly reactive and bioactive aldehydes such as acrolein, 4-hydroxy-2-nonenal, malondialdehyde and glyoxal. Mechanisms of formation of these products are now well-understood. For example, this article has highlighted that formation of MDA from linoleic acid may be easier than previously thought. The mentioned aldehydes propagate oxidative stress and inflammation by inducing insulin resistance, inhibiting sirt1 and AMPK, reducing adiponectin secretion, as well as forming AGEs and ALEs that activate the RAGE receptor. Inhibiting LPO and the LPO product-associated oxidative stress and inflammation is necessary for preventing and/or ameliorating progression of the metabolic syndrome. This may not be effectively accomplished by dietary agents that merely scavenge free radicals and/or quench singlet oxygen, but also by those that inhibit the signaling pathways that generate non-lipid ROS, or scavenge the reactive carbonyls, ALEs and AGEs. In addition, saturated fat, sugar, meat, and salt, that fuel the signaling pathways that initiate LPO should be reduced. The metabolic influence of some dietary components such as salt and n-6 PUFAs is particularly influenced by genetics, and this should be duly considered when making dietary recommendations.

References

  1. 1. Alberti, K. G. M.M, Zimmet, P.Z. for the WHO Consultation (1998). Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus. Provisional report of a WHO Consultation. Diabetic Medicine 1998; 15: 539-553
  2. 2. National Cholesterol Education Program (NCEP): Expert Panel on Detection and Treatment of High Blood Cholesterol in Adults (2002). Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation 106, 3143-3421
  3. 3. Onyango, A. N. (2018). Cellular stresses and stress responses in the pathogenesis of insulin resistance. Oxidative Medicine and Cellular Longevity, Article ID 4321714.
  4. 4. Onyango, A. N. (2016). Endogenous generation of singlet oxygen and ozone in human and animal tissues: mechanisms, biological significance, and influence of dietary components. Oxidative Medicine and Cellular Longevity, 2016.
  5. 5. Onyango, A. N. (2016). Formation of aldehydic phosphatidylcholines during the anaerobic decomposition of a phosphatidylcholine bearing the 9-hydroperoxide of linoleic acid. BioMed Research International, Article ID 8218439.
  6. 6. Onyango, A. N. (2017). The contribution of singlet oxygen to insulin resistance. Oxidative Medicine and Cellular Longevity, Article ID 8765972.
  7. 7. Girotti, A. W. (1985). Mechanisms of lipid peroxidation. Journal of Free Radicals in Biology & Medicine, 1(2), 87-95.
  8. 8. Onyango, A. N., Inoue, T., Nakajima, S., Baba, N., Kaneko, T., Matsuo, M., & Shimizu, S. (2001). Synthesis and stability of phosphatidylcholines bearing polyenoic acid hydroperoxides at the sn-2 position. Angewandte Chemie International Edition, 40(9), 1755-1757.
  9. 9. Onyango, A. N. (2012). Small reactive carbonyl compounds as tissue lipid oxidation products; and the mechanisms of their formation thereby. Chemistry and physics of lipids, 165(7), 777-786.
  10. 10. Schneider, C., Porter, N. A., & Brash, A. R. (2008). Routes to 4-hydroxynonenal: fundamental issues in the mechanisms of lipid peroxidation. Journal of Biological Chemistry, 283(23), 15539-15543.
  11. 11. Onyango, A. N., & Baba, N. (2010). New hypotheses on the pathways of formation of malondialdehyde and isofurans. Free Radical Biology and Medicine, 49(10), 1594-1600.
  12. 12. Pillon, N. J., & Soulage, C. O. (2012). Lipid peroxidation by-products and the metabolic syndrome. Lipid peroxidation, 409-436.
  13. 13. Zoico, E., Rubele, S., De Caro, A., Nori, N., Mazzali, G., Fantin, F., ... & Zamboni, M. (2019). Brown and beige adipose tissue and aging. Frontiers in endocrinology, 10, 368.
  14. 14. Castro, J. P., Grune, T., & Speckmann, B. (2016). The two faces of reactive oxygen species (ROS) in adipocyte function and dysfunction. Biological Chemistry, 397(8), 709-724.
  15. 15. Cedikova, M., Kripnerová, M., Dvorakova, J., Pitule, P., Grundmanova, M., Babuska, V., ... & Kuncova, J. (2016). Mitochondria in white, brown, and beige adipocytes. Stem cells international, 2016.
  16. 16. Andrade, J. M. O., Barcala-Jorge, A. S., Batista-Jorge, G. C., Paraíso, A. F., de Freitas, K. M., de Farias Lelis, D., ... & Santos, S. H. S. (2019). Effect of resveratrol on expression of genes involved thermogenesis in mice and humans. Biomedicine & Pharmacotherapy, 112, 108634.
  17. 17. Bargut, T. C. L., Souza-Mello, V., Aguila, M. B., & Mandarim-de-Lacerda, C. A. (2017). Browning of white adipose tissue: lessons from experimental models. Hormone molecular biology and clinical investigation, 31(1).
  18. 18. Kim, S. P., Ha, J. M., Yun, S. J., Kim, E. K., Chung, S. W., Hong, K. W., ... & Bae, S. S. (2010). Transcriptional activation of peroxisome proliferator-activated receptor-γ requires activation of both protein kinase A and Akt during adipocyte differentiation. Biochemical and Biophysical Research Communications, 399(1), 55-59.
  19. 19. Sanchez-Gurmaches, J., Calejman, C. M., Jung, S. M., Li, H., & Guertin, D. A. (2019). Brown fat organogenesis and maintenance requires AKT1 and AKT2. Molecular Metabolism, 23, 60-74.
  20. 20. Lasar, D., Rosenwald, M., Kiehlmann, E., Balaz, M., Tall, B., Opitz, L., ... & Wolfrum, C. (2018). Peroxisome proliferator activated receptor gamma controls mature brown adipocyte inducibility through glycerol kinase. Cell Reports, 22(3), 760-773.
  21. 21. Xu, Q., Fan, Y., Loor, J. J., Liang, Y., Sun, X., Jia, H., ... & Xu, C. (2021). Adenosine 5′-monophosphate-activated protein kinase ameliorates bovine adipocyte oxidative stress by inducing antioxidant responses and autophagy. Journal of Dairy Science.
  22. 22. Iside, C., Scafuro, M., Nebbioso, A., & Altucci, L. (2020). SIRT1 activation by natural phytochemicals: an overview. Frontiers in Pharmacology, 11, 1225.
  23. 23. Li, F., Li, H., Jin, X., Zhang, Y., Kang, X., Zhang, Z., ... & Sun, H. (2019). Adipose-specific knockdown of Sirt1 results in obesity and insulin resistance by promoting exosomes release. Cell Cycle, 18(17), 2067-2082.
  24. 24. Lu, Q., Mundy, M., Chambers, E., Lange, T., Newton, J., Borgas, D., ... & Rounds, S. (2017). Alda-1 protects against acrolein-induced acute lung injury and endothelial barrier dysfunction. American Journal of Respiratory Cell and Molecular Biology, 57(6), 662-673.
  25. 25. Shearn, C. T., Backos, D. S., Orlicky, D. J., Smathers-McCullough, R. L., & Petersen, D. R. (2014). Identification of 5′ AMP-activated kinase as a target of reactive aldehydes during chronic ingestion of high concentrations of ethanol. Journal of Biological Chemistry, 289(22), 15449-15462.
  26. 26. Zhang, X., Wang, Z., Li, J., Gu, D., Li, S., Shen, C., & Song, Z. (2013). Increased 4-hydroxynonenal formation contributes to obesity-related lipolytic activation in adipocytes. PLoS One, 8(8), e70663.
  27. 27. D. Demozay, J. C. Mas, S. Rocchi, E. Van Obberghen, FALDH reverses the deleterious action of oxidative stress induced by lipid peroxidation product 4-hydroxynonenal on insulin signaling in 3T3-L1 adipocytes. Diabetes 57, 1216-1226 (2008).
  28. 28. Xu, X. J., Gauthier, M. S., Hess, D. T., Apovian, C. M., Cacicedo, J. M., Gokce, N., ... & Ruderman, N. B. (2012). Insulin sensitive and resistant obesity in humans: AMPK activity, oxidative stress, and depot-specific changes in gene expression in adipose tissue. Journal of lipid research, 53(4), 792-801.
  29. 29. Kehrer, J. P., & Biswal, S. S. (2000). The molecular effects of acrolein. Toxicological Sciences, 57(1), 6-15.
  30. 30. Pillon, N. J., Croze, M. L., Vella, R. E., Soulère, L., Lagarde, M., & Soulage, C. O. (2012). The lipid peroxidation by-product 4-hydroxy-2-nonenal (4-HNE) induces insulin resistance in skeletal muscle through both carbonyl and oxidative stress. Endocrinology, 153(5), 2099-2111.
  31. 31. Kawasaki, N., Asada, R., Saito, A., Kanemoto, S., & Imaizumi, K. (2012). Obesity-induced endoplasmic reticulum stress causes chronic inflammation in adipose tissue. Scientific reports, 2(1), 1-7.
  32. 32. Lee, J. M., Park, S., Lee, D., Ginting, R. P., Lee, M. R., Lee, M. W., & Han, J. (2021). Reduction in endoplasmic reticulum stress activates beige adipocytes differentiation and alleviates high fat diet-induced metabolic phenotypes. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, 1867(5), 166099.
  33. 33. Frohnert, B. I., Long, E. K., Hahn, W. S., & Bernlohr, D. A. (2014). Glutathionylated lipid aldehydes are products of adipocyte oxidative stress and activators of macrophage inflammation. Diabetes, 63(1), 89-100.
  34. 34. Rahman, M., Steuer, J., Gillgren, P., Végvári, Á., Liu, A., & Frostegård, J. (2019). Malondialdehyde conjugated with albumin induces pro-Inflammatory activation of T cells isolated from human atherosclerotic plaques both directly and via dendritic cell–mediated mechanism. JACC: Basic to Translational Science, 4(4), 480-494.
  35. 35. Keuper, M. (2019). On the role of macrophages in the control of adipocyte energy metabolism. Endocrine Connections, 8(6), R105-R121.
  36. 36. Jiang, S. Z., Lu, W., Zong, X. F., Ruan, H. Y., & Liu, Y. (2016). Obesity and hypertension. Experimental and therapeutic medicine, 12(4), 2395-2399.
  37. 37. Kargi, A. Y., & Iacobellis, G. (2014). Adipose tissue and adrenal glands: novel pathophysiological mechanisms and clinical applications. International Journal of Endocrinology, 2014.
  38. 38. Sarzani, R., Guerra, F., Mancinelli, L., Buglioni, A., Franchi, E., & Dessì-Fulgheri, P. (2012). Plasma aldosterone is increased in class 2 and 3 obese essential hypertensive patients despite drug treatment. American Journal of Hypertension, 25, 818-826
  39. 39. Goodfriend, T. L., Ball, D. L., Egan, B. M., Campbell, W. B., & Nithipatikom, K. (2004). Epoxy-keto derivative of linoleic acid stimulates aldosterone secretion. Hypertension, 43(2), 358-363.
  40. 40. Cannavo, A., Bencivenga, L., Liccardo, D., Elia, A., Marzano, F., Gambino, G., ... & Paolocci, N. (2018). Aldosterone and mineralocorticoid receptor system in cardiovascular physiology and pathophysiology. Oxidative medicine and cellular longevity, Article ID 1204598.
  41. 41. Zhong, Z., Kotova, O., Davidescu, A., Ehren, I., Ekberg, K., Jörnvall, H., ... & Chibalin, A. V. (2004). C-peptide stimulates Na+, K+-ATPase via activation of ERK1/2 MAP kinases in human renal tubular cells. Cellular and Molecular Life Sciences CMLS, 61(21), 2782-2790.
  42. 42. Beltowski, J., Wójcicka, G., Jamroz-Wiśniewska, A., Borkowska, E., & Marciniak, A. (2005). Antioxidant treatment normalizes nitric oxide production, renal sodium handling and blood pressure in experimental hyperleptinemia. Life sciences, 77(15), 1855-1868.
  43. 43. Beltowski, J. (2010). Leptin and the regulation of renal sodium handling and renal Na+−transporting ATPases: role in the pathogenesis of arterial hypertension. Current cardiology reviews, 6(1), 31-40.
  44. 44. Packer, M. (2020). Interplay of adenosine monophosphate-activated protein kinase/sirtuin-1 activation and sodium influx inhibition mediates the renal benefits of sodium-glucose co-transporter-2 inhibitors in type 2 diabetes: A novel conceptual framework. Diabetes, Obesity and Metabolism, 22(5), 734-742.
  45. 45. Asselin, C., Bouchard, B., Tardif, J. C., & Des Rosiers, C. (2006). Circulating 4-hydroxynonenal–protein thioether adducts assessed by gas chromatography–mass spectrometry are increased with disease progression and aging in spontaneously hypertensive rats. Free Radical Biology and Medicine, 41(1), 97-105.
  46. 46. Aldini, G., Orioli, M., Rossoni, G., Savi, F., Braidotti, P., Vistoli, G., ... & Carini, M. (2011). The carbonyl scavenger carnosine ameliorates dyslipidaemia and renal function in Zucker obese rats. Journal of cellular and molecular medicine, 15(6), 1339-1354.
  47. 47. Banday, A. A., & Lokhandwala, M. F. (2008). Oxidative stress-induced renal angiotensin AT1 receptor upregulation causes increased stimulation of sodium transporters and hypertension. American Journal of Physiology-Renal Physiology, 295(3), F698-F706.
  48. 48. Luo, P., Yan, M., Frohlich, E. D., Mehta, J. L., & Hu, C. (2011). Novel concepts in the genesis of hypertension: role of LOX-1. Cardiovascular drugs and therapy, 25(5), 441.
  49. 49. Frostegård, J., Wu, R., Lemne, C., Thulin, T., Witztum, J. L., & de Faire, U. (2003). Circulating oxidized low-density lipoprotein is increased in hypertension. Clinical Science, 105(5), 615-620.
  50. 50. Jung, S. B., Kim, C. S., Kim, Y. R., Naqvi, A., Yamamori, T., Kumar, S., ... & Irani, K. (2013). Redox factor-1 activates endothelial SIRTUIN1 through reduction of conserved cysteine sulfhydryls in its deacetylase domain. PloS one, 8(6), e65415.
  51. 51. Xie, X., Yang, C., Cui, Q., Ma, W., Liu, J., Yao, Q., ... & Wang, N. (2019). Stachydrine Mediates Rapid Vascular Relaxation: Activation of Endothelial Nitric Oxide Synthase Involving AMP-Activated Protein Kinase and Akt Phosphorylation in Vascular Endothelial Cells. Journal of Agricultural and Food Chemistry, 67(35), 9805-9811.
  52. 52. Yang, L., Zhang, J., Xing, W., Zhang, X., Xu, J., Zhang, H., ... & Gao, F. (2016). SIRT3 deficiency induces endothelial insulin resistance and blunts endothelial-dependent vasorelaxation in mice and human with obesity. Scientific reports, 6(1), 1-12.
  53. 53. Vladykovskaya, E., Sithu, S. D., Haberzettl, P., Wickramasinghe, N. S., Merchant, M. L., Hill, B. G., ... & Srivastava, S. (2012). Lipid peroxidation product 4-hydroxy-trans-2-nonenal causes endothelial activation by inducing endoplasmic reticulum stress. Journal of Biological Chemistry, 287(14), 11398-11409.
  54. 54. Dehghani, T., & Panitch, A. (2020). Endothelial cells, neutrophils and platelets: getting to the bottom of an inflammatory triangle. Open Biology, 10(10), 200161.
  55. 55. Shaeib, F., Khan, S. N., Thakur, M., Kohan-Ghadr, H. R., Drewlo, S., Saed, G. M., ... & Abu-Soud, H. M. (2016). The impact of myeloperoxidase and activated macrophages on metaphase II mouse oocyte quality. PLoS One, 11(3), e0151160.
  56. 56. Delporte, C., Van Antwerpen, P., Vanhamme, L., Roumeguère, T., & Zouaoui Boudjeltia, K. (2013). Low-density lipoprotein modified by myeloperoxidase in inflammatory pathways and clinical studies. Mediators of inflammation, 2013.
  57. 57. Azcona, J. A., Tang, S., Jeitner, T., Guo, A. M., & Schwartzman, M. L. (2019). Novel Contribution of Myeloperoxidase-derived Hypochlorous Acid to 20-HETE Production Post-Ischemia. The FASEB Journal, 33(S1), 513-511.
  58. 58. Wu, C. C., & Schwartzman, M. L. (2011). The role of 20-HETE in androgen-mediated hypertension. Prostaglandins & other lipid mediators, 96(1-4), 45-53.
  59. 59. Ecobici, M., & Stoicescu, C. (2017). Arterial Stiffness and Hypertension–Which Comes First?. Maedica, 12(3), 184.
  60. 60. Soler, A., Hunter, I., Joseph, G., Hutcheson, R., Hutcheson, B., Yang, J., ... & Rocic, P. (2018). Elevated 20-HETE in metabolic syndrome regulates arterial stiffness and systolic hypertension via MMP12 activation. Journal of molecular and cellular cardiology, 117, 88-99.
  61. 61. Borregaard, N., Jensen, H. S., & Bjerrum, O. W. (1987). Prevention of tissue damage: Inhibition of Myeloperoxidase mediated inactivation of α 1 inhibitor by N-acetyl cysteine, Glutathione, and Methionine. Agents and Actions, 22(3), 255-260.
  62. 62. van der Toorn, M., Smit-de Vries, M. P., Slebos, D. J., de Bruin, H. G., Abello, N., van Oosterhout, A. J., ... & Kauffman, H. F. (2007). Cigarette smoke irreversibly modifies glutathione in airway epithelial cells. American Journal of Physiology-Lung Cellular and Molecular Physiology, 293(5), L1156-L1162.
  63. 63. Safar, M. E., Asmar, R., Benetos, A., Blacher, J., Boutouyrie, P., Lacolley, P., ... & French Study Group on Arterial Stiffness. (2018). Interaction between hypertension and arterial stiffness: an expert reappraisal. Hypertension, 72(4), 796-805.
  64. 64. Ren, R., Hashimoto, T., Mizuno, M., Takigawa, H., Yoshida, M., Azuma, T., & Kanazawa, K. (2013). A lipid peroxidation product 9-oxononanoic acid induces phospholipase A2 activity and thromboxane A2 production in human blood. Journal of Clinical Biochemistry and Nutrition, 52(3), 228-233.
  65. 65. Zhao, W. G., & Richardson, J. S. (1990). Prostacyclin, thromboxane A2, and hypertension. Clinical and investigative medicine. Medecine clinique et experimentale, 13(6), 343-352.
  66. 66. Slatter, D. A., Paul, R. G., Murray, M., & Bailey, A. J. (1999). Reactions of lipid-derived malondialdehyde with collagen. Journal of Biological Chemistry, 274(28), 19661-19669.
  67. 67. Kawamoto, R., Ninomiyax, D., Kusunoki, T., Kasai, Y., Ohtsuka, N., & Kumagi, T. (2016). Oxidative stress is associated with increased arterial stiffness in middle-aged and elderly community-dwelling persons. Journal of Clinical Gerontology and Geriatrics, 7(4), 136-140.
  68. 68. Sell, D. R., & Monnier, V. M. (2012). Molecular basis of arterial stiffening: role of glycation–a mini-review. Gerontology, 58(3), 227-237.
  69. 69. Grossin, N., Auger, F., Niquet-Leridon, C., Durieux, N., Montaigne, D., Schmidt, A. M., ... & Boulanger, E. (2015). Dietary CML-enriched protein induces functional arterial aging in a RAGE-dependent manner in mice. Molecular nutrition & food research, 59(5), 927-938.
  70. 70. Peng, Y., Kim, J. M., Park, H. S., Yang, A., Islam, C., Lakatta, E. G., & Lin, L. (2016). AGE-RAGE signal generates a specific NF-κB RelA “barcode” that directs collagen I expression. Scientific reports, 6(1), 1-10.
  71. 71. Han, J. M., Kim, H. I., Lee, Y. J., Lee, J. W., Kim, K. M., & Bae, J. C. (2019). Differing associations between fatty liver and dyslipidemia according to the degree of hepatic steatosis in Korea. Journal of Lipid and Atherosclerosis 8, 258-266.
  72. 72. Chen, Z., Tian, R., She, Z., Cai, J., & Li, H. (2020). Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radical Biology and Medicine, 152, 116-141.
  73. 73. Chen, L., Zhu, H., Su, S., Harshfield, G., Sullivan, J., Webb, C., ... & Dong, Y. (2020). High-mobility group box-1 is associated with obesity, inflammation, and subclinical cardiovascular risk among young adults: a longitudinal cohort study. Arteriosclerosis, thrombosis, and vascular biology, 40(11), 2776-2784.
  74. 74. Morita, M., Ishida, N., Uchiyama, K., Yamaguchi, K., Itoh, Y., Shichiri, M., ... & Niki, E. (2012). Fatty liver induced by free radicals and lipid peroxidation. Free radical research, 46(6), 758-765.
  75. 75. Yu, S. S., Castillo, D. C., Courville, A. B., & Sumner, A. E. (2012). The triglyceride paradox in people of African descent. Metabolic syndrome and related disorders, 10(2), 77-82.
  76. 76. Kovacova, Z., Tencerova, M., Roussel, B., Wedellova, Z., Rossmeislova, L., Langin, D., ... & Stich, V. (2012). The impact of obesity on secretion of adiponectin multimeric isoforms differs in visceral and subcutaneous adipose tissue. International Journal of Obesity, 36(10), 1360-1365.
  77. 77. Clarenbach, J. J., Vega, G. L., Adams-Huet, B., Considine, R. V., Ricks, M., & Sumner, A. E. (2007). Variability in postheparin hepatic lipase activity is associated with plasma adiponectin levels in African Americans. Journal of Investigative Medicine, 55(4), 187-194.
  78. 78. Marfella, R., Grella, R., Rizzo, M. R., Barbieri, M., Grella, R., Ferraraccio, F., ... & Nicoletti, G. (2009). Role of subcutaneous abdominal fat on cardiac function and proinflammatory cytokines in premenopausal obese women. Annals of plastic surgery, 63(5), 490-495.
  79. 79. Haas, M. J., & Mooradian, A. D. (2010). Regulation of high-density lipoprotein by inflammatory cytokines: establishing links between immune dysfunction and cardiovascular disease. Diabetes/metabolism research and reviews, 26(2), 90-99.
  80. 80. Singh, R., Wang, Y., Schattenberg, J. M., Xiang, Y., & Czaja, M. J. (2009). Chronic oxidative stress sensitizes hepatocytes to death from 4-hydroxynonenal by JNK/c-Jun overactivation. American Journal of Physiology-Gastrointestinal and Liver Physiology, 297(5), G907-G917.
  81. 81. Onyango, A. N. (2020). Mechanisms of the regulation and dysregulation of glucagon secretion. Oxidative Medicine and Cellular Longevity, Article ID 3089139
  82. 82. Rabinovitch, A. L. E. X., Suarez-Pinzon, W. L., Strynadka, K., Lakey, J. R., & Rajotte, R. V. (1996). Human pancreatic islet beta-cell destruction by cytokines involves oxygen free radicals and aldehyde production. The Journal of Clinical Endocrinology & Metabolism, 81(9), 3197-3202.
  83. 83. Femlak, M., Gluba-Brzózka, A., Ciałkowska-Rysz, A., & Rysz, J. (2017). The role and function of HDL in patients with diabetes mellitus and the related cardiovascular risk. Lipids in Health and Disease, 16(1), 1-9.
  84. 84. Shao, B., Pennathur, S., Pagani, I., Oda, M. N., Witztum, J. L., Oram, J. F., & Heinecke, J. W. (2010). Modifying apolipoprotein AI by malondialdehyde, but not by an array of other reactive carbonyls, blocks cholesterol efflux by the ABCA1 pathway. Journal of Biological Chemistry, 285(24), 18473-18484.
  85. 85. Huang, J., Yancey, P. G., Tao, H., Borja, M. S., Smith, L. E., Kon, V., ... & Linton, M. F. (2020). Reactive dicarbonyl scavenging effectively reduces MPO-mediated oxidation of HDL and restores PON1 activity. Nutrients, 12(7), 1937.
  86. 86. Plaisance, V., Brajkovic, S., Tenenbaum, M., Favre, D., Ezanno, H., Bonnefond, A., ... & Abderrahmani, A. (2016). Endoplasmic reticulum stress links oxidative stress to impaired pancreatic beta-cell function caused by human oxidized LDL. PLoS One, 11(9), e0163046.
  87. 87. Mazier, W., & Cota, D. (2017). Islet endothelial cell: friend and foe. Endocrinology, 158(2), 226-228.
  88. 88. Kowluru, A. (2020). Oxidative Stress in Cytokine-Induced Dysfunction of the Pancreatic Beta Cell: Known Knowns and Known Unknowns. Metabolites, 10(12), 480.
  89. 89. Tran, T. T. T., Postal, B. G., Demignot, S., Ribeiro, A., Osinski, C., de Barros, J. P. P., ... & Carrière, V. (2016). Short term palmitate supply impairs intestinal insulin signaling via ceramide production. Journal of Biological Chemistry, 291(31), 16328-16338.
  90. 90. Fucho, R., Casals, N., Serra, D., & Herrero, L. (2017). Ceramides and mitochondrial fatty acid oxidation in obesity. The FASEB Journal, 31(4), 1263-1272.
  91. 91. Haffar, T., Akoumi, A., & Bousette, N. (2016). Lipotoxic palmitate impairs the rate of β-oxidation and citric acid cycle flux in rat neonatal cardiomyocytes. Cellular Physiology and Biochemistry, 40(5), 969-981.
  92. 92. Wang, J., Chen, Y., Song, Q., Griffiths, A., & Song, Z. (2020). mTORC1-IRE1α pathway activation contributes to palmitate-elicited triglyceride secretion and cell death in hepatocytes. Experimental Biology and Medicine, 245(14), 1268-1279.
  93. 93. Tang, Y., Zhao, X., Antoine, D., Xiao, X., Wang, H., Andersson, U., ... & Lu, B. (2016). Regulation of posttranslational modifications of HMGB1 during immune responses. Antioxidants & Redox Signaling, 24(12), 620-634.
  94. 94. Nair, A., Ebenezer, P., & Francis, J. (2015). High-Mobility Group Box 1 (HMGB1) in Angiotensin II induced hypertension. The FASEB Journal, 29, 1059-1057.
  95. 95. Wang, Y., Zhong, J., Zhang, X., Liu, Z., Yang, Y., Gong, Q., & Ren, B. (2016). The role of HMGB1 in the pathogenesis of type 2 diabetes. Journal of diabetes research, 2016.
  96. 96. Schüler, R., Osterhoff, M. A., Frahnow, T., Seltmann, A. C., Busjahn, A., Kabisch, S., ... & Pfeiffer, A. F. (2017). High-saturated-fat diet increases circulating angiotensin-converting enzyme, which is enhanced by the rs4343 polymorphism defining persons at risk of nutrient-dependent increases of blood pressure. Journal of the American Heart Association, 6(1), e004465.
  97. 97. Nieva-Echevarría, B., Goicoechea, E., & Guillén, M. D. (2020). Food lipid oxidation under gastrointestinal digestion conditions: A review. Critical Reviews in Food Science and Nutrition, 60(3), 461-478
  98. 98. Macho-González, A., Garcimartín, A., López-Oliva, M. E., Bastida, S., Benedí, J., Ros, G., ... & Sánchez-Muniz, F. J. (2020). Can Meat and Meat-Products Induce Oxidative Stress?. Antioxidants, 9(7), 638.
  99. 99. Kanner, J., Gorelik, S., Roman, S., & Kohen, R. (2012). Protection by polyphenols of postprandial human plasma and low-density lipoprotein modification: the stomach as a bioreactor. Journal of Agricultural and Food Chemistry, 60(36), 8790-8796.
  100. 100. Cheon, H. G., & Cho, Y. S. (2014). Protection of palmitic acid-mediated lipotoxicity by arachidonic acid via channeling of palmitic acid into triglycerides in C2C12. Journal of biomedical science, 21(1), 1-10.
  101. 101. Moravcova, A., Červinková, Z., Kučera, O., Mezera, V., Rychtrmoc, D., & Lotkova, H. (2015). The effect of oleic and palmitic acid on induction of steatosis and cytotoxicity on rat hepatocytes in primary culture. Physiological research, 64.
  102. 102. Zhang, Y., Xue, R., Zhang, Z., Yang, X., & Shi, H. (2012). Palmitic and linoleic acids induce ER stress and apoptosis in hepatoma cells. Lipids in health and disease, 11(1), 1-8.
  103. 103. Mak, I. L., Lavery, P., Agellon, S., Rauch, F., Murshed, M., & Weiler, H. A. (2019). Arachidonic acid exacerbates diet-induced obesity and reduces bone mineral content without impacting bone strength in growing male rats. The Journal of nutritional biochemistry, 73, 108226.
  104. 104. Fleckenstein-Elsen, M., Dinnies, D., Jelenik, T., Roden, M., Romacho, T., & Eckel, J. (2016). Eicosapentaenoic acid and arachidonic acid differentially regulate adipogenesis, acquisition of a brite phenotype and mitochondrial function in primary human adipocytes. Molecular nutrition & food research, 60(9), 2065-2075.
  105. 105. Pisani, D. F., Ghandour, R. A., Beranger, G. E., Le Faouder, P., Chambard, J. C., Giroud, M., ... & Amri, E. Z. (2014). The ω6-fatty acid, arachidonic acid, regulates the conversion of white to brite adipocyte through a prostaglandin/calcium mediated pathway. Molecular metabolism, 3(9), 834-847.
  106. 106. Chan, P. C., Hsiao, F. C., Chang, H. M., Wabitsch, M., & Hsieh, P. S. (2016). Importance of adipocyte cyclooxygenase-2 and prostaglandin E2-prostaglandin E receptor 3 signaling in the development of obesity-induced adipose tissue inflammation and insulin resistance. The FASEB Journal, 30(6), 2282-2297.
  107. 107. Uchida, K. (2017). HNE as an inducer of COX-2. Free Radical Biology and Medicine, 111, 169-172.
  108. 108. Williams, E. S., Baylin, A., & Campos, H. (2007). Adipose tissue arachidonic acid and the metabolic syndrome in Costa Rican adults. Clinical nutrition, 26(4), 474-482.
  109. 109. Gilani, A., Agostinucci, K., Hossain, S., Pascale, J. V., Garcia, V., Adebesin, A. M., ... & Schwartzman, M. L. (2021). 20-HETE interferes with insulin signaling and contributes to obesity-driven insulin resistance. Prostaglandins & Other Lipid Mediators, 152, 106485.
  110. 110. Nelson, G. J., Schmidt, P. C., Bartolini, G., Kelley, D. S., Phinney, S. D., Kyle, D., ... & Schaefer, E. J. (1997). The effect of dietary arachidonic acid on plasma lipoprotein distributions, apoproteins, blood lipid levels, and tissue fatty acid composition in humans. Lipids, 32(4), 427-433.
  111. 111. Froyen, E., & Burns-Whitmore, B. (2020). The Effects of Linoleic Acid Consumption on Lipid Risk Markers for Cardiovascular Disease in Healthy Individuals: A Review of Human Intervention Trials. Nutrients, 12(8), 2329.
  112. 112. Iggman, D., Ärnlöv, J., Cederholm, T., & Risérus, U. (2016). Association of adipose tissue fatty acids with cardiovascular and all-cause mortality in elderly men. JAMA cardiology, 1(7), 745-753.
  113. 113. Jandacek, R. J. (2017, June). Linoleic acid: a nutritional quandary. In Healthcare (Vol. 5, No. 2, p. 25). Multidisciplinary Digital Publishing Institute.
  114. 114. Kothapalli, K. S., Ye, K., Gadgil, M. S., Carlson, S. E., O’Brien, K. O., Zhang, J. Y., ... & Brenna, J. T. (2016). Positive selection on a regulatory insertion–deletion polymorphism in FADS2 influences apparent endogenous synthesis of arachidonic acid. Molecular Biology and Evolution, 33(7), 1726-1739.
  115. 115. Li, P., Zhao, J., Kothapalli, K. S., Li, X., Li, H., Han, Y., ... & Gao, Y. (2018). A regulatory insertion-deletion polymorphism in the FADS gene cluster influences PUFA and lipid profiles among Chinese adults: a population-based study. The American Journal of Clinical Nutrition, 107(6), 867-875.
  116. 116. Goedecke, J. H., Chorell, E., van Jaarsveld, P. J., Risérus, U., & Olsson, T. (2021). Fatty acid metabolism and associations with insulin sensitivity differs between black and white South African women. The Journal of Clinical Endocrinology & Metabolism, 106(1), e140-e151.
  117. 117. Tsukamoto, I., & Sugawara, S. (2018). Low levels of linoleic acid and α-linolenic acid and high levels of arachidonic acid in plasma phospholipids are associated with hypertension. Biomedical reports, 8(1), 69-76.
  118. 118. Maingrette, F., & Renier, G. (2005). Linoleic acid increases lectin-like oxidized LDL receptor-1 (LOX-1) expression in human aortic endothelial cells. Diabetes, 54(5), 1506-1513.
  119. 119. Hao, Q., Lillefosse, H. H., Fjære, E., Myrmel, L. S., Midtbø, L. K., Jarlsby, R. H., ... & Madsen, L. (2012). High-glycemic index carbohydrates abrogate the antiobesity effect of fish oil in mice. American Journal of Physiology-Endocrinology and Metabolism, 302(9), E1097-E1112.
  120. 120. Zou, Y., Wang, Y. N., Ma, H., He, Z. H., Tang, Y., Guo, L., ... & Tang, Q. Q. (2020). SCD1 promotes lipid mobilization in subcutaneous white adipose tissue. Journal of lipid research, 61(12), 1589-1604.
  121. 121. Camell, C., & Smith, C. W. (2013). Dietary oleic acid increases M2 macrophages in the mesenteric adipose tissue. PloS one, 8(9), e75147.
  122. 122. Guo, X. F., Li, X., Shi, M., & Li, D. (2017). n-3 polyunsaturated fatty acids and metabolic syndrome risk: a meta-analysis. Nutrients, 9(7), 703.
  123. 123. Kim, J., Okla, M., Erickson, A., Carr, T., Natarajan, S. K., & Chung, S. (2016). Eicosapentaenoic acid potentiates brown thermogenesis through FFAR4-dependent up-regulation of miR-30b and miR-378. Journal of Biological Chemistry, 291(39), 20551-20562.
  124. 124. Holland, W. L., Adams, A. C., Brozinick, J. T., Bui, H. H., Miyauchi, Y., Kusminski, C. M., ... & Scherer, P. E. (2013). An FGF21-adiponectin-ceramide axis controls energy expenditure and insulin action in mice. Cell metabolism, 17(5), 790-797.
  125. 125. Dutchak, P. A., Katafuchi, T., Bookout, A. L., Choi, J. H., Ruth, T. Y., Mangelsdorf, D. J., & Kliewer, S. A. (2012). Fibroblast growth factor-21 regulates PPARγ activity and the antidiabetic actions of thiazolidinediones. Cell, 148(3), 556-567.
  126. 126. Quesada-López, T., Cereijo, R., Turatsinze, J. V., Planavila, A., Cairó, M., Gavaldà-Navarro, A., ... & Villarroya, F. (2016). The lipid sensor GPR120 promotes brown fat activation and FGF21 release from adipocytes. Nature communications, 7(1), 1-17.
  127. 127. Drouin, G., Catheline, D., Guillocheau, E., Gueret, P., Baudry, C., Le Ruyet, P., ... & Legrand, P. (2019). Comparative effects of dietary n-3 docosapentaenoic acid (DPA), DHA and EPA on plasma lipid parameters, oxidative status and fatty acid tissue composition. The Journal of nutritional biochemistry, 63, 186-196.
  128. 128. Griffin, B. A. (2008). How relevant is the ratio of dietary n-6 to n-3 polyunsaturated fatty acids to cardiovascular disease risk? Evidence from the OPTILIP study. Current opinion in lipidology, 19(1), 57-62.
  129. 129. Li, N., Jia, M., Deng, Q., Wang, Z., Huang, F., Hou, H., & Xu, T. (2020). Effect of low-ratio n-6/n-3 PUFA on blood lipid level: a meta-analysis. Hormones, 1-10.
  130. 130. Ma, T., Liaset, B., Hao, Q., Petersen, R. K., Fjære, E., Ngo, H. T., ... & Madsen, L. (2011). Sucrose counteracts the anti-inflammatory effect of fish oil in adipose tissue and increases obesity development in mice. PloS one, 6(6), e21647.
  131. 131. Varma, V., Boros, L. G., Nolen, G. T., Chang, C. W., Wabitsch, M., Beger, R. D., & Kaput, J. (2015). Metabolic fate of fructose in human adipocytes: a targeted 13 C tracer fate association study. Metabolomics, 11(3), 529-544.
  132. 132. Sodhi, K., Hilgefort, J., Banks, G., Gilliam, C., Stevens, S., Ansinelli, H. A., ... & Khitan, Z. (2016). Uric acid-induced adipocyte dysfunction is attenuated by HO-1 upregulation: potential role of antioxidant therapy to target obesity. Stem cells international, 8197325 .
  133. 133. Sautin, Y. Y., & Johnson, R. J. (2008). Uric acid: the oxidant-antioxidant paradox. Nucleosides, Nucleotides, and Nucleic Acids, 27(6-7), 608-619.
  134. 134. Zhang, J., Diao, B., Lin, X., Xu, J., & Tang, F. (2019). TLR2 and TLR4 mediate an activation of adipose tissue renin-angiotensin system induced by uric acid. Biochimie, 162, 125-133.
  135. 135. Bartlett, D. E., Miller, R. B., Thiesfeldt, S., Lakhani, H. V., Shapiro, J. I., & Sodhi, K. (2018). The role of Na/K-ATPase signaling in oxidative stress related to aging: implications in obesity and cardiovascular disease. International journal of molecular sciences, 19(7), 2139.
  136. 136. Pratt, R. D., Brickman, C., Nawab, A., Cottrill, C., Snoad, B., Lakhani, H. V., ... & Sodhi, K. (2019). The adipocyte Na/K-ATPase oxidant amplification loop is the central regulator of western diet-induced obesity and associated comorbidities. Scientific reports, 9(1), 1-11.
  137. 137. Lee, M., Sorn, S. R., Lee, Y., & Kang, I. (2019). Salt induces adipogenesis/lipogenesis and inflammatory adipocytokines secretion in adipocytes. International journal of molecular sciences, 20(1), 160.
  138. 138. Diao, Z., Asico, L. D., Van Anthony, M. V., Zheng, X., Cuevas, S., Armando, I., ... & Wang, X. (2017). Increased renal oxidative stress in salt-sensitive human GRK4γ486V transgenic mice. Free Radical Biology and Medicine, 106, 80-90.
  139. 139. Zhu, Z., Wu, F., Lu, Y., Wang, Z., Zang, J., Yu, H., ... & Ding, G. (2018). The association of dietary cholesterol and fatty acids with dyslipidemia in Chinese metropolitan men and women. Nutrients, 10(8), 961
  140. 140. Püschel, G. P., & Henkel, J. (2018). Dietary cholesterol does not break your heart but kills your liver. Porto Biomedical Journal, 3, e12.
  141. 141. Sánchez-Tapia, M., Miller, A. W., Granados-Portillo, O., Tovar, A. R., & Torres, N. (2020). The development of metabolic endotoxemia is dependent on the type of sweetener and the presence of saturated fat in the diet. Gut Microbes, 12, 1801301.
  142. 142. Bonet, M. L., Canas, J. A., Ribot, J., & Palou, A. (2015). Carotenoids and their conversion products in the control of adipocyte function, adiposity and obesity. Archives of biochemistry and biophysics, 572, 112-125.
  143. 143. Wong, S. K., Chin, K. Y., Suhaimi, F. H., Ahmad, F., & Ima-Nirwana, S. (2017). Vitamin E as a potential interventional treatment for metabolic syndrome: evidence from animal and human studies. Frontiers in pharmacology, 8, 444.
  144. 144. Puah, C., Choo, Y., Ma, A., & Chuah, C. (2007). The effect of physical refining on palm vitamin E (tocopherol, tocotrienol and tocomonoenol). American Journal of Applied Sciences, 4(6), 374-377.
  145. 145. Sotler, R., Poljšak, B., Dahmane, R., Jukić, T., Jukić, D. P., Rotim, C., ... & Starc, A. (2019). Prooxidant activities of antioxidants and their impact on health. Acta Clinica Croatica, 58(4), 726.
  146. 146. Zou, W., Chen, C., Zhong, Y., An, J., Zhang, X., Yu, Y., ... & Fu, J. (2013). PI3K/Akt pathway mediates Nrf2/ARE activation in human L02 hepatocytes exposed to low-concentration HBCDs. Environmental science & technology, 47(21), 12434-12440.
  147. 147. Wardyn, J. D., Ponsford, A. H., & Sanderson, C. M. (2015). Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways. Biochemical society transactions, 43(4), 621-626.
  148. 148. Bonet, M. L., Canas, J. A., Ribot, J., & Palou, A. (2015). Carotenoids and their conversion products in the control of adipocyte function, adiposity and obesity. Archives of biochemistry and biophysics, 572, 112-125.
  149. 149. Wong, S. K., Chin, K. Y., Suhaimi, F. H., Ahmad, F., & Ima-Nirwana, S. (2017). Vitamin E as a potential interventional treatment for metabolic syndrome: evidence from animal and human studies. Frontiers in pharmacology, 8, 444.
  150. 150. Puah, C., Choo, Y., Ma, A., & Chuah, C. (2007). The effect of physical refining on palm vitamin E (tocopherol, tocotrienol and tocomonoenol). American Journal of Applied Sciences, 4(6), 374-377.
  151. 151. Zhuang, Y., Wu, H., Wang, X., He, J., He, S., & Yin, Y. (2019). Resveratrol attenuates oxidative stress-induced intestinal barrier injury through PI3K/ Akt-mediated Nrf2 signaling pathway. Oxidative medicine and cellular longevity, 2019.
  152. 152. He, S., Guo, Y., Zhao, J., Xu, X., Wang, N., & Liu, Q. (2020). Ferulic acid ameliorates lipopolysaccharide-induced barrier dysfunction via microRNA200c-3p-mediated activation of PI3K/AKT pathway in Caco-2 cells. Frontiers in pharmacology, 11, 376.
  153. 153. Zhao, L., Zhang, Q., Ma, W., Tian, F., Shen, H., & Zhou, M. (2017). A combination of quercetin and resveratrol reduces obesity in high-fat diet-fed rats by modulation of gut microbiota. Food & function, 8(12), 4644-4656.
  154. 154. Zhao, Y., Chen, B., Shen, J., Wan, L., Zhu, Y., Yi, T., & Xiao, Z. (2017). The beneficial effects of quercetin, curcumin, and resveratrol in obesity. Oxidative medicine and cellular longevity, 2017.
  155. 155. Xu, T., Song, Q., Zhou, L., Yang, W., Wu, X., Qian, Q., ... & Li, S. (2021). Ferulic acid alleviates lipotoxicityinduced hepatocellular death through the SIRT1-regulated autophagy pathway and independently of AMPK and Akt in AML-12 hepatocytes. Nutrition & metabolism, 18(1), 1-13.
  156. 156. Ruan, H., & Dong, L. Q. (2016). Adiponectin signaling and function in insulin target tissues. Journal of molecular cell biology, 8(2), 101-109.
  157. 157. Miyata, Y., Tanaka, H., Shimada, A., Sato, T., Ito, A., Yamanouchi, T., & Kosano, H. (2011). Regulation of adipocytokine secretion and adipocyte hypertrophy by polymethoxyflavonoids, nobiletin and tangeretin. Life sciences, 88(13-14), 613-618.
  158. 158. Rezvan, N., Moini, A., GorganiFiruzjaee, S., & Hosseinzadeh-Attar, M. J. (2018). Oral quercetin supplementation enhances adiponectin receptor transcript expression in polycystic ovary syndrome patients: a randomized placebo-controlled doubleblind clinical trial. Cell Journal (Yakhteh), 19(4), 627.
  159. 159. Shahi, M., Zakerzadeh, M., Zakerkish, M., Zarei, M., & Saki, A. (2017). Effect of sesamin supplementation on glycemic status, inflammatory markers, and adiponectin levels in patients with type 2 diabetes mellitus. Journal of dietary supplements, 14(1), 65-75.
  160. 160. Tomé-Carneiro, J., Gonzálvez, M., Larrosa, M., Yáñez-Gascón, M. J., García-Almagro, F. J., Ruiz-Ros, J. A., ... & Espín, J. C. (2013). Grape resveratrol increases serum adiponectin and downregulates inflammatory genes in peripheral blood mononuclear cells: a triple-blind, placebo-controlled, one-year clinical trial in patients with stable coronary artery disease. Cardiovascular Drugs and Therapy, 27(1), 37-48.
  161. 161. Reibe-Pal, S., & Febbraio, M. A. (2017). Adiponectin serenades ceramidase to improve metabolism. Molecular metabolism, 6(3), 233.
  162. 162. Jin, J., Lu, Z., Li, Y., Cowart, L. A., Lopes-Virella, M. F., & Huang, Y. (2018). Docosahexaenoic acid antagonizes the boosting effect of palmitic acid on LPS inflammatory signaling by inhibiting gene transcription and ceramide synthesis. PloS one, 13(2), e0193343.
  163. 163. Deng, P., Hoffman, J. B., Petriello, M. C., Wang, C. Y., Li, X. S., Kraemer, M. P., ... & Hennig, B. (2020). Dietary inulin decreases circulating ceramides by suppressing neutral sphingomyelinase expression and activity in mice. Journal of lipid research, 61(1), 45-53.
  164. 164. Mihalj, M., Tadzic, R., Vcev, A., Rucevic, S., & Drenjancevic, I. (2016). Blood pressure reduction is associated with the changes in oxidative stress and endothelial activation in hypertension, regardless of antihypertensive therapy. Kidney and Blood Pressure Research, 41(6), 721-735.
  165. 165. Rao, P. V., & Gan, S. H. (2014). Cinnamon: a multifaceted medicinal plant. Evidence-Based Complementary and Alternative Medicine, 642942.
  166. 166. Lundberg, J. O., Carlström, M., & Weitzberg, E. (2018). Metabolic effects of dietary nitrate in health and disease. Cell Metabolism, 28(1), 9-22.
  167. 167. Bunbupha, S., Prasarttong, P., Poasakate, A., Maneesai, P., & Pakdeechote, P. (2021). Imperatorin alleviates metabolic and vascular alterations in high-fat/high-fructose diet-fed rats by modulating adiponectin receptor 1, eNOS, and p47phox expression. European Journal of Pharmacology, 174010.
  168. 168. Anderson, E. J., Vistoli, G., Katunga, L. A., Funai, K., Regazzoni, L., Monroe, T. B., ... & Aldini, G. (2018). A carnosine analog mitigates metabolic disorders of obesity by reducing carbonyl stress. The Journal of clinical investigation, 128(12), 5280-5293.
  169. 169. Ho, C. T., & Wang, M. (2013). Dietary phenolics as reactive carbonyl scavengers: potential impact on human health and mechanism of action. Journal of traditional and complementary medicine, 3(3), 139-141.
  170. 170. Khalifa, I., Xia, D., Dutta, K., Peng, J., Jia, Y., & Li, C. (2020). Mulberry anthocyanins exert anti-AGEs effects by selectively trapping glyoxal and structural-dependently blocking the lysyl residues of β-lactoglobulins. Bioorganic Chemistry, 96, 103615.
  171. 171. Wang, X., Chang, T., Jiang, B., Desai, K., & Wu, L. (2007). Attenuation of hypertension development by aminoguanidine in spontaneously hypertensive rats: role of methylglyoxal. American journal of hypertension, 20(6), 629-636.
  172. 172. Liu, P., Wang, Y., Yang, G., Zhang, Q., Meng, L., Xin, Y., & Jiang, X. (2021). The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacological Research, 105420.
  173. 173. Yan, H., & Ajuwon, K. M. (2017). Butyrate modifies intestinal barrier function in IPEC-J2 cells through a selective upregulation of tight junction proteins and activation of the Akt signaling pathway. PloS one, 12(6), e0179586.
  174. 174. Mani, V., Hollis, J. H., & Gabler, N. K. (2013). Dietary oil composition differentially modulates intestinal endotoxin transport and postprandial endotoxemia. Nutrition & metabolism, 10(1), 1-9.

Written By

Arnold N. Onyango

Submitted: 14 April 2021 Reviewed: 28 April 2021 Published: 20 May 2021