Open access

Lipid Peroxidation by-Products and the Metabolic Syndrome

Written By

Nicolas J. Pillon and Christophe O. Soulage

Submitted: 04 November 2011 Published: 29 August 2012

DOI: 10.5772/46019

From the Edited Volume

Lipid Peroxidation

Edited by Angel Catala

Chapter metrics overview

4,227 Chapter Downloads

View Full Metrics

1. Introduction

About twenty-one percent of the air we breathe is composed of oxygen, and our life will not be possible without it. Oxygen is however a toxic, highly reactive molecule which was originally released in atmosphere as a waste product of the first photosynthetic organisms. Its accumulation on Earth indeed led to a massive extinction of living species. Few organisms survived and some developed the ability to use this toxic oxygen to improve the production of energy from carbohydrates. This “oxidative metabolism” was however a double-edge sword as the use of intracellular oxygen generates deleterious oxidative damages. To protect themselves toward this toxicity, those organisms consequently developed several “antioxidants” protection mechanisms which helped them maintain a balance between oxidative damage and efficient use of oxygen to produce energy.

When antioxidant defences are reduced and/or oxidative mechanisms increased, uncontrolled oxidation of cell targets leads to the accumulation of reactive oxygen species (ROS) and a state of “oxidative stress”, often deleterious for the cells. This stress is involved in the pathophysiology of several human diseases, and especially in the development of metabolic diseases, even if its causative role remains questionable. A definite increase in oxidative stress biomarkers can be found in obese and diabetic humans as well as in animal model of these diseases. Accumulation of ROS can be deleterious by itself or can induce the oxidation of proteins, nucleic acids and lipids, generating secondary by-products. The specific reactivity of ROS towards polyunsaturated fatty acids (PUFAs) present in cell membranes induces lipid peroxidation, a noxious mechanism producing toxic aldehydes. Among them, malondialdehyde (MDA) and 4-hydroxy-2-nonenal (HNE) have been extensively studied. Originally simple markers of lipid peroxidation, these aldehydes have demonstrated causative roles in the impairment of cellular functions: activation of signalling pathways, apoptosis, and modification of enzyme function. In addition to being hallmarks of oxidative damage, lipid aldehydes could be mediators of oxidative insults, propagating tissue injury and activating cellular stress signalling pathways. Several studies demonstrated the association of obesity and diabetes with lipid peroxidation by-products, and the role of aldehydes in impairment of insulin function and signalling was recently pointed out.

This chapter aims to review the diverse implications of lipid peroxidation by-products in the pathophysiology of metabolic diseases, from evidence of their production during obesity and diabetes to the cellular mechanisms of their toxicity and protection against their deleterious effects.

Advertisement

2. Lipid peroxidation by-products

Under conditions of oxidative stress, excessive production of reactive oxygen species promotes the peroxidation of polyunsaturated fatty acids (PUFA). The resulting accumulation of hydroperoxides, unstable molecules, leads to their non-enzymatic degradation in many compounds, including aldehydes. Quantification of lipid peroxidation in biological samples has been extensively performed with the thiobarbituric acid (TBA) test. TBA detects malondialdehyde (MDA), an end-product of nonenzymatic PUFA oxidative degradation, which has therefore been used for decades as a marker of lipid peroxidation (Gutteridge, 1982). Another aldehyde: acrolein, first attracted attention because of its formation during tobacco combustion and its ubiquitous presence in the environment (Dong & Moldoveanu, 2004). Because of its carcinogenic potential, its role in smoking-related diseases has received extensive attention; however, acrolein is also produced endogenously though lipid peroxidation and its link with oxidative-associated pathologies is now well established. 4-hydroxy-2-alkenals are specific by-products of the oxidation of omega-3 and omega-6 fatty acids. 4-hydroxy-2-nonenal (HNE) is derived from the oxidation of polyunsaturated fatty acids of the n-6 series, mainly linoleic and arachidonic acids, while 4-hydroxy-2-hexenal (HHE) results from the peroxidation of polyunsaturated fatty acids of the n-3 series (mainly docosahexaenoic, eicosapentaenoic and linolenic acid). The peroxidation of arachidonic acid via 12-lipoxygenase leads to the formation of 4-hydroxy-2-dodecadienal (HDDE) (Guichardant et al., 2006).

NameMolecular Weight (Da)Molecular
Formula
Skeletal Formula
Acrolein (prop-2-enal)56.1C3H4O
Malondialdehyde (propanedial, MDA)72.1C3H4O2
4-hydroxy-2-hexenal (HHE)114.1C6H10O2
4-hydroxy-2-nonenal (HNE)156.2C9H16O2
4-hydroxy-2-dodecadienal (HDDE)196.0C12H20O2

Table 1.

α,β-Unsaturated aldehydes produced during polyunsaturated fatty acids oxidation

2.1. Chemistry and reactivity

Acrolein, MDA and 4-hydroxy-alkenals are α,β-unsaturated aldehydes, a class of compounds sharing the general structure C=C−C=O. They are characterised by an aldehyde group (C=O) on carbon 1 and a conjugated double bond (C=C) between carbons 2 and 3 (Table 1). In this structure, the oxygen atom of the carbonyl group increases the polarity of the double bond, which makes α,β-unsaturated aldehydes potent electrophiles. Acrolein has the simplest structure composed of 3 carbons, MDA is a dicarbonyl compound and 4-hydroxy-2-alkenals are characterized by the presence of a hydroxyl group on carbon 4. HHE, HNE and HDDE only differ by the length of their carbon chain and the presence of an additional double bond for the HDDE. In the case of MDA and 4-hydroxy-alkenals, the presence of a second oxygen atom makes the double bond even more reactive.

These aldehydes are part of the “reactive electrophile species” able to form covalent adducts with the nucleophilic groups present in DNA, proteins and phospholipids. In physiological conditions, they spontaneously react with the thiol group of glutathione to form Michael adduct by attack of the nucleophilic group of glutathione to the double bond of aldehydes. They can also react with thiol groups present on cysteine residues of certain proteins, leading to impairment of their biological activity. Under certain conditions, especially alkaline pH, aldehydes react with the amine groups present in proteins, nucleic acids and aminophospholipids, leading to Michael adducts. On the other hand, the reaction between a primary amine group and the carbonyl group of the aldehyde leads to the formation of Schiff bases (Schaur, 2003).

Aldehydes produced during lipid peroxidation are precursors of Advanced Lipoxidation End products (ALEs). Together with Advanced Glycation End products (AGEs) generated during glycoxidation, they accumulate in cells and tissues. The “carbonyl stress” is a result of this adduct accumulation, which induces protein dysfunctions and consequent pathological events such as inflammation and apoptosis (Negre-Salvayre et al., 2008).

2.2. Cellular effects

2.2.1. Cytotoxicity

Since α,β-unsaturated compounds are strong electrophiles, they exhibit a high cytotoxic and mutagenic potential and have consequently been extensively studied for their effects on cell viability. HNE, HHE and acrolein indeed induce cell death, but the lethal concentration 50 (LC50), concentration that induces the death of 50% of the cells, is subject to variation, depending on the aldehyde, exposure duration and cell type (Table 2). The LC50 for a long exposure (>16 hours) to HNE or HHE is however consistently found 20-60 µM in several cell types, including human lymphoma Jurkat cells, lens epithelial cells, hamster V79-4 cells and muscle cells (Table 2). For acrolein, the same treatment gives a range of LC50 of 5-100 µM in human fibroblasts, human neuroblastoma cells, PC12 chromaffin cells and lymphocytes. MDA-induced cell death is less documented, even if the LC50 range for MDA is found around 1 mM in cortical, endothelial cells and fibroblasts. MDA was also reported to induce cell cycle arrest, which is to relate to cell damage and death (Ji et al., 1998). HDDE appears to be the most toxic lipid aldehyde with a LC50 in endothelial cells in the submicromolar range.

NameLC50, µMReferences
Acrolein
(prop-2-enal)
5-100Poirier et al., 2002; Luo et al., 2005; Jia et al., 2009b, 2009a
Malondialdehyde
(propanedial, MDA)
600-3000Michiels & Remacle, 1991; Hipkiss et al., 1997; Cheng et al., 2011
4-hydroxy-2-hexenal
(HHE)
20-60Liu et al., 2000; Choudhary et al., 2002; Pillon et al., 2010; Li et al., 2011
4-hydroxy-2-nonenal
(HNE)
20-60Liu et al., 2000; Choudhary et al., 2002; Pillon et al., 2010; Li et al., 2011
4-hydroxy-2-dodecadienal
(HDDE)
0.22Riahi et al., 2010

Table 2.

Range of lethal concentration 50 for long-term treatment (>16 hours) with α,β-unsaturated aldehydes. LC50 values were calculated from the indicated references.

On the opposite, very little cell death is detectable for short term treatments (<4 hours), likely because aldehyde-induced cell death involves apoptosis mechanisms not yet occurring during this short period of time. Several studies indeed reported that cell death is induced by aldehydes through apoptosis for low concentration and both apoptosis and necrosis for high doses (Luo et al., 2005; Liu et al., 2010). Acrolein-induced necrosis was described in few studies (Luo et al., 2005), but mitochondrial-driven cell death seems to be the canonical road and was widely studied. Acrolein-induced apoptosis was indeed confirmed in several cell types through DNA fragmentation, phosphatidylserine externalization, poly(ADP-ribose) polymerase cleavage and activation of caspases (Pan et al., 2009; Roy et al., 2010). Last but not least, hydroxyalkenals are potent activators of apoptosis. They both induce DNA fragmentation and activation of caspases in very different cell types (Choudhary et al., 2002; Vaillancourt et al., 2008). In addition, HHE has been shown to decrease the expression/phosphorylation of Bcl-2, while increasing that of Bax, leading to apoptosis of human renal epithelial cells (Bae et al., 2011; Bodur et al., 2012).

Interestingly, the toxicity of aldehydes is highly correlated to their ability to form covalent adducts on proteins. In muscle cells, the lethal concentration 50 (LC50) for 12 different aldehydes was calculated, including HHE and HNE (Pillon et al., 2010). This LC50 was strongly correlated with their respective potency to form covalent adducts on albumin in vitro (Figure 1). This demonstrates that the cytotoxicity and likely other biological effects of aldehydes mainly occur through chemical adduction of other biomolecules.

Figure 1.

Toxicity is correlated to the adduction ability. Viability of muscle cells was measured in response to 12 different aldehydes

4-hydroxy-2-hexenal, 4-hydroxy-2-nonenal, 4-hydroxy-2-dodecenal, 4-hydroxy-2-hexenal dimethylacetal, 4-hydroxy-2-nonenal dimethylacetal, 4-hydroxy-2-dodecenal dimethylacetal, trans-2-hexenal, trans-2-nonenal, trans-2-dodecenal, hexanal, nonanal, dodecanal

. The calculated LC50 was then correlated to their ability to form covalent adducts on bovine serumalbumin (Pillon et al., 2010).

2.2.2. Oxidative stress and ROS production

The classical sequence of events is that oxidative stress triggers lipid peroxidation which in turns produces aldehydes by-products. However, an interesting paradigm was pointed out by several groups: lipid aldehydes are able to induce the production of ROS, and this is thought to be of importance in their deleterious effects. For instance, acrolein treatment produces nitric oxide (Misonou et al., 2006) and induces generation of intracellular oxidants (Luo et al., 2005; Wang et al., 2011). Similarly, accumulation of intracellular ROS was described in cells treated with MDA (Cheng et al., 2011); and unsurprisingly, the 4-hydroxyalkenals HHE and HNE share the same ability. HHE induces ROS in neurons and tubular epithelial cells (Long et al., 2008; Bae et al., 2011) and HNE induces mitochondrial oxidative stress in neurons, vascular muscle, liver and skeletal muscle cells (Uchida et al., 1999; Lee et al., 2006; Pillon et al., 2012). The source of ROS was suggested to be mitochondria, as several aldehydes have been shown to induce a significant decrease in mitochondrial membrane potential (Uchida et al., 1999; Luo et al., 2005).

Figure 2.

The vicious circle linking ROS and lipid aldehyde production

This concept is reinforced by the fact that an increase in glutathione pool can prevent the deleterious effects of HNE on both adduct formation and ROS production (Pillon et al., 2012) and that dysfunction of glutathione S-transferase, a major enzyme for aldehyde detoxification, leads to excess 4-hydroxy-2-nonenal and oxidative stress (Kostyuk et al., 2010; Curtis et al., 2010). Based on this body of evidence, lipid by-products can be seen as parts of a vicious circle in which increased ROS production generates aldehydes which further amplify the generation of oxidative species and so on (Figure 2).

2.2.3. Signalling pathways and transcription factors

Consistent with the extensive work done on cytotoxicity, the stress signalling pathways have been primarily pointed out as the main intracellular route activated by aldehydes. The mitogen-activated protein kinases (MAPKs) are indeed activated by several aldehydes including MDA, acrolein and HNE. MDA activates c-Jun N-terminal kinases (JNK) and extracellular signal-regulated kinases (ERK) (Cheng et al., 2011) and acrolein-induced apoptosis occurs through activation of p38 and ERK (Tanel & Averill-Bates, 2007). The activation of these three MAPK has also been described following treatment with HNE (Uchida et al., 1999; Zarrouki et al., 2007; Pillon et al., 2012) and HHE (Je et al., 2004; Bae et al., 2011). Overall, most studies investigated the cytotoxic effects of aldehydes and thus focused on cellular stress pathways; therefore, very little data is available regarding their potential effects on other pathways. Only very recent work shows that HNE interfere with insulin signalling pathway through oxidative stress and adduction of IRS1 and Akt (Demozay et al., 2008; Shearn et al., 2011; Pillon et al., 2012).

Aldehydes regulate gene expression by activating the signalling pathways described above, or by direct modification of transcription factors. Unsurprisingly, aldehyde production regulates the expression of several antioxidant enzymes such as NAD(P)H quinone oxidoreductase-1, Heme oxygenase-1 and glutathione S-transferase (GST). This occurs through the activation of the Nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which drives the expression of these antioxidant enzymes. Aldehyde-induced Heme Oxygenase-1 expression is indeed mediated by the Nrf2 pathway in HUVECs (Lee et al., 2011) and Nrf2 silencing significantly attenuates the induction of this same gene by acrolein (Zhang & Forman, 2008).

Beyond the expression of antioxidant enzymes, cellular growth, apoptosis and inflammatory responses can be induced by aldehydes, involving the activation of the nuclear factor kappa B (NF-κB) family. The effects of acrolein on NF-κB activation are controversial, but its activation by HNE and HHE is well documented. HHE and HNE induce NF-kappaB activation through IKK/NIK pathway, leading to IκB phosphorylation and subsequent proteolysis (Page et al., 1999; Je et al., 2004; Lee et al., 2004). HNE has moreover been shown to induce DNA-binding of NF-κB in vascular smooth muscle cells (Ruef et al., 2001).

The peroxisome proliferator-activated receptor (PPAR) family regulate the expression of genes that encode proteins involved in energy balance. They act as ligand-activated transcription factors and are responsive to the lipid status of the cell, therefore important during high fat diets and obesity. HNE is an intracellular agonist of PPARβ/δ while HHE do not activate this receptor (Coleman et al., 2007). Through this pathway, HNE significantly elevates adiponectin gene expression, concomitant with increased PPAR-γ gene expression and transactivity. Meanwhile, HDDE acts through PPARδ signalling pathways to regulate glucose transport in vascular endothelial cells subjected to hyperglycemia (Riahi et al., 2010) and HNE stimulates insulin secretion from Beta cells through interaction with PPARδ (Cohen et al., 2011).

2.3. Concentration in plasma and tissues

Little data is available concerning aldehydes levels in biological fluids, except for MDA and HNE which have been widely used as lipid peroxidation markers. The concentration of MDA in the plasma of healthy subjects is around 2-5 µM and increases up to 2-fold in type 2 diabetic patients (Figure 4). In the specific context of metabolic diseases, MDA is positively correlated with BMI and waist circumference in obese patients (Furukawa et al., 2004). Depending on the study, HNE concentration has been found to range from 50 nM to 10 µM under normal conditions. This significant variability in concentration according to the authors could be explained partly by the method used (LC/MS, GC/MS...) and also by the difficulty to measure such reactive derivatives. If HNE has been widely studied, there is however scarce data in the literature regarding the pathophysiological concentrations of HHE and HDDE. Plasma HHE concentration was however found to be around 9 nM in human, and dramatically increases to reach 90 nM after several weeks of a diet rich in omega-3 fatty acids (Calzada et al., 2010). Our group recently showed that HHE concentration was 20 nM in humans and 7 nM in rats and that it increases in both type-2 diabetes patients and type-1 diabetic rats, reinforcing existing evidence for a role of lipid aldehydes in metabolic diseases.

TissueAldehydeConcentration
(µM)
References
PlasmaMDA1 – 6See figure 4
HHE0.006 – 0.090(Calzada et al., 2010; Pillon et al, unpublished)
HNE0.007 – 11(McGrath et al., 2001; Selley, 2004; Syslova et al., 2009)
Exhaled breath condensateHNE0.25 - 5(Syslova et al., 2009; Manini et al., 2010)
MDA0.63 - 14(Syslova et al., 2009; Manini et al., 2010)
Ventricular FluidHNE0.2 - 120(Lovell et al., 1997)
Pancreatic IsletsHNE23 – 35(Miwa et al., 2000)

Table 3.

Concentration range of aldehydes in healthy plasma and tissues

One should keep in mind that all current quantification methods (HPLC, GC) only assay the free fraction (i.e. unreacted) of lipid aldehydes present in samples. Being very reactive, this is certainly not representative of the amount indeed produced from lipid peroxidation, which rapidly react with neighbour targets to form covalent adducts and thus, other non-quantitative methods estimating the amount of HNE have been used. For example, detection of protein adducts by immunohistochemistry has shown a significant increase in HHE and HNE proteins adducts in Parkinson's disease and in chronic liver disease (Yoritaka et al., 1996; Paradis et al., 1997). Of particular interest for this chapter, HNE adducts on plasma albumin are increased in type 2 diabetes (Toyokuni et al., 2000).

Advertisement

3. Insulin secretion and type-1 diabetes

3.1. Lipid aldehydes and insulin secretion

Oxidative stress associated with hyperglycemia is suspected to participate in beta cell dysfunction in terms of insulin synthesis and/or secretion. Diabetic animals display increased levels of lipoperoxidation in pancreas, and HNE concentration was reported to reach up to 35 µM in pancreatic islets of diabetic rats (Miwa et al., 2000). In addition, HNE-modified proteins are increased in the pancreatic beta-cells of Goto-Kakizaki rats, a genetic model of non obese type 2 diabetes (Ihara et al., 1999), as well as in type 2 diabetic patients (Sakuraba et al., 2002). HNE and other lipid peroxidation by-products such as 2-hexenal and 2-butenal inhibit glucose-induced insulin secretion in isolated rat islets. Both glucose utilization and glucose oxidation are blunted in islets after treatment with aldehydes suggesting that they impair glucose-induced insulin secretion through an interference with glycolytic pathway and citric acid cycle (Miwa et al., 2000). Another piece of evidence comes from the exposure of beta cells to NO donors and to interleukin-1 beta, which leads to generation of oxidative stress and lipoperoxidation by-products. MDA and HNE produced under this condition are involved in the activation of an apoptotic program, contributing to the reduction in the beta cell mass (Cahuana et al., 2003). Peroxynitrite indeed triggers lipoperoxidation in the beta-cell line RIN-5-F, and the resulting protein carbonylation is a key factor linking NO-dependent lipoperoxidation and apoptosis (Cahuana et al., 2003). Alloxan, a toxic glucose analogue, has been widely used to generate rodent models of type-1 diabetes, as it selectively destroys insulin-producing cells in the pancreas. Alloxan-induced diabetic rats exhibit increased lipid peroxidation associated with defects in insulin secretion, which can be prevented by the antioxidant S-allyl cysteine therefore restoring insulin secretion and ameliorating the glycaemic control (Augusti & Sheela, 1996).

3.2. Lipid aldehydes and the beta cells: Doctor Jeckyll or Mr Hyde?

HNE was shown to elicit various physiological or physiopathological responses: high concentrations elicit beta cell death and defect in insulin secretion, while lower concentrations act as signalling mediators. In INS-1E beta-cells, elevated glucose levels increase the release of arachidonic acid and linoleic acid from membrane phospholipids and promote their peroxidation to HNE. At non cytotoxic concentrations, HNE behaves as an endogenous ligand for nuclear receptor PPAR-δ, stimulating insulin secretion in beta-cells (Cohen et al., 2011). In rat islet beta-cell-derived RINm5F cells, a recent report demonstrates the involvement of the transient receptor potential (TRP) cation channels in the HNE-induced insulin secretion. Short-term (1h) exposure to HNE induces a transient increase in intracellular calcium concentration and triggers insulin secretion. HNE induces calcium influx through activation of TRP channels (amongst which TRPA1) which appears to be coupled with the L-type voltage-dependent calcium channel, and ultimately insulin secretion (Numazawa et al., 2012). Lipid aldehydes should therefore be considered either as detrimental (>10 µM) or as beneficial (sub micromolar range) depending on their actual tissue concentration.

3.3. Direct adduction of the insulin polypeptide

Under conditions of oxidative stress, insulin, a polypeptidic hormone composed of 51 amino acid residues, is exposed to direct oxidative insult or to modification by lipoperoxidation by-products. Several amino acids are putative sites of adduction, and thus, covalent binding of lipid aldehydes affect the biological actions of this hormone. This applies to acrolein and methylglyoxal, whose fixation on insulin has been shown to reduce both hypoglycemic effects in rats and glucose uptake in 3T3-L1 adipocytes (Jia et al., 2006; Medina-Navarro et al., 2007). HHE and HNE, toxic aldehydes generated during lipid peroxidation, also modify the B-chain of human insulin in vitro, predominantly at the His B5 and His B10 residues via Michael adduction (Figure 3). Adduct formation affects the biological activity of insulin in vivo, decreasing its hypoglycemic effect in mice and stimulation of glucose uptake in adipose and muscle cells (Pillon et al., 2011).

Figure 3.

Structure of insulin monomer displaying two HHE adducts on histidine residues. From Pillon et al, 2011.

Advertisement

4. Obesity, insulin resistance and type-2 diabetes

4.1. Lipid peroxidation by-products association with obesity

Obesity is a major factor in the development of metabolic syndrome. After consumption of an energy-dense (i.e. high-fat) diet, plasma HNE levels increase rapidly and significantly within minutes (Devaraj et al., 2008). When consumed regularly, this diet promotes obesity, which suggests a role for HNE very early in the development of obesity. On the other hand, levels of circulating HNE tend to decrease when obese people are maintained on calorie restriction (Johnson et al., 2007), demonstrating that lipid peroxidation is tightly linked to high fat diet and obesity. Furukawa et al. reported that increased oxidative stress in accumulated fat is an important pathogenic mechanism of obesity-associated metabolic syndrome. Production of ROS is indeed selectively increased in white adipose tissue of obese mice and associated with a blunted expression of antioxidant enzymes. In good agreement, fat accumulation correlates with systemic lipid peroxidation in humans (Furukawa et al., 2004), and the plasma concentration of MDA is 1.8 fold higher in subjects with a BMI above 40 kg/m2 compared to lean individuals (Olusi, 2002). Diet-induced obesity increases tissue and plasma accumulation of ALEs (protein–acrolein and protein–HNE adducts for example), suggesting that obesity is associated with an increase in the formation of lipid peroxidation-derived aldehydes (Baba et al., 2011). A significant accumulation of HNE was noticed in the white adipose tissue of obese mice, where the adipocyte fatty acid binding protein (AFABP also known as aP2) is the soluble protein most highly modified by HNE in this tissue (Grimsrud et al., 2007). In obese mice roughly 7% of the AFABP in adipose tissue is covalently modified by HNE resulting in a decreased binding affinity for fatty acids. Lipid peroxidation is however not restricted to adipose tissue since HNE is also elevated in skeletal muscles of Otsuka Long-Evans Tokushima fatty (OLETF) rat, a model for hyperphagic obesity (Morris et al., 2008). Intracellular triglyacylglycerols accumulate in the muscle of obese humans where it is considered as a pathogenic factor in the development of insulin resistance. In obese compared to endurance-trained subjects, the lipid peroxidation to intracellular triacylglycerols ratio was 4-fold higher suggesting that obesity is associated with increased muscle lipid peroxidation (Russell et al., 2003; Vincent et al., 2006).

Chronic, low grade inflammation of white adipose tissue is a hallmark of obesity and a major contributor to oxidative stress and lipid peroxidation (Wellen & Hotamisligil, 2003). In the expanding adipose tissue, hypertrophied adipocytes contribute to the inflammation by up-regulating the expression and release of pro-inflammatory cytokines. In 3T3-L1 adipocytes, HNE can dose-dependently increase the expression of the inducible cyclooxygenase (COX-2) (Zarrouki et al., 2007) and that of the plasminogen activator inhibitor-1 (PAI-1). In the meantime, HNE decreases the expression of the anti-inflammatory, insulin-sensitizing hormone adiponectin (Soares et al., 2005; Wang et al., 2012), therefore linking lipid peroxidation by-products and chronic inflammation.

4.2. Lipid peroxidation by-products association with type-2 diabetes

Epidemiological studies demonstrates that fasting glycemia is positively correlated with oxidative stress markers such as 8-epi-PGF2α and TBARs and negatively correlated with plasma glutathione (Trevisan et al., 2001; Menon et al., 2004). In type-2 diabetic individuals, 8-epi-PGF2α is positively correlated to the HOMA index for insulin resistance (Gopaul et al., 2001) and urinary acrolein correlates with glycated haemoglobin HbA1c (Daimon et al., 2003). This was confirmed in animal models of insulin resistance which exhibit increased markers of oxidative stress, such as plasma F2-isoprostanes (Laight et al., 1999a). In parallel, antioxidant defences are reduced during an oral glucose tolerance test in normal and non-insulin-dependent diabetic subjects (Ceriello et al., 1998), and diabetes is associated with decreased vitamin C and glutathione (Maxwell et al., 1997; Dierckx et al., 2003). Reciprocally, an intensive treatment of diabetes improves circulating levels of H2O2 and MDA (Wierusz-Wysocka et al., 1995); and improved insulin sensitivity resulting from exercise and/or dietary restriction is associated with reduced levels of lipid peroxidation products (Reviewed by Vincent et al., 2007). On the other hand, insulin sensitivity can be improved through antioxidant or carbonyl scavenging treatment (Kamenova, 2006; Vincent et al., 2009), demonstrating the tight link existing between oxidative stress, oxidation by-products and insulin resistance.

Figure 4.

Plasma MDA concentration (µM) and blood GSH content (mmol/g haemoglobin) in healthy and type-2 diabetic (T2D) individuals. Results are a meta-analysis from 23 different publications; one dot represents the mean value obtained in one study

Results from Wierusz-Wysocka et al., 1995; Vijayalingam et al., 1996; Feillet-Coudray et al., 1999; Rábago-Velasco et al., 2000; Rizvi & Zaid, 2001; Seghrouchni et al., 2002; Dinçer et al., 2002; Dierckx et al., 2003; Duman et al., 2003; Pasaoglu et al., 2004; Memişoğullari & Bakan, 2004; Skrha et al., 2005; Kurtul et al., 2005; Ozdemir et al., 2005; Mahboob et al., 2005; Saxena et al., 2005; Sampathkumar et al., 2005; Kuppusamy et al., 2005; Lapolla et al., 2007; Sathiyapriya et al., 2007; Singhania et al., 2008; Jain et al., 2009; Nakhjavani et al., 2010; Narasimhan et al., 2010; Shinde et al., 2011; Huang et al., 2011; Bahadoran et al., 2011; Zhang et al., 2011; Pácal et al., 2011; Calabrese et al., 2011 and Rasic-Milutinovic et al., 2012

. In average, MDA is significantly increased by 60% while GSH is decreased by 25% in T2D compared to healthy subjects (paired student t-test, n=21).

Among α,β-unsaturated aldehydes, only MDA and to some extend HNE have been studied as oxidative stress biomarkers in diabetes, and they are indeed both increased up to 2-fold in both human (Figure 4) and animal models of type-2 diabetes (Wierusz-Wysocka et al., 1995; Dierckx et al., 2003). It has moreover been demonstrated that Type-2 diabetes duration is independently associated with increased levels of lipid peroxidation (Nakhjavani et al., 2010), and our group recently showed an increase in HHE concentration in type-2 diabetes patients, reinforcing existing evidence for the specific role of lipid aldehydes in insulin resistance.

Aldehydes concentration is increased in several tissues during diabetes, as revealed by increased levels of HNE in pancreas, liver, brain and heart. Pancreatic islets from type-2 diabetic patients are positively stained with HNE, suggesting that oxidative stress through lipid peroxidation could contribute to the reduced beta-cell mass and islet-cell injury (Sakuraba et al., 2002). An increased level of HNE-modified proteins is reported in the pancreatic beta cells of Goto Kakizaki rats as a result of hyperglycaemia (Ihara et al., 1999); and accumulation of HNE is observed in liver of diabetic rats due to the impairment of HNE-metabolizing enzymes (Traverso et al., 1998, 2002). Diabetic mice under a high fat diet exhibit increased HNE adducts levels in temporal lobes relative to control (Lyn-Cook et al., 2009), and HNE conjugation of GLUT3, the glucose transporter present in neurons, is increased in the hippocampus of diabetic rats subjected to stress (Reagan et al., 2000). When db/db obese mice are fed a Western diet containing 21% fat and 0.15% cholesterol, they develop obesity, hyperglycemia, and insulin resistance. In this situation, HNE is significantly elevated in the left ventricular myocardium of diabetic mice compared to their lean littermates (Yamashita et al., 2010). Finally, hyperglycemic Zucker Diabetic Fatty (ZDF) rats exhibited a 8-fold increase in plasma HDDE concentration compared to their lean non diabetic counterparts (Riahi et al., 2010).

4.3. Causative role for lipid peroxidation by-products in the metabolic syndrome

This body of evidence linking oxidative stress with the metabolic syndrome is however only based on correlations and do not decipher the mechanisms and/or the causative role of oxidative stress in diabetes. There are consequently two main hypotheses:

  1. First is chronic hyperglycemia which then leads to oxidative stress. In this case, lipid peroxidation products would be by-products of this oxidative stress and a consequence of diabetes, even if contributing to the progression of the disease and its complications.

  2. First is oxidative stress which precedes the development of diabetes and plays a causative role in its development. In this hypothesis, deregulation of antioxidant defences and increased oxidative stress would lead to accumulation of secondary by-products consequently inducing insulin resistance.

We will focus in this chapter on the second hypothesis for which evidences have been recently accumulating from human, animal and cell culture studies.

Oxidative stress, through reactive oxygen species can positively and negatively regulate insulin signalling, depending on time, dose, model and free radical used (for review, see Bashan et al., 2009). It is however admitted that prolonged oxidative stress impairs insulin signalling, insulin-induced GLUT4 translocation and glucose uptake. This occurs through several mechanisms including, but not limited to IRS inhibitory phosphorylation, MAPK activation and endoplasmic reticulum stress. This body of evidence for the role of oxidative stress in insulin resistance has been demonstrated in adipocytes, muscle, liver and cardiac cells (Rudich et al., 1998; Bloch-Damti et al., 2006; JeBailey et al., 2007; Singh et al., 2008a; Shibata et al., 2010; Tan et al., 2011). Even in vivo, a pro-oxidant challenge provokes the onset of type-2 diabetes in insulin resistant rats (Laight et al., 1999b) and chronic methylglyoxal infusion by minipump causes pancreatic beta-cell dysfunction and induces type-2 diabetes in Sprague-Dawley rats (Dhar et al., 2011).

The role of lipid aldehydes in vivo during metabolic diseases is often indirectly assessed, and the most compelling evidence for a causative role of aldehydes comes from polymorphisms in the glutathione-S-transferase (GST) gene. This family of enzymes is responsible for the detoxification of aldehydes through conjugation to glutathione; and several deletion polymorphisms leading to blunted enzyme activity have been described. Patients carrying certain null GST polymorphisms had up to 3-fold increased incidence of type-2 diabetes mellitus compared to those with normal genotypes (Amer et al., 2011). Accordingly, the expression of the GST4A is blunted in the adipose tissue of obese insulin resistant subjects (Curtis et al., 2010).

Figure 5.

The obesity – lipid peroxidation vicious circle

This same observation was made in animals model, were it has been suggested that excessive production of HNE might be sufficient to cause obesity and the metabolic syndrome. Mice lacking the gene encoding the HNE-conjugating enzyme mGSTA4-4 develop obesity and insulin resistance, unsurprisingly associated with HNE accumulation in multiple tissues (Singh et al., 2008b). Cell culture work further shows that dysfunction of glutathione S-transferase or its ablation by siRNA leads to excess HNE accumulation, increased protein carbonylation, oxidative stress, and mitochondrial dysfunction (Kostyuk et al., 2010; Curtis et al., 2010). On the other hand, overexpression of glutathione-S-transferase A4-4 protects against oxidative stress and HNE-induced apoptosis (Vaillancourt et al., 2008). Additional findings suggest that HNE causes fat accumulation by promoting fatty acid synthesis and suppressing fatty acid beta-oxidation. Interestingly, the phenotype of mGSTA4-4 null mice is strain dependent: mGSTA4-4 null mice with 129/sv genetic background exhibit both increased accumulation of HNE and obesity while those with C57Bl6 genetic background are lean and HNE is unchanged (Singh et al., 2008b). In good agreement, silencing of the mGSTA4-4 gene in the nematode Caenorhabditis elegans also results in an accumulation of lipid peroxidation by-products and a fatty phenotype (Singh et al., 2009). When HNE is experimentally increased in the nematode, either by genetic deletion or through direct exposure, it promotes fat accumulation. The mechanism involves HNE inhibition of mitochondrial aconitase leading to an accumulation of malonyl CoA, precursor of fatty acid synthesis (Zimniak, 2010). Taken together, these data suggest that accumulation of lipid aldehydes and fat deposition could be mutually inductive leading to a vicious circle promoting fat accretion (Figure 5).

The direct causative role of lipid peroxidation by-products in insulin resistance has also been recently evidenced in cell cultures studies. A first study identified that methylglyoxal, an aldehyde by-product of glucose oxidation, can impair the insulin signalling pathways independently of the formation of intracellular reactive oxygen species (Riboulet-Chavey et al., 2006). Then, focusing on lipid peroxidation products, two studies demonstrated that HNE can induce insulin resistance in adipocytes and muscle cell through inhibition of IRS and Akt signalling, as well as insulin-induced glucose uptake (Demozay et al., 2008; Pillon et al., 2012). These two studies identified carbonyl stress (notably IRS1 adduction) and ROS production as the possible mechanisms responsible for HNE effects; a third one being possibly the adduction of Akt2, which inhibits insulin-dependent Akt signalling in HepG2 cells (Shearn et al., 2011). Altogether, these studies strongly suggest that excessive production of aldehydes might be sufficient to cause obesity, diabetes and the metabolic syndrome.

Advertisement

5. Preventing the deleterious effects of aldehydes

It has been known for decades that supplementation in α-lipoic acid in type-2 diabetic subject improves glucose tolerance and insulin sensitivity (Jacob et al., 1996). Similarly, increased intake of Vitamin E in obese and insulin resistant patients reduces fasting glycemia and the HOMA index for insulin resistance, and this is correlated with a decrease in the concentration of peroxides in plasma (Manning et al., 2004). This adds further evidence for a causative role of oxidative stress in the metabolic syndrome, and opens new therapeutic perspectives using antioxidant and/or scavenging of toxic aldehydes.

5.1. Glutathione and enzymatic detoxification

Glutathione (GSH) is a tripeptide (L-γ-glutamyl-L-cysteinyl-glycine) which is present in high concentration in the cytoplasm of living cells: 5-50 nmol/mg proteins (Jungas et al., 2002; Dominy et al., 2007). GSH is an important coenzyme of several enzymatic reactions, and exerts its antioxidant activity mainly through regeneration of vitamin E, and also through direct interaction with free radical and aldehydes. GSH is obviously necessary for the activity of glutathione-S-transferases (GSTs), a family of enzymes responsible for detoxification of electrophile by-products, such as the ones derived from lipid peroxidation, and GSH exhibits indeed a high reactivity for HNE. Though HNE is electrophile enough to spontaneously react with GSH, this reaction is dramatically accelerated via the conjugation process catalyzed by GSTs (Alin et al., 1985). As described above, mice lacking GSTs develop obesity and insulin resistance (Singh et al., 2008b), and humans carrying null polymorphisms for GST have a 3-fold increased risk of having type-2 diabetes (Amer et al., 2011), pointing out the important role of GSH in the metabolic syndrome. As addition reaction to GSH contributes to the detoxification of aldehydes, pharmacological strategies to increase glutathione pools or GST activity should be protective against aldehydes. Several studies were indeed successful in protecting cultured cells from the deleterious effects of oxidative stress through an increase in intracellular pools of reduced glutathione. This strategy was particularly efficient to protect the cells against the deleterious effects of HNE (Yadav et al., 2008; Jia et al., 2009b; Pillon et al., 2012).

In addition to glutathione-S-transferases, several enzymes are also responsible for the detoxification of aldehydes: aldehyde dehydrogenase, alcohol dehydrogenase (Hartley et al., 1995), aldose reductase (Srivastava et al., 2000) and fatty aldehyde dehydrogenase (Demozay et al., 2008). These enzymes however participate to a lower extent in the metabolism of aldehydes, compared to glutathione and GST. Recent data indicate that if HHE and HNE are both metabolized via glutathione, the effectiveness of detoxification differs for these two molecules. By extension, aldehydes may be metabolized with different affinities and efficiencies by detoxification enzymes, which could explain some differences in their respective toxicities (Long et al., 2010).

5.2. Scavenging

In chemistry, a scavenger is a chemical substance able to remove or inactivate impurities or unwanted reaction products. In living cells, a scavenger is, by extension, a molecule able to inactivate toxic compounds such as ROS and aldehydes, therefore preventing their deleterious effects. In the case of aldehydes, a scavenger would be a strong nucleophile molecule on which HHE, HNE or any other aldehyde would form a covalent adduct. Consequently, most of the aldehyde scavengers are amino- or sulphur-containing drugs such as N-acetyl-cysteine (NAC), hydralazine, S-adenosyl-methionine (SAM) aminoguanidine (AGD) and α-lipoic acid, the latter being tested in several human studies for treatment of type-2 diabetes.

5.2.1. α-lipoic acid

α-lipoic acid is a natural compound found in many foodstuffs (such as potatoes, broccoli and meat), but in rather low amount. The effect of lipoic acid was demonstrated in animal models, where it enhances insulin-stimulated glucose metabolism in skeletal muscle from insulin-resistant rat (Jacob et al., 1996). It was rapidly tested in several clinical studies which demonstrated its beneficial effects in type-2 diabetes through a decrease in fasted blood glucose, enhancement of glucose disposal, improved insulin sensitivity and decreased insulin resistance (Jacob et al., 1995). These results were confirmed in vitro, where α-lipoic acid prevents the development of glucose-induced insulin resistance in adipocytes (Greene et al., 2001) and were reproduced by many independent studies. Treatment with lipoic acid decreases oxidative stress in both adipocytes and muscle cells (Rudich et al., 1999; Maddux et al., 2001) and also decreases lipid peroxidation markers in insulin resistant rats (Thirunavukkarasu & Anuradha, 2004), suggesting that the improvement of insulin sensitivity is due to its antioxidant properties. Hence, the current literature supports the use of alpha lipoic acid for the treatment of diabetes complications and it consequently became the first antioxidant supplement used for the treatment of diabetes complications, being already approved in Germany for the treatment of diabetic neuropathy.

5.2.2. N-acetylcysteine (NAC)

N-acetylcysteine is a cysteine derivative and a potent antioxidant. Its properties are mainly due to its thiol group able to reduce free radicals as well as its role as a precursor in the formation of glutathione (Zafarullah et al., 2003). NAC exhibits highly protective scavenging properties against aldehydes and protects against MDA increase and GSH decrease in animal models of insulin resistance. NAC is able to improve insulin sensitivity in healthy rats (Figure 6) and reverses insulin resistance and aldehyde-induced hypertension in rats (Haber et al., 2003). In cell culture studies, NAC can prevent the insulin resistance induced by HNE in muscle cells (Pillon et al., 2012), as well as the one induced by advanced glycation end products in adipocytes (Unoki et al., 2007), thus confirming the important role NAC can play in improving both oxidative stress parameters and insulin resistance.

Figure 6.

Insulin sensitizing effect of N-acetylcysteine. Wistar rats were given NAC in drinking water for one week (total intake was 225 mg.kg-1.day-1). Insulin sensitivity was calculated using a standard insulin tolerance test

Animals fasted overnight were then injected intraperitoneally with 0.5 UI/kg body weight of insulin. Plasma glucose was measured from tail vein blood using a glucometer at 0, 20, 40, 60, and 120 min following the injection. Glucose disappearance rate for ITT (KITT; %/min) was calculated as: KITT=(0.693x100)/t1/2, where t1/2 was calculated from the slope of the plasma glucose concentration, considering an exponential decrement of glucose concentration during the 20 min after insulin administration. Higher insulin sensitivity index (KITT) scores mean higher response of tissues to insulin.

. Results are average ± SEM from 5 different animals per group, expressed as percent of basal glycemia. From Pillon et al. unpublished results.

Results from clinical trials in type-2 diabetic subjects show that NAC is able to decrease oxidative stress parameters, increase GSH and decrease plasma VCAM-1 (De Mattia et al., 1998); moreover, long-term N-acetylcysteine administration reduces endothelial activation and is proposed as a potential antiatherogenic therapy (Martina et al., 2008). Despite the fact that NAC can improve insulin sensitivity in women with polycystic ovary syndrome (Fulghesu et al., 2002) and this body of evidence suggesting that NAC may slow down the progression of diabetic complications, to date no clinical trial demonstrated any significant benefit of its supplementation in diabetes.

5.2.3. Aminoguanidine (AGD)

Aminoguanidine is a highly nucleophilic agent which reacts in vitro and in vivo with aldehydes, therefore protecting against the deleterious effects of ALE precursors (Peyroux & Sternberg, 2006). It is also an antioxidant able to quench hydroxyl radicals and in vivo. AGD in drinking water decreases lipid peroxidation in type-1 diabetic rats and rabbits (Ihm et al., 1999). In experimental animal models of diabetes, AGD demonstrates significant effects in protecting against pathological complications, such as diabetic nephropathies, atherosclerosis and neurovascular complications (El Shazly et al., 2009). Consequently, several clinical trials in humans have been designed to evaluate AGD efficiency but they demonstrate only mild effects and were not conclusive, partly because of side-effects, and of weak carbonyl scavenger effects in human vascular tissues (Bolton et al., 2004).

5.2.4. Hydralazine

Primarily used as an antihypertensive drug, hydralazine exhibits a pronounced nucleophilicity and is consequently very efficient in scavenging several aldehydes (acrolein, HNE) and ketones, as well as aldehyde-adducted proteins (Burcham et al., 2002). It is also a powerful antioxidant, able to inhibit the generation of ROS (Münzel et al., 1996). Its scavenging activity in vivo was demonstrated by its ability to reverse the formation of HNE and acrolein adducts on tissue proteins in atherosclerotic aortas of hypercholesterolemic animals (Vindis et al., 2006), but its effects on insulin resistance and diabetes are to date uncharacterized.

5.2.5. S-adenosyl-methionine (SAM)

In living organisms, SAM is endogenously synthesized from methionine in every cell, but the liver is the major site of its synthesis and degradation. SAM is an important precursor for cysteine and glutathione production (Lu, 2000) and its involvement in several metabolic pathways makes it essential for a wide spectrum of cellular processes. SAM inhibits both HNE production and adducts formation and efficiently prevents high-fat diet-induced non-alcoholic steatohepatitis in rats (Lieber et al., 2007), even if its direct effects on insulin resistance are still unknown.

Advertisement

6. Conclusion

Lipid peroxidation by-products are associated with metabolic diseases, but their primary role during obesity and diabetes is subject to debate. In vitro and in vivo animal studies highlighted that an aldehyde challenge, through a state of carbonyl stress, affects several steps involved in the development of obesity and type-2 diabetes (Figure 7). On insulin-sensitive tissues (muscle, adipose tissue), aldehydes lead to insulin resistance, while in pancreas, aldehydes impair insulin secretion. Together with the carbonylation of the insulin peptide itself, aldehydes could contribute to the defects in insulin action, leading to the metabolic syndrome.

Figure 7.

Lipid aldehydes effects leading to the metabolic syndrome.

Despite this evidence, the role of aldehydes is mitigated by the mild effect obtained with antioxidants and/or aldehyde scavengers in the treatment of diabetic complications in human. “Oxidative stress” and its derivatives are nevertheless important in the metabolic syndrome, and prevention or treatment of some of its associated complications could be accessed through reduction of both ROS and toxic aldehydes by-products, as it is the case with lipoic acid.

References

  1. 1. AlinP.DanielsonU. H.MannervikB.1985Hydroxyalk-2-enals are substrates for glutathione transferase. FEBS Lett 179267270
  2. 2. Amer MA, Ghattas MH, Abo-Elmatty DM & Abou-El-Ela SH2011Influence of glutathione S-transferase polymorphisms on type-2 diabetes mellitus risk. Genet Mol Res; DOI:October.31.14.
  3. 3. Augusti KT & Sheela CG1996Antiperoxide effect of S-allyl cysteine sulfoxide, an insulin secretagogue, in diabetic rats. Experientia 52115120
  4. 4. BabaS. P.HellmannJ.SrivastavaS.BhatnagarA.2011Aldose reductase (AKR1B3) regulates the accumulation of advanced glycosylation end products (AGEs) and the expression of AGE receptor (RAGE). Chem Biol Interact 191357363
  5. 5. BaeE. H.ChoS.JooS. Y.MaKimS. K.LeeS. H.J.KimS. W.2011Hydroxy-2-hexenal-induced apoptosis in human renal proximal tubular epithelial cells. Nephrol Dial Transplant 2638663873
  6. 6. BashanN.KovsanJ.KachkoI.OvadiaH.RudichA.2009Positive and negative regulation of insulin signaling by reactive oxygen and nitrogen species. Physiol Rev 892771
  7. 7. Bloch-DamtiA.PotashnikR.GualP.Le Marchand-BrustelY.TantiJ. F.RudichA.BashanN.2006Differential effects of IRS1 phosphorylated on Ser307 or Ser632 in the induction of insulin resistance by oxidative stress. Diabetologia 4924632473
  8. 8. BodurC.KutukO.TezilT.BasagaH.2012Inactivation of Bcl-2 through IκB kinase (IKK)-dependent phosphorylation mediates apoptosis upon exposure to 4hydroxynonenal (HNE). J Cell Physiol; DOI:jcp.24057.
  9. 9. BoltonW. K.CattranD. C.MEWilliamsAdler. S. G.AppelG. B.CartwrightK.FoilesP. G.FreedmanB. I.RaskinP.RatnerR. E.BSSpinowitzWhittier. F. C.Wuerth-PJ.2004Randomized trial of an inhibitor of formation of advanced glycation end products in diabetic nephropathy. Am J Nephrol 243240
  10. 10. Burcham PC, Kaminskas LM, Fontaine FR, Petersen DR & Pyke SM2002Aldehyde-sequestering drugs: tools for studying protein damage by lipid peroxidation products. Toxicology 181-182, 229-236.
  11. 11. CahuanaG. M.TejedoJ. R.JimenezJ.RamirezR.SobrinoF.BedoyaF. J.2003Involvement of advanced lipooxidation end products (ALEs) and protein oxidation in the apoptotic actions of nitric oxide in insulin secreting RINm5F cells. Biochem Pharmacol 6619631971
  12. 12. CalzadaC.ColasR.GuillotN.GuichardantM.LavilleM.VéricelE.LagardeM.2010Subgram daily supplementation with docosahexaenoic acid protects low-density lipoproteins from oxidation in healthy men. Atherosclerosis 208467472
  13. 13. CerielloA.BortolottiN.CrescentiniA.MotzE.LizzioS.RussoA.EzsolZ.TonuttiL.TabogaC.1998Antioxidant defences are reduced during the oral glucose tolerance test in normal and non-insulin-dependent diabetic subjects. Eur J Clin Invest 28329333
  14. 14. ChengJ.WangF.Yu-FD.Wu-FP.Chen-GJ.2011The cytotoxic mechanism of malondialdehyde and protective effect of carnosine via protein cross-linking/mitochondrial dysfunction/reactive oxygen species/MAPK pathway in neurons. Eur J Pharmacol 650184194
  15. 15. ChoudharyS.ZhangW.ZhouF.CampbellG. A.ChanL. L.ThompsonE. B.AnsariN. H.2002Cellular lipid peroxidation end-products induce apoptosis in human lens epithelial cells. Free Radic Biol Med 32360369
  16. 16. CohenG.RiahiY.ShamniO.GuichardantM.ChatgilialogluC.FerreriC.KaiserN.SassonS.2011Role of lipid peroxidation and PPAR-δ in amplifying glucose-stimulated insulin secretion. Diabetes 6028302842
  17. 17. Coleman JD, Prabhu KS, Thompson JT, Reddy PS, Peters JM, Peterson BR, Reddy CC & Vanden Heuvel JP2007The oxidative stress mediator 4-hydroxynonenal is an intracellular agonist of the nuclear receptor peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta). Free Radic Biol Med 4211551164
  18. 18. CurtisJ. M.GrimsrudP. A.WrightW. S.XuX.FonceaR. E.GrahamD. W.BrestoffJ. R.BMWiczerIlkayeva. O.CianfloneK.MuoioD. E.ArriagaE. A.BernlohrD. A.2010Downregulation of adipose glutathione S-transferase A4 leads to increased protein carbonylation, oxidative stress, and mitochondrial dysfunction. Diabetes 5911321142
  19. 19. DaimonM.SugiyamaK.KamedaW.SaitohT.OizumiT.HirataA.YamaguchiH.OhnumaH.IgarashiM.KatoT.2003Increased urinary levels of pentosidine, pyrraline and acrolein adduct in type 2 diabetes. Endocr J 506167
  20. 20. DemozayD.Mas-CJ.RocchiS.Van ObberghenE.2008FALDH reverses the deleterious action of oxidative stress induced by lipid peroxidation product 4-hydroxynonenal on insulin signaling in 3T3-L1 adipocytes. Diabetes 5712161226
  21. 21. DevarajS.Wang-PolagrutoJ.PolagrutoJ.KeenC. L.JialalI.2008High-fat, energy-dense, fast-food-style breakfast results in an increase in oxidative stress in metabolic syndrome. Metab Clin Exp 57867870
  22. 22. DharA.DharI.JiangB.DesaiK. M.WuL.2011Chronic methylglyoxal infusion by minipump causes pancreatic beta-cell dysfunction and induces type 2 diabetes in Sprague-Dawley rats. Diabetes 60899908
  23. 23. DierckxN.HorvathG.van GilsC.VertommenJ.van de VlietJ.De LeeuwI.Manuel-y-KeenoyB.2003Oxidative stress status in patients with diabetes mellitus: relationship to diet. Eur J Clin Nutr 579991008
  24. 24. DominyJ. E.HwangJ.StipanukM. H.2007Overexpression of cysteine dioxygenase reduces intracellular cysteine and glutathione pools in HepG2/C3A cells. Am J Physiol Endocrinol Metab 293, E6269
  25. 25. Dong J-Z & Moldoveanu SC2004Gas chromatography-mass spectrometry of carbonyl compounds in cigarette mainstream smoke after derivatization with 24dinitrophenylhydrazine. J Chromatogr A 1027, 25-35.
  26. 26. El Shazly AHM, Mahmoud AM & Darwish NS2009Potential prophylactic role of aminoguanidine in diabetic retinopathy and nephropathy in experimental animals. Acta Pharm 596773
  27. 27. FulghesuA. M.CiampelliM.MuzjG.BelosiC.SelvaggiL.AyalaG. F.LanzoneA.2002N-acetyl-cysteine treatment improves insulin sensitivity in women with polycystic ovary syndrome. Fertil Steril 7711281135
  28. 28. FurukawaS.FujitaT.ShimabukuroM.IwakiM.YamadaY.NakajimaY.NakayamaO.MakishimaM.MatsudaM.ShimomuraI.2004Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest 11417521761
  29. 29. GopaulN. K.MDManrajHebe. A.LeeKwai.YanS.JohnstonA.MJCarrierEEAnggard2001Oxidative stress could precede endothelial dysfunction and insulin resistance in Indian Mauritians with impaired glucose metabolism. Diabetologia 44706712
  30. 30. Greene EL, Nelson BA, Robinson KA & Buse MG2001alpha-Lipoic acid prevents the development of glucose-induced insulin resistance in 3T3-L1 adipocytes and accelerates the decline in immunoreactive insulin during cell incubation. Metab Clin Exp 5010631069
  31. 31. Grimsrud PA, Picklo MJ, Griffin TJ & Bernlohr DA2007Carbonylation of adipose proteins in obesity and insulin resistance: identification of adipocyte fatty acid-binding protein as a cellular target of 4-hydroxynonenal. Mol Cell Proteomics 6624637
  32. 32. GuichardantM.BacotS.MoliereP.LagardeM.2006Hydroxy-alkenals from the peroxidation of n-3 and n-6 fatty acids and urinary metabolites. Prostaglandins Leukot Essent Fatty Acids 75179182
  33. 33. Gutteridge JM1982Free-radical damage to lipids, amino acids, carbohydrates and nucleic acids determined by thiobarbituric acid reactivity. Int J Biochem 14649653
  34. 34. CAHaberLam. T. K. T.YuZ.GuptaN.GohT.BogdanovicE.GiaccaA.FantusI. G.2003N-acetylcysteine and taurine prevent hyperglycemia-induced insulin resistance in vivo: possible role of oxidative stress. Am J Physiol Endocrinol Metab 285, E744753
  35. 35. Hartley DP, Ruth JA & Petersen DR1995The hepatocellular metabolism of 4-hydroxynonenal by alcohol dehydrogenase, aldehyde dehydrogenase, and glutathione S-transferase. Arch Biochem Biophys 316197205
  36. 36. Hipkiss AR, Preston JE, Himswoth DT, Worthington VC & Abbot NJ1997Protective effects of carnosine against malondialdehyde-induced toxicity towards cultured rat brain endothelial cells. Neurosci Lett 238135138
  37. 37. IharaY.ToyokuniS.UchidaK.OdakaH.TanakaT.IkedaH.HiaiH.SeinoY.YamadaY.1999Hyperglycemia causes oxidative stress in pancreatic beta-cells of GK rats, a model of type 2 diabetes. Diabetes 48927932
  38. 38. IhmS. H.YooH. J.ParkS. W.IhmJ.1999Effect of aminoguanidine on lipid peroxidation in streptozotocin-induced diabetic rats. Metab Clin Exp 4811411145
  39. 39. JacobS.HenriksenE. J.SchiemannA. L.SimonI.ClancyD. E.TritschlerH. J.JungW. I.AugustinH. J.DietzeG. J.1995Enhancement of glucose disposal in patients with type 2 diabetes by alpha-lipoic acid. Arzneimittelforschung 45872874
  40. 40. JacobS.StreeperR. S.FogtD. L.HokamaJ. Y.TritschlerH. J.DietzeG. J.HenriksenE. J.1996The antioxidant alpha-lipoic acid enhances insulin-stimulated glucose metabolism in insulin-resistant rat skeletal muscle. Diabetes 4510241029
  41. 41. JeJ. H.LeeJ. Y.JungK. J.SungB.GoE. K.YuB. P.ChungH. Y.2004NF-kappaB activation mechanism of 4-hydroxyhexenal via NIK/IKK and 38MAPK pathway. FEBS Lett 566, 183-189.
  42. 42. JeBailey. L.WanonoO.NiuW.RoesslerJ.RudichA.KlipA.2007Ceramide- and oxidant-induced insulin resistance involve loss of insulin-dependent Rac-activation and actin remodeling in muscle cells. Diabetes 56394403
  43. 43. JiC.CARouzerMarnett. L. J.PietenpolJ. A.1998Induction of cell cycle arrest by the endogenous product of lipid peroxidation, malondialdehyde. Carcinogenesis 1912751283
  44. 44. JiaL.ZhangZ.ZhaiL.BaiY.2009aProtective effect of lipoic acid against acrolein-induced cytotoxicity in IMR-90 human fibroblasts. J Nutr Sci Vitaminol 55126130
  45. 45. JiaX.OlsonD. J. H.RossA. R. S.WuL.2006Structural and functional changes in human insulin induced by methylglyoxal. FASEB J 2015551557
  46. 46. JiaZ.MisraB. R.ZhuH.LiY.MisraH. P.2009bUpregulation of cellular glutathione by 3H-12dithiole-3-thione as a possible treatment strategy for protecting against acrolein-induced neurocytotoxicity. Neurotoxicology 30, 1-9.
  47. 47. JohnsonJ. B.SummerW.CutlerR. G.MartinB.Hyun-HD.DixitV. D.PearsonM.NassarM.TelljohannR.TellejohanR.MaudsleyS.CarlsonO.JohnS.LaubD. R.MattsonM. P.2007Alternate day calorie restriction improves clinical findings and reduces markers of oxidative stress and inflammation in overweight adults with moderate asthma. Free Radic Biol Med 42665674
  48. 48. JungasT.MottaI.DuffieuxF.FanenP.StovenV.OjciusD. M.2002Glutathione levels and BAX activation during apoptosis due to oxidative stress in cells expressing wild-type and mutant cystic fibrosis transmembrane conductance regulator. J Biol Chem 2772791227918
  49. 49. KamenovaP.2006Improvement of insulin sensitivity in patients with type 2 diabetes mellitus after oral administration of alpha-lipoic acid. Hormones (Athens) 5251258
  50. 50. KostyukV.PotapovichA.CesareoE.BresciaS.GuerraL.ValacchiG.PecorelliA.RaskovicD.De LucaC.PastoreS.KorkinaL.2010Dysfunction of Glutathione S-Transferase Leads to Excess 4Hydroxy-2-Nonenal and H2O2 and Impaired Cytokine Pattern in Cultured Keratinocytes and Blood of Vitiligo Patients. Antioxid Redox Signal; DOI:ars.2009.2976.
  51. 51. Laight DW, Desai KM, Gopaul NK, Anggard EE & Carrier MJ1999aF2-isoprostane evidence of oxidant stress in the insulin resistant, obese Zucker rat: effects of vitamin E. Eur J Pharmacol 3778992
  52. 52. Laight DW, Desai KM, Gopaul NK, Anggard EE & Carrier MJ1999bPro-oxidant challenge in vivo provokes the onset of NIDDM in the insulin resistant obese Zucker rat. Br J Pharmacol 128269271
  53. 53. Lee JY, Je JH, Jung KJ, Yu BP & Chung HY2004Induction of endothelial iNOS by 4-hydroxyhexenal through NF-kappaB activation. Free Radic Biol Med 37539548
  54. 54. Lee JY, Jung GY, Heo HJ, Yun MR, Park JY, Bae SS, Hong KW, Lee WS & Kim CD2006Hydroxynonenal induces vascular smooth muscle cell apoptosis through mitochondrial generation of reactive oxygen species. Toxicol Lett 166212221
  55. 55. LeeS. E.JeongS. I.Kim-DG.YangH.Park-SC.Jin-HY.ParkY. S.2011Upregulation of heme oxygenase-1 as an adaptive mechanism for protection against crotonaldehyde in human umbilical vein endothelial cells. Toxicol Lett 201240248
  56. 56. LiD.FerrariM.EllisE. M.2011Human aldo-keto reductase AKR7A2 protects against the cytotoxicity and mutagenicity of reactive aldehydes and lowers intracellular reactive oxygen species in hamster 79-4cells. Chemico-Biological Interactions; DOI:j.cbi.2011.09.007.
  57. 57. LieberC. S.MALeoCao. Q.MakK. M.RenC.PonomarenkoA.WangX.DecarliL. M.2007The Combination of S-adenosylmethionine and Dilinoleoylphosphatidylcholine Attenuates Non-alcoholic Steatohepatitis Produced in Rats by a High-Fat Diet. Nutr Res 27565573
  58. 58. LiuW.KatoM.AAAkhandHayakawa. A.SuzukiH.MiyataT.KurokawaK.HottaY.IshikawaN.NakashimaI.20004hydroxynonenal induces a cellular redox status-related activation of the caspase cascade for apoptotic cell death. J Cell Sci 113 ( Pt 4), 635-641.
  59. 59. LiuX.YangZ.PanX.ZhuM.XieJ.2010Crotonaldehyde induces oxidative stress and caspase-dependent apoptosis in human bronchial epithelial cells. Toxicol Lett 1959098
  60. 60. LongE. K.MurphyT. C.LeiphonL. J.WattJ.JDMorrowMilne. G. L.HowardJ. R. H.MJPicklo2008Trans-4-hydroxy-2-hexenal is a neurotoxic product of docosahexaenoic (22:6; n-3) acid oxidation. J Neurochem 105714724
  61. 61. Long EK, Rosenberger TA & Picklo MJ2010Ethanol withdrawal increases glutathione adducts of 4-hydroxy-2-hexenal but not 4-hydroxyl-2-nonenal in the rat cerebral cortex. Free Radic Biol Med 48384390
  62. 62. Lovell MA, Ehmann WD, Mattson MP & Markesbery WR1997Elevated 4-hydroxynonenal in ventricular fluid in Alzheimer’s disease. Neurobiol Aging 18457461
  63. 63. Lu SC2000S-Adenosylmethionine. Int J Biochem Cell Biol 32391395
  64. 64. LuoJ.RobinsonJ. P.ShiR.2005Acrolein-induced cell death in PC12 cells: role of mitochondria-mediated oxidative stress. Neurochem Int 47449457
  65. 65. Lyn-CookL. E.Jr LawtonM.TongM.SilbermannE.LongatoL.JiaoP.MarkP.WandsJ. R.XuH.de la MonteS. M.2009Hepatic ceramide may mediate brain insulin resistance and neurodegeneration in type 2 diabetes and non-alcoholic steatohepatitis. J Alzheimers Dis 16715729
  66. 66. BAMadduxSee. W.LawrenceJ. C.Jr GoldfineA. L.GoldfineI. D.EvansJ. L.2001Protection against oxidative stress-induced insulin resistance in rat L6 muscle cells by mircomolar concentrations of alpha-lipoic acid. Diabetes 50404410
  67. 67. ManiniP.AndreoliR.SforzaS.Dall’AstaC.GalavernaG.MuttiA.NiessenW. M. A.2010Evaluation of Alternate Isotope-Coded Derivatization Assay (AIDA) in the LC-MS/MS analysis of aldehydes in exhaled breath condensate. J Chromatogr B Analyt Technol Biomed Life Sci 87826162622
  68. 68. Manning PJ, Sutherland WHF, Walker RJ, Williams SM, De Jong SA, Ryalls AR & Berry EA2004Effect of high-dose vitamin E on insulin resistance and associated parameters in overweight subjects. Diabetes Care 2721662171
  69. 69. MartinaV.MashaA.GigliardiV. R.BrocatoL.ManzatoE.BerchioA.MassarentiP.SettanniF.Della CasaL.BergaminiS.IannoneA.2008Long-term N-acetylcysteine and L-arginine administration reduces endothelial activation and systolic blood pressure in hypertensive patients with type 2 diabetes. Diabetes Care 31940944
  70. 70. De MattiaG.BraviM. C.LaurentiO.Cassone-FaldettaM.ProiettiA.De LucaO.ArmientoA.FerriC.1998Reduction of oxidative stress by oral N-acetyl-L-cysteine treatment decreases plasma soluble vascular cell adhesion molecule-1 concentrations in non-obese, non-dyslipidaemic, normotensive, patients with non-insulin-dependent diabetes. Diabetologia 4113921396
  71. 71. MaxwellS. R.ThomasonH.SandlerD.LeguenC.MABaxterThorpe. G. H.JonesA. F.BarnettA. H.1997Antioxidant status in patients with uncomplicated insulin-dependent and non-insulin-dependent diabetes mellitus. Eur J Clin Invest 27484490
  72. 72. Mc GrathL. T.BMMc GleenonBrennan. S.Mc CollD.Mc LroyI.S.PassmoreA. P.2001Increased oxidative stress in Alzheimer’s disease as assessed with 4-hydroxynonenal but not malondialdehyde. QJM 94485490
  73. 73. Medina-NavarroR.Guzmán-GrenfellA. M.Díaz-FloresM.Duran-ReyesG.Ortega-CamarilloC.Olivares-CorichiI. M.HicksJ. J.2007Formation of an adduct between insulin and the toxic lipoperoxidation product acrolein decreases both the hypoglycemic effect of the hormone in rat and glucose uptake in 3T3 adipocytes. Chem Res Toxicol 2014771481
  74. 74. MenonV.RamM.DornJ.ArmstrongD.MutiP.FreudenheimJ. L.BrowneR.SchunemannH.TrevisanM.2004Oxidative stress and glucose levels in a population-based sample. Diabet Med 2113461352
  75. 75. MichielsC.RemacleJ.1991Cytotoxicity of linoleic acid peroxide, malondialdehyde and 4-hydroxynonenal towards human fibroblasts. Toxicology 66225234
  76. 76. MisonouY.AsahiM.YokoeS.MiyoshiE.TaniguchiN.2006Acrolein produces nitric oxide through the elevation of intracellular calcium levels to induce apoptosis in human umbilical vein endothelial cells: implications for smoke angiopathy. Nitric Oxide 14180187
  77. 77. MiwaI.IchimuraN.SugiuraM.HamadaY.TaniguchiS.2000Inhibition of glucose-induced insulin secretion by 4-hydroxy-2-nonenal and other lipid peroxidation products. Endocrinology 14127672772
  78. 78. Morris RT, Laye MJ, Lees SJ, Rector RS, Thyfault JP & Booth FW2008Exercise-induced attenuation of obesity, hyperinsulinemia, and skeletal muscle lipid peroxidation in the OLETF rat. J Appl Physiol 104708715
  79. 79. MünzelT.KurzS.RajagopalanS.ThoenesM.BerringtonW. R.ThompsonJ. A.BAFreemanHarrisonD. G.1996Hydralazine prevents nitroglycerin tolerance by inhibiting activation of a membrane-bound NADH oxidase. A new action for an old drug. J Clin Invest 9814651470
  80. 80. NakhjavaniM.EsteghamatiA.NowrooziS.AsgaraniF.RashidiA.KhalilzadehO.2010Type 2 diabetes mellitus duration: an independent predictor of serum malondialdehyde levels. Singapore Med J 51582585
  81. 81. Negre-SalvayreA.CoatrieuxC.IngueneauC.SalvayreR.2008Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br J Pharmacol 153620
  82. 82. NumazawaS.TakaseM.AhikoT.IshiiM.Shimizu-IS.YoshidaT.2012Possible Involvement of Transient Receptor Potential Channels in Electrophile-Induced Insulin Secretion from RINm5F Cells. Biol Pharm Bull 35346354
  83. 83. Olusi SO2002Obesity is an independent risk factor for plasma lipid peroxidation and depletion of erythrocyte cytoprotectic enzymes in humans. Int J Obes Relat Metab Disord 2611591164
  84. 84. PageS.FischerC.BaumgartnerB.HaasM.KreuselU.LoidlG.HaynM.Ziegler-HeitbrockH. W.NeumeierD.BrandK.1999Hydroxynonenal prevents NF-kappaB activation and tumor necrosis factor expression by inhibiting IkappaB phosphorylation and subsequent proteolysis. J Biol Chem 2741161111618
  85. 85. PanJ.KefferJ.EmamiA.MaLanX.GoldmanR.R.Chung-LF.2009Acrolein-derived DNA adduct formation in human colon cancer cells: its role in apoptosis induction by docosahexaenoic acid. Chem Res Toxicol 22798806
  86. 86. ParadisV.KollingerM.FabreM.HolstegeA.PoynardT.BedossaP.1997In situ detection of lipid peroxidation by-products in chronic liver diseases. Hepatology 26135142
  87. 87. PeyrouxJ.SternbergM.2006Advanced glycation endproducts (AGEs): Pharmacological inhibition in diabetes. Pathol Biol 54405419
  88. 88. PillonN. J.CrozeM. L.VellaR. E.SoulèreL.LagardeM.SoulageC. O.2012The Lipid Peroxidation By-Product 4-Hydroxy-2-Nonenal (4-HNE) Induces Insulin Resistance in Skeletal Muscle through Both Carbonyl and Oxidative Stress. Endocrinology 15320992111
  89. 89. PillonN. J.SoulèreL.VellaR. E.CrozeM.CaréB. R.SoulaH. A.DoutheauA.LagardeM.SoulageC. O.2010Quantitative structure-activity relationship for 4-hydroxy-2-alkenal induced cytotoxicity in L6 muscle cells. Chem Biol Interact 188171180
  90. 90. PillonN. J.VellaR. E.SoulèreL.BecchiM.LagardeM.SoulageC. O.2011Structural and functional changes in human insulin induced by the lipid peroxidation byproducts 4-hydroxy-2-nonenal and 4-hydroxy-2-hexenal. Chem Res Toxicol 24752762
  91. 91. PoirierM.FournierM.BrousseauP.MorinA.2002Effects of volatile aromatics, aldehydes, and phenols in tobacco smoke on viability and proliferation of mouse lymphocytes. J Toxicol Environ Health Part A 6514371451
  92. 92. ReaganL. P.MagariñosA. M.YeeD. K.SwzedaL. I.Van BuerenA.Mc CallA. L.BSMc Ewen2000Oxidative stress and HNE conjugation of GLUT3 are increased in the hippocampus of diabetic rats subjected to stress. Brain Res 862292300
  93. 93. RiahiY.Sin-MaliaY.CohenG.AlpertE.GruzmanA.EckelJ.StaelsB.GuichardantM.SassonS.2010The natural protective mechanism against hyperglycemia in vascular endothelial cells: roles of the lipid peroxidation product 4-hydroxydodecadienal and peroxisome proliferator-activated receptor delta. Diabetes 59808818
  94. 94. Riboulet-ChaveyA.PierronA.DurandI.MurdacaJ.GiudicelliJ.Van ObberghenE.2006Methylglyoxal impairs the insulin signaling pathways independently of the formation of intracellular reactive oxygen species. Diabetes 5512891299
  95. 95. RoyJ.PallepatiP.BettaiebA.Averill-BatesD. A.2010Acrolein induces apoptosis through the death receptor pathway in A549 lung cells: role of 53Can J Physiol Pharmacol 88, 353-368.
  96. 96. RudichA.TiroshA.PotashnikR.HemiR.KanetyH.BashanN.1998Prolonged oxidative stress impairs insulin-induced GLUT4 translocation in 3T3-L1 adipocytes. Diabetes 4715621569
  97. 97. RudichA.TiroshA.PotashnikR.KhamaisiM.BashanN.1999Lipoic acid protects against oxidative stress induced impairment in insulin stimulation of protein kinase B and glucose transport in 3T3-L1 adipocytes. Diabetologia 42949957
  98. 98. RuefJ.MoserM.BodeC.KublerW.MSRunge2001hydroxynonenal induces apoptosis, NF-kappaB-activation and formation of 8-isoprostane in vascular smooth muscle cells. Basic Res Cardiol 96143150
  99. 99. RussellA. P.GastaldiG.Bobbioni-HarschE.ArboitP.GobeletC.DériazO.GolayA.WitztumJ. L.Giacobino-PJ.2003Lipid peroxidation in skeletal muscle of obese as compared to endurance-trained humans: a case of good vs. bad lipids? FEBS Lett 551104106
  100. 100. SakurabaH.MizukamiH.YagihashiN.WadaR.HanyuC.YagihashiS.2002Reduced beta-cell mass and expression of oxidative stress-related DNA damage in the islet of Japanese Type II diabetic patients. Diabetologia 458596
  101. 101. Schaur RJ2003Basic aspects of the biochemical reactivity of 4-hydroxynonenal. Mol Aspects Med 24149159
  102. 102. Selley ML2004Increased (E)-4-hydroxy-2-nonenal and asymmetric dimethylarginine concentrations and decreased nitric oxide concentrations in the plasma of patients with major depression. J Affect Disord 80249256
  103. 103. ShearnC. T.FritzK. S.ReiganP.PetersenD. R.2011Modification of Akt2 by 4-hydroxynonenal inhibits insulin-dependent Akt signaling in HepG2 cells. Biochemistry 5039843996
  104. 104. ShibataM.HakunoF.YamanakaD.OkajimaH.FukushimaT.HasegawaT.OgataT.ToyoshimaY.ChidaK.KimuraK.SakodaH.TakenakaA.AsanoT.Takahashi-IS.2010Paraquat-induced oxidative stress represses phosphatidylinositol 3-kinase activities leading to impaired glucose uptake in 3T3-L1 adipocytes. J Biol Chem 2852091520925
  105. 105. SinghI.CareyA. L.WatsonN.MAFebbraioHawleyJ. A.2008aOxidative stress-induced insulin resistance in skeletal muscle cells is ameliorated by gamma-tocopherol treatment. Eur J Nutr 47387392
  106. 106. SinghS. P.NiemczykM.SainiD.AwasthiY. C.ZimniakL.ZimniakP.2008bRole of the electrophilic lipid peroxidation product 4-hydroxynonenal in the development and maintenance of obesity in mice. Biochemistry 4739003911
  107. 107. SinghS. P.NiemczykM.ZimniakL.ZimniakP.2009Fat accumulation in Caenorhabditis elegans triggered by the electrophilic lipid peroxidation product 4-hydroxynonenal (4-HNE). Aging (Albany NY) 16880
  108. 108. SoaresA. F.GuichardantM.CozzoneD.Bernoud-HubacN.Bouzaidi-TialiN.LagardeM.GeloenA.2005Effects of oxidative stress on adiponectin secretion and lactate production in 3T3-L1 adipocytes. Free Radic Biol Med 38882889
  109. 109. SrivastavaS.DixitB. L.CaiJ.SharmaS.HurstH. E.BhatnagarA.SrivastavaS. K.2000Metabolism of lipid peroxidation product, 4-hydroxynonenal (HNE) in rat erythrocytes: role of aldose reductase. Free Radic Biol Med 29642651
  110. 110. SyslovaK.KacerP.KuzmaM.NajmanovaV.FenclovaZ.VlckovaS.LebedovaJ.PelclovaD.2009Rapid and easy method for monitoring oxidative stress markers in body fluids of patients with asbestos or silica-induced lung diseases. J Chromatogr B Analyt Technol Biomed Life Sci; DOI:j.jchromb.2009.06.008.
  111. 111. TanY.IchikawaT.LiJ.SiQ.YangH.ChenX.GoldblattC. S.MeyerC. J.LiX.CaiL.CuiT.2011Diabetic downregulation of Nrf2 activity via ERK contributes to oxidative stress-induced insulin resistance in cardiac cells in vitro and in vivo. Diabetes 60625633
  112. 112. TanelA.Averill-BatesD. A.200738and ERK mitogen-activated protein kinases mediate acrolein-induced apoptosis in Chinese hamster ovary cells. Cell Signal 19, 968-977.
  113. 113. ThirunavukkarasuV.AnuradhaC. V.2004Influence of alpha-lipoic acid on lipid peroxidation and antioxidant defence system in blood of insulin-resistant rats. Diabetes Obes Metab 6200207
  114. 114. ToyokuniS.YamadaS.KashimaM.IharaY.YamadaY.TanakaT.HiaiH.SeinoY.UchidaK.2000Serum 4-hydroxy-2-nonenal-modified albumin is elevated in patients with type 2 diabetes mellitus. Antioxid Redox Signal 2681685
  115. 115. TraversoN.MeniniS.CossoL.OdettiP.AlbanoE.MAPronzatoMarinariU. M.1998Immunological evidence for increased oxidative stress in diabetic rats. Diabetologia 41265270
  116. 116. TraversoN.MeniniS.OdettiP.MAPronzatoCottalasso. D.MarinariU. M.2002Diabetes impairs the enzymatic disposal of 4-hydroxynonenal in rat liver. Free Radic Biol Med 32350359
  117. 117. TrevisanM.BrowneR.RamM.MutiP.FreudenheimJ.CarosellaA. M.ArmstrongD.2001Correlates of markers of oxidative status in the general population. Am J Epidemiol 154348356
  118. 118. UchidaK.ShiraishiM.NaitoY.ToriiY.NakamuraY.OsawaT.1999Activation of stress signaling pathways by the end product of lipid peroxidation. 4-hydroxy-2-nonenal is a potential inducer of intracellular peroxide production. J Biol Chem 27422342242
  119. 119. UnokiH.BujoH.YamagishiS.TakeuchiM.ImaizumiT.SaitoY.2007Advanced glycation end products attenuate cellular insulin sensitivity by increasing the generation of intracellular reactive oxygen species in adipocytes. Diabetes Res Clin Pract 76236244
  120. 120. VaillancourtF.FahmiH.ShiQ.LavigneP.RangerP.FernandesJ. C.BenderdourM.20084Hydroxynonenal induces apoptosis in human osteoarthritic chondrocytes: the protective role of glutathione-S-transferase. Arthritis Res Ther 10, R107.
  121. 121. VincentH. K.BourguignonC.VincentK. R.2006Resistance training lowers exercise-induced oxidative stress and homocysteine levels in overweight and obese older adults. Obesity (Silver Spring) 1419211930
  122. 122. VincentH. K.BourguignonC. M.WeltmanA. L.VincentK. R.BarrettE.InnesK. E.TaylorA. G.2009Effects of antioxidant supplementation on insulin sensitivity, endothelial adhesion molecules, and oxidative stress in normal-weight and overweight young adults. Metab Clin Exp 58254262
  123. 123. Vincent HK, Innes KE & Vincent KR2007Oxidative stress and potential interventions to reduce oxidative stress in overweight and obesity. Diabetes Obes Metab 9813839
  124. 124. VindisC.Escargueil-BlancI.ElbazM.MarcheixB.Grazide-HM.UchidaK.SalvayreR.Nègre-SalvayreA.2006Desensitization of platelet-derived growth factor receptor-beta by oxidized lipids in vascular cells and atherosclerotic lesions: prevention by aldehyde scavengers. Circ Res 98785792
  125. 125. WangL.SunY.AsahiM.OtsuK.2011Acrolein, an environmental toxin, induces cardiomyocyte apoptosis via elevated intracellular calcium and free radicals. Cell Biochem Biophys 61131136
  126. 126. WangZ.DouX.GuD.ShenC.YaoT.NguyenV.BraunschweigC.SongZ.2012Hydroxynonenal differentially regulates adiponectin gene expression and secretion via activating PPARγ and accelerating ubiquitin-proteasome degradation. Mol Cell Endocrinol 349222231
  127. 127. Wellen KE & Hotamisligil GS2003Obesity-induced inflammatory changes in adipose tissue. J Clin Invest 11217851788
  128. 128. Wierusz-WysockaB.WysockiH.ByksH.ZozulinskaD.WykretowiczA.KazmierczakM.1995Metabolic control quality and free radical activity in diabetic patients. Diabetes Res Clin Pract 27193197
  129. 129. Yadav UCS, Ramana KV, Awasthi YC & Srivastava SK2008Glutathione level regulates HNE-induced genotoxicity in human erythroleukemia cells. Toxicol Appl Pharmacol 227257264
  130. 130. YamashitaC.HayashiT.MoriT.MatsumotoC.KitadaK.MiyamuraM.SohmiyaK.UkimuraA.OkadaY.YoshiokaT.KitauraY.MatsumuraY.2010Efficacy of olmesartan and nifedipine on recurrent hypoxia-induced left ventricular remodeling in diabetic mice. Life Sci 86322330
  131. 131. YoritakaA.HattoriN.UchidaK.TanakaM.StadtmanE. R.MizunoY.1996Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease. Proc Natl Acad Sci U S A 9326962701
  132. 132. ZafarullahM.LiW. Q.SylvesterJ.AhmadM.2003Molecular mechanisms of N-acetylcysteine actions. Cell Mol Life Sci 60620
  133. 133. ZarroukiB.SoaresA. F.GuichardantM.LagardeM.GeloenA.2007The lipid peroxidation end-product 4-HNE induces COX-2 expression through 38MAPKactivation in 3T3-L1 adipose cell. FEBS Lett 581, 2394-2400.
  134. 134. ZhangH.FormanH. J.2008Acrolein induces heme oxygenase-1 through PKC-delta and PI3K in human bronchial epithelial cells. Am J Respir Cell Mol Biol 38483490
  135. 135. ZimniakP.2010Hydroxynonenal and fat storage: A paradoxical pro-obesity mechanism? Cell Cycle 933933394

Notes

  • 4-hydroxy-2-hexenal, 4-hydroxy-2-nonenal, 4-hydroxy-2-dodecenal, 4-hydroxy-2-hexenal dimethylacetal, 4-hydroxy-2-nonenal dimethylacetal, 4-hydroxy-2-dodecenal dimethylacetal, trans-2-hexenal, trans-2-nonenal, trans-2-dodecenal, hexanal, nonanal, dodecanal
  • Results from Wierusz-Wysocka et al., 1995; Vijayalingam et al., 1996; Feillet-Coudray et al., 1999; Rábago-Velasco et al., 2000; Rizvi & Zaid, 2001; Seghrouchni et al., 2002; Dinçer et al., 2002; Dierckx et al., 2003; Duman et al., 2003; Pasaoglu et al., 2004; Memişoğullari & Bakan, 2004; Skrha et al., 2005; Kurtul et al., 2005; Ozdemir et al., 2005; Mahboob et al., 2005; Saxena et al., 2005; Sampathkumar et al., 2005; Kuppusamy et al., 2005; Lapolla et al., 2007; Sathiyapriya et al., 2007; Singhania et al., 2008; Jain et al., 2009; Nakhjavani et al., 2010; Narasimhan et al., 2010; Shinde et al., 2011; Huang et al., 2011; Bahadoran et al., 2011; Zhang et al., 2011; Pácal et al., 2011; Calabrese et al., 2011 and Rasic-Milutinovic et al., 2012
  • Animals fasted overnight were then injected intraperitoneally with 0.5 UI/kg body weight of insulin. Plasma glucose was measured from tail vein blood using a glucometer at 0, 20, 40, 60, and 120 min following the injection. Glucose disappearance rate for ITT (KITT; %/min) was calculated as: KITT=(0.693x100)/t1/2, where t1/2 was calculated from the slope of the plasma glucose concentration, considering an exponential decrement of glucose concentration during the 20 min after insulin administration. Higher insulin sensitivity index (KITT) scores mean higher response of tissues to insulin.

Written By

Nicolas J. Pillon and Christophe O. Soulage

Submitted: 04 November 2011 Published: 29 August 2012