Elsevier

Free Radical Biology and Medicine

Volume 65, December 2013, Pages 978-987
Free Radical Biology and Medicine

Review Article
Signaling properties of 4-hydroxyalkenals formed by lipid peroxidation in diabetes

https://doi.org/10.1016/j.freeradbiomed.2013.08.163Get rights and content

Highlights

  • Hyperglycemia activates PLA2 and the release of PUFA from membrane phospholipids.

  • The diabetic environment intensifies the peroxidation of PUFA to 4-hydroxyalkenals.

  • Under these conditions, vascular endothelial cells generate 4-HDDE and β-cells 4-HNE.

  • Both compounds act as hormetic agents in the early compensatory stages of diabetes.

  • Diabetic complications are associated with long-term exposure to these compounds.

Abstract

Peroxidation of polyunsaturated fatty acids is intensified in cells subjected to oxidative stress and results in the generation of various bioactive compounds, of which 4-hydroxyalkenals are prominent. During the progression of type 2 diabetes mellitus, the ensuing hyperglycemia promotes the generation of reactive oxygen species (ROS) that contribute to the development of diabetic complications. It has been suggested that ROS-induced lipid peroxidation and the resulting 4-hydroxyalkenals markedly contribute to the development and progression of these pathologies. Recent findings, however, also suggest that noncytotoxic levels of 4-hydroxyalkenals play important signaling functions in the early phase of diabetes and act as hormetic factors to induce adaptive and protective responses in cells, enabling them to function in the hyperglycemic milieu. Our studies and others′ have proposed such regulatory functions for 4-hydroxynonenal and 4-hydroxydodecadienal in insulin secreting β-cells and vascular endothelial cells, respectively. This review presents and discusses the mechanisms regulating the generation of 4-hydroxyalkenals under high glucose conditions and the molecular interactions underlying the reciprocal transition from hormetic to cytotoxic agents.

Introduction

Type 2 diabetes mellitus (T2DM) is a chronic disease that has recently reached an epidemic proportion worldwide. Three major stages in the development and progression of the disease have been identified: the first “prediabetes” asymptomatic phase commences with progressive peripheral insulin resistance, impaired glucose tolerance, and subsequently a gradual increase in blood glucose level, which remain below the diabetic threshold due to a compensatory augmented insulin secretion from pancreatic β-cells [1]. Following this overstimulation period, the second phase is characterized by β-cell dysfunction and decompensation that leads to hypoinsulinemia and frank hyperglycemia. At this phase, some peripheral tissues react to the hyperglycemic environment by initiating compensatory defense mechanisms that limit or slow the development of diabetic complications. For instance, skeletal muscles and vascular endothelial and smooth muscle cells reduce the rate of glucose influx by downregulating the expression of their glucose transporters and their abundance in the plasma membrane [2], [3], [4]. In the late stages of T2DM, the chronic nutrient overload (hyperglycemia and/or hyperlipidemia) promotes the secretion of metabolites and factors from adipose depots (adipokines), the liver (hepatokines), and skeletal muscles (myokines) that interfere with these protective mechanisms and cause a progressive damage to peripheral tissues and organs, culminating in cardiovascular disease, nephropathy, neuropathy, or impaired wound healing [5], [6], [7]. Beta-cell apoptosis and death that results in a severe decrease in β-cell mass in islets of Langerhans is also common in the advanced stage of the disease. While these detrimental interactions in the late stages of diabetes have been well characterized and extensively investigated, less is known about the molecular mechanisms underlying the adaptive responses in the early stages. Both peripheral diabetic complications and β-cell failure are associated with an increased production of ROS [8], [9]. Among many direct interactions with macromolecules in cells, ROS also induce nonenzymatic peroxidation of polyunsaturated fatty acids (PUFA) to generate α,β-unsaturated aldehydes that belong to the 4-hydroxyalkenal family (e.g., 4-hydroxy-2E-nonenal; 4-HNE, 4-hydroxy-2E,6Z-dodecadienal; 4-HDDE) [9], [10], [11]. These electrophilic aldehydes avidly bind covalently to nucleophilic moieties in macromolecules and compromise cell functions and structures [12], [13]. Nonetheless, at low and noncytotoxic levels, these aldehydes can function as signaling molecules by interacting with transduction pathways and receptors and thereby promote compensatory defense mechanisms. In many respects these advanced lipid peroxidation products are similar to advanced glycation end products (AGEs) that can evoke cellular regulatory mechanisms by interacting with specific cell surface receptors (RAGEs), whereas their excessive generation and accumulation following chronic hyperglycemia are detrimental to cells and tissues and contribute further to the development of diabetic complications [8], [9], [14], [15], [16], [17]. This review presents recent studies on the dual function of 4-hydroxyalkenals in β-cells and vascular endothelial cells (VEC), where they function as hormetic factors in the prediabetes phase and become cytotoxic agents as the disease progresses.

Section snippets

Compensatory mechanisms and cell decompensation in the progression of T2DM

Hyperglycemia in diabetes results from a combination of peripheral insulin resistance and progressive deterioration of the pancreatic β-cell mass and function, resulting in reduced insulin secretion and circulating levels of the hormone that fail to support adequate metabolism in peripheral insulin-sensitive tissues. Diabetes is also characterized by the development of end-organ complications [18]. The underlying risk factor in the etiology of these complications is hyperglycemia, which alters

PUFA metabolism and peroxidation in diabetes

Arachidonic acid (AA) is a conditionally essential fatty acid available to mammals directly from the diet and/or through enzymatic elongation and desaturation of the essential linoleic acid (LA) [39]. The combined action of fatty acyl-CoA synthase and lysophospholipid acyltransferase promotes the incorporation of AA and LA at the sn-2 position of phospholipids [40], whereas enzymes of the phospholipase A2 (PLA2) family hydrolyze this bond to liberate them. Free AA and LA are transformed to

Cellular targets for 4-hydroxyalkenals

It has been repeatedly shown that exogenous 4-HNE alters vital functions and signaling pathways in cells by covalent interactions with key proteins in signal transduction and regulatory pathways [36], [37], [51], [74], [75], [76], [77], [78], [79], [80]. Among these, 4-HNE affects p38 mitogen-activated protein kinases (p38MAPK) [81], c-Jun N-terminal kinase (JNK) [82], protein kinase Cβ and -δ (PKCβ and PKCδ) [83], [84], cell cycle regulators [85], epithelial growth factor receptor (EGRF) [86],

Role of 4-hydroxyalkenals in VEC and β-cells adaptation to hyperglycemia

As noted above, the rate of 4-HDDE and 4-HNE production in VEC and β-cells, respectively, increases in a glucose-dependent manner. We hypothesized that this phenomenon was primarily due to an increased availability of free AA and LA to peroxidation processes. To investigate this, we performed lipidomic analyses of the composition of the fatty acids in membrane phospholipids of both types of cells following incubation with increasing glucose concentrations. Table 1 shows that abundance of AA and

Are 4-hydroxynonenals involved in VEC and β-cells dysfunction in T2DM?

Similar to other cells [37], in vitro exposure of VEC, INS-1E cells, and freshly isolated rat islets to high and supraphysiological levels of 4-HDDE or 4-HNE induces apoptosis and cell death [9], [30], [31], [138]. This is due primarily to covalent adduct formation with macromolecules and failure of the cellular enzymatic and nonenzymatic neutralization systems to cope with the 4-hydroxyalkenal overload. Some studies on different types of cells and tissues suggest that endogenous 4-HNE may

Conclusions

Lipid peroxidation is enhanced under hyperglycemic conditions and leads to the generation of 4-hydroxyalkenals, such as 4-HNE and 4-HDDE. The tendency of these bioactive aldehydes to form adducts with proteins, phospholipids, and nucleic acid has long been linked to their cytotoxic activities. Indeed, in vitro treatments of VEC and β-cells with these agents induced marked apoptosis and cell death. However, the endogenous levels of 4-HDDE in VEC and of 4-HNE in β-cells under high glucose

Acknowledgments

The study was supported by a grants from the Israel Science Foundation (44/10), the Brettler Center for Research of Molecular Pharmacology and Therapeutics in the Hebrew University and by COST (European Cooperation in Science and Technology) Action CM1201 on “Biomimetic Radical Chemistry.” S. Sasson is the Adolf D. and Horty Storch Chair in Pharmaceutical Sciences, at the Faculty of Medicine, The Hebrew University of Jerusalem, Israel. He is affiliated with the David R. Bloom Center for

References (144)

  • C. Schneider et al.

    Routes to 4-hydroxynonenal: fundamental issues in the mechanisms of lipid peroxidation

    J. Biol. Chem.

    (2008)
  • E. Niki et al.

    Lipid peroxidation: mechanisms, inhibition, and biological effects

    Biochem. Biophys. Res. Commun.

    (2005)
  • K. Uchida

    4-Hydroxy-2-nonenal: a product and mediator of oxidative stress

    Prog. Lipid Res.

    (2003)
  • W.A. Pryor et al.

    Suggested mechanisms for the production of 4-hydroxy-2-nonenal from the autoxidation of polyunsaturated fatty acids

    Free Radic. Biol. Med.

    (1990)
  • M. Guichardant et al.

    Hydroxy-alkenals from the peroxidation of n-3 and n-6 fatty acids and urinary metabolites. Prostaglandins Leukot

    Essent. Fatty Acids

    (2006)
  • S. Bacot et al.

    Covalent binding of hydroxy-alkenals 4-HDDE, 4-HHE, and 4-HNE to ethanolamine phospholipid subclasses

    J. Lipid Res.

    (2003)
  • B.L. Trumpower

    The protonmotive Q cycle. Energy transduction by coupling of proton translocation to electron transfer by the cytochrome bc1 complex

    J. Biol. Chem.

    (1990)
  • P. Thornalley et al.

    The autoxidation of glyceraldehyde and other simple monosaccharides under physiological conditions catalysed by buffer ions

    Biochim. Biophys. Acta

    (1984)
  • E. Niki

    Lipid peroxidation: physiological levels and dual biological effects

    Free Radic. Biol. Med.

    (2009)
  • D.R. Petersen et al.

    Reactions of 4-hydroxynonenal with proteins and cellular targets

    Free Radic. Biol. Med.

    (2004)
  • N. Zarkovic

    4-Hydroxynonenal as a bioactive marker of pathophysiological processes

    Mol. Aspects Med.

    (2003)
  • T. Grune et al.

    The proteasomal system and HNE-modified proteins

    Mol. Aspects Med.

    (2003)
  • B. Zarrouki et al.

    The lipid peroxidation end-product 4-HNE induces COX-2 expression through p38MAPK activation in 3T3-L1 adipose cell

    FEBS Lett.

    (2007)
  • F. Biasi et al.

    c-Jun N-terminal kinase upregulation as a key event in the proapoptotic interaction between transforming growth factor-beta1 and 4-hydroxynonenal in colon mucosa

    Free Radic. Biol. Med.

    (2006)
  • L. Castello et al.

    4-Hydroxynonenal signalling to apoptosis in isolated rat hepatocytes: the role of PKC-delta

    Biochim. Biophys. Acta

    (2005)
  • I. Nakashima et al.

    4-Hydroxynonenal triggers multistep signal transduction cascades for suppression of cellular functions

    Mol. Aspects Med.

    (2003)
  • S. Page et al.

    4-Hydroxynonenal prevents NF-kappaB activation and tumor necrosis factor expression by inhibiting IkappaB phosphorylation and subsequent proteolysis

    J. Biol. Chem.

    (1999)
  • Z.H. Chen et al.

    Adaptive response induced by lipid peroxidation products in cell cultures

    FEBS Lett.

    (2006)
  • Z.H. Chen et al.

    4-Hydroxynonenal induces adaptive response and enhances PC12 cell tolerance primarily through induction of thioredoxin reductase 1 via activation of Nrf2

    J. Biol. Chem.

    (2005)
  • M. Tanito et al.

    Upregulation of thioredoxin system via Nrf2-antioxidant responsive element pathway in adaptive-retinal neuroprotection in vivo and in vitro

    Free Radic. Biol. Med.

    (2007)
  • Y.M. Go et al.

    Reactive aldehyde modification of thioredoxin-1 activates early steps of inflammation and cell adhesion

    Am. J. Pathol.

    (2007)
  • J.A. Doorn et al.

    Covalent adduction of nucleophilic amino acids by 4-hydroxynonenal and 4-oxononenal

    Chem. Biol. Interact.

    (2003)
  • M. Benderdour et al.

    Cardiac mitochondrial NADP+-isocitrate dehydrogenase is inactivated through 4-hydroxynonenal adduct formation: an event that precedes hypertrophy development

    J. Biol. Chem.

    (2003)
  • J. Chen et al.

    Inhibition of cytochrome c oxidase activity by 4-hydroxynonenal (HNE). Role of HNE adduct formation with the enzyme subunits

    Biochim. Biophys. Acta

    (1998)
  • G.C. Weir et al.

    Five stages of evolving beta-cell dysfunction during progression to diabetes

    Diabetes

    (2004)
  • E. Alpert et al.

    Delayed autoregulation of glucose transport in vascular endothelial cells

    Diabetologia

    (2005)
  • E. Alpert et al.

    A natural protective mechanism against hyperglycaemia in vascular endothelial and smooth-muscle cells: role of glucose and 12-hydroxyeicosatetraenoic acid

    Biochem. J.

    (2002)
  • B.K. Pedersen

    Muscles and their myokines

    J. Exp. Biol.

    (2011)
  • H. Sell et al.

    Skeletal muscle insulin resistance induced by adipocyte-conditioned medium: underlying mechanisms and reversibility

    Am. J. Physiol. Endocrinol. Metab.

    (2008)
  • N. Stefan et al.

    The role of hepatokines in metabolism

    Nat. Rev. Endocrinol.

    (2013)
  • F. Giacco et al.

    Oxidative stress and diabetic complications

    Circ. Res.

    (2010)
  • Cohen, G.; Riahi, Y.; Sasson, S. Free radicals and metabolic disorders. In: Encyclopedia of radicals in chemistry,...
  • M. Jaganjac et al.

    Reactive aldehydes—second messengers of free radicals in diabetes mellitus

    Free Radic. Res.

    (2013)
  • G. Cohen et al.

    Lipid peroxidation of poly-unsaturated fatty acids in normal and obese adipose tissues

    Arch. Physiol. Biochem.

    (2011)
  • A. Negre-Salvayre et al.

    Pathological aspects of lipid peroxidation

    Free Radic. Res.

    (2010)
  • A. Negre-Salvayre et al.

    Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors

    Br. J. Pharmacol.

    (2008)
  • A. Rouhiainen et al.

    RAGE-mediated cell signaling

    Methods Mol. Biol.

    (2013)
  • A. Negre-Salvayre et al.

    Hyperglycemia and glycation in diabetic complications

    Antioxid. Redox Signal.

    (2009)
  • W.T. Cade

    Diabetes-related microvascular and macrovascular diseases in the physical therapy setting

    Phys. Ther.

    (2008)
  • N. Kaiser et al.

    Failure of beta-cell adaptation in type 2 diabetes: lessons from animal models

    Front. Biosci.

    (2009)
  • Cited by (0)

    View full text