pdf Diabetic retinopathy

Embed Size (px)

Citation preview

  • 7/27/2019 pdf Diabetic retinopathy

    1/13

    Role of Aldose Reductase and Oxidative Damage inDiabetes and the Consequent Potential for

    Therapeutic Options

    Satish K. Srivastava, Kota V. Ramana, and Aruni Bhatnagar

    Department of Human Biological Chemistry and Genetics (S.K.S., K.V.R.), University of Texas Medical Branch, Galveston,Texas 77555; and Division of Cardiology (A.B.), Department of Medicine, University of Louisville, Louisville, Kentucky40202

    Aldose reductase (AR) is widely expressed aldehyde-metabo-lizing enzyme. The reduction of glucose by the AR-catalyzedpolyol pathway has been linked to the development of sec-ondary diabetic complications. Although treatment with ARinhibitors has been shown to prevent tissue injury in animal

    modelsof diabetes,the clinical efficacy of these drugs remainsto be established. Recent studies suggest that glucose may bean incidental substrate of AR,whichappears to be more adeptin catalyzing the reduction of a wide range of aldehydes gen-erated from lipid peroxidation. Moreover, inhibition of theenzyme has been shown to increase inflammation-inducedvascular oxidative stress and prevent myocardial protectionassociated with the late phase of ischemic preconditioning.On the basis of these studies, several investigators have as-cribed an importantantioxidant role to theenzyme. Addition-ally, ongoing work indicates that AR is a critical componentof intracellular signaling, and inhibition of the enzyme pre-

    vents high glucose-, cytokine-, or growth factor-induced ac-tivation of protein kinase C and nuclear factor--binding pro-tein. Thus, treatment with AR inhibitors prevents vascularsmooth muscle cell growth and endothelial cell apoptosis inculture and inflammation and restenosis in vivo. Additional

    studies indicate that theantioxidantand signalingrolesof ARare interlinked and that AR regulates protein kinase C andnuclear factor-B via redox-sensitivemechanisms. These dataunderscore the need for reevaluating anti-AR interventionsfor the treatment of diabetic complications. Potentially, thedevelopment of newer drugs that selectively inhibit AR-mediated glucose metabolismand signaling, without affectingaldehyde detoxification, may be useful in preventing inflam-mation associated with the development of diabetic compli-cations, particularly micro- and macrovascular diseases. (En-docrine Reviews 26: 380392, 2005)

    I. Introduction: Aldose Reductase and Diabetic Complications

    II. AR and Antioxidant ProtectionIII. Redox Regulation of AR ActivityIV. Regulation of Intracellular Signaling by ARV. AR Inhibitors and the Treatment of Secondary Diabetic

    ComplicationsVI. Conclusion

    I. Introduction: Aldose Reductase and Diabetic

    Complications

    ALDOSE REDUCTASE (AR) is a monomeric reducednicotinamide adenine dinucleotide (NAD) phosphate(NADPH)-dependent enzyme and a member of aldo-keto

    reductase superfamily. The enzyme was described in 1956 byHers (1) as a glucose-reducing activity. It was subsequentlyreported by van Heyningen (2) that high levels of AR activity

    are present in the rat lens and that during diabetic and

    galactosemic cataractogenesis, AR-derived polyolssorbitoland galactitolaccumulate in the ocular lens. Building onthis observation, Kinoshita et al. (3) and Varma and Kinoshita(4) demonstrated that treatment with pharmacological in-hibitors of AR ameliorated cataractogenesis in diabetic ratsand galactose-exposed rabbits. Based on these observationsit was proposed that accumulation of sorbitol in the lens, dueto AR-catalyzed reduction of glucose, causes osmotic swell-ing resulting in ionic imbalance and protein insolubilizationleading to cataractogenesis (57). A similar sequence ofevents could also account for hyperglycemic injury associ-ated with diabetic retinopathy, nephropathy, and neuropa-thy (Fig. 1).

    The osmotic hypothesis that diabetic complications aredue to sorbitol accumulation in tissues has engendered ex-tensive investigations over the last three decades. Severaldrugs with varying AR-inhibiting efficacy (e.g., sorbinil, sta-til, tolrestat, and zopolrestat) have been synthesized andtested (811). Initial trials in animal models showed signif-icant protection against diabetic complications (1214). ARinhibitors, in addition to preventing diabetic and galac-tosemic cataracts (3, 4), ameliorated some of the features ofdiabetic nephropathy (15, 16) and neuropathy (1719). How-ever, clinical trials with AR inhibitors have yielded uncertainresults, in part due to the high nonspecific toxicity of thisclass of drugs.

    In the mid-1980s we demonstrated that most of the inhib-

    First Published Online April 6, 2005Abbreviations: AR, Aldose reductase; DAG, diaceylglycerol; DHN,

    1,4-dihydroxy-2-nonene; FGF, fibroblast growth factor; GSNO, nitroso-glutathione; GSSG, oxidized glutathione; HNE, 4-hydroxynonenal;NAD, nicotinamide adenine dinucleotide; NADPH, reduced NADphosphate; NF-B, nuclear factor--binding protein; NO, nitric oxide;PKC, protein kinase C; PLC, phospholiase C; VSMC, vascular smoothmuscle cells.

    Endocrine Reviews is published bimonthly by The Endocrine Society(http://www.endo-society.org), the foremost professional society serv-ing the endocrine community.

    0163-769X/05/$20.00/0 Endocrine Reviews 26(3):380392Printed in U.S.A. Copyright 2005 by The Endocrine Society

    doi: 10.1210/er.2004-0028

    380

  • 7/27/2019 pdf Diabetic retinopathy

    2/13

    itors synthesized by pharmaceutical companies were notselective for AR but also inhibited other members of the

    aldo-keto reductase superfamily such as aldehyde reductase(20). In addition, we also demonstrated that increased os-motic pressure due to accumulation of polyols in galac-tosemic and diabetic cataractogenesis could not be the maincause of cataractogenesis (2123). Antioxidants such as bu-tylated hydroxytoluene and 6-hydroxy-2,5,7,8-tetramethe-nyl-chroman-2-carboxylic acid (Trolox) prevented diabeticcataractogenesis in rats and significantly attenuated lensopacity in galactose-fed rats even though the polyol levels inthe lens were extremely high (8090 mm), indicating thatpolyol accumulation per se is not sufficient for cataractogen-esis and that other metabolic changes accompanying ARactivation may be critical and important modulators of the

    cataractous effects of hyperglycemia and diabetes (Fig. 2).Because AR utilizes NADPH to catalyze glucose reduc-tion, we, as well as other investigators, suggested that tissueinjury associated with high glucose may be due, in part, toincreased NADPH utilization by the reduction of glucose byAR (24, 25). In the presence of normal glucose (5.5 mm),AR-catalyzed reduction represents less than 3% of total glu-cose utilization, whereas in the presence of high glucose (20mm), more than 30% of the glucose is used by AR (26),suggesting that the profound increase in the AR-catalyzedreductive pathway may impose a significant strain onNADPH supply. Because NADPH is used for several criticalreductive metabolic steps, such as the detoxification of re-active oxygen species and hydroperoxides (e.g., by the glu-

    tathione reductase/glutathione peroxidase system), a largedrain on the NADPH pool could compromise the ability of

    the cell to protect itself from oxidative stress.Another reason for the inconsistent effects of AR inhibitors

    may relate to posttranslation modification of the enzyme. Asdocumented extensively in our previous publications (2628), the oxidation state of a single cysteine residue located atthe active site of the enzyme regulates both substrate andinhibitor binding and thus, given the extensive oxidativechanges in diabetes, it is likely that in diabetic tissues AR is

    FIG. 2. Oxidative stress is the main contributor to diabetic catarac-togenesis. Digital image analysis was performed on lensescultured innormal glucose (5.5 mM; NG), high glucose (50 mM; HG) without orwith antioxidants on d 8. Sorbitol content was measured by using gaschromatography. The data represent mean SD (n 4). **,P 0.001;#, Pnonsignificant in HG-treated group compared withNG-treatedgroup. BHT, Butylated hydroxytoluene.

    FIG. 1. Involvement of polyol pathwayin diabetic complications. During hy-perglycemia, reduction of glucose tosorbitol by AR constitutes the first andtherate-limitingstep of thepolyolpath-way that converts glucose to fructose

    via sorbitol dehydrogenase (SDH). Inthis pathway both NADPH and NAD

    are consumed as cofactors for the en-zymes AR andSDH. Osmotic stressdueto accumulation of sorbitol and oxida-tive stress due to changes in the ratio ofNADPH/NADP and reduced NAD(NADH)/NAD are the major cause of

    various complications of secondary di-abetes. ROS, Reactive oxygen species.

    Srivastava et al. Aldose Reductase and Diabetic Complications Endocrine Reviews, May 2005, 26(3):380392 381

  • 7/27/2019 pdf Diabetic retinopathy

    3/13

    insensitive to inhibitors due to oxidative modification. Insupport of this view, we have demonstrated that AR inhuman erythrocytes exists in two forms, activated and un-activated,and in hyperglycemiatotalactivity of AR increases(29). Multiple forms of AR have also been reported by otherinvestigators (30, 31), and our in vitro data suggest that ox-idants can induce a variety of changes in the catalytic activityof the enzyme, although which of these forms occur in vivoand how they are interconverted remains unclear. Never-theless, the view that the hyperglycemia changes the sensi-tivity of AR to inhibition is further supported by the obser-vation that prolonged culture of endothelial cells in highglucose progressively decreases the efficacy of sorbinil inpreventing sorbitol accumulation (32).

    In contrast to inhibitor data, newer molecular approacheshave provided more unambiguous evidence for the involve-ment of AR in diabetic complications. In these studies trans-genic overexpression of AR gene selectively in the lens wasfound to accelerate diabetic and galactosemic cataract for-mation in mice (33). Additionally, ubiquitous overexpression

    of the AR gene increased the rate of neuropathic changes indiabetic animals (3436). Collectively, these data arguestrongly that AR is an important component of high glucose-induced metabolic changes that underlie the development ofsecondary diabetic complications. Nevertheless, despite thisevidence, the exact mechanism by which AR contributes tothe development of diabetic complications remains unclear.Persistent increase in extracellular glucose levels inducespleiotrophic changes in metabolism that elicit polygenic re-sponses (Fig. 3). Extensive changes in the activation of pro-tein kinases and the accumulation of advanced glycation endproducts have been reported (3739), although their rela-tionships with modulatory influences on AR remain unclear.

    In principle, AR could contribute to both protein kinaseactivation and advanced glycation end product accumula-tion. In addition, AR could catalyze the formation of potentprotein glycating agents and induce oxidative stress.

    Reduction of glucose by AR leads to the formation ofsorbitol, which, in some tissues, is further oxidized to fruc-tose upon sorbitol dehydrogenase-catalyzed oxidation. Theconversion of glucose to fructose (the polyol pathway)results not only in the utilization of NADPH, but also NAD.As a result, increased activity of the polyol pathway duringhyperglycemia could lead to a depletion of NADPH andaccumulation of reduced NAD. This shift in the redox stateof pyridine coenzymes recapitulates the metabolic pheno-type of hypoxia and has been proposed to induce a state ofpseudohypoxia resulting in hypoxia-like responses (4042).Polyol pathway-mediated alterations in pyridine nucleotideshave been linked to diverse metabolic changes such as thesynthesis of nitric oxide (NO) and activation of protein ki-nases (39). Specifically, it has been proposed that the increasein NADH due to elevated polyol pathway activity couldincrease the synthesis of diacylglycerol (DAG) from dihy-droxyacetone phosphate (43). DAG levels could also increase

    by stress that activates phospholipase C (PLC in Fig. 4). DAGis an essential activator of the classical and novel members

    of the protein kinase C family, and DAG-dependent activa-tion ofthese kinases is thought to play a key rolein mesangialexpansion and smooth muscle cells proliferation induced byhigh glucose (4446). Inhibitors of specific protein kinase C(PKC) isoforms are currently under clinical trial for the treat-ment of diabetic complications (47, 48).

    II. AR and Antioxidant Protection

    Due to its ability to reduce glucose, AR is involved inseveral tissue-specific metabolic pathways. For instance, inthe kidney, AR participates in osmoregulation (49, 50) and,in seminal vesicles, in the generation of fructose (1). The

    FIG. 3. Pathways of hyperglycemia-induced oxidative stress. In-creased superoxide production in the mitochondria during hypergly-cemia causes increased oxidative stress. During hyperglycemia theratio NADPH/NAD decreases due to excessive use of NADPH for thereduction of glucose to sorbitol. Consequently, the NADPH availabil-ity to maintain glutathione in the reduced form catalyzed by gluta-thione reductase decreases,which causes oxidative stress. Increase inadvanceglycationend products (AGEs)formationand bindingof AGEto its cognate receptor (RAGE) also increases oxidative stress. In-creased DAG formation in hyperglycemia activates PKC and subse-quently induces oxidative stress via a PKC-dependent activation ofNAD(P)H oxidase.

    FIG. 4. Pathways of increased DAG in hyperglycemia. Increasedstress conditions because of autooxidation of glucose, osmotic stress,oxidative stress, and hypoxia during polyol pathway hyperactivitycan contribute to increase in DAG levels through a cascade of signalsthat activate phospholipase C (PLC) or directly through glycolysis.

    AGE, Advance glycation end product; ROS, reactive oxygen species;SDH, sorbitol dehydrogenase.

    382 Endocrine Reviews, May 2005, 26(3):380 392 Srivastava et al. Aldose Reductase and Diabetic Complications

  • 7/27/2019 pdf Diabetic retinopathy

    4/13

    observation that AR and related proteins are up-regulated byfibroblast growth factor (FGF) and other mitogens suggeststhat AR may be involved in cell growth or growth factor-induced changes in cellular metabolism (5161). Our studiesshow that in vitro homogenous AR catalyzes the reduction ofa large series of saturated and unsaturated aldehydes with103- to 104-fold higher efficiency than glucose (62). The en-zyme is particularly efficient in reducing medium- to long-chain (C-6 to C-18) aldehydes such as those generated in highabundance during lipid peroxidation (63, 64). The enzymealso catalyzes the reduction of the glutathione conjugates ofunsaturated aldehydes, in many cases with efficiency higherthan that of the parent free aldehyde (6567). The reductionof glutathione conjugates by AR may be a protective mech-anism that may be useful in minimizing the reactivity of thealdehyde function unquenched by glutathiolation. In addi-tion, our recent studies show that AR is also an efficientcatalyst for the reduction of core aldehydesor aldehydesgenerated in oxidized phospholipids (Refs. 6570 and Fig. 5).

    The role of AR in detoxification is supported by structural

    studies showing that the active site of AR lacks ionic residuecharacteristic of carbohydrate-binding proteins (7176). In-stead, x-ray analyses of AR crystals reveal a highly plasticand hydrophobic active site (73, 74). This site could poten-tially accommodate a wide range of structures if the alde-hyde function were to orient itself appropriately between thetwo ionic active site residues, His-110 and Tyr-48, whichparticipate in acid-base catalysis. The structure of the binarycomplex of the enzyme with NADP also indicates a pro-found conformational change upon NADPH binding (72). Ithas been suggested that the energy released as a result of thisinteraction is used for stabilization of the transition state withlittle demand from energy stabilization due to substrate

    binding (77). Hence, as a result of high-affinity interactionwith NADPH, AR functions as an unusually promiscuousaldehyde removase, features that seem to be critically re-quired of a detoxification enzyme involved in the removal ofa wide range of aldehydes and glutathione aldehyde adductsgenerated during lipid peroxidation.

    Based on our substrate specificity studies showing high-

    affinity reduction of glutathione conjugates by AR (66, 67),we reasoned that the active site of the enzyme must conformto a specificglutathione-binding domain. Consistent with thepresence of specific interactions between the amino acid res-idues of glutathione and the AR active site, we found thatalterations in the structure of glutathione diminished thecatalytic efficiency for the reduction of glutathione-aldehydeconjugates and that nonaldehydic conjugates of glutathioneor glutathione analogs displayed active site inhibition (67).Molecular dynamics calculations suggest that the conjugatesadopt a specific low-energy configuration at the active site(Fig. 6). Mutations of the active site residues identified bythese calculations selectively decreased catalysis of the glu-tathione-aldehyde conjugates, without affecting reduction ofthe free aldehyde. The high specificity and selectivity withwhich the enzyme catalyzes the reduction of glutathioneconjugates suggest that such conjugates may be in vivo sub-strates of the enzyme. Indeed, our metabolic studies with4-hydroxynonenal (HNE) show AR-dependent reduction ofits glutathione adduct in heart, smooth muscle, and eryth-

    rocytes (7881).Reduction of aldehyde-glutathione conjugates by AR may

    be of physiological significance, particularly under condi-tions of oxidative stress when other antioxidant mechanismsare overwhelmed. Under these conditions, AR, by reducing

    both free aldehydes and their glutathione conjugates, couldpromote efficient removal and detoxification of unsaturatedaldehydes, which are the major electrophilic end products oflipid peroxidation. Thus, investigations into the structureand kinetics of AR point toward a general detoxification roleof the enzyme, indicating that lipid aldehydes and theirglutathione conjugates are physiological substrates of theenzyme and that glucose is perhaps an incidental substrate,

    which is used only when it is high in concentration or oc-casionally for tissue-specific metabolism.

    Consistent with its role as a detoxification enzyme, AR hasbeen found to be regulated by aldehydes generated fromlipid peroxidation (64, 70, 82), thiol-reducing agents (83 88),metal ions (8991), and NO (9297). Tissue levels of AR arealso increased under conditions of high oxidative stress such

    FIG. 5. Reduction of lipid aldehydes byaldose reductase. The bold numbers in

    parentheses represent reference num-bers.

    Srivastava et al. Aldose Reductase and Diabetic Complications Endocrine Reviews, May 2005, 26(3):380392 383

  • 7/27/2019 pdf Diabetic retinopathy

    5/13

    as iron overload (98), alcoholic liver disease (99), heart failure(100), and vascular inflammation (53, 101). Moreover, AR isalso specifically up-regulated during vasculitis, specificallyin the areas of high HNE formation, and inhibition of ARincreases the concentration of free HNE and protein-HNEadducts, accompanied by a 3-fold increase in the number ofapoptotic cells (102). Our studies show that AR is also up-regulated in the heart by brief episodes of ischemia (103).This increase in AR was prevented by inhibitors of PKC andNO synthase, indicating that the up-regulation of AR is reg-ulated by cardioprotective signaling associated with the latephaseof ischemicpreconditioning. Furthermore, a posttrans-lational activation of AR during myocardial ischemia has

    been reported (104). The functional significance of the in-crease in AR in the ischemic heart is underscored by theobservation that inhibition of AR abolishes the cardiopro-tective effects of late preconditioning (105107). Collectively,these data provide support to the view that AR is a compo-nent of an antioxidant defense mechanism, which protectstissues from the harmful effect of lipid peroxidation productssuch as HNE that accumulate during conditions of oxidativestress such as ischemia and inflammation.

    Although HNE and related aldehydes are metabolized byseveral different pathways, metabolism via AR could rep-resent a true detoxification mechanism. In most cells, HNEand related aldehydes readily form glutathione conjugates;

    however, the formation of glutathione-HNE itself may not besufficient for detoxification. The glutathione-aldehyde con-jugates are toxic. They induce DNA damage and stimulateradical formation (108110). Therefore, reduction of the glu-tathione-aldehyde conjugates by AR may be necessary tosubstantially annul the reactivity of the conjugate and todecrease free radical generation. Nonetheless, the mecha-nisms by which AR-dependent aldehyde metabolism regu-lates the development of diabetic complications remain un-clear, and the contribution of AR to cell growth andantioxidant defense requires further elucidation. In particu-lar, it remains to be clarified how AR expression and activityare regulated and which components of intracellular signal-ing are regulated by AR.

    III. Redox Regulation of AR Activity

    As discussed above, a component of the hyperglycemicinjury is due to an increased flux of glucose through thepolyol pathway because the inhibition of AR prevents someof the diabetic complications. Although inefficacy of AR in-hibitors could be due to their nonspecificity and hypersen-sitivity of selected individuals, the limited long-term efficacyof the AR inhibitors could be attributed to the posttransla-tional modification in AR that alters ligand binding andcatalysis. In our earlier studies we found that AR isolatedfrom individuals with different levels of hyperglycemia dis-played altered kinetic properties and was less sensitive tohydantoin inhibitors such as sorbinil compared with theenzyme isolated from the tissues of normoglycemic subjects(29). Similar changes were observed at various stages ofpurification of AR in the absence of a reducing reagent suchas dithiothreitol or -mercaptoethanol from the tissues ofnormal subjects or from recombinant bacteria overexpress-ing AR (30, 31). Both theMichaelis-Menten constant (Km)andinhibition constant (Ki) values of the enzyme significantlyincreased upon its in vitro thiol modification by HNE (64, 82),nitrosoglutathione (GSNO) (92), and oxidized glutathione(GSSG) (83).

    The high sensitivity of AR to oxidants such as H2O2 andNO was attributed to a highly reactive cysteine (Cys-298)residue present at the active site of the enzyme (88). OxidantssuchasH2O2 cause enzyme inactivation. Also glutathiolationof the Cys-298 results in a significant inactivation of theenzyme (83, 88, 92). However, depending on the conditionsof the reaction and the nature of the NO donor used, AR iseither S-thiolated (inactivated enzyme) or S-nitrosated (ac-tivated enzyme). On the basis of these observations, we hy-pothesized that NO regulates the intracellular activity of ARand consequently the flux of glucose via the polyol pathway(9298). Because, in hyperglycemia, NO synthesis is signif-icantly less compared with normoglycemic subjects, it has

    been reasoned that the AR activity may be up-regulated indiabetic tissues (111, 112).

    Cardiovascular complications are one of the major causes

    FIG. 6. Molecular modeling showing two possible orien-

    tations of glutathione-propanal bound to AR. Orienta-tions 1 and 2 are shown in panels A and B, respectively.Carbon, nitrogen, sulfur, and oxygen atoms are colored

    gray, blue,yellow, and red, respectively. Potential hydro-gen bonds are indicated with dashed lines. [Reproducedwith permission from B. L. Dixit et al.: J Biol Chem275:2158721595, 2000 (67).]

    384 Endocrine Reviews, May 2005, 26(3):380 392 Srivastava et al. Aldose Reductase and Diabetic Complications

  • 7/27/2019 pdf Diabetic retinopathy

    6/13

    of mortality in patients with prolonged diabetes (113). Therole of AR in cardiovascular complications, especially inatherosclerosis (100), restenosis subsequent to balloon injury(53, 57), and cardiac preconditioning (103106), has beenextensively investigated. Vascular endothelial cells are themain source of NO for vascular smooth muscle cells becausein the vasculature, NO synthase is present mainly in theendothelium (114). The NO secreted by endothelial cellscould readily form GSNO with glutathione that is abundantin vascular smooth muscle cells (VSMC). The GSNO formedcould, in turn, readily S-glutathiolate AR at Cys-298 (92).Although GSSG (50100 m) also S-glutathiolates AR, it isunlikely that the physiological source of S-glutathiolation ofAR (or other proteins) could be GSSG because the concen-tration of GSSG in cells rarely exceeds 2025 m. BecauseS-glutathiolation inactivates AR, under normoglycemic con-ditions, it appears likely that a significant fraction of AR invascular tissues is present in an inactive form, whereas inhyperglycemia a decrease in NADPH/NADP ratio andother factors would decrease NO and the AR would be in the

    active form. Indeed, we have demonstrated that, in diabeticrat aorta, both AR activity and sorbitol levels are more than20-fold higher than nondiabetic aorta (95). Daily ip injectionsof NO synthase inhibitor, NG-nitro-l-argininemethyl ester orapplication of nitroglycerin patch significantly increased theactivity of aorta AR as well as sorbitol formation in bothnondiabetic and streptozotocin-diabetic rats. On the otherhand, daily injections of NO synthase substrate, l-arginine,significantly decreased AR activity and sorbitol content inaortas of both diabetic and nondiabetic rats. Similar resultswere obtained when aortic rings from normal and diabeticmice were incubated with l-arginine and NG-nitro-l-argin-inemethyl ester, whereas, both NO synthase substrate and

    inhibitor had no effect on AR activity or sorbitol content ofaorta from endothelial NO synthase-null mice (97). Interest-ingly, it was observed that S-glutathiolation of AR in VSMC

    by NO donors such as GSNO, which inactivates the enzyme,is reversible. Thus, the changes in glucose levels that alter thelevels of NO would change the AR activity, sorbitol levels,and related effects in diabetic subjects. Hence, in addition toglycemic control, NO donors or drugs that increase NO lev-els could represent one treatment modality for the preven-tion or treatment of diabetic complications. Because hyper-glycemia causes increased generation of reactive oxygenspecies by autooxidation of glucose and other metabolicpathways, antioxidants would also have beneficial effects.

    Diabetic complications such as cataractogenesis, retinopa-thy, and cardiovascular effects have been shown to be ame-liorated by antioxidants such as butylated hydroxytoluene,

    butylated hydroxyanisole, Trolox, ascorbate, vitamin E, N-acetyl cysteine, pyruvate, etc. (115120).

    IV. Regulation of Intracellular Signaling by AR

    Diabetes is a major risk factor for the development ofcardiovascular disease. It is associated with a 2- to 4-foldhigher risk of cardiovascular disease, and it accelerates theprogression and increases the severity of atherosclerotic le-sion formation in peripheral, coronary, and cerebral arteries

    (121125). Moreover, diabetics have a higher propensity forrestenosis after percutaneous transluminal coronary angio-plasty (126). Even though coronary stenting significantlyreduces restenosis, diabetes remains a powerful predictor ofin-stent restenosis (127). Processes that lead to an increase insmooth muscle cell growth in diabetic as well as nondiabeticrestenotic vessels have not been identified but, given theobservation that high levels of protein-HNE adducts areassociated with proliferative vascular lesion, it appears thatproducts of lipid peroxidation, such as HNE, play a signif-icant role in modulating the growth of vascular lesion. Thisis supported by our observation that treatment with lowconcentrations (2 m) of HNE increases VSMC growth inculture, although at higher concentrations HNE induced ap-optotic cell death (53). To test the role of HNE in vascularproliferation, we determined how changes in its metabolismvia AR would affect its mitogenic activity. Surprisingly, wefound that inhibition of AR prevented VSMC growth inculture (53). Serum-starved VSMC cultured in 1 m HNEshowed increased proliferation compared with cells cultured

    without HNE, as determined by MTT (3-[4,5-dimethylthia-zol-2-yl]-2,5-diphenyl tetrazolium bromide) assay and cellcount, whereas under similar conditions, HNE caused apo-ptosis in vascular endothelial cells and lens epithelial cells(Fig. 7). Both proliferation and apoptosis were attenuated byinhibiting AR using two structurally different AR inhibitors,sorbinil andtolrestat, andalso by using antisenseAR or smallinterfering RNA. In addition to HNE-stimulated growth,smooth muscle cells growing in response to serum FGF,TNF, or high glucose were also sensitive to AR inhibitors(128). Moreover, inhibition of AR also prevented smoothmuscle cell growth in vivo: we balloon-injured normal anddiabetic rat carotid artery and followed restenosis by quan-

    tifying the neointima formation (101). In both normal anddiabetic rats, AR inhibitor significantly (45%) prevented neo-intimal hyperplasia (Fig 8). Interestingly, the neointimal hy-

    FIG. 7. Inhibition of AR attenuates proliferation of VSMC and apo-ptosisof human umbilical vein endothelial cells (HUVEC)and humanlens epithelial cells (HLEC). Growth-arrested cells were pretreatedwith or without AR inhibitors, sorbinil and tolrestat (10 M) for 24 hfollowed by stimulation with TNF (2 nM) for an additional 24 h. Cell

    viability was determined by measuring the incorporation of [3H]thy-midine (10 Ci/ml), added 6 h before the end of the experiment. Theextent of cell proliferation or apoptosis is expressed as percentagechange in cell viability. **, P 0.001; *, P 0.01; and #, P 0.001in highglucose (HG)AR inhibitor group compared with HG-treatedgroup alone.

    Srivastava et al. Aldose Reductase and Diabetic Complications Endocrine Reviews, May 2005, 26(3):380392 385

  • 7/27/2019 pdf Diabetic retinopathy

    7/13

    perplasia was associated with increased nuclear factor--bind-ing protein (NF-B) activation (128). This was significantlyprevented by sorbinil, suggesting that AR is required for sig-naling pathways responsible for NF-B activation and for theproliferation of smooth muscle cells in vascular lesions. To-gether, these observations suggest that AR is essential forsmooth muscle cell growth induced by several mitogenic path-ways and that inhibition of AR interrupts their growth in cul-ture and in vivo (53, 57, 128).

    Although the mechanism by which AR regulates growthremains unclear, several of the key signaling pathways re-lated to cell growth are sensitive to AR inhibition. Signifi-cantly, our studies reveal that inhibition of AR preventsactivation of the transcription factor NF-B stimulated witheither TNF (Fig. 9),FGF, angiotensinII, or high glucose (101,128). Inhibition of AR, however, does not seem to directlyinterfere with the DNA binding of NF-B, but preventsNF-B activation by extinguishing signaling events up-stream to dissociation of NF-B from IB and the nucleartranslocation of NF-B. This appears to be due to the inhi-

    bition of phosphorylation and degradation of IB in cellstreated with AR inhibitors. Significantly, inhibition of ARdoes not prevent NF-B when the cells are stimulated byphorbol esters, indicating that inhibition of AR does notinterfere with the IB-NF-B signaling pathway but preventssignaling events upstream to PKC (128).

    Similar to the effects observed with TNF, inhibition orablation of AR also interrupts the activation of the PKC-NF-B by high glucose (101). The sensitivity of both highglucose- and TNF-mediated cell growth to AR inhibitorssuggests that both stimuli might activate overlapping path-ways. Indeed, previous studies have shown that treatmentwith anti-TNF antibodies prevents accelerated restenosis indiabetic vessels (129), and in our laboratory treatment withanti-TNF antibodies was effective in preventing high glu-cose-induced smooth muscle cell growth in culture (K. V.Ramana, R. Tammali, A. Bhatnagar, and S. K. Srivastava,unpublished observations). Hence, it appears that the mito-genic effects of high glucose may be mediated, in part, bystimulation of cytokine release and autocrine activation of

    FIG. 8. AR inhibitor attenuates NF-B activation in bal-loon-injured normal and diabetic rat arteries. Sections ofballoon-injured arteries were obtained from nondiabeticand diabetic rats 10 and 21 d after balloon injury and werestained with anti-p65 antibodies. Immunoreactivity is ev-ident asa dark brown stain, whereas the nonreactive areasdisplay only the background color. The bar graph showsmean SEM values of the percent of neointima stained bythe anti-p65 antibody. *,P 0.05 in tolrestat vs. control;,

    P 0.05 in diabetic vs. control; and #, P 0.05 diabetic tolrestat vs. diabetic without tolrestat. [Reprinted with per-mission from K. V. Ramana et al.: Diabetes 53:29102920,2004 (101). 2004 American Diabetes Association.]

    FIG. 9. Inhibition of AR attenuatesTNF-induced activation of NF-B inVSMC and human umbilical vein en-dothelial cells (HUVEC). Quiescent

    VSMC (A) and HUVEC (B) were pre-incubated without or withthe indicatedconcentrations of sorbinil for 24 h andthen stimulated with 0.1 nM TNF for1 h. The nuclear extracts were pre-pared, and NF-B activity was mea-sured by EMSA. [A, Reproduced withpermission from K. V. Ramana et al.:

    J Biol Chem 277:3206332070, 2002(128); and B, reproduced with permis-sion from K. V. Ramana et al.: FASEB

    J 18:12091218, 2004 (134).]

    386 Endocrine Reviews, May 2005, 26(3):380 392 Srivastava et al. Aldose Reductase and Diabetic Complications

  • 7/27/2019 pdf Diabetic retinopathy

    8/13

    the cell growth pathways. How glucose could induce therelease of cytokines is unclear. However, the recent obser-vationthat thereleaseof TNF is regulated by PKC (130 132)suggests that activation of PKC by high glucose could triggerTNF release and stimulate growth. Because inhibition/ablation of AR prevents both PKC activation (Fig. 10) andTNF signaling, it could interrupt the initial trigger events aswell as subsequent autocrine stimulation of mitogenic sig-naling in cells exposed to high glucose (101, 133).

    Studies delineating the role of AR in intracellular signalingsuggest an alternative paradigm for understanding the con-tribution of this enzyme to the development of diabetic com-plications and the efficacy of AR inhibitors against secondarydiabetic complications, despite the detoxification role of theenzyme. Hence, inhibition of inflammation could representone mechanism by which beneficial effects could be derivedfrom inhibiting AR. Our studies show that key steps in theinflammatory process, such as NF-B activation and the in-crease in the expression of adhesion molecules ICAM-1 andVCAM-1 and monocyte adhesion, could be prevented by

    inhibiting AR (Fig. 11) (134). Moreover, inhibition of AR alsoprevents the cytotoxic effects of TNF on endothelial cells.

    Our studies have also shown that inhibition of AR bysorbinil or tolrestat prevents TNF-induced increase in Baxand Bad and the down-regulation of Bcl-2 (135, 136). Inhi-

    bition of AR also abrogates activator protein 1 DNA bindingactivity and prevents the activation of caspase-3, c-Jun N-terminal kinase, and p38 MAPK in cells stimulated by TNF,suggesting that AR could be a critical regulator of TNF-induced apoptotic signaling in endothelial cells (136). Giventhe key role of the endothelium in regulating atherogenesisand restenosis, the salutary effects of AR inhibitors on thesecells could further contribute to the ability of these drugs to

    limit inflammation and vascular adhesion. Although theoverall effects of inhibiting AR during atherosclerotic lesionformation remain to be studied, they arelikely to be complex.Given that inhibition of AR could prevent inflammatorychanges as well as increase the accumulation of lipid per-oxidation products, inhibition of AR could yield highly con-text-dependent results, and the benefits of inhibiting AR may

    be specific to the extent of lesion progression and hypergly-cemia and/or may require concurrent administration of an-tioxidants. Such duality and complexity of effects is, how-

    ever, expected. AR regulates oxidative and inflammatoryresponses, both of which could be beneficial or harmful de-pending upon the context and the extent to which they arestimulated and the rate at which they are resolved. Regard-less, much work remains to be done to further our under-standing of the enigmatic role of AR and its contribution ofantioxidant defenses as well as the development of second-ary diabetic complications.

    V. AR Inhibitors and the Treatment of Secondary

    Diabetic Complications

    Despite the initial promise, outcomes of clinical trials withAR inhibitors have been disappointing. Most studies revealonly modest improvement with multiple side effects. How-ever, despite such negative data, it would be unfortunate ifanti-AR therapy were to be relegated to a historical footnote.Clinical trials with AR inhibitors were designed and con-ducted with only limited information about the enzyme and

    with little or no understanding of its physiological role inprocesses other than the reduction of glucose. Moreover,most clinical trials were designed to examine only a limitedset of physiological and pathological end points and weremostly focused on diabetic neuropathy. However, subse-quent to initial clinical trials, much has been learned aboutthe enzyme and its role in glucose metabolism, detoxifica-tion, inflammation, and growth that will be critical in rede-signing clinical trials with a larger number of pathologicalindices and end points.

    Given the extensive data showing that by catalyzing thereduction of lipid-derived aldehydes and their glutathioneconjugates (62, 66, 67), AR protects against oxidative stress

    (101, 128), it may be beneficial to try a combination therapywith AR inhibitor and antioxidants. Because long-term di-abetes is associated with increased oxidative stress, the ben-eficial effects of AR inhibitors may be diminished by a con-current accumulation of lipid peroxidation products.Therefore, to prevent this, it may be necessary to concur-rently administer antioxidants that prevent lipid peroxida-tion or enhance the expression of antioxidant enzymes thatcan compensate for the lack of AR. Treatment with antioxi-dants may also keep the enzyme in the reduced form and

    FIG. 10. Ablation of AR attenuates cytokines,growthfactors, high glucose, HNE butnot phor-bol 12-myristate 13-acetate (PMA)-induced ac-tivation of PKC in VSMC. The VSMC weretransiently transfected with AR antisense orscrambled control oligonucleotides. Subse-quently, the cells were stimulated with TNF(0.1nM), basic FGF (bFGF) (5 ng/ml),platelet-derived growth factor (PDGF)-AB (5 ng/ml),angiotensin II (Ang-II; 2 M),HNE(1 M), highglucose (HG, 50 mM),orPMA(10nM)for4h.Themembrane-bound PKC activity was determinedby using the Promega Signa TECT PKC assaysystem (Promega Corp., Madison, WI). Barsrepresent mean SE (n 4). *,P 0.001in ARantisense group compared with control group.[A portion of the data was reproduced with per-mission from K. V. Ramana et al.: J Biol Chem277:3206332070, 2002 (128).]

    Srivastava et al. Aldose Reductase and Diabetic Complications Endocrine Reviews, May 2005, 26(3):380392 387

  • 7/27/2019 pdf Diabetic retinopathy

    9/13

    thus prevent drug resistance that develops due to AR mod-ification under conditions of chronic hyperglycemia. Addi-tionally, treatment with AR inhibitors could be restricted toa specific stage of disease development. Thus, for instance,inhibition of AR may be more beneficial during early stagesof diabetes, when oxidative stress is low, than during laterstages of the disease when, due to increased oxidative stressand oxidative modification of the enzyme, it may be difficultto inhibit AR or to accrue significant benefits from anti-ARinterventions without further increasing oxidative stress.

    Although changes in cellular metabolism and tissue injury

    have been monitored in both animal studies and clinicaltrials, changes in inflammation and inflammatory markershave not been carefully examined. In view of recent studiesshowing that inhibition of AR prevents multiple inflamma-

    tory pathways (Refs. 101, 128, and 134; Fig. 12), it may benecessary to examine how AR inhibitors affect systemic andlocal inflammation during diabetes. This appears to be par-ticularly critical because chronic inflammation has emergedto be one of the critical features of diabetic complications,particularly cardiovascular disease. Moreover, based on datashowing that inhibition of AR prevents restenosis, vascularsmooth muscle growth, and endothelial cell apoptosis (101,134136), it may be possible to design animal and clinicalstudies to test the efficacy of anti-AR therapy against micro-and macrovascular complications of diabetes, which appear

    to be the most serious and prevalent outcomes of prolongeddiabetes. Because protection against vascular changes wasaccompanied by an inhibition of cell signaling involved ininflammation, it appears likely that AR inhibitors may be

    FIG. 11. Inhibition of AR attenuates TNF-induced monocyte adhesion and cell surface expression of intercellular adhesion molecule (ICAM)-1and vascular cell adhesion molecule (VCAM)-1 in human umbilical vein endothelial cells (HUVEC). Quiescent HUVEC were preincubatedwithout or with 10 M sorbinil/tolrestat for 24 h, and then stimulated with 2 nM TNF for 24 h. After 24 h of incubation, THP-1 (monocytes)cells were added, and the incubation continued for another 12 h. Cell adhesion assays were performed using light microscope. To determinecellsurface expressionof adhesion moleculesafter stimulation with TNF, thecellswere fixed in 4% formaldehyde,and theexpression of ICAM-1and VCAM-1 was measured by ELISA using anti-VCAM-1 and anti-ICAM-1 antibodies, respectively. Bars represent mean SE (n 4). ***,

    P 0.001 in TNF- AR inhibitors-treated group compared with TNF-treated group. [Reproduced with permission from K. V. Ramana et

    al.: FASEB J 18:12091218, 2004 (134).]

    FIG. 12. Schematic representation ofAR involvement in cell signaling. Cyto-kines, growthfactors,and hyperglycemiagenerate ROS and cause peroxidation oflipids resulting in the generation of toxiclipid aldehydes such as HNE. AR effi-ciently reduces HNE and its conjugatewith glutathione (GSH) to 14-dihy-droxy-2-nonene (DHN) and GS-DHN, re-spectively. The reduced products of alde-hydes may be involved in the cytotoxicsignaling leading to cell death or growth

    via activation of phospholipase C (PLC)/PKC/NF-B pathway. Paracrine effectsof cytokines such as TNF amplify hy-perglycemia and other signals as shownin the pathway.

    388 Endocrine Reviews, May 2005, 26(3):380 392 Srivastava et al. Aldose Reductase and Diabetic Complications

  • 7/27/2019 pdf Diabetic retinopathy

    10/13

    effective against diseases other than diabetes, particularlythose that are associated with high levels of cytokine gen-eration and inflammation such as rheumatoid arthritis andsepsis. Acute intervention with AR inhibitors during sepsisappears to be particularly attractive because short-term treat-ment will promote recovery by inhibiting both cytokine sig-naling and production, without the risk of long-term treat-ment with AR inhibitors that may chronically increaseoxidative stress.

    Finally, to derive maximal benefits from AR inhibition, itis imperative to have specific and selective inhibitors. Al-though the currently available AR inhibitors bind to theenzyme with high affinity, they also display high levels ofnonspecific toxicity. Moreover, in the absence of detailedpharmacokinetic studies, it is unclear whether the dose of ARinhibitors used for clinical studies was effective in inhibitingAR. Hence, further studies are warranted to carefully deter-mine the extent of enzyme inhibition in human tissues for agiven dose of AR inhibitor and whether their efficacy persistsin diabetic tissues. More importantly, however, it appears

    that it may be necessary to design more specific inhibitors ofAR that could selectively inhibit the ability of the enzyme tocatalyze the reduction of glutathione conjugates and glucose,without inhibiting aldehyde detoxification. Our structure-activity studies (66, 67) with free and glutathione-conjugatedaldehydes suggest that there are distinct glutathione- andaldehyde-binding domains on the enzyme, and selectivemodification of the enzyme active site could prevent recog-nition and reduction of glutathione conjugates without af-fecting aldehyde reduction. These results suggest the inter-esting possibility that the signaling and detoxification rolesof AR could be regulated independently of each other andthat more selective inhibitors could be designed to selectively

    prevent cell injury without compromising antioxidant defense.

    VI. Conclusion

    Due to its ability to reduce a wide variety of aldehydes,ranging from membrane phospholipids to glucose, AR playsa complex role in cellular metabolism and signaling. Al-though identified initially as a glucose-reducing enzyme, theenzyme is now believed to be an important component ofantioxidant defense involved in the removal and detoxifi-cation of reactive aldehydes generated by lipid peroxidation.Inhibition of AR has been shown to prevent the developmentof diabetic complications in animal models; however, a crit-

    ical evaluation of the clinical efficacy of AR inhibitors awaitsa clearer understanding of the role of AR in regulating in-flammation and cell growth. More selective and effectiveinhibitors are needed to specifically inhibit the cytotoxic roleof AR in cell signaling without affecting its detoxificationrole. Such inhibitors are likely to be more effective in treatingsecondary diabetic complications by preventing inflamma-tion due to chronic hyperglycemia.

    Acknowledgments

    Address all correspondence and requests for reprints to: Satish K.Srivastava, Department of Human Biological Chemistry and Genetics,

    University of Texas Medical Branch, Galveston, Texas 77555. E-mail:[email protected]

    This work was supported, in part, by NIH Grants DK36118, EY01677,HL55477, and HL59378.

    References

    1. Hers HG 1956 The mechanism of the transformation of glucose infructose in the seminal vesicles. Biochim Biophys Acta 22:202203

    2. van Heyningen R 1959 Formation of polyols by the lens of the ratwith sugar cataract. Nature 468:194195

    3. Kinoshita JH, Dvornik D, Kraml M, Gabbay KH 1968 The effectof an aldose reductase inhibitor on the galactose-exposed rabbitlens. Biochim Biophys Acta 158:472475

    4. Varma SD, Kinoshita JH 1976 Inhibition of lens aldose reductaseby flavonoidstheir possible role in the prevention of diabetic cat-aracts. Biochem Pharmacol 25:25052513

    5. Varma SD, Mizuno A, Kinoshita JH 1977 Diabetic cataracts andflavonoids. Science 195:205206

    6. Kinoshita JH, Fukushi S, Kador P, Merola LO 1979 Aldose re-ductase in diabetic complications of the eye. Metabolism 28:462469

    7. Kinoshita JH,KadorP, CatilesM 1981 Aldose reductasein diabetic

    cataracts. JAMA 246:2572618. Kador PF, Robison Jr WG, Kinoshita JH 1985 The pharmacology

    of aldose reductase inhibitors. Annu Rev Pharmacol Toxicol 25:691714

    9. Dvornik D 1992 Aldose reductase inhibitors as pathobiochemicalprobes. J Diabetes Complications 6:2534

    10. Tsai SC, Burnakis TG 1993 Aldose reductase inhibitors:an update.Ann Pharmacother 27:751754

    11. Pfeifer MA, Schumer MP, Gelber DA 1997 Aldose reductase in-hibitors: the end of an era or the need for different trial designs?Diabetes 46:S82S89

    12. Pitts NE,GundersenK, Mehta DJ,Vreeland F, Shaw GL,PetersonMJ, Collier J 1986 Aldose reductase inhibitors in clinical practice.Preliminary studies on diabetic neuropathy and retinopathy.Drugs 32:3035

    13. Stribling D 1990 Clinical trials with aldose reductase inhibitors.

    Exp Eye Res 50:62162414. Hotta N, Kakuta H, Ando F, Sakamoto N 1990 Current progressin clinical trials of aldose reductase inhibitors in Japan. ExpEye Res50:625628

    15. Ranganathan S, Krempf M, Feraille E, Charbonnel B 1993 Shortterm effect of an aldose reductase inhibitor on urinary albuminexcretion rate (UAER) and glomerular filtration rate (GFR) in type1 diabetic patients with incipient nephropathy. Diabetes Metab19:257261

    16. Passariello N, Sepe J, Marrazzo G, De Cicco A, Peluso A, PisanoMC, Sgambato S, Tesauro P, DOnofrio F 1993 Effect of aldosereductase inhibitor (tolrestat) on urinary albumin excretion rateand glomerular filtration rate in IDDM subjects with nephropathy.Diabetes Care 16:789795

    17. Handelsman DJ, Turtle JR1981 Clinical trial ofan aldosereductaseinhibitor in diabetic neuropathy. Diabetes 30:459 464

    18.Jaspan J, Maselli R, Herold K, Bartkus C

    1983 Treatment of se-verely painful diabetic neuropathy with an aldose reductase in-hibitor: relief of pain and improved somatic and autonomic nervefunction. Lancet 2:758762

    19. Jaspan JB, Towle VL, Maselli R, Herold K 1986 Clinical studieswith an aldose reductase inhibitor in the autonomic and somaticneuropathies of diabetes. Metabolism 35:8392

    20. Srivastava SK, Petrash JM, Sadana IJ, Ansari NH, Partridge CA1982 Susceptibility of aldehyde and aldose reductases of humantissues to aldose reductase inhibitors. Curr Eye Res 2:407410

    21. Srivastava SK, Ansari NH 1988 Prevention of sugar-induced cata-ractogenesis in rats by butylated hydroxytoluene. Diabetes37:15051508

    22. Ansari NH, Srivastava SK 1990 Allopurinol promotes and buty-lated hydroxy toluene prevents sugar-induced cataractogenesis.Biochem Biophys Res Commun 168:939943

    23. Ansari NH, Bhatnagar A, Fulep E, Khanna P, Srivastava SK 1994

    Srivastava et al. Aldose Reductase and Diabetic Complications Endocrine Reviews, May 2005, 26(3):380392 389

  • 7/27/2019 pdf Diabetic retinopathy

    11/13

    Trolox protects hyperglycemia-induced cataractogenesis in cul-tured rat lens. Res Commun Chem Pathol Pharmacol 84:93104

    24. Bhatnagar A, Srivastava SK 1992 Aldose reductase: congenial andinjurious profiles of an enigmatic enzyme. Biochem Med MetabBiol 48:91121

    25. Sheetz MJ, King GL 2002 Molecular understanding of hypergly-cemias adverse effects for diabetic complications. JAMA 288:25792588

    26. Gonzalez RG, Barnett P, Aguayo J, Cheng HM, Chalack LT 1984Direct measurement of polyol pathway activity in the ocular lens.Diabetes 33:196199

    27. Liu SQ, Bhatnagar A, Ansari NH, Srivastava SK Identification ofthe reactive cysteine residue in human placenta aldose reductase.Biochim Biophys Acta 1993 1164:268272

    28. Bhatnagar A, LiuSQ, Ueno N, Chakrabarti B, Srivastava SK1994Human placental aldose reductase: role of Cys-298 in substrate andinhibitor binding. Biochim Biophys Acta 1205:207214

    29. Srivastava SK, Hair GA, Das B 1985 Activated and unactivatedforms of human erythrocyte aldose reductase. Proc Natl Acad SciUSA 82:72227226

    30. Del Corso A, Barsacchi D, Camici M, Garland D, Mura U 1989Bovine lens aldose reductase: identification of two enzyme forms.Arch Biochem Biophys 270:604 610

    31. Grimshaw CE 1990 Chromatographic separation of activated and

    unactivated forms of aldose reductase. Arch Biochem Biophys 278:27327632. Lorenzi M, Toledo S, Boss GR, Lane MJ, Montisano DF 1987 The

    polyol pathway and glucose 6-phosphate in human endothelialcells cultured in high glucose concentrations. Diabetologia 30:222227

    33. Lee AY, Chung SK, Chung SS 1995 Demonstration that polyolaccumulation is responsible for diabetic cataract by the use oftransgenic mice expressing the aldose reductase gene in the lens.Proc Natl Acad Sci USA 92:27802784

    34. Yagihashi S, Yamagishi S, Wada R, Sugimoto K, Baba M, WongHG, Fujimoto J, Nishimura C, Kokai Y 1996 Galactosemic neu-ropathy in transgenic mice for human aldose reductase. Diabetes45:5659

    35. Yagihashi S, Yamagishi SI, Wada Ri R, Baba M, Hohman TC,Yabe-Nishimura C, Kokai Y 2001 Neuropathy in diabetic mice

    overexpressing human aldose reductase and effects of aldose re-ductase inhibitor. Brain 124:2448245836. Song Z, Fu DT, Chan YS, Leung S, Chung SS, Chung SK 2003

    Transgenic mice overexpressing aldose reductase in Schwann cellsshow more severe nerve conduction velocity deficit and oxidativestress under hyperglycemic stress. Mol Cell Neurosci 23:638647

    37. Leto G, Pricci F, Amadio L, Iacobini C, Cordone S, Diaz-Horta O,Romeo G, Barsotti P, Rotella CM, di Mario U, Pugliese G 2001Increased retinal endothelial cell monolayer permeability induced

    by the diabetic milieu: role of advanced non-enzymatic glycationand polyol pathway activation. Diabetes Metab Res Rev 17:448458

    38. Yan SF, Ramasamy R, Naka Y, Schmidt AM 2003 Glycation, in-flammation, and RAGE: a scaffold for the macrovascular compli-cations of diabetes and beyond. Circ Res 93:11591169

    39. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T,Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP, GiardinoI, Brownlee M 2000 Normalizing mitochondrial superoxide pro-duction blocks three pathways of hyperglycaemic damage. Nature404:787790

    40. WilliamsonJR, Chang K, Frangos M, Hasan KS,Ido Y, KawamuraT, Nyengaard JR, van den Enden M, Kilo C, Tilton RG 1993Hyperglycemic pseudohypoxia and diabetic complications. Dia-

    betes 42:80181341. Ido Y, Williamson JR 1997 Hyperglycemic cytosolic reductive

    stress pseudohypoxia: implications for diabetic retinopathy. In-vest Ophthalmol Vis Sci 38:14671470

    42. Obrosova IG, Stevens MJ, Lang HJ 2001 Diabetes-inducedchanges in retinal NAD-redox status: pharmacological modulationand implications for pathogenesis of diabetic retinopathy. Phar-macology 62:172180

    43. Thomas TP, Porcellati F, Kato K, Stevens MJ, Sherman WR,Greene DA 1994 Effects of glucose on sorbitol pathway activation,

    cellular redox, and metabolism of myo-inositol, phosphoinositide,and diacylglycerol in cultured human retinal pigment epithelialcells. J Clin Invest 93:27182724

    44. Derubertis FR, Craven PA 1994 Activation of protein kinase C inglomerular cells in diabetes. Mechanisms and potential links to thepathogenesis of diabetic glomerulopathy. Diabetes 43:18

    45. KoyaD, KingGL Protein kinase C activation and the developmentof diabetic complications. Diabetes 47:859 866

    46. Graier WF, Grubenthal I, Dittrich P, Wascher TC, Kostner GM1995 Intracellular mechanism of high D-glucose-induced modula-tion of vascular cell proliferation. Eur J Pharmacol 294:221229

    47. Frank RN 2002 Potential new medical therapies for diabetic reti-nopathy: protein kinase C inhibitors. Am J Ophthalmol 133:693698

    48. Tuttle KR, Anderson PW 2003 A novel potential therapy for di-abetic nephropathy and vascular complications: protein kinase C inhibition. Am J Kidney Dis 42:456465

    49. Burg MB 1988 Role of aldose reductase and sorbitol in maintainingthe medullary intracellular milieu. Kidney Int 33:635641

    50. Burg MB 1995 Molecular basis of osmotic regulation. Am J Physiol268:F983F996

    51. Donohue PJ, Alberts GF, Hampton BS, Winkles JA 1994 Adelayed-early gene activated by fibroblast growth factor-1 encodesa protein related to aldose reductase. J Biol Chem 269:86048609

    52. Iwata T, Sato S, Jimenez J, McGowan M, Moroni M, Dey A,Ibaraki N, Reddy VN, Carper D 1999 Osmotic response elementis required for the induction of aldose reductase by tumor necrosisfactor-. J Biol Chem 274:79938001

    53. Ruef J, Liu SQ, Bode C, Tocchi M, Srivastava S, Runge MS,Bhatnagar A 2000 Involvement of aldose reductase in vascularsmooth muscle cell growth and lesion formation after arterial in-

    jury. Arterioscler Thromb Vasc Biol 20:1745175254. Spycher SE, Tabataba-Vakili S, ODonnell VB, Palomba L, Azzi

    A 1997 Aldose reductase induction: a novel response to oxidativestress of smooth muscle cells. FASEB J 11:181188

    55. Tawata M, Ohtaka M, Hosaka Y, Onaya T 1992 Aldose reductasemRNA expression and its activity are induced by glucose in fetalrat aortic smooth muscle (A10) cells. Life Sci 51:719726

    56. Ramana KV, Chandra D, Srivastava S, Bhatnagar A, SrivastavaSK 2003 Aldose reductase mediates the mitogenic signals of cy-

    tokines. Chem Biol Interact 143144:58759657. Bhatnagar A, Ruef J, Liu S, Srivastava S, Srivastava SK 2001

    Regulation of vascular smooth muscle cell growth by aldose re-ductase. Chem Biol Interact 130132:627 636

    58. SibbittJr WL, Mills RG,BiglerCF, EatonRP, Griffey RH,VanderJagt DL 1989 Glucose inhibition of human fibroblast proliferationand response to growth factors is prevented by inhibitors of aldosereductase. Mech Ageing Dev 47:265279

    59. Derylo B, Babazono T, Glogowski E, Kapor-Drezgic J, HohmanT, Whiteside C 1998 High glucose-induced mesangial cell alteredcontractility: role of the polyol pathway. Diabetologia 41:507515

    60. Asnaghi V, GerhardingerC, Hoehn T, AdebojeA, Lorenzi M 2003A role for the polyol pathway in the early neuroretinal apoptosisand glial changes induced by diabetes in the rat. Diabetes 52:506511

    61. Price SA, Agthong S, Middlemas AB, Tomlinson DR 2004 Mi-

    togen-activated protein kinase p38 mediates reduced nerve con-duction velocity in experimental diabetic neuropathy: interactionswith aldose reductase. Diabetes 53:18511856

    62. Srivastava S, Watowich SJ, Petrash JM, Srivastava SK, BhatnagarA 1999 Structural and kinetic determinants of aldehyde reduction

    by aldose reductase. Biochemistry 38:425463. Srivastava S, Liu SQ, Conklin DJ, Zacarias A, Srivastava SK,

    Bhatnagar A 2001 Involvement of aldose reductase in the metab-olism of atherogenic aldehydes. Chem Biol Interact 130132:563571

    64. Srivastava S, Chandrasekar B, Bhatnagar A, Prabhu SD 2002Lipid peroxidation-derived aldehydes and oxidative stress in thefailing heart: role of aldose reductase. Am J Physiol Heart CircPhysiol 283:H2612H2619

    65. RamanaKV, Dixit BL, SrivastavaS, Bhatnagar A, Balendiran GK,Watowich SJ, Petrash JM, Srivastava SK 2001 Characterization of

    390 Endocrine Reviews, May 2005, 26(3):380 392 Srivastava et al. Aldose Reductase and Diabetic Complications

  • 7/27/2019 pdf Diabetic retinopathy

    12/13

    the glutathione binding site of aldose reductase. Chem Biol Interact130132:537548

    66. Ramana KV, Dixit BL, Srivastava S, Balendiran GK, SrivastavaSK, Bhatnagar A 2000 Selective recognition of glutathiolated al-dehydes by aldose reductase. Biochemistry 39:1217212180

    67. Dixit BL,Balendiran GK,Watowich SJ,SrivastavaS, RamanaKV,Petrash JM, Bhatnagar A, Srivastava SK 2000 Kinetic and struc-tural characterization of the glutathione-binding site of aldose re-

    ductase. J Biol Chem 275:215872159568. Srivastava S, Spite M, Trent JO, West MB, Ahmed Y, Bhatnagar

    A 2004 Aldose reductase-catalyzed reduction of aldehyde phos-pholipids. J Biol Chem 279:5339553406

    69. Bhatnagar A, Srivastava S, Wang LF, Chandra A, Ansari NH,Srivastava SK 1999 Cardiac metabolism of enals. Adv Exp MedBiol 463:223229

    70. Srivastava SK, Chandra A, Srivastava S, Petrash JM, BhatnagarA 1999 Regulation of aldose reductase by aldehydes and nitricoxide. Adv Exp Med Biol 463:501507

    71. Nakano T, Petrash JM 1996 Kinetic and spectroscopic evidence foractive site inhibition of human aldose reductase. Biochemistry 35:1119611202

    72. Schade SZ, Early SL, Williams TR, Kezdy FJ, Heinrikson RL,Grimshaw CE, Doughty CC 1990 Sequence analysis of bovine lensaldose reductase. J Biol Chem 265:36283635

    73. Wilson DK, Bohren KM, Gabbay KH, Quiocho FA 1992 An un-likely sugar substrate site in the 1.65 A structure of the humanaldosereductaseholoenzyme implicated in diabetic complications.Science 257:8184

    74. Wilson DK, Tarle I, Petrash JM, Quiocho FA 1993 Refined 1.8 Astructure of human aldose reductase complexed with the potentinhibitor zopolrestat. Proc Natl Acad Sci USA 90:98479851

    75. Tarle I, Borhani DW, Wilson DK, Quiocho FA, Petrash JM 1993Probing the active site of human aldose reductase. Site-directedmutagenesis of Asp-43, Tyr-48, Lys-77, and His-110. J Biol Chem268:2568725693

    76. Petrash JM, Tarle I, Wilson DK, Quiocho FA 1994 Aldose reduc-tase catalysis and crystallography. Insights from recent advances inenzyme structure and function. Diabetes 43:955959

    77. Borhani DW, Harter TM, Petrash JM 1992 The crystal structure ofthe aldose reductase. NADPH binary complex. J Biol Chem 267:

    248412484778. Srivastava S, Chandra A, Ansari NH, Srivastava SK, Bhatnagar A1998 Identification of cardiac oxidoreductase(s) involved in themetabolism of the lipid peroxidation-derived aldehyde-4-hydrox-ynonenal. Biochem J 329:469475

    79. Srivastava S, Chandra A, Wang LF, Seifert Jr WE, DaGue BB,Ansari NH, Srivastava SK, Bhatnagar A 1998 Metabolism of thelipid peroxidation product, 4-hydroxy-trans-2-nonenal, in isolatedperfused rat heart. J Biol Chem 273:1089310900

    80. Srivastava S, Dixit BL, Cai J, Sharma S, Hurst HE, Bhatnagar A,Srivastava SK 2000 Metabolism of lipid peroxidation product,4-hydroxynonenal (HNE) in rat erythrocytes: role of aldose reduc-tase. Free Radic Biol Med 29:642651

    81. Srivastava S, Conklin DJ, Liu SQ, Prakash N, Boor PJ, SrivastavaSK, Bhatnagar A 2001 Identification of biochemical pathways forthe metabolism of oxidized low-density lipoprotein derived alde-hyde-4-hydroxy trans-2-nonenal in vascular smooth muscle cells.Atherosclerosis 158:339350

    82. Del Corso A, Dal Monte M, Vilardo PG, Cecconi I, Moschini R,Banditelli S, Cappiello M, Tsai L, Mura U 1998 Site-specific in-activation of aldose reductase by 4-hydroxynonenal. Arch BiochemBiophys 350:245248

    83. Cappiello M, Voltarelli M, Giannessi M, Cecconi I, Camici G,Manao G, Del Corso A, Mura U 1994 Glutathione dependentmodification of bovine lens aldose reductase. Exp Eye Res 58:491501

    84. Cappiello M, Amodeo P, Mendez BL, Scaloni A, Vilardo PG,Cecconi I, Dal Monte M, Banditelli S, Talamo F, Micheli V,GiblinFJ, CorsoAD, Mura U 2001 Modulation of aldose reductaseactivity through S-thiolation by physiological thiols. Chem BiolInteract 130132:597 608

    85. Cappiello M, Vilardo PG, Cecconi I, Leverenz V, Giblin FJ, DelCorso A, Mura U 1995 Occurrence of glutathione-modified aldose

    reductasein oxidatively stressed bovine lens.Biochem Biophys ResCommun 207:775782

    86. Cappiello M, Voltarelli M, Cecconi I, Vilardo PG, Dal Monte M,Marini I, DelCorso A, Wilson DK,Quiocho FA, Petrash JM,MuraU 1996 Specifically targeted modification of human aldose reduc-tase by physiological disulfides. J Biol Chem 271:3353933544

    87. Del Corso A, Camici M, Mura U 1987 In vitro modification ofbovine lens aldose reductase activity. Biochem Biophys Res Com-

    mun 148:36937588. Petrash JM, Harter TM, Devine CS, Olins PO, Bhatnagar A, Liu

    S, Srivastava SK 1992 Involvement of cysteine residues in catalysisand inhibition of human aldose reductase. Site-directed mutagen-esis of Cys-80, -298, and -303. J Biol Chem 267:2483324840

    89. Cecconi I, Scaloni A, Rastelli G, Moroni M, Vilardo PG, Costan-tino L, Cappiello M, Garland D, Carper D, Petrash JM, Del CorsoA, Mura U 2002 Oxidative modification of aldose reductase in-duced by copper ion. Definition of the metal-protein interactionmechanism. J Biol Chem 277:4201742027

    90. Halder AB, James M, Crabbe C 1985 Chemical modification stud-ies on purified bovine lens aldose reductase. Ophthalmic Res 17:185188

    91. Cecconi I, Moroni M, Vilardo PG, Dal Monte M, Borella P, Ras-telliG, Costantino L,Garland D, Carper D, Petrash JM, DelCorsoA, Mura U 1998 Oxidative modification of aldose reductase in-

    duced by copper ion. Factors and conditions affecting the process.Biochemistry 37:141671417492. Chandra A, Srivastava S, Petrash JM, Bhatnagar A, Srivastava SK

    1997 Active site modification of aldose reductase by nitric oxidedonors. Biochim Biophys Acta 1341:217222

    93. Dixit BL, Ramana KV, Chandra D, Jackson EB, Srivastava S,Bhatnagar A, Srivastava SK 2001 Metabolic regulation of aldosereductase activity by nitric oxide donors. Chem Biol Interact 130132:573581

    94. Srivastava S, Dixit BL, Ramana KV, Chandra A, Chandra D,Zacarias A, Petrash JM, Bhatnagar A, Srivastava SK 2001 Struc-tural and kinetic modifications of aldose reductase by S-nitroso-thiols. Biochem J 358:111118

    95. Chandra D, Jackson EB, Ramana KV, Kelley R, Srivastava SK,Bhatnagar A 2002 Nitric oxideprevents aldose reductase activationand sorbitol accumulation during diabetes. Diabetes 51:30953101

    96. Srivastava SK, Ramana KV, Chandra D, Srivastava S, BhatnagarA 2003 Regulation of aldose reductase and the polyol pathwayactivity by nitric oxide. Chem Biol Interact 143144:333340

    97. Ramana KV, Chandra D, Srivastava S, Bhatnagar A, SrivastavaSK 2003 Nitric oxide regulates the polyol pathway of glucosemetabolism in vascular smooth muscle cells. FASEB J 17:417425

    98. Barisani D, Meneveri R, Ginelli E, Cassani C, Conte D2000 Ironoverload and gene expression in HepG2 cells: analysis by differ-ential display. FEBS Lett 469:208212

    99. Oconnor T, Ireland LS, Harrison DJ, Hayes JD 1999 Major dif-ferences exist in the function and tissue-specific expression of hu-man aflatoxin B1 aldehyde reductase and the principal humanaldo-keto reductase AKR1 family members. Biochem J 343:487504

    100. Srivastava S, Chandrasekar B, Bhatnagar A, Prabhu SD 2002Lipid peroxidation-derived aldehydes and oxidative stress in thefailing heart: role of aldose reductase. Am J Physiol 283:H2612H2619

    101. Ramana KV, Friedrich B, Srivastava S, Bhatnagar A, SrivastavaSK 2004 Activation of nuclear factor-B by hyperglycemia in vas-cular smooth muscle cells is regulated by aldose reductase. Dia-

    betes 53:29102920102. Rittner HL, Hafner V, Klimiuk PA, Szweda LI, Goronzy JJ,

    Weyand CM 1999 Aldose reductase functions as a detoxificationsystem for lipid peroxidation products in vasculitis. J Clin Invest103:10071013

    103. Shinmura K, Bolli R, Liu SQ, Tang XL, Kodani E, Xuan YT,Srivastava S, Bhatnagar A 2002 Aldose reductase is an obligatorymediator of the late phase of ischemic preconditioning. Circ Res91:240246

    104. Hwang YC, Sato S, Tsai JY, Yan S, Bakr S, Zhang H, Oates PJ,Ramasamy R 2002 Aldose reductase activation is a key componentof myocardial response to ischemia. FASEB J 16:243245

    105. Ramasamy R, Oates PJ, Schaefer S 1997 Aldose reductase inhi-

    Srivastava et al. Aldose Reductase and Diabetic Complications Endocrine Reviews, May 2005, 26(3):380392 391

  • 7/27/2019 pdf Diabetic retinopathy

    13/13

    bition protects diabetic and nondiabetic rat hearts from ischemicinjury. Diabetes 46:292300

    106. Kaneko M, Ramasamy R 2004 Aldose reductase: a key player inmyocardial ischemic injury. Exerc Sport Sci Rev 32:1923

    107. Hwang YC, Kaneko M, Bakr S, Liao H, Lu Y, Lewis ER, Yan S,Ii S, Itakura M, RuiL, Skopicki H, Homma S, Schmidt AM, OatesPJ, Szabolcs M, Ramasamy R 2004 Central role for aldose reduc-tase pathway in myocardial ischemic injury. FASEB J 18:11921199

    108. Horvath JJ, Witmer CM, Witz G 1992 Nephrotoxicity of the 1:1acrolein-glutathione adduct in the rat. Toxicol Appl Pharmacol117:200207

    109. Chakrabarti S, Malick MA 1991 In vivo nephrotoxic action of anisomeric mixture of S-(1-phenyl-2-hydroxyethyl)glutathione andS-(2-phenyl-2-hydroxyethyl)glutathione in Fischer-344 rats. Toxi-cology 67:1527

    110. Boon PJ, Marinho HS, Oosting R, Mulder GJ 1999 Glutathioneconjugation of 4-hydroxy-trans-2,3-nonenal in the rat in vivo, theisolated perfused liver and erythrocytes. Toxicol Appl Pharmacol159:214223

    111. Kicic E, Palmer TN 1996 Increased white cell aldose reductasemRNA levels in diabetic patients. Diabetes Res Clin Pract 33:3136

    112. Shimizu H, Ohtani KI, Tsuchiya T, Sato N, Tanaka Y, TakahashiH, Uehara Y, Inukai T, Mori M 2000 Aldose reductase mRNAexpression is associated with rapid development of diabetic mi-

    croangiopathy in Japanese type 2 diabetic (T2DM) patients. Dia-betes Nutr Metab 13:7579113. Spallarossa P, Barsotti A, Cordera R, Ghigliotti G, Maggi D,

    Brunelli C 2004 Reduction of cardiovascular morbidity and mor-tality in type 2 diabetes. A rational approach to hypoglycemictherapy. J Endocrinol Invest 27:485495

    114. Moncada S, Palmer RM, Higgs EA 1991 Nitric oxide: physiology,pathophysiology, and pharmacology. Pharmacol Rev 43:109142

    115. Wu SY, Leske MC 2000 Antioxidants and cataract formation: asummary review. Int Ophthalmol Clin 40:71 81

    116. Richer S 2000 Antioxidants and the eye. Int Ophthalmol Clin 40:116

    117. Kowluru RA, Kennedy A 2001 Therapeutic potential of anti-oxi-dants and diabetic retinopathy. Expert Opin Investig Drugs 10:16651676

    118. Dickinson PJ, Carrington AL, Frost GS, Boulton AJ 2002 Neuro-

    vascular disease, antioxidants and glycation in diabetes. DiabetesMetab Res Rev 18:260272119. Vaziri ND 2004 Roles of oxidative stress and antioxidant therapy

    in chronic kidney disease and hypertension. Curr Opin NephrolHypertens 13:9399

    120. Ruhe RC, McDonald RB 2001 Use of antioxidant nutrients in theprevention and treatment of type 2 diabetes. J Am Coll Nutr 20:363S369S

    121. Basta G, Schmidt AM, De Caterina R 2004 Advanced glycationend products and vascular inflammation: implications for accel-erated atherosclerosis in diabetes. Cardiovasc Res 63:582592

    122. Dandona P, Chaudhuri A, Aljada A 2004 Endothelial dysfunctionand hypertension in diabetes mellitus. Med Clin North Am 88:911931

    123. Wilson Tang WH, Maroo A, Young JB 2004 Ischemic heart diseaseand congestive heart failure in diabetic patients. Med Clin NorthAm 88:10371061

    124. Lim HS, Lip 2003 GY Diabetes, the renin-angiotensin system andheart disease. Curr Vasc Pharmacol 1:225238

    125. Lim HS, MacFadyen RJ, Lip GY 2004 Diabetes mellitus, the renin-angiotensin-aldosterone system, and the heart. Arch Intern Med164:17371748

    126. Morgan KP, Kapur A, Beatt KJ 2004 Anatomy of coronary disease

    in diabetic patients: an explanation for poorer outcomes after per-cutaneous coronary intervention and potential target for interven-tion. Heart 90:732738

    127. Indolfi C, Mongiardo A, Curcio A, Torella D 2003 Molecularmechanisms of in-stent restenosis and approach to therapy witheluting stents. Trends Cardiovasc Med 3:142148

    128. Ramana KV, Chandra D, Srivastava S, Bhatnagar A, AggarwalBB, Srivastava SK 2002 Aldose reductase mediates mitogenic sig-

    naling in vascular smooth muscle cells. J Biol Chem 277:3206332070

    129. Zhou Z, Lauer MA, Wang K, Forudi F, Zhou X, Song X, SolowskiN, Kapadia SR, Nakada MT, Topol EJ, Lincoff AM 2002 Effect ofanti-tumornecrosisfactor-polyclonalantibody on restenosis after

    balloon angioplasty in a rabbit atherosclerotic model. Atheroscle-rosis 161:153159

    130. Catley MC, CambridgeLM, Nasuhara Y, Ito K, Chivers JE,BeatonA, Holden NS, Bergmann MW, Barnes PJ, Newton R 2004 Inhib-itors of protein kinase C (PKC) prevent activatedtranscription: roleof events downstream of NF-B DNA binding. J Biol Chem 279:1845718466

    131. Foey AD, Brennan FM 2004 Conventional protein kinase C andatypical protein kinase Cdifferentially regulate macrophage pro-duction of tumornecrosisfactor- and interleukin-10. Immunology112:4453

    132. Satoh A, Gukovskaya AS, Nieto JM, Cheng JH, Gukovsky I,Reeve Jr JR, Shimosegawa T, Pandol SJ2004 PKC- and- regulateNF-B activation induced by cholecystokinin and TNF- in pan-creatic acinar cells. Am J Physiol Gastrointest Liver Physiol 287:G582G591

    133. RamanaKV, Friedrich B, Bhatnagar A, SrivastavaSK 2003 Aldosereductase mediates cytotoxic signals of hyperglycemia and TNF-in human lens epithelial cells. FASEB J 17:315317

    134. Ramana KV, Bhatnagar A, SrivastavaSK 2004 Inhibition of aldosereductase attenuates TNF--induced expression of adhesion mol-ecules in endothelial cells. FASEB J 18:12091218

    135. Chandra D, Ramana KV, Friedrich B, Srivastava S, Bhatnagar A,Srivastava SK 2003 Role of aldose reductase in TNF--inducedapoptosis of vascular endothelial cells. Chem Biol Interact 143144:605612

    136. Ramana KV, Bhatnagar A, Srivastava SK 2004 Aldose reductase

    regulates TNF-

    -induced cell signaling and apoptosis in vascularendothelial cells. FEBS Lett 570:189194137. Srivastava S, Ramana KV, Srivastana SK, DSouza SE, Bhatnagar

    A 2004 Role of aldose reductase in the detoxification of oxidizedphospholipids. ACS Symposium Series 865:49 64

    138. Vander Jagt DL, Hassebrook RK, Hunsaker LA, Brown WM,Royer RE 2001 Metabolism of the 2-oxoaldehyde methylglyoxal byaldose reductase and by glyoxalase-I: roles for glutathione in bothenzymes and implications for diabetic complications. Chem BiolInteract 130132:549562

    139. Wermuth B, Monder C 1983 Aldose and aldehyde reductase ex-hibit isocorticosteroid reductase activity. Eur J Biochem 131:423426

    140. Matsuura K, Deyashiki Y, Bunai Y, Ohya I, Hara A 1996 Aldosereductase is a major reductase for isocaproaldehyde, a product ofside-chain cleavage of cholesterol, in human and animal adrenalglands. Arch Biochem Biophys 328:265271

    141. Petrash JM, Harter TM, Murdock GL 1996 A potential role foraldose reductase in steroid metabolism. In: Weiner H, Lindahl R,Crabb DW, Flynn TG, eds. Enzymology and molecular biology ofcarbonyl metabolism. Vol 6. New York: Plenum Press; 465473

    142. Srivastava S, Liu SQ, Bhatnagar A 2003 Cellular metabolism ofbase propanols. FASEB J 17:A175A176

    Endocrine Reviews is published bimonthly by The Endocrine Society (http://www.endo-society.org), the foremost professional societyserving the endocrine community.

    392 Endocrine Reviews, May 2005, 26(3):380 392 Srivastava et al. Aldose Reductase and Diabetic Complications