Vous êtes sur la page 1sur 10

Free Radical Biology & Medicine, Vol. 34, No. 2, pp.

186 195, 2003


Copyright 2003 Elsevier Science Inc.
Printed in the USA. All rights reserved
0891-5849/03/$see front matter

PII S0891-5849(02)01195-4

Original Contribution
EARLY OXIDATIVE STRESS IN THE DIABETIC KIDNEY: EFFECT OF
DL--LIPOIC ACID

IRINA G. OBROSOVA,* LAMIA FATHALLAH,* EDWIN LIU, and JAFFAR NOUROOZ-ZADEH


*Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA; and Department of Medicine,
University College London, Royal Free and University College Medical School, London, UK

(Received 14 August 2002; Revised 20 September 2002; Accepted 2 October 2002)

AbstractOxidative stress is implicated in the pathogenesis of diabetic nephropathy. The attempts to identify early
markers of diabetes-induced renal oxidative injury resulted in contradictory findings. We characterized early oxidative
stress in renal cortex of diabetic rats, and evaluated whether it can be prevented by the potent antioxidant, DL--lipoic
acid. The experiments were performed on control rats and streptozotocin-diabetic rats treated with/without DL--lipoic
acid (100 mg/kg i.p., for 3 weeks from induction of diabetes). Malondialdehyde plus 4-hydroxyalkenal concentration
was increased in diabetic rats vs. controls (p .01) and this increase was partially prevented by DL--lipoic acid. F2
isoprostane concentrations (measured by GCMS) expressed per either mg protein or arachidonic acid content were not
different in control and diabetic rats but were decreased several-fold with DL--lipoic acid treatment. Both GSH and
ascorbate (AA) levels were decreased and GSSG/GSH and dehydroascorbate/AA ratios increased in diabetic rats vs.
controls (p .01 for all comparisons), and these changes were completely or partially (AA) prevented by DL--lipoic
acid. Superoxide dismutase, glutathione peroxidase, glutathione reductase, glutathione transferase, and NADH oxidase,
but not catalase, were upregulated in diabetic rats vs. controls, and these activities, except glutathione peroxidase, were
decreased by DL--lipoic acid. In conclusion, enhanced oxidative stress is present in rat renal cortex in early diabetes,
and is prevented by DL--lipoic acid. 2003 Elsevier Science Inc.

KeywordsRenal cortex, F2-isoprostanes, Malondialdehyde, 4-Hydroxyalkenals, Glutathione, Ascorbate, Antioxida-


tive enzymes, Rat, Streptozotocin diabetes, Free radicals

INTRODUCTION [3,4], tubulo-interstitium [5], and vasculature [6], and


has been implicated in mesangial expansion of extra-
The epidemiological studies [1] and Diabetes Control
cellular matrix [7,8] and other early and late events of
and Complication Trial [2] point to a strong consistent
diabetic nephropathy, i.e., increased glomerular filtra-
relationship between hyperglycemia and the incidence
tion rate [9], urinary albumin excretion and proteinuria
and progression of diabetic nephropathy in patients with
[7,8,10], glomerulosclerosis [7,11], and tubulo-inter-
both Type 1 and Type 2 diabetes mellitus. One of the
stitial fibrosis [11]. Studies in animal models of dia-
most important consequences of glucose toxicity in
betes revealed that some of these functional and mor-
tissue-sites for diabetic complications is enhanced oxi-
phological abnormalities can be prevented by
dative stress, i.e., increased production of reactive oxy-
antioxidants [711]. However, in general, the findings
gen species combined with downregulation or insuffi-
are inconclusive because several reports indicated the
cient upregulation of antioxidant defense mechanisms.
absence of improvement [12] and even worsening [13]
Enhanced oxidative stress has been documented in all
of diabetic nephropathy with antioxidant treatment.
three compartments of the renal cortex, i.e., glomeruli
The latter could result from insufficient understanding
of the mechanisms underlying renal oxidative damage
Address correspondence to: Dr. Irina Obrosova, Division of Endo- and, in particular, very contradictory information on
crinology and Metabolism, Department of Internal Medicine, Univer- the effects of diabetes on major components of free
sity of Michigan Medical Center, 1150 West Medical Center Drive,
MSRB II, Rm 5560C, Ann Arbor, MI 48109-0678, USA; Tel: (734) radical production and antioxidative defense, which
763-3055; Fax: (734) 975-1849; E-Mail: iobrosso@umich.edu. hampers the selection of specific targets for antioxi-

186
Renal oxidative stress in diabetes 187

dant therapy as well as proper antioxidants and anti- istration prevented renal insufficiency, glomerulat mes-
oxidant combinations. angial matrix expansion, and glomerulosclerosis. Fur-
Among all existing animal models of diabetes, the thermore, a pilot clinical study has demonstrated a
streptozotocin (STZ)-induced diabetic rat model was beneficial effect of DL--lipoic acid on urinary albumin
characterized best, and was found to develop all major concentration in patients with diabetes mellitus [42].
nephropathic changes described in patients with diabetes
mellitus. The attempts to identify early markers of renal
cortex oxidative injury in STZ-diabetic rats resulted in MATERIALS AND METHODS
contradictory findings. Whereas most of the studies agree
The experiments were performed in accordance with
that enhanced oxidative stress and its major tissue man-
regulations specified by The Guiding Principles in the
ifestation, i.e., lipid peroxidation (LPO) are present in the
Care and Use of Animals (DHEW Publication, NIH
kidney cortex shortly after induction of diabetes [14 16],
80-23) and the University of Michigan Protocol for An-
no consensus has been achieved regarding the major
imal Studies.
mechanisms of free radical production as well as the
extent of compromise of the key antioxidative defense Animals
mechanisms. One group suggested the importance of
intramitochondrial superoxide production [17], whereas Male Wistar rats (Charles River, Wilmington, MA,
others point to the contribution of extramitochondrial, in USA), body weight 250 300 g, were fed a standard rat
addition to [18] or instead of [19] intramitochondrial chow diet (ICN Biomedicals, Cleveland, OH, USA) and
mechanisms, to diabetes-induced kidney oxidative dam- had ad libitum access to water. Diabetes was induced by
age. The concentrations of the key biological nonenzy- a single injection of streptozotocin (Upjohn, Kalamazoo,
matic antioxidant, reduced glutathione (GSH), were re- MI, USA; 55 mg/kg body weight, i.p.). Blood samples
ported decreased [20,21] or unchanged [10,22] in the for glucose measurements were taken from the tail vein
renal cortex of diabetic rats, and the major antioxidative ca. 48 h after streptozotocin injection and the day before
defense enzyme activities, i.e., superoxide dismutase the rats were killed. Rats with blood glucose 13.9
(SOD), catalase, glutathione peroxidase (GSHPx), gluta- mmol/l were considered as diabetic. The experimental
thione reductase (GSSGRed), and glutathione transferase groups included control rats and diabetic rats treated
(GSHTrans) were found decreased, unchanged, or in- with/without DL--lipoic acid (Sigma, St. Louis, MO,
creased [16,19,2328]. The information on other com- USA; 100 mg kg1 day1, i.p., for 3 weeks after
ponents of free radical generation and antioxidative de- induction of diabetes).
fense, e.g., the ascorbate system and NADH oxidase in Experimental procedure
the diabetic kidney, is either limited [29] or unavailable.
The purpose of this study was to characterize early Rats were sedated with carbon dioxide and killed by
changes in LPO and antioxidative defense in the renal cervical dislocation. Both kidneys were immediately dis-
cortex of diabetic rats, and to evaluate whether these sected and frozen in liquid nitrogen. Four different sets
changes can be prevented by DL--lipoic acid. DL-- of 30 mg renal cortex samples were either (i) extracted
lipoic acid is a universalantioxidant that combines free with 15.4 mol/l methanesulphonic acid and used for
radical scavenging and metal chelating properties with measurements of malondialdehyde (MDA) plus 4-hy-
an ability (after conversion to dihydrolipoic acid) to droxyalkenals (4-HA); (ii) extracted with 5% metaphos-
regenerate the levels of other nonenzymatic and enzy- phoric acid and used for measurements of dehydroascor-
matic antioxidants, i.e., GSH, ascorbate, -tocopherol, bate (DHAA) and total ascorbate (tAA); (iii) extracted
catalase, and GSH peroxidase [30,31]. DL--lipoic acid with 6% perchloric acid, neutralized [32], and used for
effectively counteracts oxidative stress in the lens [32], measurements of reduced (GSH) and oxidized (GSSG)
retina [33,34], and peripheral nerve [35,36] of streptozo- glutathione; or (iv) homogenized in 1 ml 0.1 mol/l
tocin-diabetic rats, and prevents diabetes-induced endo- sodium-phosphate buffer, pH 6.5, and used for mea-
thelial dysfunction [37] and manifestations of diabetic surements of superoxide dismutase, catalase, GSH
neuropathy [35,36,38 40], retinopathy [34], and cataract peroxidase, GSSG reductase, GSH transferase, and
[41]. In the same animal model, DL--lipoic acid was NADH oxidase activities and protein content. Renal
found to prevent early glomerular injury manifested by cortex samples of 200 mg were homogenized in 2 ml
increased urinary albumin excretion, fractional albumin 10 mmol/l phosphate buffer, pH 7.4, containing 100
clearance, glomerular volume, and glomerular content of mmol/l butylated hydroxytoluene (set 5). The homog-
immunoreactive transforming growth factor- and colla- enates were used for measurements of total (i.e., free
gen 1(IV) [10]. Recent studies from the same group [8] plus esterified) F2-isoprostane [43 45] and arachi-
revealed that chronic 7 month DL--lipoic acid admin- donic acid [46] concentrations.
188 I. G. OBROSOVA et al.

Biochemical measurements Maidenhead, UK). Quantitative analysis was performed


using selected ion monitoring (SIM) of the carboxylate
MDA plus 4-HA, GSH, and GSSG concentrations,
anion [M-181]- at m/z 569 and 573 for F2-isoprostanes
SOD, catalase, GSH peroxidase, GSSG reductase, GSH
and PGF2-d4 as the internal standard, respectively.
transferase, and NADH oxidase activities, and protein
For fatty acid analysis [46], 200 l aliquots of ho-
content were assayed as described [33].
mogenate (set 5) were mixed with 250 l ethanol con-
For measurements of DHAA and AA, the renal cortex
taining 100 g of heptadecanoic acid as an internal
homogenates in 5% meta-phosphoric acid were centri-
standard. Five hundred microliters of ethyl acetate and
fuged at 4000 g for 10 min. In the DHAA assay, 0.1
300 l water were then added to the homogenate, vor-
ml of the supernatant was mixed with 0.9 ml 2 mol/l
texed, and centrifuged at 1000 g for 5 min. The upper
Na-acetate buffer, pH 6.2. The reaction was started by
organic layer was then transferred to a new set of glass
addition of 0.02 ml 92.5 mM O-phenylenediamine (Sig-
vials. Five hundred microliters of ethyl acetate was
ma). The initial and final readings were taken at exci-
added to the remaining aqueous phase and the mixture
tation: 350 nm, emisssion: 430 nm. For measurements
was vortexed and centrifuged as described above. The
of tAA, 0.1 ml of the supernatant was mixed with 0.9 ml
upper organic layers were pooled and dried under a
2 M Na-acetate buffer, pH 6.2 and 10 U of ascorbate
stream of nitrogen. Fatty acid methyl esters were pre-
oxidase (Sigma) to convert ascorbate (AA) to DHAA.
pared by adding 500 l of 14% boron trifluoride solution
After completion of the reaction, DHAA was quantified
in methanol to the dried final lipid extract and incubated
as described above. The differences in initial and final
at 60C for 30 min. The samples were allowed to cool
readings in DHAA and tAA assays were compared with
before water (1 ml) and hexane (1 ml) were added. The
those of AA standards processed in the same run. AA
mixtures were vortexed and centrifuged. The hexane
levels were calculated as the difference between tAA and
(upper) layer was collected and dried. The residue was
DHAA.
redissolved in 100 l of hexane. Fatty acid analyses were
For measurements of total F2-isoprostanes [43 45], 1
carried out on a Fisons MFC 800 gas chromatograph
ml aliquots of homogenate from step 5 were transferred
equipped with a flame ionization detector and a splitless
into glass tubes and 500 l of 4 mol/l KOH were added.
injector. Fatty acids methyl esters were separated on an
After incubation at 45C for 45 min, the pH was adjusted
Omegawax 320 column (0.32 m 30 m, film thickness
to 3 by adding 500 l of 4 mol/l HCI. The internal
0.25 m; Supelco) using a temperature gradient of 120
standard 3,3',4,4'-tetradeuterated 9,11-prostaglandin
240C at 6C/min. One microliter samples were injected
F2 (PGF2-d4, 5 ng in 100 l ethanol) was added and the
into the gas chromatograph.
samples were vortex mixed. Then 10 ml ethyl acetate
was added, and the samples were vortexed for 20 s and Statistical analysis
centrifuged at 1000 g for 5 min. The upper organic
The results are expressed as mean SEM. Data were
layer was transferred into a new set of glass tubes. Total
subjected to equality of variance F test, and then to log
lipid extract was then applied onto an aminopropyl
transformation, if necessary, before one-way analysis of
(NH2) cartridge. The cartridge was sequentially washed
variance. When overall significance (p .05) was at-
with 10 ml of hexane/ethyl acetate (30/70, v/v), acetoni-
tained, individual between group comparisons were
trile/water (90/10, v/v) and acetonitrile. Isoprostanes
made using the Student- Newman-Keuls multiple range
were eluted by washing the cartridge with 5 ml of ethyl
test. Significance was defined at p .05. When between-
acetate/methanol/acetic acid (85/10/5, v/v/v). The final
group variance differences could not be normalized by
eluate from the NH2-cartridge was dried under nitrogen
log transformation, the data were analyzed by the non-
at 45C and was then converted to pentafluoro-benzyl
parametric Kruskal-Wallis one-way analysis of variance,
ester/trimethylsylil ether derivatives. Gas-chromatogra-
followed by the Fishers PLSD test for multiple compar-
phy-mass spectrometry/negative ion chemical ionization
isons.
(NICI) was carried out on a Hewlett Packard 5890 GC
linked to a VG70SEQ using the NICI with ammonia as
reagent gas as described elsewhere [44]. Briefly, samples RESULTS
(2 l) were injected onto a SPB-1701 (30 m 0.25 mm
I.D.; 0.25 m film thickness; Supelco, Dorset, England). The final body weights were lower in diabetic rats
Separation was carried out using a temperature program: than in the control group (Table 1). The initial body
initial temperature: 175C; initial time: 2 min; rate: weights (not shown) were similar in control and diabetic
30C/min; final temperature: 270C; final time: 30 min. groups. The final body weights in DL--lipoic acid-
Samples (2 l) were injected into a temperature pro- treated diabetic rats were slightly but significantly lower
grammed Gerstel injector (Thames Chromatography, than in the untreated diabetic group.
Renal oxidative stress in diabetes 189

Table 1. Final Body Weights and Blood Glucose Concentrations in tion, normalized per g of arachidonic acid (Fig. 2C),
Control Rats and Diabetic Rats Treated With/Without DL--lipoic
Acid (Mean SEM)
tended to increase in diabetic rats compared with the
control group, but the difference between the groups did
Blood not achieve statistical significance (p .12). It was 5.3
glucose lower in DL--lipoic acid-treated diabetic rats than in the
Body concentration
weight (g) (mmol/l) untreated diabetic group (p .01).
Renal cortex GSH concentration was slightly, but
Control (n 19) 352 4 3.61 0.11 significantly, decreased in diabetic rats compared with
Diabetic (n 20) 293 7* 20.6 0.61*
Diabetic DL--lipoic acid (n 16) 268 5*,** 19.7 0.61* the control group (Fig. 3A, p .01), and this decrease
was overcorrected by DL--lipoic acid treatment (p
The number of observations is indicated in parentheses. .01 vs. untreated diabetic group). GSSG concentrations
* Significantly different compared with controls (p .01).
** Significantly different compared with untreated diabetic group were similar among the experimental groups (Fig. 3B).
(p .01). GSSG/GSH ratio was 1.6-fold higher in diabetic rats
than in the control group (p .01, Fig. 3C), and this
increase was completely prevented by DL--lipoic acid
Blood glucose concentration was increased 5.5-fold in treatment (p .01 vs. untreated diabetic group).
diabetic rats compared with the control group, and was Renal cortex AA concentration was 2.2-fold lower in
not affected by DL--lipoic acid treatment. diabetic rats than in the control group (p .01, Fig. 4A),
Renal cortex MDA plus 4-HA concentration was 2.4- and this decrease was partially prevented by DL--lipoic
fold higher in diabetic rats compared with the control acid (p .01 vs. either control or untreated diabetic
group (p .01, Fig. 1). This increase was partially groups). DHAA concentrations were similar in control
prevented by DL--lipoic acid (p .05 vs. untreated and diabetic groups, but was significantly (p .05)
diabetic group, and p .05 vs. controls). lower in DL--lipoic acid-treated diabetic group com-
Renal cortex F2-isoprostane concentrations normal- pared with the nondiabetic control group (Fig. 4B).
ized per mg protein were similar in control and diabetic DHAA/AA ratio (Fig. 4C) was 2.2-fold higher in dia-
rats (Fig. 2A), but were 4-fold lower in DL--lipoic betic rats than in the control group (p .01), and this
acid-treated diabetic rats than in the corresponding un- increase was completely prevented by DL--lipoic acid
treated group (p .01). Renal cortex arachidonic acid (p .01 vs. untreated diabetic group).
concentration was 1.4-fold lower in diabetic rats than in Renal cortex SOD activity was 1.4-fold higher in
the control group (p .01, Fig. 2B), and this decrease diabetic rats than in the control group, and this upregu-
was prevented by DL--lipoic acid treatment (p .05 lation was completely prevented by DL--lipoic acid
vs. untreated diabetic group). F2-isoprostane concentra- (Table 2). Catalase activities were similar in control and
diabetic groups but were lower in DL--lipoic acid-
treated diabetic group compared with nondiabetic con-
trols. GSH peroxidase activity was increased 1.4-fold in
diabetic rats compared with the control group, and this
increase was not prevented by DL--lipoic acid treat-
ment. GSSG reductase and GSH transferase activities
were 1.2- and 1.3-fold higher in diabetic rats than in the
control group, and diabetes-associated upregulation of
both enzymes was prevented by DL--lipoic acid treat-
ment. NADH oxidase activity was 1.6-fold higher in
diabetic rats compared with the control group. This in-
crease was completely arrested by DL--lipoic acid;
furthermore, NADH oxidase activity was 2.8-fold lower
in DL--lipoic acid-treated diabetic rats than in the non-
diabetic control rats.

DISCUSSION

Fig. 1. Renal cortex MDA plus 4-HA concentrations in control rats and Our findings of increased LPO, upregulated NADH
diabetic rats treated with/without DL--lipoic acid (Mean SEM, n oxidase, compromised concentrations of the key nonen-
10). **Significantly different compared with the control group (p
.01); #Significantly different compared with the untreated diabetic zymatic antioxidants GSH and AA, as well as the gluta-
group (p .05). thione and ascorbate redox states, and increased SOD,
190 I. G. OBROSOVA et al.

Fig. 2. Renal cortex (A) F2-isoprostane concentrations normalized per mg protein; (B) arachidonic acid concentrations; (C) F2-
isoprostane concentrations normalized per g of arachidonic acid in control rats and diabetic rats treated with/without DL--lipoic acid
(Mean SEM, n 9 10). **Significantly different compared with the control group (p .01); #,##Significantly different compared
with the untreated diabetic group (p .05 and .01, respectively).

GSH peroxidase, GSSG reductase, and GSH transferase stress (i.e., lipid peroxidation [52,53] and GSH depletion
activities clearly indicate that enhanced oxidative stress [5355]) in diabetic tissues are corrected by aldose re-
is present in the renal cortex at a very early stage of ductase inhibitors, i.e., agents that do not interfere with
diabetes. Several lines of evidence suggest that renal the STZ effect on -cells but arrest hyperglycemia-in-
oxidative stress in STZ-diabetic rats is a consequence of duced sorbitol pathway activation in other tissues, at the
the diabetic state, and, primarily, hyperglycemia, rather doses far below the effective doses of the most effective
than well-established [47] pro-oxidant effect of STZ antioxidants, including DL--lipoic acid.
itself. First, the pro-oxidant effect of STZ is limited to Diabetes-induced LPO in the kidney is manifested by
pancreatic -cells, whereas tissue targets for diabetic a 2.4-fold increased concentration of MDA plus 4-HA,
complications, e.g., retina and peripheral nerve, do not and a trend towards an accumulation of F2-isoprostane,
develop oxidative changes during the first 10 d after the specific product of arachidonic acid peroxidation. It
induction of STZ-diabetes (Obrosova, unpublished). is important to realize that neither MDA plus 4-HA nor
Second, the half-life of STZ is very short (15 min), F2-isoprostane are ideal markers of LPO, and that both
whereas oxidative stress in the diabetic tissues, e.g. renal variables are affected by changes in other metabolic
glomeruli, is more advanced in chronic than in short- pathways present in diabetes. In particular, MDA is the
term STZ-diabetes [48]. Third, impaired antioxidative side product of prostaglandin metabolism [56,57], and
defense [49], DNA oxidative damage [50], and other tissue MDA concentrations are changed with inhibition
markers of oxidative stress (e.g., increased expression of of cyclooxygenase 1 [58], e.g., in diabetes [56]. In a
heme oxygenase-1 [51]) in the diabetic kidney are cor- similar fashion, tissue F2-isoprostane concentrations de-
rected by insulin administered to animals with estab- pend not only on the rate of LPO, but on arachidonic acid
lished STZ-induced diabetes. And fourth, oxidative abundance, the rate of arachidonate metabolism to pros-

Fig. 3. Renal cortex (A) GSH concentrations; (B) GSSG concentrations; (C) GSSG/GSH ratios, in control rats and diabetic rats treated
with/without DL--lipoic acid (Mean SEM, n 710). *,**Significantly different compared with the control group (p .05 and
.01, respectively); ##Significantly different compared with the untreated diabetic group (p .01).
Renal oxidative stress in diabetes 191

Fig. 4. Renal cortex (A) AA concentrations; (B) DHAA concentrations; (C) DHAA/AA ratios, in control rats and diabetic rats treated
with/without DL--lipoic acid (Mean SEM, n 8). *,**Significantly different compared with the control group (p .05 and .01,
respectively); ##Significantly different compared with the untreated diabetic group (p .01).

taglandin F2- and the prostaglandin clearance. Both NADH oxidase but not downregulation of major antioxi-
arachidonate biosynthesis and metabolism [56,59], and dative defense enzymes. The response of the glutathione
prostaglandin F2- clearance [60] are compromised in and ascorbate systems of antioxidative defense in the
diabetes. Thus, it is quite possible that both MDA and diabetic kidney is similar to the one in two other tissue
F2-isoprostane measurements in diabetic tissues, includ- sites for diabetic complications, i.e., lens and peripheral
ing the renal cortex, underestimate the rate of LPO. In nerve [32,35,36]. In all three tissues, GSH depletion
any case, it is preferable to use several markers of LPO parallels the loss of total glutathione, whereas accumu-
rather than MDA or F2-isoprostane alone. Our findings lation of the oxidized form, GSSG, is either minor or
of increased renal LPO in short-term diabetes are con- absent. At the same time, a decrease in the renal cortex
sistent with elevated lipid peroxide, malondialdehyde, GSH concentration in diabetes of a 3 week duration is
and fluorescent chromolipid concentrations modest compared with the corresponding changes in
[14 16,18,22] as well as increased rate of NADPH- peripheral nerve [62] and particularly, lens [63]. The
dependent LPO [61] in other reports. One group has latter is quite consistent with relatively low sorbitol path-
found LPO to depend on duration of diabetes with way activity in the renal cortex compared with two other
clearly manifested LPO 1 week after induction of diabe- tissues [29,32,36,62 64]. The inverse relationship be-
tes, its drop to baseline at 2 4 weeks, and second pro- tween the sorbitol pathway activity and GSH concentra-
gressive increase after 5 and 6 weeks [19]. These obser- tions in tissue sites for diabetic complications has been
vations are different from ours because we found revealed in a number of studies, performed not only with
increased LPO in rats with 3 week duration of STZ- structurally diverse aldose reductase (AR) inhibitors
diabetes. [52,54,55,65 67], but also in AR-overexpressing trans-
Enhanced renal oxidative stress in short-term diabetes genic [68] and AR-knockout [69] mice. The recent study
is associated with the loss of two major nonenzymatic of our group [70], however, indicates that the mecha-
antioxidants, GSH and AA, compromised glutathione nisms underlying GSH depletion in diabetic tissues are
and ascorbate redox states, upregulation of cytoplasmic complex and involve downstream pathways, i.e., poly-

Table 2. Antioxidative Defense Enzyme and NADH Oxidase Activities (nmol/min per mg Protein) in the
Renal Cortex of Control Rats and Diabetic Rats Treated With/Without DL--lipoic Acid (Mean SEM)

Diabetic DL --lipoic
Control Diabetic acid

SOD 719 52 (11) 974 93* (10) 663 49*,## (10)


Catalase 3775 323 (8) 2906 488 (7) 2071 197* (8)
GSH peroxidase 558 30 (10) 754 29** (10) 724 32** (8)
GSSG reductase 110 10 (10) 135 6* (10) 113 5# (10)
GSH tranferase 181 10 (10) 228 15** (9) 156 6## (7)
NADH oxidase 4.11 0.34 (11) 6.73 0.98** (7) 2.41 0.24## (9)

The number of observations is indicated in parentheses.


* ,** Significantly different compared with controls (p .05 and .01, respectively).
# ,##
Significantly different compared with untreated diabetic group (p .05 and .01, respectively).
192 I. G. OBROSOVA et al.

(ADP-ribosyl)ation. Renal AA depletion in streptozoto- effect of the compound in our study is also manifested by
cin-diabetic rats in our study is consistent with another a 2.8-fold decrease in extramitochondrial NADH oxidase
report [29]. In contrast to the study [29], we found activity and a blunted upregulation of the key antioxida-
DHAA/AA ratio increased in the diabetic renal cortex. tive defense enzymes, i.e., SOD, GSSG reductase, and
The latter is in agreement with compromised GSH con- GSH transferase in DL--lipoic acid-treated diabetic
centration and GSSG/GSH ratios considering that GSH group. The effect on NADH oxidase is probably related
and other cellular thiols, e.g., dihydrolipoic acid, play an to the important role of DL--lipoic acid in redox sig-
important role in vitamin C homeostasis by regenerating naling [76,77]. In biological tissues, DL--lipoic acid is
AA from DHAA and semiascorbyl radicals [30]. AA rapidly reduced to dihydrolipoic acid by intramitochon-
concentrations in the lens have been found reduced by drial NADH-dependent dihydrolipoamide dehydroge-
L-buthionine (S,R)-sulfoximine, an inhibitor of glutathi- nase, thus shifting NAD/NADH ratios towards a more
one biosynthesis [31]. oxidized state in mitochondrial and secondary cytosolic
Diabetes-induced changes in antioxidative defense compartments. An increase in NAD/NADH ratios with
enzyme activities are known to be tissue-specific. All DL--lipoic acid treatment has been described in several
major antioxidative enzymes, i.e., SOD, GSH peroxi- studies including those in the diabetic tissues
dase, GSSG reductase, and GSH transferase are upregu- [69,32,36,78]. Note that DL--lipoic acid has a number
lated in the diabetic lens [54,71], but are downregulated of important metabolic effects, i.e., acts as a co-factor for
in the diabetic retina [33]. SOD and catalase activities are pyruvate and 2-oxoglutarate complexes, increases glu-
decreased in the diabetic peripheral nerve [36,72], cose transport [79,80] and utilization [32,81]. Beneficial
whereas the enzymes of glutathione metabolism remain effects on diabetic complications [32 41] and, in partic-
unaffected [72]. Of interest, in the kidney, SOD, GSH ular, on diabetic nephropathy [8,10,42], are likely to
peroxidase, GSSG reductase, and GSH transferase, but result from the whole complex of antioxidant and meta-
not catalase, appeared upregulated in short-term diabe- bolic effects of the compound rather than from antioxi-
tes, which is consistent with a number of reports dant properties alone. Several studies suggest that anti-
[19,24,25]. In contrast, in several other studies SOD oxidant properties of -lipoic acid can further be
activity was found decreased [4,16] or unchanged increased by the use of R--lipoic acid instead of the
[26,7375]. Whereas the vast majority of studies in dia- racemic form [82 84]. Furthermore, R-enantiomer is
betes models evaluated total SOD [4,16,23,24,26,73 likely to have a better pharmacological profile than DL-
75], the report [19] describes different responses of ex- -lipoic acid [82]. Thus, it is quite possible that R-
tamitochondrial Cu,Zn-SOD and intramitochondrial Mn- enantiomer will be effective on diabetic complications at
SOD. Of interest, only Cu,Zn-SOD activity was found the doses far lower than those required for DL--lipoic
upregulated in the kidney of diabetic rats, consistent with acid. Further preclinical studies are needed to evaluate
the increased Cu,Zn-SOD gene expression in the study R--lipoic acid as a candidate for drug development in
[9]. The results [9,19] are in agreement with our findings diabetic complications, and, in particular, diabetic ne-
of a 1.6-increase in extramitochondrial NADH oxidase phropathy.
activity, which is one of the major extramitochondrial In conclusion, enhanced oxidative stress is present in
sources of superoxide anion radicals, and support the the renal cortex at a very early stage of diabetes, and is
primary importance of extramitochondrial, rather than associated with the loss of two major nonenzymatic
intramitochondrial, superoxide production in diabetes- antioxidants, GSH and AA, and upregulation of extram-
induced renal oxidative damage. itochondrial NADH oxidase rather than downregulation
Most of the diabetes-induced changes observed in the of major antioxidative defense enzymes. The potent an-
present study were completely or partially prevented by tioxidant DL--lipoic acid effectively counteracts anti-
the potent antioxidant DL--lipoic acid, which is quite oxidant loss and lipid peroxidation in the diabetic kidney.
consistent with the effects of this compound in other The findings justify the rationale for evaluation of R--
tissues of diabetic animals [3236,40] as well as in other, lipoic acid as well as more narrow spectrum agents, i.e.,
nondiabetic models of oxidative stress [30,31]. DL-- stimulators of glutathione biosynthesis (N-acetyl-L-cys-
lipoic acid caused a dramatic 5-fold decrease in renal teine) and inhibitors of NADH oxidase (apocynin), on
cortex F2-isoprostane concentration, partially prevented
diabetic nephropathy.
accumulation of MDA plus 4-HA, arrested diabetes-
induced GSH and AA depletion and increase in GSSG/
GSH and DHAA/AA ratios. Similar effects of DL-- REFERENCES
lipoic acid on renal cortex MDA and GSH
[1] Klein, R. Hyperglycemia and microvascular and macrovascular
concentrations have been previously reported in the disease in diabetes. Diabetes Care 18:258 268; 1995.
chronic STZ-diabetic rat model [8,11]. The antioxidant [2] The effect of intensive treatment of diabetes on the development
Renal oxidative stress in diabetes 193

and progression of long-term complications in insulin-dependent Oxidative stress in experimental diabetes induced by streptozo-
diabetes mellitus. The Diabetes Control and Complications Trial tocin. Acta Physiol. Hung. 85:29 38; 199798.
Research Group. N. Engl. J. Med. 329:977986; 1993. [21] Aragno, M.; Tamagno, E.; Gatto, V.; Brignardello, E.; Parola, S.;
[3] Horie, K.; Miyata, T.; Maeda, K.; Miyata, S.; Sugiyama, S.; Danni, O.; Boccuzzi, G. Dehydroepiandrosterone protects tissues
Sakai, H.; Strihou, C. Y.; Monnier, V. M.; Witztum, J. L.; of streptozotocin-treated rats against oxidative stress. Free Radic.
Kurokawa, K. Immunohistochemical colocalization of glycoxida- Biol. Med. 26:14671474; 1999.
tion products and lipid peroxidation products in diabetic renal [22] Braunlich, H.; Marx, F.; Stein, G. Glutathione status, lipid per-
glomerular lesions. Implication for glycoxidative stress in the oxidation and kidney function in streptozotocin diabetic rats. Exp.
pathogenesis of diabetic nephropathy. J. Clin. Invest. 100:2995 Toxicol. Pathol. 46:143147; 1994.
3004; 1997. [23] Volkovova, K.; Chorvathova, V.; Jurcovicova, M.; Koszeghyova,
[4] Reddi, A. S.; Bollineni, J. S. Renal cortical expression of mRNAs L.; Bobek, P. Antioxidative state of the myocardium and kidneys
for antioxidant enzymes in normal and diabetic rats. Biochem. in acute diabetic rats. Physiol. Res. 42:251255; 1993.
Biophys. Res. Commun. 235:598 601; 1997. [24] Irizar, A.; Ioannides, C. Extrahepatic expression of P450 proteins
[5] Dobashi, K.; Asayama, K.; Hayashibe, H.; Uchida, N.; Koba- in insulin-dependent diabetes mellitus. Xenobiotica 25:941949;
yashi, M.; Kawaoi, A.; Kato, K. Effect of diabetes mellitus 1995.
induced by streptozotocin on renal superoxide dismutases in the [25] Mak, D. H.; Ip, S. P.; Li, P. C.; Poon, M. K.; Ko, K. M.
rat. A radioimmunoassay and immunohistochemical study. Vir- Alterations in tissue glutathione antioxidant system in streptozo-
chows Archiv. B. Cell Pathol. Incl. Mol. Pathol. 60:6772; 1991. tocin-induced diabetic rats. Mol. Cell. Biochem. 162:153158;
[6] Schnackenberg, C. G.; Wilcox, C. S. The SOD mimetic tempol 1996.
restores vasodilation in afferent arterioles of experimental diabe- [26] Rauscher, F. M.; Sanders, R. A.; Watkins, J. B. 3rd Effects of
tes. Kidney Int. 59:1859 1864; 2001. piperine on antioxidant pathways in tissues from normal and
[7] Cooper, M. E. Interaction of metabolic and haemodynamic factors streptozotocin-induced diabetic rats. J. Biochem. Mol. Toxicol.
in mediating experimental diabetic nephropathy. Diabetologia 14:329 334; 2000.
44:19571972; 2001. [27] Kurusu, A.; Shou, I.; Nakamura, S.; Fukui, M.; Shirato, I.; To-
[8] Melhem, M. F.; Craven, P. A.; Liachenko, J.; DeRubertis, F. R. mino, Y. Effects of the new hydroxy-3-methylglutaryl coenzyme
Alpa-lipoic acid attenuates hyperglycemia and prevents glomer- a reductase inhibitor fluvastatin on anti-oxidant enzyme activities
ular mesangial expansion in diabetes. J. Am. Soc. Nephrol. 13: and renal function in streptozotocin-induced diabetic rats. Clin.
108 116; 2002. Exp. Pharmacol. Physiol. 27:767770; 2000.
[9] Lal, M. A.; Korner, A.; Matsuo, Y.; Zelenin, S.; Cheng, S. X.; [28] Reddi, A. S.; Bollineni, J. S. Selenium-deficient diet induces renal
Jaremko, G.; DiBona, G. F.; Eklof, A. C.; Aperia, A. Combined oxidative stress and injury via TGF-beta1 in normal and diabetic
antioxidant and COMT inhibitor treatment reverses renal abnor- rats. Kidney Int. 59:13421353; 2001.
malities in diabetic rats. Diabetes 49:13811389; 2000. [29] Lindsay, R. M.; Jamieson, N. S.; Walker, S. A.; McGuigan, C. C.;
[10] Melhem, M. F.; Craven, P. A.; DeRubertis, F. R. Effects of Smith, W.; Baird, J. D. Tissue ascorbic acid and polyol pathway
dietary supplementation of alpha-lipoic acid on early glomerular metabolism in experimental diabetes. Diabetologia 41:516 523;
injury in diabetes mellitus. J. Am. Soc. Nephrol. 12:124 133; 1998.
2001. [30] Packer, L.; Witt, E. H.; Tritschler, H. J. -Lipoic acid as a
[11] Trachtman, H.; Futterweit, S.; Maesaka, J.; Ma, C.; Valderrama, biological antioxidant. Free Radic. Biol. Med. 19:227250; 1995.
E.; Fuchs, A.; Tarectecan, A. A.; Rao, P. S.; Sturman, J. A.; Boles, [31] Maitra, I.; Serbinova, E.; Trischler, H.; Packer, L. -Lipoic acid
T. H. Taurine ameliorates chronic streptozocin-induced diabetic prevents buthionine-sulfoximine-induced cataract formation in
nephropathy in rats. Am. J. Physiol. 269:F429 F438; 1995. newborn rats. Free Radic. Biol. Med. 18:823 829; 1995.
[12] Soulis-Liparota, T.; Cooper, M. E.; Dunlop, M.; Jerums, G. The [32] Obrosova, I.; Cao, X.; Greene, D. A.; Stevens, M. J. Diabetes-
relative roles of advanced glycation, oxidation and aldose reduc- induced changes in lens antioxidant status, glucose utilization and
tase inhibition in the development of experimental diabetic ne- energy metabolism: effect of DL-alpha-lipoic acid. Diabetologia
phropathy in the Sprague-Dawley rat. Diabetologia 38:387394; 41:14421450; 1998.
1995. [33] Obrosova, I. G.; Fathallah, L.; Greene, D. A. Early changes in
[13] Agardh, C. D.; Senram, U.; Torffvit, O.; Agardh, E. Effects of lipid peroxidation and antioxidative defense in diabetic rat retina:
inhibition of glycation and oxidative stress on the development of effect of DL-alpha-lipoic acid. Eur. J. Pharmacol. 398:139 146;
diabetic nephropathy in rats. J. Diabetes Complications In press. 2000.
[14] Rungby, J.; Flyvbjerg, A.; Andersen, H. B.; Nyborg, K. Lipid [34] Obrosova, I. G.; Minchenko, A. G.; Marinescu, V.; Fathallah, L.;
peroxidation in early experimental diabetes in rats: effects of Kennedy, A.; Stockert, C. M.; Frank, R. N.; Stevens, M. J.
diabetes and insulin. Acta Endocrinol. (Copenh.) 126:378 380; Antioxidants attenuate early up regulation of retinal vascular
1992. endothelial growth factor in streptozotocin-diabetic rats. Diabe-
[15] Lubec, B.; Hermon, M.; Hoeger, H.; Lubec, G. Aromatic hy- tologia 44:11021110; 2001.
droxylation in animal models of diabetes mellitus. FASEB J. [35] Nagamatsu, M.; Nickander, K. K.; Schmelzer, J. D.; Raya, A.;
12:15811587; 1998. Wittrock, D. A.; Tritschler, H.; Low, P. A. Lipoic acid improves
[16] Kedziora-Kornatowska, K. Effect of angiotensin convertase in- nerve blood flow, reduces oxidative stress, and improves distal
hibitors and AT1 angiotensin receptor antagonists on the devel- nerve conduction in experimental diabetic neuropathy. Diabetes
opment of oxidative stress in the kidney of diabetic rats. Clin. Care 18:1160 1167; 1995.
Chim. Acta 287:19 27; 1999. [36] Stevens, M. J.; Obrosova, I.; Cao, X.; Van Huysen, C.; Greene,
[17] Nishikawa, T.; Edelstein, D.; Brownlee, M. The missing link: a D. A. Effects of DL-alpha-lipoic acid on peripheral nerve con-
single unifying mechanism for diabetic complications. Kidney Int. duction, blood flow, energy metabolism, and oxidative stress in
58(Suppl. 77):26 30; 2000. experimental diabetic neuropathy. Diabetes 49:1006 1015; 2000.
[18] Mano, T.; Shinohara, R.; Nagasaka, A.; Nakagawa, H.; Uchimura, [37] Keegan, A.; Cotter, M. A.; Cameron, N. E. Effects of diabetes and
K.; Hayashi, R.; Nakano, I.; Tsugawa, T.; Watanabe, F.; Koba- treatment with the antioxidant alpha-lipoic acid on endothelial
yashi, T.; Fujiwara, K.; Nakai, A.; Itoh, M. Scavenging effect of and neurogenic responses of corpus cavernosum in rats. Diabe-
nicorandil on free radicals and lipid peroxide in streptozotocin- tologia 42:343350; 1999.
induced diabetic rats. Metabolism 49:427 431; 2000. [38] Garrett, N. E.; Malcangio, M.; Dewhurst, M.; Tomlinson, D. R.
[19] Kakkar, R.; Mantha, S. V.; Radhi, J.; Prasad, K.; Kalra, J. Anti- alpha-Lipoic acid corrects neuropeptide deficits in diabetic rats
oxidant defense system in diabetic kidney: a time course study. via induction of trophic support. Neurosci. Lett. 222:191194;
Life Sci. 60:667 679; 1997. 1997.
[20] Matkovics, B.; Kotorman, M.; Varga, I. S.; Hai, D. Q.; Varga, C. [39] Cameron, N. E.; Cotter, M. A.; Horrobin, D. H.; Tritschler, H. J.
194 I. G. OBROSOVA et al.

Effects of alpha-lipoic acid on neurovascular function in diabetic [57] Bose, R.; Sutherland, G. R.; Pinsky, C. Biological and method-
rats: interaction with essential fatty acids. Diabetologia 41:390 ological implications of prostaglandin involvement in mouse
399; 1998. brain lipid peroxidation measurements. Neurochem. Res. 14:217
[40] Coppey, L. J.; Gellett, J. S.; Davidson, E. P.; Dunlap, J. A.; Lund, 220; 1989.
D. D.; Yorek, M. A. Effect of antioxidant treatment of strepto- [58] Dannhardt, G.; Flemmer, L.; Hartmann, R. W.; Kleber, A.;
zotocin-induced diabetic rats on endoneurial blood flow, motor Schulze, E. Spectrofluorimetric quantification of malondialde-
nerve conduction velocity, and vascular reactivity of epineurial hyde for evaluation of cyclooxygenase-1/thromboxane synthase
arterioles of the sciatic nerve. Diabetes 50:19271937; 2001. inhibition. Arch. Pharm. (Weinheim) 331:359 364; 1998.
[41] Borenshtein, D.; Ofri, R.; Werman, M.; Stark, A.; Tritschler, [59] Miinea, C.; Kuruvilla, R.; Merrikh, H.; Eichberg, J. Altered
H. J.; Moeller, W.; Madar, Z. Cataract development in diabetic arachidonic acid biosynthesis and antioxidant protection mecha-
sand rats treated with alpha-lipoic acid and its gamma-linolenic nisms in Schwann cells grown in elevated glucose. J. Neurochem.
acid conjugate. Diabetes Metab. Res. Rev. 17:44 50; 2001. 81:12531262; 2002.
[42] Morcos, M.; Borcea, V.; Isermann, B.; Gehrke, S.; Ehret, T.; [60] Tajiri, Y.; Umeda, F.; Inoguchi, T.; Nawata, H. Effects of throm-
Henkels, M.; Schiekofer, S.; Hofmann, M.; Amiral, J.; Tritschler, boxane synthetase inhibitor (OKY-046) on urinary prostaglandin
H.; Ziegler, R.; Wahl, P.; Nawroth, P. P. Effect of alpha-lipoic excretion and renal function in streptozotocin-induced diabetic
acid on the progression of endothelial cell damage and albumin- rat. J. Diabetes Complications 8:126 132; 1994.
uria in patients with diabetes mellitus: an exploratory study. [61] Raza, H.; Ahmed, I.; John, A.; Sharma, A. K. Modulation of
Diabetes Res. Clin. Pract. 52:175183; 2001. xenobiotic metabolism and oxidative stress in chronic streptozo-
[43] Gopaul, N. K.; Nourooz-Zadeh, J.; Mallet, A. I.; Anggard, E. E. tocin-induced diabetic rats fed with Momordica charantia fruit
Formation of F2-isoprostanes during aortic endothelial cell-medi- extract. J. Biochem. Mol. Toxicol. 14:131139; 2000.
ated oxidation of low density lipoprotein. FEBS Lett. 348:297 [62] Obrosova, I. G.; Van Huysen, C.; Fathallah, L.; Cao, X.; Stevens,
300; 1994. M. J.; Greene, D. A. Evaluation of 1-adrenoceptor antagonist on
[44] Nourooz-Zadeh, J.; Gopaul, N. K.; Barrow, S.; Mallet, A. I.; nerve function, metabolism and antioxidative defense. FASEB J.
Anggard, E. E. Analysis of F2-isoprostanes as indicators of non- 14:1548 1558; 2000.
enzymatic lipid peroxidation in vivo by gas chromatography-mass [63] Obrosova, I. G.; Fathallah, L.; Lang, H. J. Interaction between
spectrometry: development of a solid-phase extraction procedure. osmotic and oxidative stress in diabetic precataractous lens. Stud-
J. Chromatogr. B Biomed. Appl. 667:199 208; 1995. ies with a sorbitol dehydrogenase inhibitor. Biochem. Pharmacol.
[45] Nourooz-Zadeh, J.; Pereira, P. F(2) isoprostanes, potential spe- 58:19451954; 1999.
cific markers of oxidative damage in human retina. Ophthalmic [64] Kador, P. F. The role for aldose reductase in the development of
Res. 32:133137; 2000. diabetic complications. Med. Res. Rev. 8:325352; 1988.
[46] Nourooz-Zadeh, J.; Pereira, P. Age-related accumulation of free [65] Obrosova, I.; Faller, A.; Burgan, J.; Ostrow, E.; Williamson, J. R.
polyunsaturated fatty acids in human retina. Ophthalmic Res. Glycolytic pathway, redox state of NAD(P)-couples and energy
31:273279; 1999. metabolism in lens in galactose-fed rats: effect of an aldose
[47] Ho, E.; Chen, G.; Bray, T. M. Alpha-phenyl-tert-butylnitrone reductase inhibitor. Curr. Eye Res. 16:34 43; 1997.
(PBN) inhibits NFkappaB activation offering protection against [66] Obrosova, I. G.; Fathallah, L. Evaluation of an aldose reductase
chemically induced diabetes. Free Radic. Biol. Med. 28:604 inhibitor on lens metabolism, ATP-ases and antioxidative de-
614; 2000. fense: an intervention study. Diabetologia 43:1048 1055; 2000.
[48] Chen, H. C.; Guh, J. Y.; Shin, S. J.; Tsai, J. H.; Lai, Y. H. Insulin [67] Obrosova, I. G.; Van Huysen, C.; Fathallah, L.; Cao, X.; Greene,
and heparin suppress superoxide production in diabetic rat glo- D. A.; Stevens, M. J. An aldose reductase inhibitor reverses early
meruli stimulated with low-density lipoprotein. Kidney Int. diabetes-induced changes in peripheral nerve function, metabo-
78(Suppl.):S124 S127; 2001. lism, and antioxidative defense. FASEB J. 16:123125; 2002.
[49] Van Dam, P. S.; Bravenboer, B.; van Asbeck, B. S.; van Oirschot, [68] Lee, A. Y. W.; Chung, S. S. M. Contribution of polyol pathway
J. F.; Marx, J. J.; Gispen, W. H. Effects of insulin treatment on to oxidative stress in diabetic cataract. FASEB J. 13:2330; 1999.
endoneurial and systemic oxidative stress in relation to nerve [69] Ho, E. C. M.; Lam, K. S. L.; Chung, S. S. M. Aldose reductase-
conduction in streptozotocin-diabetic rats. Eur. J. Clin. Invest. deficient mice are protected from reduction of nerve conduction
26:11431149; 1996. associated with diabetes. J. Periph. Nerv. Syst. 5:169 188; 2000.
[50] Kakimoto, M.; Inoguchi, T.; Sonta, T.; Yu, H. Y.; Imamura, M.; [70] Obrosova, I. G.; Li, F.; White, L.; Abatan, O. I.; Szabo, C.;
Etoh, T.; Hashimoto, T.; Nawata, H. Accumulation of 8-hydroxy- Stevens, M. J. Poly(ADP-ribose)synthetase-knockout mice are
2'-deoxyguanosine and mitochondrial DNA deletion in kidney of protected from functional and biochemical deficits associated
diabetic rats. Diabetes 51:1588 1595; 2002. with early diabetes-like neuropathy. Diabetes 51(Suppl. 2):A206;
[51] Hayashi, K.; Haneda, M.; Koya, D.; Maeda, S.; Isshiki, K.; 2002.
Kikkawa, R. Enhancement of glomerular heme oxygenase-1 ex- [71] Khanna, P.; Wang, L.; Perez-Polo, R. J.; Ansari, N. H. Oxidative
pression in diabetic rats. Diabetes Res. Clin. Pract. 52:8596; defense enzyme activity and mRNA levels in lenses of diabetic
2001. rats. J. Toxicol. Environ. Health 51:541555; 1997.
[52] Saito, H. Biochemical changes in lens, aqueous humor and vitre- [72] Obrosova, I. G.; Greene, D. A.; Lang, H. J. Antioxidative defense
ous body and effects of aldose reductase inhibitor (TAT) on rats in diabetic peripheral nerve: effect of DL--lipoic acid, aldose
with experimental diabetes. Nippon Ika Daigaku Zasshi 62:339 reductase inhibitor and sorbitol dehydrogenase inhibitor. In:
350; 1995. Packer, L.; Roesen, P.; Tritschler, H.; King, G.; Azzi, A., eds.
[53] Lowitt, S.; Malone, J. I.; Salem, A. F.; Korthals, J.; Benford, S. Antioxidants in diabetes management. New York: Marcel Dekker;
Acetyl-L-carnitine corrects the altered peripheral nerve function 2000:93110.
of experimental diabetes. Metabolism 44:677 680; 1995. [73] Kakkar, R.; Kalra, J.; Mantha, S. V.; Prasad, K. Lipid peroxida-
[54] Lou, M. F.; Dickerson, J. E. Jr.; Garadi, R.; York, B. M. Jr. tion and activity of antioxidant enzymes in diabetic rats. Mol.
Glutathione depletion in the lens of galactosemic and diabetic Cell. Biochem. 151:113119; 1995.
rats. Exp. Eye Res. 46:517530; 1988. [74] Rauscher, F. M.; Sanders, R. A.; Watkins, J. B. 3rd Effects of
[55] Cameron, N. E.; Cotter, M. A.; Jack, A. M.; Basso, M. D.; isoeugenol on oxidative stress pathways in normal and streptozo-
Hohman, T. C. Protein kinase C effects on nerve function, per- tocin-induced diabetic rats. J. Biochem. Mol. Toxicol. 15:159
fusion, Na(), K()-ATPase activity and glutathione content in 164; 2001.
diabetic rats. Diabetologia 42:1120 1130; 1999. [75] Sanders, R. A.; Rauscher, F. M.; Watkins, J. B. 3rd Effects of
[56] Naveh-Floman, N.; Weissman, C.; Belkin, M. Arachidonic acid quercetin on antioxidant defense in streptozotocin-induced dia-
metabolism by retinas of rats with streptozotocin-induced diabe- betic rats. J. Biochem. Mol. Toxicol. 15:143149; 2001.
tes. Curr. Eye Res. 3:11351139; 1984. [76] Roy, S.; Sen, C. K.; Tritschler, H. J.; Packer, L. Modulation of
Renal oxidative stress in diabetes 195

cellular reducing equivalent homeostasis by alpha-lipoic acid. [81] Jacob, S.; Streeper, R. S.; Fogt, D. L.; Hokama, J. Y.; Tritschler,
Mechanisms and implications for diabetes and ischemic injury. H. J.; Dietze, G. J.; Henriksen, E. J. The antioxidant alpha-lipoic
Biochem. Pharmacol. 53:393399; 1997. acid enhances insulin-stimulated glucose metabolism in insulin-
[77] Sen, C. K. Redox signaling and the emerging therapeutic potential resistant rat skeletal muscle. Diabetes 45:1024 1029; 1996.
of thiol antioxidants. Biochem. Pharmacol. 55:17471758; 1998. [82] Maitra, I.; Serbinova, E.; Tritschler, H. J.; Packer, L. Stereospe-
[78] Obrosova, I. G.; Stevens, M. J.; Lang, H. J. Diabetes-induced cific effects of R-lipoic acid on buthionine sulfoximine-induced
changes in retinal NAD-redox status: pharmacological modula- cataract formation in newborn rats. Biochem. Biophys. Res. Com-
tion and implications for pathogenesis of diabetic retinopathy. mun. 221:422 429; 1996.
Pharmacology 62:172180; 2001. [83] Kilic, F.; Handelman, G. J.; Traber, K.; Tsang, K.; Packer, L.;
[79] Konrad, D.; Somwar, R.; Sweeney, G.; Yaworsky, K.; Hayashi, Trevithick, J. R. Modelling cortical cataractogenesis XX. In vitro
M.; Ramlal, T.; Klip, A. The antihyperglycemic drug alpha-lipoic effect of alpha-lipoic acid on glutathione concentrations in lens in
acid stimulates glucose uptake via both GLUT4 translocation and model diabetic cataractogenesis. Biochem. Mol. Biol. Int. 46:585
GLUT4 activation: potential role of p38 mitogen-activated pro- 595; 1998.
tein kinase in GLUT4 activation. Diabetes 50:1464 1471; 2001. [84] Lockhart, B.; Jones, C.; Cuisinier, C.; Villain, N.; Peyroulan, D.;
[80] Moini, H.; Tirosh, O.; Park, Y. C.; Cho, K. J.; Packer, L. R-alpha- Lestage, P. Inhibition of L-homocysteic acid and buthionine sul-
lipoic acid action on cell redox status, the insulin receptor, and phoximine-mediated neurotoxicity in rat embryonic neuronal cul-
glucose uptake in 3T3-L1 adipocytes. Arch. Biochem. Biophys. tures with alpha-lipoic acid enantiomers. Brain Res. 855:292
397:384 391; 2002. 297; 2000.

Vous aimerez peut-être aussi