Vous êtes sur la page 1sur 24

Free Radical Biology & Medicine, Vol. 19, No. 2, pp.

227-250, 1995
Copyright 1995 Elsevier Science Ltd
Pergamon Printed in the USA. All rights reserved
0891-5849/95 $29.00 + .00

0891-5849(95)00017-8

Review Article
ALPHA-LIPOIC ACID AS A BIOLOGICAL ANTIOXIDANT

LESTER PACKER,* ERIC H. W l T r , * and HANS JORGEN TRITSCHLERt


*Membrane Bioenergetics Group and Department of Molecular & Cell Biology, University of California, Berkeley, CA,
USA; and *ASTA Medica, Frankfurt Am Main, Germany

(Received 24 October 1994; Revised 16 December 1994; Accepted 20 December 1994)

Abstract---a-Lipoic acid, which plays an essential role in mitochondrial dehydrogenase reactions, has recently gained consider-
able attention as an antioxidant. Lipoate, or its reduced form, dihydrolipoate, reacts with reactive oxygen species such as
superoxide radicals, hydroxyl radicals, hypochlorous acid, peroxyl radicals, and singlet oxygen. It also protects membranes by
interacting with vitamin C and glutathione, which may in turn recycle vitamin E. In addition to its antioxidant activities,
dihydrolipoate may exert prooxidant actions through reduction of iron. a-Lipoic acid administration has been shown to be
beneficial in a number of oxidative stress models such as ischemia-reperfusion injury, diabetes (both ot-lipoic acid and dihydroli-
poic acid exhibit hydrophobic binding to proteins such as albumin, which can prevent glycation reactions), cataract formation,
HIV activation, neurodegeneration, and radiation injury. Furthermore, lipoate can function as a redox regulator of proteins such
as myoglobin, prolactin, thioredoxin and NF-KB transcription factor. We review the properties of lipoate in terms of (l) reactions
with reactive oxygen species; (2) interactions with other antioxidants; (3) beneficial effects in oxidative stress models or clinical
conditions.

Keywords---Antioxidant, Dihydrolipoate, Dihydrolipoic acid, a-Lipoate, ot-Lipoic acid, Oxidative stress, Redox regulation,
Review, Thioctic acid, Free radicals

INTRODUCTION c o w o r k e r s as an acetate r e p l a c i n g f a c t o r 1'2 and is insol-


u b l e in water, b u t s o l u b l e in o r g a n i c solvents. It is
T h e m e t a b o l i c r o l e o f a - l i p o i c a c i d (Fig. 1) has b e e n
k n o w n b y a v a r i e t y o f n a m e s , i n c l u d i n g t h i o c t i c acid; 1,
k n o w n f o r d e c a d e s . It w a s first i s o l a t e d b y R e e d and
2 - d i t h i o l a n e - 3 p e n t a n o i c acid; 1, 2 - d i t h i o l a n e - 3 valeric
acid; and 6 , 8 - t h i o c t i c acid. A s l i p o a m i d e , it f u n c t i o n s
as a c o f a c t o r in the m u l t i e n z y m e c o m p l e x e s that cata-
Dr. Lester Packer is a professor in the Department of Molecular
and Cell Biology at the University of California at Berkeley. He l y z e the o x i d a t i v e d e c a r b o x y l a t i o n o f a - k e t o acids such
received a Ph.D. in Microbiology and Biochemistry from Yale Uni- as p y r n v a t e , ct-ketoglutarate, and b r a n c h e d c h a i n a -
versity in 1956. At Berkeley he has been Professor of Physiology k e t o a c i d s ) c~-Lipoic a c i d w a s t e n t a t i v e l y classified as
since 1961, Director of the Membrane Bioenergetics Group at the
University of California at Berkeley, and a Senior Scientist at the a v i t a m i n after its isolation, 1'4 but it w a s later f o u n d to
Lawrence Berkeley Laboratory since 1972. His research, in the area b e s y n t h e s i z e d b y a n i m a l s and humansS; h o w e v e r , the
of biological oxidation and bioenergetics has emphasized studies on c o m p l e t e e n z y m e p a t h w a y that is r e s p o n s i b l e for the
the role of oxidants and antioxidants in biological systems.
He is one of the world's leading researchers on vitamin E and d e n o v o s y n t h e s i s has n o t yet b e e n e l u c i d a t e d . S e v e r a l
biological antioxidants. His recent work has elucidated new areas studies i n d i c a t e that o c t a n o a t e s e r v e s as the i m m e d i a t e
in the biochemistry of vitamin E--the vitamin E cycle. This is p r e c u r s o r f o r the 8 - c a r b o n fatty a c i d chain, and cys-
relevent to the understanding of new enzymic reactions of vitamin t e i n e a p p e a r s to be the s o u r c e o f sulfur. 6
E, the vitamin E radical, and the biological consequences of the
action of vitamin E. Recent work concerns the prevention of oxida- M o r e r e c e n t l y , a great d e a l o f attention has b e e n
tively induced injury in biological systems by antioxidants and how g i v e n to p o s s i b l e a n t i o x i d a n t f u n c t i o n s f o r a - l i p o i c
antioxidants such as vitamin E and lipoic acid affect gene expression a c i d a n d its r e d u c e d f o r m , d i h y d r o l i p o i c a c i d ( D H L A ,
and cell regulation. These investigations are helping to develop a
new broader understanding of biological antioxidant defense mecha- Fig. 1). In this r e v i e w w e are c o n c e r n e d w i t h the anti-
nisms. o x i d a n t p r o p e r t i e s o f t h e s e c o m p o u n d s and the p r e v e n -
Dr. Packer has edited more than 50 books, authored over 500 t i v e and t h e r a p e u t i c i m p l i c a t i o n s and applications that
articles, is a member of 8 professional societies and 3 editorial boards
of scientific journals, and has organized numerous conferences in arise f r o m this n e w v i e w o f an o l d cofactor.
the area of his research interests. Currently he is President of The
Oxygen Club of California, President Elect of International Society Antioxidant criteria
for Free Radical Research, and Vice President of UNESCO's Global M a n y criteria m u s t b e c o n s i d e r e d w h e n e v a l u a t i n g
Network on Molecular and Cell Biology.
Address correspondence to: Lester Packer, 251 Life Sciences Ad- the a n t i o x i d a n t p o t e n t i a l o f a c o m p o u n d . S o m e o f t h e s e
dition, University of California, Berkeley, CA 94720, USA. c o n c e r n c h e m i c a l a n d b i o c h e m i c a l aspects:
227
228 L. PACKER et al.

~-Lipoic acid Dihydrolipoic acid

s s SH SH

/ \C .2 I
C
I
C C
.2
c
C C C
/ COOH COOH
H2 H2 H2
1-12 H2 H2

Fig. 1. The structures of ot-lipoic acid and dihydrolipoic acid, shown as chemical structures (top), ball-and-stick models (middle),
and space-filling models (bottom). The naturally occurring R-enantiomers are shown. Synthetic a-lipoic acid is a racemic mixture
of the R- and the S-enantiomer.

Specificity of free radical quenching ies are confirming its role in the prevention of numer-
Metal chelating activity ous oxidant-related diseases, such as heart disease. 7"s
Interaction with other antioxidants An " i d e a l " antioxidant would fulfill all of the
Effects on gene expression above criteria. The c~-lipoic acid/dihydrolipoic acid re-
dox couple approaches the ideal; it has been called
Other criteria are important when considering pre- " a universal antioxidant. ''9 a-Lipoic acid is readily
ventive or therapeutic applications: absorbed from the diet. It is probably rapidly converted
Absorption and bioavailability to D H L A in many tissues, as recent advances in assay
Concentration in tissues, cells, and extracellular technique have made evident) 'H One or both of the
fluid components of the redox couple effectively quench a
Location (in aqueous or membrane domains or in number of free radicals in both lipid and aqueous do-
mains. Both D H L A 21'32'33 and a-lipoic acid 13'19"23'24
both?)
have metal-chelating activity. D H L A acts synergisti-
A substance need not excel in meeting all these cally with other antioxidants, indicating that it is capa-
criteria to be considered a good antioxidant. For exam- ble of regenerating other antioxidants from their radical
ple, vitamin E acts only in the membrane or lipid do- or inactive forms. Finally, there is evidence that they
mains, its dominant action is to quench lipid peroxyl may have effects on regulatory proteins and on genes
radicals, and it has little or no activity against radicals involved in normal growth and metabolism.
in the aqueous phase, yet it is considered one of the Because of these antioxidant attributes, a number
central antioxidants of the body. Epidemiological stud- of experimental and clinical studies have been carried
Antioxidant properties of lipoic acid 229

out which show a-lipoic acid to be useful or potentiaily may be due to the somewhat strained conformation of
useful as a therapeutic agent in such conditions as the 5-membered ring in the intramolecular disulfide
diabetes, ischemia-reperfusion injury, heavy-metal form of a-lipoic acid; there is no such strain on the
poisoning, radiation damage, neurodegeneration, and intermolecular disulfide of glutathione disulfide, per-
HIV infection. haps explaining its lack of reactivity in this system.
c~-Lipoic acid has been reported to scavenge singlet
oxygen in at least four different systems. Two early
ANTIOXIDANT ACTIONS
studies showed that a-lipoic acid reacted with singlet
An antioxidant function for tz-lipoic acid was sug- oxygen generated by rubrene autooxidation 15 or by
gested as early as 1959 by Rosenberg and Culik, 4 who photosensitized oxidation of methylene16; these
observed that administration of ot-lipoic acid prevented experiments were carried out in organic solvents. Later
scurvy symptoms in vitamin C-deficient guinea pigs studies conducted under more physiological conditions
as well as preventing symptoms of vitamin E defi- also indicate that cz-lipoic acid is an effective scaven-
ciency in rats fed a diet lacking ot-tocopherol. It has ger of singlet oxygen. Kaiser et al. 17 generated singlet
only been recently, however, that the specific effects oxygen by thermolysis of endoperoxide and detected
of ol-lipoic acid and DHLA in free radical quenching, it by chemiluminescence; in this system tz-lipoic acid
metal chelation, antioxidant recycling, and gene ex- reacted with singlet oxygen with a rate constant of
pression have been investigated. 1.38 108 M-Is -1. In experiments in which singlet
oxygen was generated by thermolysis of endoperoxide
Reactive oxygen species quenching and metal and detected by single strand DNA breaks, c~-lipoic
chelation acid was confirmed to be a scavenger of singlet
oxygen. ~8.19
Lipoic acid. There is general agreement about the anti- Early chemical studies also indicated that ct-lipoic
oxidant properties of a-lipoic acid. It scavenges hy- acid reacts with hydrogen peroxide. 2 However, in
droxyl radicals, hypochlorous acid, and singlet oxygen. these studies high concentrations of H202 (30%) and
It does not appear to scavenge hydrogen peroxide or nonphysiological conditions (e.g., one day reaction in
superoxide radical and probably does not scavenge per- acetone) were used. When tz-lipoic acid was tested
oxyl radicals (Table 1). It may chelate transition against H/O2 in an aqueous environment, ~3no reaction
metals. was found, using a peroxidase-based assay system for
Two studies indicate that tr-lipoic acid is a potent H202, for concentrations of a-lipoic acid up to 6 mM.
hydroxyl radical scavenger. In one, 12 hydroxyl radical
Two separate studies have failed to show that a-
was generated by 2 mM HzO2 + 0.2 mM FeSO4. The
lipoic acid can scavenge superoxide radical. Both used
radical was detected by electron spin resonance (ESR)
xanthine or hypoxanthine and xanthine oxidase to gen-
using the spin-trapping agent 5,5-Dimethylpyrroline-
erate superoxide radical. In one, in which superoxide
N-oxide (DMPO). 1 mM ot-lipoic acid completely
was detected by ESR using the DMPO spin trap, 12 no
eliminated the DMPO-OH adduct signal. Another
reaction was seen (Fig. 2). Similarly, Scott et al., 13
study, 13 using a similar hydroxyl radical-generating
using cytochrome c reduction to detect superoxide,
system (2.8 mM HzO2, 0.05 mM FeC13, 0.1 mM EDTA,
also found no effect of c~-lipoic acid.
and 0.1 mM ascorbate) but a different assay for the
radical (deoxyribose degradation) also found ct-lipoic The situation regarding the ability of ot-lipoic acid
acid to be a hydroxyl radical scavenger. In this study, to scavenge peroxyl radicals is not so clear-cut. One
a rate constant of 4.7 101 M-is -~ was calculated; group 9 reported that o~-lipoic acid does not react with
this is an essentially diffusion-limited reaction rate. peroxyl radicals in either aqueous or lipid environ-
Hence, ot-lipoic acid appears to be a highly effective ments. Peroxyl radicals were generated using thermo-
scavenger of hydroxyl radical. labile azo initiators, either 2,2' azobis (2-amidinopro-
There is similar agreement about the ability of ot- pane)-dihydrochloride (AAPH) to generate peroxyl
lipoic acid to scavenge hypochlorous acid. Haenen and radicals in the aqueous phase or 2,2'-azobis (2,4 di-
Bast 14and Scott et al.13 both found that 50 #M t~-lipoic methyvaleronitrile) (AMVN) to generate peroxyl radi-
acid almost completely abolished the inactivation of cals in lipids. Phycoerythrin fluorescence decay was
a 1-antiproteinase by 50/zM HOC1. This is in contrast used to detect peroxyl radicals in the aqueous environ-
to glutathione, whose reduced form is a potent scaven- ment, and in lipids (liposomes or rat liver microsomes)
ger of HOCI, comparable to ct-lipoic acid, but whose they were detected by TBARS or conjugated diene
oxidized form is almost completely ineffective. ~4 The assays. In no case was ct-lipoic acid effective in scav-
authors of this study speculate that the greater reactiv- enging peroxyl radicals. In contrast, another group, ~3
ity of c~-lipoic acid compared to oxidized glutathione using only an aqueous system and generating peroxyl
230 L. PACKER et al.

Table 1. Antioxidant Effects of a-Lipoic Acid

Scavenged by
Oxidant a-Lipoate? Test System Reference

Superoxide radical No 0 2 generated by xanthine-xanthineoxidase and detected by ESR using spin traps 12
No O~- generated by hypoxanthine-xanthiueoxidase and detected by O7 dependent 13
cytochrome c reduction
Hydrogen peroxide No H202 added directly and detected by peroxidase-based assay system 13
Hydroxyl radical Yes OH" generated by H202 + FeSO4 and detected by ESR using spin traps or by 12
chemiluminescenceusing luminol
Yes OH" generated by H202 + FeCI3 + ascorbate and detected by deoxyribose 13
degradation. Rate constant for reaction = 4.71 10~M-j s-~
Hypochlorous radical Yes HOCi added directly and detected by effect on al-antiproteinase activity 14
Yes Same 13
Peroxyl radical No Peroxyl radicals generated in aqueous phase by thermal decomposition of 9
2,2'azobis(2-amidinopropane)-dihydrochlorideand detected by fluorescence
quenching of phycoerythrin. Peroxyl radicals generated in lipids (iiposomes or
microsomes) by thermal decomposition of 2,2'-azobis (2,4
dimethylvaleronitrile)
Yes CC130"= generated by linear accelerator and detected spectrophotometrically. Rate 13
constant of reaction 1.8 l0 s M t s
Singlet oxygen Yes ~O2generated by rubrene autoperoxidationin air-saturated benzene when 15
stimulated at 546 nm and detected by following disappearance of rubrene
spectrophotometrically. Rate constant of the reaction = 1 108 M-j s ~.
DHLA not tested
Yes ~O2generated by photosensitized oxidation of methylene in chloroform or 16
methanol; reaction detected by analyzing oxidation products of the methyl ester
of LA. DHLA not tested
Yes ~O2generated by thermolysis of endoperoxide and detected by infrared 17
chemiluminescence.Rate constant of the reaction = 1.38 l0 s M-j s-j
Yes ~O2generated by thermolysis of endoperoxide and detected by single strand DNA 18, 19
breaks
Transition metals Chelator o-Lipoic acid formed stable complexes with Cu2+, Mn2+, and Zn2+ in aqueous 23
solution. Bisnorlipoate and tetranoflipoate formed more stable complexes
Chelator Same as system to test J02. When EDTA was added, the protective effect of LA 19
decreased, suggesting that it was at least partially due to metal chelation
Possible chelator LA decreased Cd2 toxicity in hepatocytes, but much less effectively than DHLA. 21
The authors speculate that LA is taken up and converted to DHLA, which is
the compound exerting the protective effect
No chelation In rat liver microsomes + FeSO4 + ascorbate, LA had no effect on accumulation 22
of TBARS
Chelator LA decreased site-specific iron-induced degradation of deoxyribose, suggesting 13
that it chelated the iron
Chelator Decreased Cu2+-inducedoxidation of ascorbate, increased partitioning of Cu2 24
into octanol, inhibited Cu2+-inducedlipid peroxidation

radical (CC130~) by radiolysis of CC14 and propan-2- was added to the system, indicating that part of the
01, found that a - l i p o i c acid reacted with this radical effect was due to iron chelation. Scott et al. ~3 found
with a rate constant of 1.8 10 s M - ~ s - l ; the reaction that a - l i p o i c acid inhibited the site-specific degradation
was followed spectrophotometrically. Although the ex- of deoxyribose by FeCl4I-I202/ascorbate, indicating
planation for the disagreement m a y lie in the different that it was able remove iron ion b o u n d to deoxyribose.
methodologies of the two groups, further work is nec- In contrast, in a rat liver microsomal system in which
essary to clarify this question. lipid peroxidation was induced by FeSO4, 22 a - l i p o i c
a - L i p o i c acid may also exert an antioxidant effect acid did not inhibit peroxidation. Hence, in this area
in biological systems through transition metal chela- also, further work is needed.
tion. It has been found to reduce Cd2+-induced toxicity a - L i p o a t e was found to form stable complexes with
in isolated hepatocytes, although the authors speculate M n 2+, Cu e+, Z n 2, with the complex b e i n g almost en-
that the effect was due to the conversion of a-lipoic tirely with the carboxylate group. 23 These investigators
acid to DHLA, which was the true chelating agent. 21 also e x a m i n e d the complexes formed with bisnorli-
Two studies indicate that a - l i p o i c acid m a y chelate poate and tetralipoate, two and four carbon shorter
iron, while one indicates that it does not. Devasagayam homologues, respectively, of a - l i p o i c acid. These were
et al., ~9 who found a - l i p o i c acid to be effective against found to be more stable than those with a - l i p o i c acid,
singlet oxygen-induced D N A single strand breaks, p r e s u m a b l y because the shorter chain allows the dithio-
found that its effectiveness was lower when E D T A lane ring to also participate in chelation. The effects
Antioxidant properties of lipoic acid 231

X/XO/SOD

X/XO/TA

X/XO/DHLA

Mn 0 ~'~ Mn 0
Fig. 2. ESR study of the effects of a-lipoic acid and dihydrolipoic acid on superoxide anion radicals generated by xanthine plus
xanthine oxidase. DMPO spin adducts generated by 5 mM xanthine (X) plus 100 #g xanthine oxidase (XO) in the presence of
40 #M DETAPAC in 150 mM KH2POa-KOH (pH 7.4) in total volume of 1 ml [DHPO] = 45 mM; [SOD] = 200 U; [a-lipoic
acid (TA) = 5 raM; [dihydrolipoic acid (DHLA) = 5 mM. ESR signals were recorded 3 min after the initiation of xanthine
oxidase reaction.

of ot-lipoic acid on copper have recently been extended with hydrogen peroxide or with singlet oxygen. How-
to oxidation situations, a-Lipoic acid was effective in ever, there is disagreement among studies as to whether
preventing Cu2+-catalyzed ascorbic acid oxidation, in- DHLA also is capable of scavenging superoxide radi-
creased the partitioning of Cu 2 into n-octanol from cal and whether it acts as an antioxidant or a prooxidant
aqueous solution, and inhibited Cu2-catalyzed liposo- in its interactions with iron ion.
mal peroxidation. 24 These observations indicate that a- In the systems in which a-lipoic acid was tested for
lipoic acid is a copper chelator. reaction with hypochlorous ion, DHLA was also found
In summary, a-lipoic acid scavenges hydroxyl radi- to be effective, with about the same scavenging
cals, HOCI, and singlet oxygen, but is ineffective ability. 14
against hydrogen peroxide and superoxide radical. It DHLA is an effective peroxyl radical scavenger in
chelates iron, copper, and other transition metals. Fur- several different systems. Kagan et al. 9 used AAPH or
ther work is required to determine whether a-lipoic AMVN to generate peroxyl radicals in aqueous or lipid
acid is effective against peroxyl radicals. systems, as described for a-lipoic acid, but, unlike ot-
Dihydrolipoic acid.Dihydrolipoic acid. With a redox lipoic acid, DHLA was found to scavenge peroxyl radi-
potential of - 0 . 3 2 V (ref. 25) for the DHLA/a-lipoic cals in these systems. It was also found to scavenge
acid redox couple, dihydrolipoic acid is a potent reduc- peroxyl radicals generated by AMVN in liposomes and
tant; for comparison, the redox potential of the GSH/ detected by fluorescence decay of parinaric acid, 28 and
GSSG couple is - 0 . 2 4 V (ref. 26)-DHLA will reduce CC1302 radicals generated in aqueous solution as de-
GSSG to GSH, but GSH is incapable of reducing ct- scribed for a-lipoic acid13; in the latter system a rate
lipoic acid to DHLA. 27 constant of 2.7 10 7 M-is -1 was calculated.
Like a-lipoic acid, DHLA is a potent antioxidant, DHLA has been shown to have both antioxidant ~2
although there is more uncertainty as to its effects and prooxidant t3 effects in systems in which hydroxyl
(Table 2). There is agreement that DHLA scavenges radical was generated; however, the prooxidant effects,
hypochlorous acid and peroxyl radicals and probably if real, are probably due to DHLA's effects on iron
scavenges hydroxyl radicals. It does not appear to react (see below). Both groups used iron salts and hydrogen
232 L. PACKER e t al.

Table 2. Antioxidant Effects of Dihydrolipoic Acid

Oxidant Scavenged by DHLA? Test System Reference

Superoxide radical Yes 03- generated by xanthine-xanthine oxidase and detected by ESR using 12
spin traps. Sulfhydryl content of DHLA decreased during reaction and
[HzO2] increased. Rate constant of reaction = 3.3 l0 s M -~ s -~
Yes O~- generated by xanthine-xanthine oxidase and detected by epinephrine 28
oxidation. Rate constant of reaction = 7.3 105 M -j s -~
No O~ generated by hypoxanthine-xanthine oxidase and detected by 03-- 13
dependent nitro-blue tetrazolium reduction.
Hydrogen peroxide No H202 added directly and detected by iron-thiocyanate. 29
No H202 added directly. Reaction monitored by measuring thiol content of 13
DHLA
Hydroxyl radical Yes OH" generated by H202 + FeSO4 and detected by ESR using spin traps 12
No (promoted oxidation) OH" generated by H202 + FeC13 + ascorbate and detected by 13
deoxyribose degradation. Prooxidant effect postulated due to reduction
of ferrous ion and/or regeneration of ascorbate by DHLA
Hypochlorous radical Yes HOCI added directly and detected by effect on ct 1-antiproteinase activity 14
Yes Same 13
Peroxyl radical Yes Peroxyl radicals generated in aqueous phase by thermal decomposition of 9
2,2 azobis(2-amidinopropane)-dihydrochloride and detected by
fluorescence quenching of phycoerythrin. Peroxyl radicals generated in
lipids (liposomes or microsomes) by thermal decomposition of 2,2'-
azobis (2,4 dimethylvalcronitrile). Stoichiometry: 1.5 mol peroxyl
radicals quenched per mol DHLA
Yes Same as Kagan et al. 1992. In addition, peroxyl radicals generated in 28
liposomes by AMVN and detected by fluorescence decay of parinaric
acid.
Yes CC130~ generated by linear accelerator and detected 13
spectrophotometrically. Rate constant of reaction 2.7 107 M -~ s -j
Singlet oxygen No IO2 generated by thermolysis of endoperoxide and detected by infrared 17
chemiluminescence.
Yes (?) IO2 generated by thermolysis of endoperoxide and detected by single 18, 19
strand DNA breaks. The protective effect of DHLA may not be due to
direct quenching of ~O2
Transition metals Chelation DHLA decreased CD 2 toxicity in isolated hepatocytes. Also decreased 21
TBARS in Cd 2 exposed hepatocytes, indicating that, in the absence of
added iron, DHLA's activity is overall antioxidant against peroxidation
(contrast to Bast and Haenen, and Scott et al.)
Chelation DHLA bound iron from ferritin in the ferrous and ferric states. 32, 33
Prooxidant In FeCI3-EDTA + H202 system, accelerated deoxyribose degration 13
Accelerated reaction also when no EDTA in system, suggesting that its
iron-binding is not as effective as its iron-reduction
However, no prooxidant effect when DHLA was used alone in an Fe
(llI)-bleomycin-DNA system
Prooxidant Rat liver microsomes + FeSO4 _+ ascorbate. DHLA accelerated 22
peroxidation as measured by TBARS
No prooxidant effect No electron transfer from DHLA to Fe 3+ as measured by Fe 2- 12
phenanthroline complex. Also, DHLA did not potentiate formation of
OH" in Fe SO4-H202 system, as measured by ESR (DMPO spin trap)
Some prooxidant effect In lipid peroxidation in microsomes, induced by AMVN, in the absence 28
of iron (chelated by defreroxaamine), DHLA decreased peroxidation
(as measured by TBARS) about 50%. In the presence of iron, DHLA
did not decrease peroxidation (although there was no pro-oxidant
effect; i.e., the antioxidant and pro-oxidant effects of DHLA balanced
in this system). In this system Tetranor-DHLA greatly increased (5X)
peroxidation in the presence of iron
Prooxidant Micromolar DHLA, in the presence of Cu 2 ions, caused single-strand 34
nicks in pSP64 plasmid DNA. Other ions were tested in the same
system and had no effect: Co 2, Cr3+, Fe 3, Fe 2+, Ni 2+, Mn 2, and
Zn 2+. Other thiols also found to cleave DNA in this system in the
presence of Cu 2.

p e r o x i d e to g e n e r a t e h y d r o x y l r a d i c a l s . S u z u k i et al.12 a d d i t i o n o f D H L A e n h a n c e d t h e p r o c e s s . It is p o s s i b l e
d e t e c t e d h y d r o x y l r a d i c a l s b y E S R , u s i n g D M P O to t h a t t h e d i f f e r e n c e i n t h e r e s u l t s is d u e t o t h e f a c t t h a t
produce spin-adducts and found that DHLA abolished S c o t t et al. u s e d a s c o r b a t e t o r e d u c e f e r r i c i r o n w h e r e a s
t h e D M P O - O H s i g n a l ; S c o t t e t al. ~3 d e t e c t e d h y d r o x y l S u z u k i e t al. u s e d a f e r r o u s i r o n s a l t w i t h o u t a s c o r b a t e ;
radicals by deoxyribose degradation and found that D H L A is k n o w n t o r e c y c l e a s c o r b a t e ( s e e b e l o w ) a n d
Antioxidant properties of lipoic acid 233

thus may have increased ascorbate's effectiveness as reduce bound F e 3+ to Fe2+. 3'31 D H L A also appears to
a reductant in this system. Scott et al. speculate that be capable of removing iron from the storage protein
this mechanism of direct reduction of ferric iron is ferritin in both the ferrous and ferric states, 32'33 though
the cause of D H L A ' s prooxidant effect. However, if it does not appear capable of removing iron from
D H L A were causing the effect through direct reduction heme. ~3 If it is indeed capable of reducing iron and if
of ferric iron, then Suzuki et al. should also have seen the iron is available for reaction, then its prooxidant
a prooxidant effect, and they did not. At any rate, it effects may be of serious concern in biological sys-
appears that D H L A scavenges hydroxyl radical, and tems. Indeed, D H L A was found to act as prooxidant
prooxidant effects may be seen in some systems for in peroxidation of rat liver microsomes 22 and in pro-
producing hydroxyl radical. duction of hydroxyl radicals. ~3 In another study in
In systems similar to those used to test a-lipoic acid which peroxidation in microsomes was initiated by
for reaction with hydrogen peroxide, 13"29 no reaction AMVN, 2s D H L A decreased peroxidation by 50% when
was found for DHLA. In addition, Kaiser et al. 17 did desferrioxamine was included in the system but did
not detect scavenging of singlet oxygen by DHLA, not decrease peroxidation when desferrioxamine was
using infrared chemiluminescence to detect singlet ox- omitted, indicating that, in the presence of iron, the
ygen. In contrast, D H L A does protect against single prooxidant effect of D H L A balanced its antioxidant
strand DNA breaks induced by singlet oxygen (unlike effect. In this system a homologue of D H L A that is
GSH, which increased breaks), but the effect was not four carbons shorter, tetranor-DHLA, exerted strong
necessarily due to quenching of singlet oxygen since prooxidant effects, increasing lipid peroxidation in the
there may be several steps in the process at which presence of iron over five-fold. In addition, DHLA, as
D H L A could exert an effect. 18'~9Since the more direct well as a number of other thiols and dithiols, has also
method of detecting singlet oxygen, infrared chemilu- been shown to induce single-strand breaks in plasmid
minescence, did not detect a decrease in its concentra- DNA in the presence of Cu 2, but not in the presence
tion, D H L A probably does not directly scavenge sin- of Fe 2 or Fe3+. 34 This is in contrast to the antioxidant
glet oxygen. Cu2+-chelating effect of ot-lipoic acid. 24
There is major, and as yet unexplained, disagree- On the other hand, in another system in which iron
ment on the effects of D H L A on superoxide radical.
was used to generate hydroxyl radical, the effect of
In a study in which O2"- was generated by xanthine-
D H L A was clearly antioxidant, 12 and in this study no
xanthine oxidase and detected by ESR using DMPO
electron transfer from D H L A to Fe 3+, as measured by
spin trap, ~2 D H L A was found to react with superoxide
formation of Fe/+-phenanthroline complex, was ob-
radical with a rate constant of 3.3 105 M - I s -t (Fig.
served. In Cd 2 toxicity of isolated hepatocytes, addi-
2). Reaction was confirmed by assaying the sufhydryl
tion of D H L A decreased TBARS, 2~ so at least in this
content of DHLA, which decreased, and by detecting
system, also, the antioxidant effect of D H L A against
an increase in H202, a probable product, in the reaction
lipid peroxidation was greater than any prooxidant ef-
medium. These results were confirmed by this group
fect.
in a later study 28 using the same system for generating
superoxide radical and competition with epinephrine Hence, the question of whether D H L A acts as pro-
oxidation to assess D H L A ' s scavenging ability. In this oxidant in biological systems is yet to be resolved.
study a rate constant of 7.3 105 M-~s -~ was found, D H L A probably increases formation of hydroxyl radi-
in good agreement with the previous work. In contrast cal in vitro when ferric ion is included in the system,
to these studies, another group, using a similar super- but the physiological significance of this is unknown.
oxide generating system (hypoxanthine-xanthine oxi- Ascorbate also reduces ferric ion to ferrous ion in vitro;
dase) and detecting superoxide by O2"--dependent ni- indeed, ascorbate + Fe 3+ is a standard initiator of lipid
tro-blue tetrazolium reduction, found no scavenging peroxidation in in vitro systems, but in vivo under
effect of DHLA. It is difficult to explain the discrep- normal conditions, where iron is very tightly bound,
ancy in the results of the two groups, and further work this effect of ascorbate is probably unimportant. The
is clearly indicated. same may be true for DHLA, although the possibility
Perhaps the most crucial question surrounding that D H L A can remove bound iron from ferritin is
D H L A is whether it acts as a prooxidant under certain cause for concern. Furthermore, the interaction of
circumstances. There are at least two ways this might D H L A with other antioxidants may negate a prooxi-
happen. First, D H L A may act as a reductant for transi- dant effect in a physiological system. For example, in
tion metals, especially iron, and second, D H L A may the study of Bast and Haenen z3 D H L A increased lipid
act to regenerate ascorbate, which is known to reduce peroxidation induced by ferrous ion, as did ascorbate.
iron. However, D H L A exerted an antioxidant effect when
D H L A binds both Fe 2+ and Fe 3+, and possibly may included in a system of ascorbate, GSSG, and ferrous
234 L. PACKERet al.

ion, when compared to the same system without through other antioxidants. Therefore, in this system
DHLA. it seems likely that DHLA, which partitions mainly in
These results for t~-lipoic acid and DHLA make the aqueous phase, is directly reducing tocopheroxyl
clear several important areas for future research: (1) radicals at the membrane/water interface. However, the
The antioxidant actions of ot-lipoic acid against peroxyl effect is weak, and the major recycling of vitamin E by
radical; (2) The action of DHLA against superoxide DHLA in biological systems probably occurs through
radical; (3) The effect of DHLA on iron and other other antioxidants.
transition metals. Furthermore, the uptake and reduc- Bast and Haenen 39 have proposed that DHLA pre-
tion of c~-lipoic acid in various tissues must be clarified, vents lipid peroxidation by reducing glutathione, which
since t~-lipoic acid and DHLA have different antioxi- in turn recycles vitamin E. This proposal is based on
dant effects. the observation that the combination of DHLA and
oxidized glutathione, but not DHLA alone, prevented
lipid peroxidation induced by Fe2+/ascorbate. 22 Alter-
Interactions with other antioxidants
natively, Kagan et al. 9 proposed that DHLA protects
DHLA appears to be able to regenerate other antiox- membranes against oxidation by recycling ascorbate,
idants, such as ascorbate and (indirectly) vitamin E, which in turn recycles vitamin E. Their ESR studies
from their radical forms. Vitamin E is the major chain- indicated DHLA-mediated reduction of ascorbyl radi-
breaking antioxidant that protects membranes from cals generated in the course of ascorbate oxidation
lipid peroxidation. 35 Vitamin E exists in biological by chromanoxyl (vitamin E or short-chain vitamin E
membranes in a low molar ratio to unsaturated phos- homologues) radicals in DOPC liposomes and also
pholipids, usually less than 0.1 nmol per mg of mem- showed that DHLA interacts with NADPH- or NADH-
brane protein, or, in other words, one molecule per dependent electron transport chains to recycle vitamin
1000 to 2000 membrane phospholipid molecules, E. Ascorbate-dependent recycling of vitamin E by
which are the main target of oxidation in membranes. DHLA has also been observed in human low density
Lipid peroxyl radicals can be generated in membranes lipoproteins 4 and erythrocyte membranes. 4~ In addi-
at the rate of 1-5 nmol per mg of membrane protein tion, there is evidence that a-lipoic acid administration
per minute, yet destructive oxidation of membrane lip- in vivo can increase the level of ubiquinol in the face
ids does not normally occur, nor is vitamin E rapidly of oxidative stress, 41a and ubiquinol is known to recycle
depleted. Furthermore, deficiency states for vitamin E v i t a m i n E. 41b Hence, current evidence points to the
are remarkably difficult to induce in adult animals. notion that DHLA can recycle vitamin E via glutathi-
These apparent paradoxes can be explained by "vita- one, vitamin C, ubiquinol, NADPH or NADH, but
min E recycling," in which the antioxidant ability of the relative contributions of each avenue are not well
vitamin E is continuously restored by other antioxi- defined.
dants. Those antioxidants that recycle vitamin E are In fact, protection of other antioxidants by a-lipoic
vitamin C, ubiquinols, and thiols (Fig. 3). 36,37 was suggested as early as 1959, by Rosenberg and
Evidence for vitamin E recycling by DHLA has Culik, 4 who stated, with startling prescience, "ot-L-
come from a number of studies. DHLA protects against poic acid, and even more so its dihydro derivative into
microsomal lipid peroxidation, but only in the presence which it is converted rapidly after entering cellular
of vitamin E38; it prolonged the lag phase prior to the metabolism, might act as an antioxidant for ascorbic
onset of low-level chemiluminescence, loss of vitamin acid and tocopherols." In the studies of Rosenberg and
E, and the rapid accumulation of thiobarbituric acid Culik, a-lipoic acid was found to prevent symptoms
reactive substances in normal, but not in vitamin E- of both vitamin E and vitamin C deficiency. Recently,
deficient, microsomes, a-Lipoic acid was not effective we have found similar protective effects of a-lipoic
in either system. DHLA could have exerted its effect acid administration in tocopherol-deficient hairless
in this system by directly reducing tocopheroxyl radi- mice42 (Fig. 4). Such results are consistent with a recy-
cal or by reducing other antioxidants (such as ascor- cling of tocopherol and/or ascorbate by a-lipoic acid,
bate), which then regenerated vitamin E. There may but could also be explained by the ability of ct-lipoic
be a weak direct interaction between DHLA and the acid to spare ascorbate and vitamin E through its sepa-
tocopheroxyl radical; we have found that the presence rate but overlapping radical-scavenging effects.
of DHLA reduces the tocopheroxyl radical ESR signal a-Lipoic acid also causes an increase in intracellular
in liposomes exposed to UV light (unpublished data). glutathione. Busse et al. 43 added a-lipoic acid to mu-
The use of UV light, which directly produces tocopher- rine neuroblastoma and melanoma cell lines, and ob-
oxyl radicals, eliminates the possiblity of iron chelation served a dose-dependent increase in GSH content of
or other antioxidant effects of DHLA, and the use of 30-70% compared to untreated controls. These inves-
liposomes eliminates the possibility of DHLA acting tigators found similar elevations in lung, liver, and
Antioxidant properties of lipoic acid 235

Crad

Semiascorbate Ascorbate
Glutathione disulfide
k ~ Lipoicacid
~ f I ~ NADH,
/ " ~ Lipoamidedehydrogenase
I ~" NADPH,
Glutathione ~ ~' Glutathione
reductase
Dihydrolipoicacid NAD, NADP+
Fig. 3. Vitamin E recycling. The tocopheroxyl radical, formed during oxidation of ~-tocopherol (shown in the membrane), may
be reduced back to a-tocopherol by a number of compounds, including ubiquinol, cytochrome c, and ascorbate. Ascorbate can
be regenerated through reaction with thiols such as glutathione or lipoic acid. These can be returned to their reduced forms by
various mechanisms, drawing on the reducing power of NADH or NADPH. Adapted from Scientific American.

kidney cells of mice injected (IP) daily with doses of redox regulation of protein thiols under normal condi-
4, 8, or 16 mg/kg a-lipoic acid for 11 days. These tions, a-Lipoic acid and DHLA do not appear to be
results have been confirmed in human Jurkat cell lines, present in the unbound state under normal physiologi-
in which intracellular concentrations of GSH increase cal conditions, but after dietary supplementation both
approximately 50% in 5 h after addition of a-lipoic forms appear in various tissues in the unbound form. 4z
acid to the culture medium (D. Han, G. Handelmann, DHLA also has a lower redox potential than GSH, as
personal communication). Such elevations in GSH well as being of lower molecular weight. These factors
cannot be explained by reduction of GSSG, since lead to the possibility that administration of exogenous
GSSG is normally present at less than 10% the concen- c~-lipoic acid may influence intracellular function not
tration of GSH. 4~ These intriguing observations are yet only through antioxidant actions but also through af-
to be explained. fecting the redox status of thiol-containing proteins,
Thus, it appears that o~-lipoic acid and DHLA act such as throredoxin, enzymes, and transport proteins.
as antioxidants not only directly, through radical Dihydrolipoate and dihydrolipoamide can reduce thio-
quenching and metal chelation, but indirectly as well, redoxin, 47"48a small ubiquitous protein that functions to
through recycling of other antioxidants and possibly transfer electrons in various biochemical processes/9
though induction of increased intracellular levels of Spector et al.50 proposed that thioredoxin may be a
glutathione. physiological electron acceptor from lipoamide. Glu-
cose transport has also been shown to be enhanced by
DHLA in a number of systems, 76-78 and it is thought
Redox regulation of proteins and influence on that this stimulation may be due to reduction of sulfhy-
protein folding
dryl groups involved in the regulation of insulin-stimu-
Thiolation of proteins has been reported to be a lated glucose transport. 8r
protective mechanism against oxidative stress, as well DHLA has also been found to influence physiologi-
as affecting the function of some thiol-containing pro- cally important proteins through mechanisms other
teins. 44aGlutathione is the most abundant thiol in mam- than disulfide reduction. DHLA reduces metmyoglobin
malian cells 44 and may be a primary agent involved in and ferrylmyoglobin to oxymyoglobin.45 DHLA also
236 L. PACKERet al.

transcription factors NF-KB, 51 which regulates the ex-


pression of genes such as human immunodeficiency
virus type 1 and those involved in inflammatory re-
sponses. 52 The effects of a-lipoic acid and D H L A on
the expression of c-fos have also been studied.
N F - r B is regulated through redox mechanisms, 53'54
and sulfhydryl groups such as Cys62 in the p50 subunit
have been identified as important in this regulationY
There are two steps in the process of NF-KB activation
8 and action that may be influenced by thiol antioxidant
such as a-lipoic acid. Early steps involve the activation
of NF-KB and dissociation from an inhibitory subunit,
IKB. These are apparently at least partly under redox
control, with oxidation stimulating activation and dis-
sociation. The binding of activated NF-KB to DNA
involves cysteine residues whose redox status is also
important, with reduced cysteine apparently enhancing
binding. Hence, the effects of thiol-containing antioxi-
dants can be complex. For example, overexpression
b of thioredoxin inhibits TPA-induced activation of NF-
KB, 55a whereas in an in vitro system thioredoxin en-
hanced binding of NF-KB to DNA. sS'55bSimilarly, incu-
bation of human Jurkat T-cells in medium containing
4 mM a-lipoic acid completely inhibits NF-KB activa-
tion induced by tumor necrosis factor or phorbol myris-
tate 13-acetate. 56 Recently, the D N A binding activity
of NF-KB was found to be enhanced by D H L A and
inhibited by a-lipoic acid. Inhibition of NF-KB D N A
binding induced by a nonreducing environment or by
exposure to a thiol-oxidizing agent, diamide, was re-
versed by DHLA. 57 The interplay of these compounds
and their effects on NF-KB in a physiological cellular
C environment is difficult to predict.
Fig. 4 Adult 12-week old hairless mice after 6 weeks of (a) normal The effects of both a-lipoic acid and D H L A on the
control diet, (b) vitamin E deficient diet, and (c) vitamin E deficient, expression of the growth-regulating gene c-fos have
c~-Iipoate-supplementeddiet. The animal on the vitamin E deficient also been investigated. 58 Jurkat T cells preincubated
diet shows symptomsof vitamin E deficiency with muscular dystro-
phy and weight loss. with either a-lipoic acid or D H L A were exposed to
TPA, an activator of c-fos expression. Cells incubated
in D H L A exhibited less c-fos m R N A expression, com-
elicits glandular kallikrein-induced prolactin proteoly- pared to controls, whereas those preincubated with a -
sis by acting upon prolactin to refold the molecule into lipoic acid exhibited greater expression compared to
conformations that are glandular kallikrein sub- controls. Active oxygen species are produced by cul-
strates. 46 Hence, D H L A may exert an influence over tured cells stimulated by TPA, 59-6~ and superoxide pro-
intracellular metabolism by means other than antioxi- duction is enhanced in TPA-stimulated leukocytes. 62
dant effects, but the physiological significance of this The suppression of c-fos expression by D H L A but not
remains to be elucidated. by a-lipoic acid may be due to the fact that D H L A
scavenges superoxide, whose production may be in-
volved in enhancing c-fos expression, whereas a-lipoic
Effects on gene expression acid does not.
Apoptosis is another process in which intracellular
There has recently been a great deal of interest in oxidation is thought to play a role. In rat thymocytes
the effects of oxidants and antioxidants on signal trans- exposed to methylprednisolone or etoposide, inducers
duction and gene expression in both normal and abnor- of apoptosis, preincubation with D H L A or lipoamide
mal conditions. In this regard a-lipoic acid and D H L A inhibited apoptosis. Lipoic acid had no effect (S. Or-
have been investigated in terms of their effect on the renius personal communication).
Antioxidant propertiesof lipoic acid 237

Although it appears that DHLA and/or a-lipoic acid of both ~-lipoic acid and DHLA in the cells and in
may influence gene expression at one or more levels, the culture medium were determined by HPLC with
the exact mechanisms and significance have yet to be electrochemical detection at various times up to 2 h.
elucidated. This area holds great promise. The intracellular concentration of DHLA in the Jurkat
cells reached 1.5 mM within 10 min. It was also found
that the cells released DHLA into the medium.
EXPERIMENTAL AND CLINICAL
The results indicate that normal mammalian cells
THERAPEUTIC STUDIES
are capable of taking up a-lipoic acid, reducing it to
a-Lipoic acid administration has been shown to be DHLA, and releasing DHLA. Hence, the effects of
effective in preventing pathology in various experi- both a-lipoic acid and DHLA may be present both
mental models in which reactive oxygen species have intracellularly and extracellularly when a-lipoic acid
been implicated. Before considering these studies, alone is administered extracellularly. This has im-
however, it is necessary to discuss whether a-lipoic portant implications in, for example, the use of a-lipoic
acid is absorbed as a dietary supplement, to what de- acid supplementation to prevent LDL oxidation.
gree it is taken up by tissues, whether it is reduced to Finally, a recent study, which repeated and extended
DHLA, and whether it is metabolized to shorter chain the experiments of Rosenberg and Culik, 4 involving
homologues. vitamin E-deficient rats, confirmed that o~-lipoic acid
is absorbed and is converted to DHLA in tissues. 42 The
hairless mice were used as a model for studying the
Absorption, uptake, intracellular reduction, and
effects of E deficiency with or without a-lipoic acid
metabolism of a-lipoic acid
supplementation, because hairless mice display obvi-
There is no doubt that administration of dietary ot- ous symptoms of deficiency within 5 weeks (Fig. 4).
lipoic acid has an effect on whole-body physiology. Mice were fed either a normal diet, a vitamin E-defi-
The work of Rosenberg and Culik 4 made this clear cient diet, or an E-deficient diet supplemented with
because a-lipoic acid, supplemented in the diet of rats 1.65 g a-lipoic acid/kg diet. a-Lipoic acid supplemen-
and guinea pigs, protected against the symptoms of tation completely prevented symptoms of vitamin E
vitamin E or vitamin C deficiency. In later experi- deficiency. Both a-lipoic acid and DHLA (unbound)
ments, 63 animals were fed a diet supplemented with were measured after 5 weeks on the various diets in
ot-lipoic acid for 12 days, then liver, skin, and brain liver, kidney, heart, and skin. Only the mice fed the
homogenates were tested for susceptibility to lipid per- diet supplemented with a-lipoic acid displayed any
oxidation. The amount of peroxidation induced by unbound a-lipoic acid or DHLA in these tissues. Total
AMVN (measured as TBARS after 40 min of incuba- a-lipoic acid was highest in heart (3.42 ___2.20 nmol/
tion with AMVN) decreased 79% in liver, 64% in skin, g wet weight) and lowest in liver (0.60 0.33 nmol/
and 50% in brain. These experiments indicate that a- g). In all tissues DHLA was detected as well as ot-
lipoic acid in the diet has an antioxidant effect, both lipoic acid, and represented from 21% to 45% of the
at the tissue level and at the level of the entire animal, total. Metabolites of a-lipoic acid were not measured.
but they do not prove that ot-lipoic acid is, in fact, even This is the strongest evidence to date that a-lipoic
absorbed intact. acid, administered in the diet, accumulates in tissues,
Experiments using radiolabeled a-lipoic acid indi- and that a substantial portion of the t~-lipoic acid is
cate that the compound is absorbed. When 14C-labeled converted to DHLA. However, the absorption and re-
lipoic acid was administered to rats, either as an IP duction of a-lipoic acid in humans is not as well-
injection or orally, via stomach tube, 80% of the ad- studied; endogenous plasma levels of a-lipoic acid
ministered radioactivity was either excreted or found were 1 - 2 5 ng/ml and of DHLA 3 3 - 1 4 5 ng/ml in 6
in the tissues. 64 This was true even in animals whose healthy volunteers, 66 but plasma levels of ~-lipoic acid
intestinal flora had been killed, so it is unlikely that and DHLA in individuals supplemented with a-lipoic
the animals were absorbing metabolites produced by acid remains to be thoroughly investigated.
bacteria in the gut. Another area that requires further study is the extent
The next question is how much of the ot-lipoic acid to which exogenously supplied a-lipoic acid is metabo-
is converted to DHLA intracellularly. lized to shorter chain homologues, which may have
ot-Lipoic acid administered to a variety of cell and different effects than a-lipoic acid itself. In studies of
tissue systems appears in the medium as DHLA. 65 administration of radiolabeled a-lipoic acid t o r a t s , 67
However, the intracellular fate was unknown. In a much of the excreted a-lipoic acid was in altered
more recent study, l lipoic acid was added to the cul- forms, including the /~-oxidation products bisnorli-
ture medium for human fibroblasts or Jurkat T-cells, poate, tetranorlipoate, and beta-hydroxybisnorlipioc
at concentrations from 1 to 4 mM. The concentrations acid. It is not known what proportion of administered
238 L. PACKER et al.

a-lipoic acid is converted to these metabolites; in one properties of a-lipoic acid and dihydrolipoic acid is of
study of c~-lipoic acid administered to cultured T-lym- particular interest. Glycation of proteins may also play
phocytes, no formation of shorter chain-length beta- a role in these and other pathologies accompanying
oxidation products of lipoic acid was observed over 2 the disease, a-Lipoic acid has been shown to have
h. 1 Hence, the question of how much a-lipoic acid promise for preventing glycation reactions.
is converted to its shorter chain homologues is still
unanswered. However, it is worth considering their Effects in animal models of type i diabetes. Type I
effects. diabetes results from/3-cell destruction by immunolog-
Reduced forms of bisnorlipoic acid and tetranorli- ical or inflammatory attack. One animal model for this
poic acid react with superoxide and are more reactive type of diabetes is the nonobese diabetic mouse. When
than dihydrolipoate. Bimolecular kinetic rate constants diabetes development was accelerated in nonobese dia-
were 17.7 105 M -1 s -~ and 10.9 103 M -~. s -~ for betic mice by cyclophosphamide administration, 60%
the superoxide reaction for bisnorlipoate and tetranorli- of the mice developed diabetes within 1 - 3 w e e k s . TM If
poate, respectively, compared to 7.3 x 105 M - t . s -1 a-lipoic acid was given (10 mg/kg, IP) for 10 days
for DHLA. 28 Reduced forms of bisnorlipoate and tetra- before and 10 days after cyclophosphamide adminis-
norlipoate were also found to scavenge peroxyl radi- tration, only 30% of the mice developed diabetes. This
cals both in aqueous solution and in liposomes, z8 effect could be due to suppression of nitric oxide re-
Reduced forms of bisnorlipoate and tetranorlipoate lease by macrophages, which has been shown to occur
also have marked prooxidant effects. In the absence in the presence of DHLA, 75 as well as to scavenging
of iron chelator, reduced forms of bisnorlipoate and of other reactive oxygen species released by inflam-
tetranorlipoate potentiated hepatic microsomal lipid matory cells which attack the pancreatic/3-cells. The
peroxidation induced by AMVN-generated peroxyl large suppression of diabetic induction by a-lipoic acid
radicals. 28 The reduced form of tetranorlipoate was ex- should prompt further research in this area.
tremely potent in inducing lipid peroxidation.
In summary, a-lipoic acid is absorbed from the diet, Effects on glucose uptake in type ii diabetes. One of
transported to the tissues, and taken up by cells where the major features in the pathogenesis of Type II diabe-
a large proportion is rapidly converted to DHLA. tes is insulin resistance. Most Type II diabetics are
Hence the effects of both a-lipoic acid and DHLA hyperinsulinemic, hence no insulin therapy is war-
must be considered when proposing mechanisms for ranted. Improvement of insulin action on glucose trans-
therapeutic effects. The extent to which a-lipoic acid port activity in skeletal muscle has therefore been the
and DHLA are metabolized, and the exact antioxidant/ basis for a number of studies seeking to find therapeutic
prooxidant balance of their metabolites, is yet to be interventions that will be successful. As skeletal mus-
established. These metabolites may also play a signifi- cle tissue is the major sink in the body for glucose
cant role in the observed effects of treatment with a- following a meal, agents that enhance glucose uptake
lipoic acid. by skeletal muscle are potentially useful in the long-
term treatment of Type II diabetes.
a-Lipoic acid enhances glucose utilization in iso-
Diabetes
lated rat diaphragm, 76 h e a r t , 77 and cultured myotubes. 78
a-Lipoic acid has potential applications for many In the latter study, the authors report that the naturally
aspects of the pathology of diabetes. In Type I (insulin- occurring R-form stereoisomer of lipoic acid is more
dependent) diabetes, destruction of pancreatic/3-cells effective than the S-form in stimulating glucose up-
causes loss of insulin secretion, whereas in Type II take. R-ct-lipoic acid-stimulated glucose transport is
(noninsulin-dependent) diabetes insulin resistance of associated with the translocation from the cytosol of
peripheral tissues is the major problem, a-Lipoic acid GLUT-1 and GLUT-4 glucose transporters to the
has potential preventive or ameliorative effects in both plasma membrane; this has an additive effect on insulin
Type I and Type II diabetes. stimulated glucose transport. S-a-Lipoic acid stimu-
Many of the complications induced by diabetes, in- lates transport to a lesser extent, but does not promote
cluding polyneuropathy and cataract formation, appear translocation and actually inhibits insulin action on
to be mediated by oxygen free radical generation68; glucose uptake.
diabetic patients have elevated serum levels of thiobar- Using the obese Zucker rat as an animal model of
bituric acid reactive substances compared to nondiabet- obesity in insulin resistance, Henriksen and col-
iCS. 69 Since a-lipoic acid and DHLA have been shown leagues 79 demonstrated that ot-lipoic acid treatment
to prevent diabetes-induced biological alterations, 7-73 markedly increased the uptake of glucose [as 2-deoxy-
the relationship between the role of oxidative stress glucose] in the absence or presence of insulin. Acute
in diabetes-induced complications and the antioxidant treatment with a-lipoic acid as a single dose of 100
Antioxidantproperties of lipoic acid 239

mg/kg body weight for 1 h, or chronic treatment (30 ting the importance of the hydrophobic interactions
mg/kg for 10 days) resulted in improved insulin-stimu- between albumin and a-lipoate in protection against
lated 2-DG uptake in epitrochlearis muscles by over glycation.
50% in both cases. Glycated human serum albumin binds fewer fatty
Jacob and colleagues 8 have also demonstrated in acids than does nonglycated albumin, 87 suggesting that
humans with Type II Diabetes that a-lipoic acid ad- glycation sites are adjacent to fatty acid binding sites.
ministration (1000 mg given intravenously) enhanced In an early step of albumin glycation, a-lipoic acid
insulin-stimulated whole body glucose disposal by may protect BSA from glycation by masking the glyca-
about 50%. These results correlate well with the find- tion site through hydrophobic binding. Although cova-
ings of Bashan et al. 78 and Henriksen et al. 79 in the lent interaction with serum albumin has been reported
myotube and rat epitrochlearis muscle studies reported to inhibit the protein glycation, noncovalent interaction
above. of Diclofenac (Voltaren) with human serum albumin
A biochemical explanation for these findings is re- was also reported to inhibit glucose attachment to hu-
quired. It has been suggested that the exogenously sup- man serum albumin. 88 a-Lipoic acid may act in a simi-
plied a-lipoic acid may bring endogenous levels of a- lar manner.
lipoic acid, known to be low in diabetic animals 7 back Protection of human low density lipoprotein [LDL]
to normal. Alternatively, a-lipoic acid may react with against glycation by a-lipoate was also investigated.
cellular sulflaydryl groups, believed to be involved in If a-lipoate inhibits LDL glycation, this could have
the regulation of insulin-stimulated glucose transport.s~ clinical significance because diabetic patients have a
The effect may also be due to LA's antioxidant func- higher frequency of atherosclerosis than nondiabetic
tion. At present it is not possible to distinguish among individuals, s9 LDL is a large particle of more than 1.5
these possibilities. Although the mode of action of million kD with a cholesterol: phospholipid surface, a
lipoic acid stimulation of glucose disposal remains to core composed primarily of cholesterol ester, and a
be elucidated, these studies are consistent with work single protein (apo B100), which is interdigitated in
in animal and cell model systems, and suggest the need the surface. Physically, apo B 100 has both hydrophilic
for a larger scale clinical study. and hydrophobic domains; the hydrophobic portion of
apo B in LDL is submerged in the lipid structure, and
Glycation reactions and atherosclerosis. Glycation of in this respect it differs from albumin, a-Lipoic acid
proteins may be an underlying factor in a number of was found not to protect LDL from glycation in v i t r o 86
the pathologies of diabetes, 82 and free radicals may be (Fig. 5). These findings also emphasize the significance
involved in the process. 68 Although several mecha- of the hydrophobicity of a-lipoate in protection against
nisms have been postulated for the pathogenesis of glycation. Schepkin et al. 9 also reported using NMR
chronic diabetic complications, protein glycation and techniques that ~t-lipoate binds to albumin, but not
oxidation by glucose (glycoxidation) are plausible to LDL.
working hypotheses. 83'84"68 However, although a-lipoic acid was not found to
a-Lipoic acid has been tested in a number of model protect LDL in the short term, the long-term effects of
systems, particularly serum albumin and lysozyme, glycation involve further reaction to Amadori products,
where the inhibition of enzyme activity or the extent and eventually the formation of advanced glycosolated
of glycosylation has been found to be protected against end-products (AGE). These steps may involve oxida-
by incubating in glucose together with a-lipoic acid. 85 tive mechanisms. It is worth noting in this regard that
In another study, Kawabata and Packer 86 found that a-lipoic acid has been found to be protective of human
noncovalent hydrophobic binding of a-lipoic acid to LDL exposed to oxidative stress 4'91 (Fig. 6). Hence,
albumin was involved in its protective effects (Fig. 5). over the long-term, a-lipoic acid may help protect dia-
Also in this in vitro test system, both the R and S betics from the atherosclerosis that is a common com-
enantiomer forms of ot-lipoic acid behaved similarly. ponent of the disease.
In addition, both a-lipoic acid and DHLA protected
albumin from glycation in the same manner, demon- Diabetic polyneuropathy. Endoneural blood flow and
strating that the preventive effect is independent of its oxygen tension are reduced in experimental diabetic
redox state. Since a-lipoate has a hydrophobic carbon neuropathy. This process of endoneural hypoxia is as-
chain, it is likely that a-lipoate binds to albumin by sociated with an increase of oxidative stress and an
hydrophobic interactions similar to fatty acids. By impairment of the nerve conduction velocity.92
comparing a-lipoate with a shorter chain homologue, Antioxidants such as glutathione have been shown
tetranor-lipoate, it was shown that ot-lipoate is much to prevent neuropathy in animal models, 93 and a-lipoic
more protective against glycation. In fact, tetranor-li- acid induces sprouting of neurites in culture. 94
poate did not show any protective effect, demonstra- Clinical trials have been carried out on the effects
240 L. PACKER et al.

100.0"
15.0 T
go.o]~

io.o : 60.0]

40.0[
5.0]~(/

20.0t

o,o' ~ . . . . . . . 0.0 . , . . . , .

0 I 2 3 4 5 6 0.0 5.0 10.0 15.0 20.0


Time (days) ct-Lipoate bound (raM)
Fig. 5. (A) Effect of tr-lipoate on LDL glycation. LDL (1 mg protein/ml) was incubated with 200 mM glucose in the presence
(open circles) or absence (filled squares) of ,~-lipoate (20 mM) in Hepes-saline (pH 7.4) at 37C. At the indicated times,
glycated LDL were separated by affinity chromatography and protein concentrations measured. Values are averages of duplicate
observations. LDL was also incubated without glucose as a negative control (filled circles). (B) The binding of ot-lipoate to
BSA. BSA (1 mM) was incubated with various concentrations of a-lipoate in Hepes-saline (pH 7.4) at 37(2 for 18 h. The
sample was centrifuged in a centricon 30 and the a-lipoate concentration in the filtrate was measured by absorption spectroscopy
(e = 150 M-' cm-').

of a-lipoic acid supplementation on diabetic neuropa- patients had no pain, four had moderate pain, and only
thy, and its use for this condition is approved in Ger- one had severe pain. Despite these dramatic clinical
many. improvements, neurophysiological measures, such as
In one placebo-controlled double blind study,95 21 vibration sense and nerve conduction velocity, showed
days of intravenous administration of a-lipoic acid no change with IV lipoic acid administration. The au-
(200 mg daily) caused alleviation of some clinical thors note that 21 days may have been too short a time
symptoms: before treatment six patients had moderate in which to observe changes in neuronal characteris-
pain and four had severe pain, and after treatment five tics.

,,o i 1,oi po od B

3O
~ 0 , ,
i 130 0.0 0,6 1,2 118 0,0 1.0 2.0 3,0
DHLA, ram
. Vitamin E Ascorbate, m M + Ascorbate + DHLA

l
50 o Light on

+ Ascorbate + DHI~ . j ~
.-.L4I~ -

-I0 0 30 60 90 0 30 60 90

TIME, m i n
Fig. 6. Time course of UV-induced vitamin E chromanoxyl and ascorbyl ESR signals in LDL suspensions. (A) Effect of
ascorbate: ascorbate, 500 #M. Insert: dependence of the lag period (during which the vitamin E chromanoxyl radical ESR signal
was not observed) on the concentration of ascorbate. (B) Effect of dihydrolipoic acid plus ascorbate, ascorbate 500/zM; DHLA
2 mM. Insert: dependence of the lag period (during which vitamin E chromanoxyl radical ESR signal was not observed) on the
concentration of adihydrolipoic acid (the ascorbate concentration was 500 #M). Incubation medium (100 #1) contained: LDL
samples with endogenous vitamin E (6.2 nmol/mg protein, 14 mg protein/ml), ascorbate, 500 #M inphosphate buffer, pH 7.4,
at 250(2. All values are given as a percentage of the maximum magnitude obtained. Vitamin E chromanoxyl radicals were
generated by irradiation with UV light (290-400 nm).
Antioxidantproperties of lipoic acid 241

In a longer study, ot-lipoic was compared to vitamin Also, in vitro diabetic cataractogenesis in rat lens
B 1 as treatment for diabetic neuropathy in a single- cell cultures exposed to high concentrations of glucose
blind study, ct-Lipoic acid (600 mg/day) or vitamin was prevented by t~-lipoic acid 99 (Fig. 7). Our group l
B 1 (400 mg/day) were administered intravenously and has hypothesized that a-lipoic acid prevents oxidative
intramuscularly, respectively, to diabetics for 3 weeks, stress in diabetic conditions by sparing vitamin C,
followed by 12 weeks of oral administration of the whose transport is affected by the disease. Since vita-
s a m e d o s e . 96 Pain and parathesia were reduced in the min C and glucose share the same carrier in noninsulin
a-lipoic acid patients when compared with the vitamin dependent tissues, the elevated blood glucose in diabe-
B 1 patients, but again in this study no improvement tes competitively inhibits the cell entry of vitamin C,
in motor or sensory nerve conduction velocity could thus resulting in localized intracellular vitamin C defi-
be demonstrated due to lipoic acid administration. ciency (Fig. 8). Exogenously supplemented ot-lipoic
However, these authors point out that a period of acid utilizes other transport systems to enter the cells,
months or years is required to observe improvement is converted to dihydrolipoate, and recycles vitamin
in diabetic neuropathy due to normalization of blood C. This would explain its protective effect in diabetic
glucose levels, so it is not surprising that no measurable cataractogenesis, as well as other complications of dia-
neurophysiological changes occurred over a period of betes.
15 weeks of a-lipoic acid supplementation. However,
clinical improvement, evaluated by blinded observers, Conclusions. a-Lipoic acid has been shown to have a
has clearly occurred in these studies. number of beneficial effects, both in prevention and
In a nonblinded study in which several parameters treatment of diabetes, c~-Lipoic acid may act in a num-
were studied, 97 diabetic patients (both type I and type ber of ways that are especially protective in diabetes.
II) were given oral supplements of either t~-lipoic acid It prevents/~-cell destruction leading to Type I diabe-
(600 mg/day for 2 weeks, then 300 mg/day for 10 tes. It enhances glucose uptake in Type II diabetes.
weeks), a-tocopherol (1575 IU daily), or selenium It prevents glycation reactions in some proteins. Its
(100/zg daily), and compared to controls (no supple- antioxidant effects may be particularly useful in slow-
ments, no placebo) after 12 weeks. Blood malondialde- ing the development of diabetic neuropathy and cata-
hyde decreased from 14.4 #mol/1 to 10.9 #mol/l in the ractogenesis, and this may be especially significant in
a-lipoic acid group. Similar reductions were seen in alleviating diabetes-induced reduction in intracellular
the a-tocopherol and selenium groups. No change was vitamin C levels.
seen in the control group. Albuminuria decreased 50%, Very few of these effects will manifest as objective
46%, and 26% in the a-lipoic acid, a-tocopherol sup- improvement over the course of weeks or even months.
plemented, and selenium supplemented groups, respec- This is demonstrated in trials for treatment of neuropa-
tively. There was clinical improvement in neurological thy, which lasted up to 12 weeks, in which objective
symptoms in all supplemented groups but not in the improvement was not observed but clear subjective
control group. In addition, while retinopathy worsened improvement was present, even in double-blinded
in 5 of 9 in the control group, there was only one case studies. Given the array of beneficial effects of ct-lipoic
of worsening of retinopathy in any of the antioxidant acid that have been shown in both animal and human
supplemented groups. This study showed that mitigat- diabetes, large, long-term, well-controlled studies seem
ing oxidation, whether by a-lipoic acid supplementa- warranted. It is unrealistic to expect dramatic effects
tion or supplementation with other antioxidants, im- in weeks, since diabetic complications develop over
proved biochemical and functional parameters in dia- years and decades.
betics; however, the study suffered from lack of
placebo control and lack of double-blind.
Ischemia-reperfusion injury
Cataracts. Another common complication of diabetes Ischemia-reperfusion injury occurs when a burst of
is cataract formation, a-Lipoic acid dietary supplemen- free radicals is produced during reoxygenation of tissue
tation has been shown to prevent cataract formation that has become hypoxic. It is important in cardiac
caused by BSO-induced inhibition of GSH synthesis in tissue (especially with the introduction of clot-dissolv-
newborn rats, while sparing lens ascorbate, tocopherol, ing drugs for the treatment of heart attack) and in brain.
and GSH. 98 100% of BSO-treated rats developed cata- Agents that prevent ischemia-reperfusion injury may
racts, but if they also received a-lipoic acid, the inci- therefore prove important in the treatment of cardiac
dence of cataract formation dropped to 40%. Signifi- infarct, during open-heart surgery, and in the treatment
cant protection of lens ascorbate, tocopherol, and gluta- of stroke and other conditions that cause interruption
thione was also observed in the lipoate-supplemented of blood flow to the brain.
rats compared to nonsupplemented. In vitro, DHLA prevents ischemia-reperfusion in-
242 L. PACKER e t al.

Fig. 7. Effect of a-lipoate on glucose induced lens opacity. Lenses as seen through a dissection microscope, after 8 days of
incubation. Rat lenses with intact capsules were incubated in Modified Eagle Medium 199, with (A) normal glucose (5.56 mM)
and 1 mM R-a-lipoic acid, (B) normal glucose (5.56 mM) and i mM S-ot-lipoic acid, (C) normal glucose (5.56 raM) and 1
mM racemic a-lipoic acid, (D) elevated glucose (55.6 mM) and I mM R-a-lipoic acid, (E) elevated glucose (55.6 mM) and 1
mM S-a-lipoic acid, and (F) elevated glucose (55.6 mM) and 1 mM racemic a-lipoic acid.

duced changes in fluidity and polarity of rat heart mito- head trauma, subarachnoid hemorrhage, stroke, or cardiac
chondria, jj Several studies extend these experiments arrest. Prehn et al., in a study with middle cerebral artery
to more closely match in vivo situations. occlusion in mice, demonstrated that in this model of
Serbinova et al. j2 reported that hearts from rats fed a- focal ischemia, treatment with DHLA, but not a-fipoic
lipoic acid were protected against ischemia-reperfusion acid, reduced the size of the infarct. 15"16 The authors
injury induced in an isolated perfused Langendorff heart also noted a hypoglycemic effect at the doses used (100
system, a-Lipoic acid preserved vitamin E in heart tissue, mg/kg body weight). In another model of rat brain isch-
improved postischemic left ventricular functional recov- emia-reperfusion injury, in which the carotid arteries
ery, and decreased lipid pemxidation and lactate dehydro- are occluded, pretreatment with a-lipoic acid decreased
genase leakage (a marker of membrane damage). Ha- mortality from 80% to 20%; this marked decrease in
ramaki et al., ~3 using Langendorff and working heart mortality was accompanied by protection of glutathione
systems, further observed that the protective effects of (which was depleted in controls) and decreased levels of
dihydrolipoic acid against rat myocardial ischemia-re- lipid peroxidation product formation.
perfusion injury are dependent on vitamin E, suggesting In other ischemia-reperfusion systems, Boveris et
that a-lipoic acid functions in this system by regenerating al. 17 reported that a-lipoic acid administration pre-
tocopherol from the tocopheroxyl radical. Assadnazari et vented rat intestinal short-term ischemia-reperfusion
ai., ~4 using a working heart system in an NMR magnet, induced overshoot of chemiluminescence, which is in-
found that DHLA added to the reperfusion buffer acceler- dicative of an increased steady-state level of singlet
ated the recovery of aortic flow during reperfusion. oxygen and an increased rate of lipid peroxidation.
DHLA also appeared to increase ATP synthesis in the Boveris also observed that ot-lipoic acid can inhibit
heart. xanthine oxidase activity; xanthine oxidase is postu-
The brain is another area where ischemia-reperfusion lated to play an important role in the mechanism of
injury can have serious consequences; for example, in ischemia-reperfusion injury.
Antioxidant properties of lipoic acid 243

"Starvation Amidst Plenty" Thioctic (o<-Lipoic)Acid SupplementationI


I Insulin insufficiency-~ Diabetes->. Glucose ElevationI
L~oate Elevation
J Enhanced Cellular Uptake** .. Lipoate uses fattyacid
Glucosylation Impaired Cellular transporter system
Reactions Uptake of
Dehydroascorbate (DHA)*
Accumulation of Dihydrolipoate(DHLA)
AGE (Advanced
Glucosylation Ascorbate Deficiency
End Products) Increased efficiency of the
Vitamin C and Vitamin E Cycles
"Aging" "Localiz!d Scurvy"
~ [
\
ROO o ~ Vitamin E ~ - DHA ~ _ DHLA~ NAD+
* Since glucose and dehydroascorbate use the
same sugar transporter system, high glucose
competitively inhibits vitamin C uptake. ROOH VitaminE Ascorbate Lipoate NADH
Free Radical

Fig. 8. Schematic diagram of the pathobiochemical situation in diabetes mellitus and the possible correction by oral administration
of a-lipoic acid. Since glucose competitively inhibits the absorption of ascorbic acid into the cells, a-lipoic acid increases the
efficiency of the vitamin C cycle and thereby also activates the vitamin E cycle.

Thus, in a number of ischemia-reperfusion model its elimination from a treatment regimen for this condi-
systems, DHLAJct-lipoic acid has been shown to be tion. "
effective in ameliorating or preventing damage. In
Alcoholic liver disease. Several studies indicated that
none of these studies were the cellular contents of
a-lipoic acid administration might be beneficial in liver
DHLA or c~-lipoic acid measured, so it is difficult to
disease, especially alcoholic liver disease, m - m How-
say with certainty exactly what the mechanism of pro-
ever, these studies suffered one or more of the follow-
tection was. In the cardiac system the effect appears ing flaws: lack of control groups, lack of statistical
due to recycling.
analysis, or the use of other treatments in addition to
~r-lipoic acid.
In a controlled, double-blind, long-term study, a-
Liver diseases
lipoic acid had no effect on the course of alcohol-
a-Lipoic acid is often used for therapy in conditions related liver disease. 1~4 Twenty patients with precir-
that involve liver pathology. The basis for such treat- rhotic liver disease received a-lipoic acid (300 mg/
ment is the metabolic role of a-lipoic acid, not its daily, orally) for 6 months, and 20 patients received
antioxidant properties. The two most extensively stud- placebo. Twenty-two of the patients abstained from
ied conditions are mushroom poisoning and alcoholic alcohol, and the other 18 reduced daily alcohol intake.
liver degeneration. There is little evidence that a-lipoic Significant improvements in blood parameters (serum
acid is useful in either of these conditions. aspartate transaminase, serum gamma glutamyl trans-
peptidase, and mean corpuscular volume) occurred in
the abstainers, as well as histological improvement in
Mushroom poisoning. There have been many case 17; 5 of the reduced intake group also showed histolog-
studies in which treatment with a-lipoic acid has been ical improvement but there was no significant improve-
associated with complete recovery from mushroom ment in blood characteristics. ~-Lipoic acid did not
(Amanita) poisoning. ~8'~9 However, 10-50% of vic- influence the course of the disease.
tims recover without a-lipoic acid treatment, so such
reports may be coincidental. In animal studies, a-lipoic Nf-Kb activation, hiv, and aids
acid has proved completely ineffective as a treatment Oxidative stress may play a role in several aspects
for Amanita poisoning, and some groups recommend of HIV infection, including activation of virus replica-
244 L. PACKERet al.

tion, 115 immunosuppression,~ 16.117and tumor initiation a-lipoic acid has in various neurodegenerative disor-
and promotion. 118 HIV-infected patients have been re- ders.
ported to be deficient in various antioxidants. H9-~21 Studies are underway in animals. For example, a
Hence, there may be a role for antioxidant therapy in recent study examined the effect of a-lipoic acid on
retarding the progress of the disease. memory loss in aging mice. ~35Many species, including
Following this reasoning, a pilot study was recently man, monkeys, rats, and mice, exhibit aging-related
conducted on the effects of a-lipoic acid supplementa- cognitive deficits that may be caused, at least in part,
tion on blood antioxidant status and blood peroxidation by oxidative stress. In mice, a-lipoic acid (100 mg/kg
products in HIV-positive individuals. 122 Twelve pa- body weight for 15 days) improved performance in an
tients (HIV positive, classified CDC IV) received oral open-field memory t e s t - - i n fact, the a-lipoic acid-
doses of 150 mg of lipoate three times daily for a treated animals performed better than young animals
period of 14 days. At the end of that time, 9 of 10 24 h after the first test (though the difference was not
patients exhibited increases in plasma ascorbate, 7 of significant). 135 Treatment with a-lipoic acid did not
7 showed increased plasma glutathione, 8 of 9 had improve memory in young animals. The a-lipoic acid-
decreased plasma malondialdehyde, and 7 of 9 bad treated animals exhibited decreased age-related N-
decreased 4-hydroxynonenal. Accompanying these methyl-o-aspartate (NMDA) receptor deficits (Bmax)
changes, the number of T-helper cells increased in 6 compared to controls, but showed no improvement in
of 9 patients, and the T-helper/T-suppresser cell ratio muscarinic, benzodiazepine, or aE-adrenergic receptor
improved in 6 of 10 patients. deficits. The authors concluded that a-lipoic acid's free
Also, in cultured cells, a-lipoic acid and dihydroli- radical scavenging ability may improve NMDA recep-
poic acid prevented HIV replication, 123 the activation tor density, leading to improved memory. These intri-
of NF-KB transcription factor, 56 which are regulated guing results suggest further tests in other species, in-
by oxidative stress. cluding humans.
These studies suggest that further research is war- Recently, Greenamyre et al. ~36observed that the in-
ranted on the effect of a-lipoic acid in HIV infection. traperitoneal administration of a-lipoic acid or DHLA
reduced the rat striatum lesions induced by excitotox-
ins which affect NMDA receptors, which may lead to
Neurodegenerative diseases calcium influx and generation of nitric oxide and other
free radicals. ~37 Both a-lipoic acid and DHLA treat-
Tissues of the central nervous system may be espe- ment decreased the area of lesion by 50%.
cially vulnerable to oxidative stress because of their Another way in which mitochondria may be im-
constant high rate of oxygen consumption and high portant in neurodegeneration is through alterations in
mitochondrial density. Mitochondria inevitably pro- their effects on calcium homeostasis. In this regard,
duce free radicals as "byproducts" of normal oxida- the recent report of Christof Richter (personal commu-
tive metabolism, 124'125 and these free radicals damage nication) that a-lipoic acid inhibits mitochondirial cal-
the mitochondrial DNA. 126 The defective proteins cium transport may be relevant to its beneficial effects
coded for by the damaged DNA can lead to synthesis noted in neurodegenerative disorders noted by Greena-
of mitochondria in which components of the electron myre. 136
transport chain preceding the damaged protein become Ischemia-reperfusion injury, induced by head
reduced, leading to greater free radical production ~27- trauma, subarachnoid hemorrhage, stroke, or cardiac
J29 and more mitochondrial damage, in a vicious cycle. arrest, is another potential source of free radical dam-
Such a vicious cycle may be responsible, in part for age to neuronal structures. The protective effect of a-
neurodegenerative diseases. lipoic acid in this condition has already been discussed
Support for this view comes from the observation of (see Ischemia-Reperfusion Injury). In addition, a-li-
high degrees of oxidative damage as well as damaged poic acid treatment of rats exposed to inhalation of n-
mitochondria in tissues from patients with neurodegen- hexane (constant exposure to 700 ppm) delayed the
erative disease. 129-134 Logical therapy or prevention onset of severe neuropathy by 3 weeks: control rats
would therefore involve antioxidant treatment. displayed severe motor neuropathy by 6 weeks, while
a-Lipoic acid is a good candidate as an antioxidant in rats which also received a-lipoic acid, severe neu-
agent in neurodegenerative diseases. It can interrupt ropathy did not appear until the ninth week. m
the chain at several points: by competing for free tran-
sition metals as a chelator, 19'2~by scavenging hydroxyl
Radiation injury
or superoxide radicals, ~2 and by scavenging peroxyl
radicals. 9 Few other antioxidants possess this kind of Irradiation is known to produce a cascade of free
versatility, and it will be interesting to see what effect radicals, and antioxidant compounds have long been
Antioxidant propertiesof lipoic acid 245

used to treat irradiation injury, a-Lipoic acid, but not tein carbonyls (a marker of protein oxidation) ap-
DHLA, protected against radiation injury to hemato- pear. 143"145Exogenously supplied DHLA is protective
poietic tissues in mice, and increased the LDso from against CS-induced oxidation of antioxidants, proteins,
8.67 to 10.93 Gy. 139 c~-Lipoic acid administration to and lipids.144 DHLA also partially protected polymor-
murine neroblastoma cells 43 increased cell survival phonuclear leukocytes from the damaging effects of
after irradiation from 2% to about 10%, and the effect CS. 146This effect may be due to scavenging of oxidants
correlated with an increase of the intracellular GSH/ in the aqueous or lipid phases, or to regeneration of
GSSG ratio induced by c~-lipoate. These results ex- ascorbic acid that has been converted to ascorbyl radi-
tended to mice irradiated with 8 Gy; a-lipoic acid ad- cal as it is oxidized by CS components. In this regard,
ministration (16 mg/kg) increased survival rates from DHLA could play a role in minimizing the pathologic
35% in untreated animals to 90% in oL-lipoic acid- consequences of smoking. To extend these studies to
treated animals. the in vivo situation, it will be necessary to know what
The worst recent widespread human radiation expo- levels of DHLA are achieved in plasma after dietary
sure occurred after the Chernobyl accident. People supplementation with a-lipoic acid.
continue to live in areas where the soil is contaminated
with 15-40 Ci/km2, and are thus exposed to constant, Heavy metal poisoning
low-level radiation. In an effort to determine whether
antioxidant treatment might help prevent damage due The possible chelating effects of a-lipoic acid,
to such constant exposure, Korkina, Afanas'ef, and together with its antioxidant effects, make it a good
Diplock recently examined the effects of 28 days of candidate for the treatment of heavy metal poison-
antioxidant treatment on a variety of blood and urinary ing. For arsenite, cadmium, and mercury, especially,
parameter in children living in areas affected by the it may be effective. Grunert t47 demonstrated that a-
Chernobyl accident. 14 Using spontaneous chemilumi- lipoic acid administration completely protected mice
nescence as a marker for blood peroxidation, they and dogs from arsenite poisoning, if the ratio of a-
found that treatment with c~-lipoic acid alone lowered lipoic acid to arsenite was at least 8:1. This was
this value to that seen in nonradiation-exposed chil- true even if the ot-lipoic acid was administered after
dren; a-lipoic acid + vitamin E treatment further low- severe symptoms of poisoning were already seen.
ered chemiluminescence to below-normal values; vita- Similar complete protection was seen for Hg 2 poi-
min E alone was without effect. Urinary excretion of soning in mice, although at low doses (2:1 molar
radioactive metabolites was also lowered by a-lipoic ratio) ot-lipoic acid appeared to potentiate the lethal
acid but not by E, presumably due to chelation by effect of Hg 2. a-Lipoic acid was ineffective against
a-lipoic acid. Liver and kidney functions were also lead and gold poisoning in mice in this study. Expo-
normalized by a-lipoic acid treatment. sure of isolated hepatocytes to a-lipoic acid or
Hence it appears that a-lipoic acid, alone or together DHLA resulted in the amelioration of cadmium 2-
with vitamin E, is an effective treatment for radiation induced membrane damage, lipid peroxidation, and
exposure, lessening indices of oxidative damage and the depletion of cellular glutathione. 2~ These findings
normalizing organ function. were extended to a rat model, in which o~-lipoic acid
at a dose of 30 mg completely prevented cadmium-
induced lipid peroxidation in brain, heart, and tes-
Cigarette smoke effects on plasma proteins
tes. 148 In this study it also completely abolished cad-
Although the exact mechanisms by which cigarette mium-induced decreases in the activities of Ca 2-,
smoke (CS) exposure causes such pathologies as ath- Na -, and Mg+-ATPases in these organs, a-Lipoic
erosclerosis, pulmonary emphysema, and cancer have acid administration to rats also increased biliary ex-
not yet been elucidated, it is known that CS contains cretion of injected Hg 2 (the major route of excre-
a number of free radical species. 141'~42Since many of tion) by 12-37-fold, L49 but dramatically decreased
the diseases caused by smoking involve, at least in biliary excretion of Cd 2, methyl mercury, Zn 2, and
part, free-radical-mediated processes, it has been pro- Cu 2. It is not clear whether this was due to chelation
posed that the free radicals in CS contribute to these of these metals in the circulation by ot-lipoic acid or
diseases. The effects of CS on lung lining fluids has other effects of a-lipoic acid. a-lipoic acid adminis-
been investigated using plasma as a model system. tration has also been found to greatly increase the
Freshly obtained human plasma is exposed to " p u f f s " rate of elimination of radiomercury in rabbits. 15
of gas-phase or whole CS, and antioxidants and mark- Hence, a-lipoic acid may be of clinical value in the
ers of oxidative damage are measured. In this system, treatment of mercury and cadmium poisoning. Its
ascorbic acid is consumed first, followed by protein effect on other heavy metals is not yet completely
thiol groups. 143.144Lipid hydroperoxides as well as pro- clear.
246 L. PACKER et aL

Chagas disease tions in both membrane and aqueous phases. 9 Both


a-lipoic acid and DHLA have substantial antioxidant
Chagas disease is common in South America and
properties. These include their ability to directly
is caused by the protozoan Trypanosoma cruzi. It is
quench a variety of reactive oxygen species, inhibit
treated with benznidazole, but side effects in up to
reactive oxygen-generators, and spare other antioxi-
50% of cases are a major problem. In a double blind
dants.
study of 50 patients with Chagas disease, tS~ 25 were
A number of experimental as well as clinical studies
treated with benznidazole + a-lipoic acid (20 mg/day)
point to the usefulness of ot-lipoic acid as a therapeutic
and 25 with benznidazole + placebo. In the benznida-
agent for such diverse disease conditions as myocardial
zole + a-lipoic acid group, only 8% of the patients
and cerebral ischemia-reperfusion injury, heavy-metal
developed side effects and all of the patients were able
poisoning, radiation damage, diabetes, neurodegenera-
to complete treatment, whereas in the benznidazole +
tive diseases, and AIDS. High doses of ot-lipoic acid
placebo group 44% of the patients developed side ef-
are approved in Germany for treatment of diabetic
fects and two had to abandon treatment, a-Lipoic acid
polyneuropathy. Furthermore, the interesting antioxi-
is a beneficial adjuvant therapy in the treatment of this
dant properties of a-lipoic acid and its interaction with
disease.
other important antioxidants like vitamin E, ascorbate,
and glutathione will provide a fertile field for continued
Side effects research.

Neither animal nor human studies to date have Acknowledgements - - This research was supported by the National
shown serious side effects with administration of a- Institutes of Health (CA47597), Council for Tobacco Research
lipoic acid. 152The LDs0 is approximately 4 0 0 - 5 0 0 mg/ (33446R2), and the State of California Tobacco Related Disease
Research Program through the University of California (#4RT-
kg following intravenous administration in rats and 0065). The authors thank Dr. Y. J. Suzuki for preliminary discussions
4 0 0 - 5 0 0 mg/kg after oral dosing in dogs. In long-term at the outset of this project.
oral supplementation at doses sufficient to reduce body
weight gain, no functional or laboratory adverse effects
were seen in animals. No evidence suggests carcino- REFERENCES
genic or teratogenic effects, but it is recommended that I. Reed, L. J.; DeBusk, B. G.; Gunsalus, I. C.; Hornberger, Jr.,
pregnant women avoid taking supplemental a-lipoic C. S. Crystalline ~t-lipoic acid: a catalytic agent associated with
acid until more data are available. 152 Although few pyrnvate dehydrogenase. Science 114:93-94; 1951.
2. Reed, L. J. The chemistry and function of lipoic acids. Adv.
adverse effects are noted in studies of administration Enzymol. 18:319-347; 1957.
of a-lipoic acid, it has been observed in thiamine defi- 3. Reed, L. J. Multienzyme complex. Ace. Chem. Res. 7:40-46;
cient rats that a high dose of lipoic acid (20 mg/kg), 1974.
4. Rosenberg, H. R.; Culik, R. Effect of a-lipoic acid on vitamin
delivered intraperitoneally, caused fatal complications; C and vitamin E deficiencies. Arch. Biochem. Biophys. 80:86-
thiamine-sufficient rats suffered no adverse effects 93; 1959.
from c~-lipoic acid supplementation, and the action of 5. Carreau, J. P. Biosynthesis of lipoic acid via unsaturated fatty
acids. Meth. Enzymol. 62:152-158; 1979.
lipoic acid in the deficient rats could be prevented 6. Dupre, S.; Spoto, G.; Matarese, R. M.; Orlando, M.; Cavallini,
by administration of thiamine just prior to lipoic acid D. Biosynthesis of lipoic acid in the rat: Incorporation of 3sS-
administration. ~53 The animals in this study were se- and J4C-labeled precursors. Arch. Biochem. Biophys. 202:361-
365; 1980.
verely thiamine-deficient, showing frank polyneuritis 7. Rimm, E. B.; Stampfer, M. J.; Ascherio, A.; Giovannucci, E.;
from thiamine deficiency before lipoic acid was admin- Colditz, G. A.; Willett, W. C. Vitamin E consumption and the
istered, and the dose of lipoic acid was large; neverthe- risk of coronary heart disease in men. N. Engl. J. Med.
328:1450-1456; 1993.
less, it may be prudent that any group likely to be 8. Stampfer, M. J.; Hennekens, C. H.; Manson, J. E.; Colditz,
severely thiamine-deficient, for example, alcoholics, G. A.; Rosner, B.; Willett, W. C. Vitamin E consumption and
should receive supplemental thiamine if c~-lipoic acid the risk of coronary disease in women. N. Engl. J. Med.
328:1444-1449; 1993.
is given. 9. Kagan, V. E.; Shvedova, A.; Serbinova, E.; Khan, S.; Swanson,
In humans, side effects include allergic skin reac- C.; Powell, R.; Packer, L. Dihydrolipoic acid: A universal
tions and possible hypoglycemia in diabetic patients antioxidant both in the membrane and in the aqueous phase.
Biochem. Pharmacol. 44:1637-1649; 1992a.
as a consequence of improved glucose utilization with 10. Handelman, G. J.; Han, D.; Tritschler, H.; Packer, L. c~-Lipoic
high doses of ot-lipoic acid. tS: acid reduction by mammalian cells to the dithiol form and
release into the culture medium. Biochem. Pharmacol. in press;
1994.
11. Podda, M.; Han, D.; Koh, B.; Fuchs, J.; Packer, L. Conversion
CONCLUSIONS of lipoic acid to dihydrolipoic acid in human keratinocytes.
Clin. Res. 42:41A; 1994.
a-Lipoic acid and its reduced form, DHLA, have 12. Suzuki, Y. J.; Tsuchiya, M.; Packer, L. Thioctic acid and dihy-
been referred to as " a universal antioxidant" that func- drolipoic acid are novel antioxidants which interact with reac-
Antioxidant properties of lipoic acid 247

tive oxygen species. Free Rad. Res. Comms. 15:255-263; Cu(II) and thiols: A powerful chemical DNA-cleaving system.
1991. Bitch. Biophys. Res. Comm. 162:1 i 11 - 1117; 1989.
13. Scott, B. C.; Aruoma, O. I.; Evans, P. J.; O'Neill, C.; van der 35. Burton, G. W.; Ingold, K. U. Autoxidation of biological mole-
Vliet, A.; Cross, C. E.; Tritschler, H.; Halliwell, B. Lipoic and cules: 1. The antioxidant activity of vitamin E and related
dihydrolipoic acids as antioxidants: A critical evaluation. Free chain-breaking phenolic antioxidant in vitro. J. Am. Chem. Soc.
Rad. Res. 20:119-133; 1994. 103:6472-6477; 1981.
14. Haenen, G. R.; Bast, A. Scavenging of hypochlorous acid by 36. Sies, H. Strategies of antioxidant defense. Eur. J. Biochem.
lipoic acid. Biochem. PharmacoL 42:2244-2246; 1991. 215:213-219; 1993.
15. Stevens, B.; Perez, S. R.; Small, R. D. The photoperoxidation 37. Packer, L. New horizons in antioxidant research: Action of the
of unsaturated organic molecules: IX. Lipoic acid inhibition thioctic acid/dihydrolipoic acid couple in biological systems.
of rubrene autoperoxidation. Photochem. Photobiol. 19:315- In: Schmidt, K.; Ulrich, H., eds. Thioctsdure: 2. International
316; 1974. thioctic acid workshop. Frankfurt: Universimed Verlag GmbH;
16. Stary, F. E.; Jindal, S. J.; Murray, R. W. Oxidation of a-lipoic 1992:35-44.
acid. Z Org. Chem. 40:58-62; 1975. 38. Scholich, H.; Murphy, M. E.; Sies, H. Antioxidant activity of
17. Kaiser, S.; Di Mascio, P.; Sies, H. Lipoat und Singulettsauer- dihydrolipoate against microsomal lipid peroxidation and its
stoff. In: Borbe, H. O.; Ulrich, H., eds. Thioctsbure. Frankfurt: dependence on ot-tocopherol. Biochim. Biophys. Acta.
pmi Verlag GmbH; 1989:69-76. 1001:256-261; 1989.
18. Devasagayam, T. P.; Di Mascio, P.; Kaiser, S.; Sies, H. Singlet 39. Bast, A.; Haenen, G. R. M. M. Regulation of lipid peroxidation
oxygen induced single-strand breaks in plasmid pBR322 DNA: of glutathione and lipoic acid: Involvement of liver microsomal
The enhancing effect of thiols. Biochim. Biophys. Acta. vitamin E free radical reductase. In: Emerit, I.; Packer, L.;
1088:409-412; 1991. Auclair, C., eds. Antioxidant in therapy and preventive medi-
19. Devasagayam, T. P. A.; Subramanian, M.; Pradhan, D. S.; cine. New York: Plenum Press; 1990:111 - ! 16.
Sies, H. Prevention of singlet oxygen-induced DNA damage 40. Kagan, V. E.; Serbinova, E. A.; Forte, T.; Scita, G.; Packer,
by lipoate. Chem.-Biol. Interactions 86:79-92; 1993. L. Recycling of vitamin E in human low density lipoproteins.
20. Saito, I.; Fukui, S. Studies on the oxidation products of lipoic J. Lipid Res. 33:385-397; 1992b.
acid. J. Vitaminology 13" 115-121; 1967. 41. Constantinescu, A.; Han, D.; Packer, L. Vitamin E recycling
21. Miiller, L.; Menzel, H. Studies on the efficacy of lipoate and in human erythrocyte membranes. J. Biol. Chem. 268:10906-
dihydrolipoate in the alteration of cadmium 2 toxicity in iso- 10913; 1993.
lated hepatocytes. Biochim. Biophys. Acta. 1052:386-391; 41a. Gotz, M. E.; Dirr, A.; Burger, R.; Janetzky, B.; Weinmuller,
1990. M.; Chan, W. W.; Chen, S. C.; Reichmann, H.; Rausch,
22. Bast, A.; Haenen, G. R. M. M. Interplay between lipoic acid W. D.; Riederer, P. Effect of lipoic acid on redox state of
and glutathione in the protection against microsomal lipid per- coenzyme Q in mice treated with 1-methyl-4-phenyl-l,2,3,6-
oxidation. Biochim. Biophys. Acta. 963:558-561; 1988. tetrahydropyridine and diethyldithiocarbamate. Eur. J. Phar-
23. Sigel, H.; Prijs, B.; McCormick, D. B.; Shih, J. C. H. Stability macol. 266:291-300; 1994.
of binary and ternary complexes of a-lipoate and lipoate deriv- 4lb. Kagan, V.; Serbinova, E.; Packer, L. Antioxidant effects of
atives with Mn 2+, Cu 2+, and Zn 2+ in solution. Arch. Biochem. ubiquinones in microsomes and mitochondria are mediated by
Biophys. 187:208-214; 1978. tocopherol recycling. Bitch. Biophys. Res. Comm. 169:851-
24. Ou, P.; Tritschler, H. J.; Wolff, S. P. Thioctic (Lipoic) acid: 857; 1990.
A therapeutic metal-chelating antioxidant? Biochem. Pharma- 42. Podda, M.; Tritschler, H. J.; Ulrich, H.; Packer, L. ot-Lipoic
col. in press; 1994. acid supplementation prevents symptoms of vitamin E defi-
25. Searls, R. L.; Sanadi, D. R. ot-Ketoglutaric dehydrogenase. 8. ciency. Biochem. Biophys. Res. Comms. submitted; 1994.
Isolation and some properties of a flavoprotein component. J. 43. Busse, E.; Zimmer, G.; -gchopohl, B.; Komhuber, B. Influence
Biol. Chem. 235:2485-2491; 1960. of alpha-lipoic acid on intracellular glutathione in vitro and in
26. Scott, I.; Duncan, W.; Ekstrand, V. Purification and properties vivo. Arzneimittel-Forschung 42:829- 831; 1992.
of glutathione reductase of human erythrocytes. J. Biol. Chem. 44. Halliwell, B.; Gutteridge, J. M. C., eds. Free radicals in biology
238:3928-3939; 1963. and medicine. Oxford: Clarendon Press; 1989.
27. Jocelyn, P. C. The standard redox potential of cysteine-cystine 44a. Thomas, J. A.; Cha;., Y. C.; Jung, C. H. Protein S-thiolation
from the thiol-disulphide exchange reaction with glutathione and dethiolation. Meth. Enzymol. 233:385-395; 1994.
and lipoic acid. Eur. J. Biochem. 2:327-331; 1967. 45. Romero, F. J.; Ordonez, I.; Arduini, A.; Cadenas, E. The reac-
28. Suzuki, Y. J.; Tsuchiya, M.; Packer, L. Antioxidant activities tivity of thiols and disulfides with different redox states of
of dihydrolipoic acid and its structural homologues. Free Rad. myoglobin: Redox and addition reactions and formation of
Res. Comms. 18:115-122; 1993. thiyl radical intermediates. J. Biol. Chem. 267:1680-1688;
29. Haenen, G. R.; de Rooij, B. M.; Vermeulen, N. P. E.; Bast, 1992.
A. Mechanism of the reaction of ebselen with endogenous 46. Hatala, M. A.; DiPippo, V. A.; Powers, C. A. Biological thiols
thiols: Dihydrolipoate is a better cofactor than glutathione in elicit prolactin proteolysis by glandular kallikrein and permit
the peroxidase activity of ebselen. Mol. Pharmacol. 37:412- regulation by biochemical pathways linked to redox control.
422; 1990. Biochemistry 30:7666-7672; 199 I.
30. Bonomi, F.; Werth, M. T.; Kurtz, Jr., D. M. Assembly of 47. Gleason, F. K.; Holmgren, A. Thioredoxin and related protein
[FenSn(SR)4]2- (n = 2,4) in aqueous media from irons, salts, in procaryotes. FEMS Microbiol. Rev. 54:271-297; 1988.
thiols, and sulfur, sulfide, or thiosulfate plus rhodanese. Inorg. 48. Holmgren, A. Thioredoxin catalyzes the reduction of insulin
Chem. 24:4331-4335; 1985. disulfides by dithiothreitol and dihydrolipoamide. J. Biol.
31. Bonomi, F.; Pagani, S.; Cariati, F.; Pozzi, A.; Crisponi, G.; Chem. 254:9627-9632; 1979.
Cristiani, F.; Nurchi, V.; Russo, U.; Zanoni, R. Synthesis and 49. Holmgren, A. Thioredoxin and glutaredoxin systems. J. Biol.
characterization of iron derivatives of dihydrolipoic acid and Chem. 264:13963-13966; 1989.
dihydrolipoamide. Inorgan. Chim. Acta. 195:109-115; 1992. 50. Spector, A.; Huang, R.-R. C.; Yan, G.-Z.; Wang, R.-R. Thiore-
32. Bonomi, F.; Pagani, S. Removal of ferritin-bound iron by DL- doxin fragment 31-36 is reduced by dihydrolipoamide and
dihydrolipoate and DL-dihydrolipoamide. Eur. J. Biochem. reduces oxidized protein. Biochem. Biophys. Res. Commun.
155:295-300; 1986. 150:156-162; 1988.
33. Bonomi, F.; Cerioli, A.; Pagani, S. Molecular aspects of the 51. Hayashi, T.; Ueno, Y.; Okamoto, T. Oxidoreductive regulation
removal of ferritin-bound iron by DL-dihydrolipoate. Biochim. of nuclear factor kB: Involvement of a cellular reducing cata-
Biophys. Acta. 994:180-186; 1989. lyst thioredoxin. J. Biol. Chem. 268:11380-11388; 1993.
34. Reed, C. J.; Douglas, K. T. Single-strand cleavage of DNA by 52. Baeuerle, P. A. The inducible transcription activator NF-kB:
248 L. PACKEI~et al.

Regulation by distinct protein subunits. Biochim. Biophys. K. K. G. Lipoic acid and diabetes: Part III. Metabolic role of
Acta. 1072:63-80; 1991. acetyl dihydrolipoic acid. J. Biosci. 10:171-179; 1986.
53. Toledano, M. B.; Leonard, W. J. Modulation of transcription 73. Wagh, S. S.; Natraj, C. V.; Menon, K. K. G. Mode of action
factor NF-kB binding activity by oxidation-reduction in vitro. of lipoic acid in diabetes. J. Biosci. 11:59-74; 1987.
Proc. Natl. Acad. Sci. USA 88:4328-4332; 1991. 74. Faust, A.; Burkart, V.; Ulrich, H.; Weischer, C. H.; Kolb, H.
54. Molitor, J. A.; Ballard, D. W.; Greene, W. C. kB-Specific Effect of lipoic acid on cyclophosphamide-induced diabetes
DNA binding proteins are differentially inhibited by enhancer and insulitis in nonobese diabetic mice. Int. J. Immunophar-
mutations and biological oxidation. New Biol. 3:987-996; mac. 16:61-66; 1994.
1991. 75. Burkart, V.; Koike, T.; Brenner, H. H.; Imai, Y.; Kolb, H.
55. Matthews, J. R.; Wakasugi, N.; Virelizier, J.-L.; Yodoi, J.; Dihydrolipoic acid protects pancreatic islet cells from inflam-
Hay, R. T. Thioredoxin regulates the DNA binding activity of matory attack. Agents and Actions 38:60-65; 1993.
NF-kB by reduction of a disulphide bond involving cysteine 76. Haugaard, N.; Haugaard, E. S. Stimulation of glucose utiliza-
62. Nucleic Acid Res. 20:3821-3830; 1992. tion by thioctic acid in rat diaphragm incubated in vitro. Bio-
55a. Schenk, H., Klein, M., Erdbrugger, W.; Droge, W. Distinct chim. Biophys. Acta 222:583-586; 1970.
effects of thioredoxin and antioxidant on the activation of tran- 77. Singh, H. P. P.; Bowman, R. H. Effect of D,L-alpha lipoic acid
scription factors NF-kB and AP-1. Proc. Natl. Acad. ScL USA on the citrate concentration and phosphofructokinase activity of
91:1672-1676; 1994. peffused hearts from normal and diabetic rats. Biochem. Bio-
55b. Hayashi, T.; Ueno, Y.; Okamoto, T. Oxidoreductive regulation phys. Res. Commun. 41:555-561 ; 1970.
of nuclear factor kappa B: Involvement of a cellular reducing 78. Bashan, N.; Burdett, E.; Guma, A.; Klip, A. Effect of thioctic
catalyst thioredoxin. J. Biol. Chem. 268:11380 - 11308; 1993. acid on glucose transport. In: Giles, F. A.; Wessel, K., eds. The
56. Suzuki, Y. J.; Aggarwal, B. B.; Packer, L. ot-Lipoic acid is a role of antioxidants in diabetes mellitus. PMI-Verl.-Gruppe;
potent inhibitor of NF-kB activation in human T cells. Bio- 1993:221-229.
chem. Biophys. Res. Commun. 189:1709-1715; 1992. 79. Henriksen, E. J.; Jacob, S.; Tritschler, H.; Wellel, K; Augustin,
57. Suzuki, Y. J.; Ulrich, H.; Packer, L. Redox regulation of NF-kB H. J.; Dietze, G. J. Chronic thioctic acid treatment increases
DNA binding activity by dihydrolipoate and tr-lipoate. Arch. insulin-stimulated glucose transport activity in skeletal muscle
Biochem. Biophys. submitted; (1994). of obese Zucker rats. Diabetes. SuppL 1:122A abstr.; 1994.
58. Mizuno, M.; Packer, L. Effects of t~-Iipoic acid and dihydroli- 80. Jacob, S.; Henriksen, E. J.; Tritschler, H. J.; Clancy, D. E.;
poic acid on expression of the proto-oncogene c-fos. Biochem. Simon, I.; Schiemann, A. L.; Ulrich, H.; Jung, I.; Dietze,
Biophys. Res. Comm. 220:1136-1142; 1994. G. J.; Augustin, H. J. Thioctic acid enhances glucose-disposal
59. Fischer, S. M.; Adams, L. M. Suppression of tumor promoter- in patients with Type 2 diabetes. Horm Metab. submitted.;
induced chemiluminescence in mouse epidermal ceils by sev- 1994.
eral inhibitors of arachidonic acid metabolism. Cancer Res. 81. Henriksen, E. J.; Hoiloszy, J. O. Effects of phenylarsine oxide
45:3130-3136; 1985. on stimulation of glucose transport in rat skeletal muscle. Am.
60. Bonser, R. W.; Dawson, J.; Thompson, N. T.; Hodson, H. F.; J. Physiol. 258:C648-C653; 1990.
Garland, L. G. Inhibition of phorbol ester stimulated superox- 82. Renold, A. E.; Mintz, D. H.; Muller, W. A.; Cahili, Jr.,
ide production by 1-oleoyl-2-acetyl-sn-gtycerol (OAG); fact or G. F. Diabetes mellitus. In: Stanbury, J. B.; Wyngaarden,
artefact? FEBS Letters 209:134-138; 1986. J. B.; Fredrickson, D. S., eds. The metabolic basis o f inherited
61. Shibanuma, M.; Kuroki, T.; Nose, K. Effects of the protein diseases. New York: McGraw-Hill Book Co.; 1978:80-109.
kinase C inhibitor H-7 and calmodulin antagonist W-7 on su- 83. Baynes, J. W. Role of oxidative stress in development of com-
peroxide production in growing and resting human histiocytic plications in diabetes. Diabetes 40:405-412; 1991.
leukemia cells (U937). Biochem. Biophys. Res. Comm. 84. Brownlee, M.; Cerami, A.; Vlassara, H. Advanced glycosyla-
144:1317-1323; 1987. tion end products in tissue and the diabetic basis of diabetic
62. Semba, M.; Inui, N.; Yamada, M. A. Inhibitory effects of complications. N. Engl. J. Med. 318:1315-1321; 1988.
antipromoters and radical scavengers on the promotion process 85. Suzuki, Y. J.; Tsuchiya, M.; Packer, L. Lipoate prevents glu-
in two-stage cell transformation. Tohoku J. Exptl. Med. cose-induced protein modifications. Free Rad. Res. Comms.
168:133-136; 1992. 17:211-217; 1992.
63. Packer, L. New horizons in vitamin E research-the vitamin E 86. Kawabata, T.; Packer, L. Alpha-lipoate can protect against
cycle, biochemistry and clinical applications. In: Ong, glycation of serum albumin, but not low density lipoprotein.
A. S. H.; Packer, L., eds. Lipid-soluble antioxidants: Biochem- Biochem. Biophys. Res. Comms. 203:99-104; 1994.
istry and clinical applications. Boston: Birkhanser Verlag; 87. Keita, Y.; Kratzer, W.; Worner, W.; Rietbrock, N. Effect of
1992a: 1- 16. free fatty acids on the binding kinetics at the benzodiazepine
64. Harrison, E. H.; McCormick, D. B. The metabolism ofdl-[l,6- binding site of glycated serum albumin. Int. J. Clin. Pharmacol.
J4C] lipoic acid in the rat. Arch. Biochem. Biophys. 160:514- Ther. Toxicol. 31:337-342; 1993.
522; 1974. 88. van Boekel, M. A.; van den Bergh, P. J.; Hoenders, H. J.
65. Peinado, J.; Sies, H.; Akerboom, T. P. M. Hepatic lipoate Glycation of human serum albumin: Inhibition by Diclofenac.
uptake. Arch. Biochem. Biophys. 273:389-395; 1989. Biochim. Biophys. Acta. 1120:201-204; 1992.
66. Teichert, J.; Preiss, R. HPLC-methods for determination of 89. Lyons, T. Lipoprotein glycation and its metabolic conse-
lipoic acid and its reduced form in human plasma. Int. J. Clin. quences. Diabetes 41, Suppl. 2:67-73; 1992.
Pharmacol., Therapy, and Toxicol. 30:511-512; 1992. 90. Schepkin, V.; Kawabata, T.; Packer, L. NMR study of lipoic
67. Spence, J. T.; McCormick, D. B. Lipoic acid metabolism in acid binding to bovine serum albumin. Biochem. Mol. Biol.
the rat. Arch. Biochem. Biophys. 174:13-19; 1976. Int. 33:879-886; 1994.
68. Wolff, S. P.; Jiang, Z.-Y.; Hunt, J. V. Protein glycation and 91. Kagan, V.; Freisleben, H.-J.; Tsuchiya, M.; Forte, T.; Packer,
oxidative stress in diabetes mellitus and ageing. Free Rad. L. Generation of probucol radicals and their reduction by ascot-
Biol. Med. 10:339-352; 1991. bate and dihydrolipoic acid in human low density lipoproteins.
69. Kuklinski, B.; Pietschmann, A. Oxidativer Strel3 und Alteru. Free Rad. Res. Comms. 15:265-276; 1991.
Z. Geriatrie. 4:224-226; 1991. 92. Low, P. A.; Nickander, K. K. Oxygen free radical effects in
70. Natraj, C. V.; Gandhi, V. M.; Menon, K. K. G. Lipoic acid sciatic nerve in experimental diabetes. Diabetes 40:873-877;
and diabetes: Effect of dihydrolipoic acid administration in 1991.
diabetic rats and rabbits. J. Biosci. 6:37-46; 1984. 93. Bravenboer, B.; Kappelle, A. C.; Hamers, F. P. T.; van Buren,
71. Gandhi, V. M.; Wagh, S. S.; Natraj, C. V.; Menon, K. K. G. T.; Erkelens, D. W.; Gispen, W. H. Potential use of glutathione
Lipoic acid and diabetes II: Mode of action of lipoic acid. J. for the prevention and treatment of diabetic neuropathy in the
Biosci. 9:117-127; 1985. streptozotocin-induced diabetic rat. Diabetologia 35:813- 817;
72. Wagh, S. S.; Gandhi, V. M.; Natraj, C. V.; Menon, 1992.
Antioxidant properties of lipoic acid 249

94. Dimpfel, W.; Spuler, M.; Pierau, F.-K.; Ulrich, H. Thioctic Treatment of alcohol-related liver disease with thioctic acid: A
acid induces dose-dependent sprouting of neurites in cultured six month randomised double-blind trial. Gut 23:1088-1093;
rat neuroblastoma cells. Dev. Pharmacol. Ther 14:193-199; 1982.
1990. 115. Legrand-Poels, S.; Vaira, D.; Pincemail, J.; van de Vorst, A.;
95. Sachse, G.; Willms, B. Efficacy of thioctic acid in the therapy Piette, J. Activation of human immunodeficiency virus type 1
of peripheral diabetic neuropathy. In: Gries, F. A.; Freund, by oxidative stress. Aids Res. Hum. Retrovir. 6:1389-1397;
H. J.; Rabe, F.; Berger, H., eds. Aspects of autonomic neuropa- 1990.
thy in diabetes. Horm. Metab. Res. Suppl. Series 9. 1980:105- 116. Freed, B. M.; Rapoport, R.; Lempert, N. Inhibition of early
108. events in the human T-lymphocyte response to mitogens and
96. Ziegler, D.; Mayer, P.; Muhlen, H.; Gries, F. A. Effeckte einer alloantigens by hydrogen peroxide. Arch. Surg. 122:99-104;
Therapie mit ot-Liponsanre gegentiber Vitamin B 1 bei der dia- 1987.
betischen Neuropathie. Diab. Stoffw. 2:443-448; 1993. 117. Aune, T. M.; Pierce, C. W. Conversion of soluble immune
97. K~hler, W.; Kuklinski, B.; Ruhlmann, C.; Plotz, C. Diabetes response suppressor to
mellitus-a free radical-associated disease: Effects of adjuvant 118. Cerutti, P. A. Prooxidant states and tumor promotion. Science
supplementation of antioxidants. In: Gries, F. A.; Wessel, K., 227:375-81 ; 1985.
eds. The role of anti-oxidants in diabetes mellitus: Oxygen 119. Buhl, R.; Jaffe, H. A.; Holroyd, K. J.; Wells, F. B.; Mastrangeli,
radicals and anti-oxidants in diabetes. Frankfurt am Main: pmi A.; Saltini, C.; Cantin, A. M.; Crystal, R. G. Glutathione defi-
Verl.-Gruppe; 1993:33-53. ciency and HIV. Lancet 335:546; 1990.
98. Maitra, I.; Serbinova, E.; Tritschler, H.; Packer, L. a-Lipooic 120. Dworkin, B. M.; Wormser, G. P.; Axelrod, F.; Pierre, N.;
acid prevents buthionine sulfoximine-induced cataract forma- Schwarz, E.; Schwartz, E.; Seaton, T. Dietary intake in patients
tion in newborn rats. Free Rad. Biol. Med. in press; 1994. with acquired immunodeficiency syndrome (AIDS), patients
99. Kilic, F.; Packer, L.; Trevethick, J. R. Modeling cortical cata- with AIDS-related complex, and serologically positive human
ractogenesis 17: In vitro effect of o~-lipoic acid on glucose- immunodeficiency virus patients: Correlations with nutritional
induced lens membrane damage, a model of diabetic cataracto- status. Jpen. J. Parent Ent. Nutr. 14:605-609; 1990.
genesis. Exptl. Eye Res. submitted; 1994. 121, Bohl, R.; Jaffe, H. A.; Holroyd, K. J.; Wells, F. B.; Mastranseli,
100. Packer, L. The role of anti-oxidative treatment of diabetes A.; Saltini, C.; Cantin, A. M.; Crystal, R. G. Systemic glutathi-
mellitus. Diabetologia. 36:1212-1213; 1993. one deficiency in symptom-free HIV-seropositive individuals.
101. Scheer, B.; Zimmer, G. Dihydrolipoic acid prevents hypoxic/ Lancet 2:1294-1298; 1989.
reoxygenation and peroxidative damage in rat heart mitochon- 122. Fuchs, J.; Schofer, H.; Milbradt, R.; Freisleben, H. J.; Buhl,
dria. Arch. Biochem. Biophys. 302:385-390: 1993. R.; Siems, W.; Grune, T. Studies on iipoate effects on blood
102. Serbinova, E.; Khwaja, S.; Reznick, A. Z.; Packer, L. Thioctic redox state in human immunodeficiency virus infected patients.
acid protects against ischemia-reperfusion injury in the isolated Arzneimittel-Forschung 43:1359-1362; 1993.
perfused Langendorff heart. Free Rad. Res. Comms. 17:49- 123. Banr, A.; Harrer, T.; Peukert, M.; Jahn, G.; Kalden, J. R.;
58; 1992. Fleckenstein, B. Alpha-lipoic acid is an effective inhibitor of
103. Haramaki, N.; Packer, L.; Assadnazari, H.; Zimmer, G. Cardiac human immuno-deficiency virus (HIV-1) replication. Klin.
recovery during postischemic reperfusion is improved by com- Wochenschr. 69:722-724; 1991.
bination of vitamin E with dihydrolipoic acid. Biochem. Bio- 124. Boveris, A.; Chance, B. The mitochondrial generation of hydro-
phys. Res. Commun. 196:1101 - 1107; 1993. gen peroxide: General properties and the effect of hyperbaric
104. Assadnazari, H.; Zimmer, G.; Freisleben, H. J.; Werk, W.; oxygen. Bioehem. J. 134:707-716; 1973.
Leibfritz, D. Cardioprotective efficiency of dihydrolipoic acid 125. Nohl, H.; Jordan, W. The mitochondrial site of superoxide
in working rat hearts during hypoxia and reoxygenation. 31p formation. Biochem. Biophys. Res. Commun. 138:533-539;
nuclear magnetic resonance investigations. Arzneimittel- 1986.
Forschung 43:425-432; 1993. 126. Richter, C.; Park, J. W.; Ames, B. N. Normal oxidative damage
105. Prehn, J. H.; Peruche, B.; Karkoutly, C.; Rogberg, C.; Mennel, to mitochondrial and nuclear DNA is extensive. Proc. Natl.
H. D.; Krieglstein, J. Dihydrolipoic acid protects neurons Acad. Sci. USA 85:6465-6467; 1988.
against ischemic/hypoxic damage. In: Krieglstein, J.; Ober- 127. Takeshige, K.; Minakami, S. NADH- and NADPH-dependent
pichler, H., eds. Pharmacology of cerebral ischemia. Stuttgart: formation of superoxide amines by bovine heart submitochon-
Wissenschaftliche Verlagsgesellscheaft mbH; 1990:357-362. drial particles. Biochem. J. 180:129-135; 1979.
106. Prehn, J. H.; Karkoutly, C.; Nuglisch, J.; Peruche, B.; 128. Zaccarato, F.; Cavallini, L.; Deana, R.; Alexandre, R. Pathways
Krieglstein, J. Dihydrolipoate reduces neuronal injury after ce- of hydrogen-peroxide generation in guinea-pig cerebral cortex
rebral ischemia. J. Cereb. Blood Flow Metab. 12:78-87; 1992. mitochondria. Biochem. Biophys. Res. Commun. 154:727-734;
107. Boveris, A.; Roldan, E. J. A.; Alvarez, S. Effect of lipoic acid 1988.
on short-term ischemia-reperfusion in rat intestine. In: Packer, 129. Saggu, H.; Cooksey, J.; Dexter, D.; Wells, F. R.; Lees, A.;
L.; Cadenas, E., eds. Biological oxidants" and antioxidants. Jenner, P.; Marsden, C. D. A selective increase in particulate
Stuttgart: Hippokrates Verlag; 1994:77-85. superoxide dismutase activity in parkinsonian substantia nigra.
108. Teutsch, C.; Brennan, R. W. Amanita mushroom poisoning J. Neurochem. 53:692-697; 1989.
with recovery from coma: A case report. Ann. Neurol. 3" 177- 130. Dexter, D. T.; Carayon, A; Javoy-Agid, F.; Agid, Y.; Wells,
179; 1978. F. R.; Daniel, S. E.; Lees, A. J.; Jenner, P.; Marsden, C. D.
109. Plotzker, R.; Jensen, D. M.; Payne, J. A. Case report: Amanita Alterations in the levels of iron, ferritin and other trace metals
virosa acute hepatic necrosis: Treatment with thioctic acid. in Parkinson's disease and other neurodegenerative diseases
Am. J. Meal. Sci. 283:79-82; 1982. affecting the basal ganglia. Brain 114:1953-1975; 1991.
110. Vesconi, S.; Langer, M.; Iapichino, G.; Costantino, D.; Busi, 131. Jenner, P.; Dexter, D. T.; Sian, J.; Schapira, A. H. V.; Marsden,
C.; Fuime, L. Therapy of cytotoxic mushroom intoxication. C. D. Oxidative stress as a cause of nigral cell death in Parkin-
Crit. Care Med. 13:402-406; 1985. son's disease and incidental Levy body disease. Ann. Neurol.
111. Bani, G. Sperimentazione clinica di un complesso coenzymatico 32:$87-$97; 1992.
in epatopazienti. CI. Terap. 52:267-280; 1970. 132. Shoffner, J. M.; Watts, R. L.; Juncos, J. L.; Torroni, A.; Wal-
112. Colombi. A.; ThOlen, H.; Huber, F. Einfluss yon Conenzym lace, D. C. Mitochondrial oxidative phosphorylation defects in
A, Nicotinamid-adenin-dinucleotid (NAD), ot-Liponsbure und Parkinson's disease. Ann. Neurol. 30:332-339; 1991.
Cocarboxylase auf die akute Hepatits (Doppelblindversuch). 133. Volicer, L.; Crino, P. B. Involvement of free radicals in demen-
Int. J. Clin. Pharmacol. 2:133-138; 1969. tia of the Alzheimer typer: A hypothesis. Neurobiol. Aging.
I 13. Mtiller, E.; Schmitt, R. Ein Beitrag zur Behandlung chronischer 11:567-571 ; 1990.
Lebererkrankungen. Med. Kiln. 71:1831-1835; 1976. 134. Wallace, D. C.; Singh, G.; Lott, M. T.; Hodge, J. A.; Schurr,
114. Marshall, A. W.; Graul, R. S.; Morgan, M. Y.; Sherlock, S. T. G.; Lezza, A. M. S.; Elsas, L. J. Mitochondrial DNA muta-
250 L. PACKER et al.

tion associated with Leber's hereditary optic neuropathy. Sci- 144. Cross, C. E.; O'Neill, C. A.; Reznick, A. Z.; Hu, M.; Packer,
ence 242:1427-1430; 1988. L.; Frei, B. Cigarette smoke oxidation of human plasma constit-
135. Stoll, S.; Hartmann, H.; Cohen, S. A.; MUller, W. E. The potent uents. In: Daina, J. N.; Pryor, W. A., eds. Tobacco, smoking
free radical scavenger a-lipoic acid improves memory in aged and nutrition: Influence o f nutrition on tobacco associated
mice: Putative relationship to NMDA receptor deficits. Phar- health risks. Vol. 686. Annals N Y Acad. Sci. 1993:72-90.
macol. Biochem. Behav. 46:799-805; 1993. 145. Reznick, A. Z.; Cross, C. E.; Hu, M.-L., Suzuki, Y. J.; Kwaja,
136. Greenamyre, J. T.; Garcia-Osuna, M.; Greene, J. G. The endog- S.; Safadi, A.; Motchnik, P. A.; Packer, L.; Halliwell, B. Modi-
enous cofactors, thioctic acid and dihydrolipoic acid, are neuro- fication of plasma proteins by cigartette smoke as measured by
protective against NMDA and malonic acid lesions of striatum. protein carbonyl formation. Biochem. J. 286:607-611; 1992.
Neurosci. Lett. 171:17-20; 1994. 146. Tsuchiya, M.; Thompson, D. F.; Suzuki, Y. J.; Cross, C. E.;
137. Choi, D. W. Glutamate neurotoxicity and diseases of the ner- Packer, L. Superoxide formed from cigarette smoke impairs
vous system. Neuron 1:623-634; 1988. polymorphonuclear leukocyte active oxygen generation activ-
138. Altenkirch, H.; Stoltenburg-Didinger, H.; Wagner, M.; Her- ity. Arch. Biochem. Biophys. 299:30-37; 1992.
mann, J.; Walter, G. Effects of lipoic acid in hexacarbon- 147. Grunert, R. R. The effect of DL-a-lipoic acid on heavy-metal
intoxication in mice and dogs. Arch. Biochem. Biophys.
induced neuropathy. Neurotoxicol. Teratol. 12:619-622; 1990.
86:190-194; 1960.
139. Ramakrishnan, N.; Wolfe, W. W.; Catravas, G. N. Radioprotec- 148. Sumathi, R.; Devi, V. K.; Varalakshmi, P. DL o~-lipoic acid
tion of hematopoietic tissues in mice by lipoic acid. Radiation
protection against cadmium-induced tissue lipid peroxidation.
Res. 130:360-365; 1992. Med. Sci. Res. 22:23-25; 1994.
140. Korkina, L. G.; Afanas'ef, I. B.; Diplock, A. T. Antioxidant 149. Gregus, Z.; Stein, A. F.; Varga, F.; Klaassen, C. D. Effect of
therapy in children affected by irradiation from the Chernobyl lipoic acid on biliary excretion of glutathione and metals. Tox-
nuclear accident. Bioch. Soc. Trans. 21:314S; 1993. icol. Appl. PharmacoL 114:88-96; 1992.
141. Pryor, W. A. Cigarette smoke and the involvement of free 150. Cardinale, A.; De Simone, G. F. The use of sulphydryl con-
radical reactions in chemical carcinogenesis. Br. J. Cancer. taining drugs and chelating agents to reduce renal irradiation
Suppl. 85:19-23; 1987. during 203Hg chlormerodrin scintigraphy. J. Nucl. Biol. Med.
142. Church, D. F.; Pryor, W. A. Free-radical chemistry of cigarette 12:121-127; 1968.
smoke and its toxicological implications. Environ. Health Per- 151. Carpintero, D. J. Uso del acido tioctico para la prevencion
spect. 64:111-126; 1985. de los efectos secondarios provacodos por el benznidazol en
143. Frei, B.; Forte, T. M.; Ames, B. N.; Cross, C. E. Gas phase pacientes con infeccion de Chaga chronica. Medicina (Buenos
oxidants of cigarette smoke induce lipid peroxidation and Aires) 43:285-290; 1983.
changes in lipoprotein properties in human blood plasma: Pro- 152. Asta Medica, a-Lipoic acid prescribing information.
tective effects of ascorbic acid. Biochem. J. 277:133-138; 153. Gal, E. M. Reversal of selective toxicity of (-)-ct-lipoic acid by
1991. thiamine in thiamine-deficient rats. Nature 207:535; 1965.

Vous aimerez peut-être aussi