ArticlePDF Available

Efficient Reduction of Lipoamide and Lipoic Acid by Mammalian Thioredoxin Reductase

Authors:

Abstract and Figures

Reduction of the antioxidant lipoic acid has been proposed to be catalyzed in vivo by lipoamide dehydrogenase (LipDH) or glutathione reductase (GR). We have found that thioredoxin reductase (TR) from calf thymus, calf liver, human placenta, and rat liver efficiently reduced both lipoic acid and lipoamide with Michaelis-Menten type kinetics in NADPH-dependent reactions. In contrast to LipDH, lipoic acid was reduced almost as efficiently as lipoamide. Under equivalent conditions at 20 degrees C, pH 8.0, mammalian TR reduced lipoic acid by NADPH 15 times more efficiently than the corresponding NADH dependent reduction catalyzed by LipDH (297 min-1 for TR vs. 20.3 min-1 for LipDH). Moreover, TR was 2.5 times faster in reducing lipoic acid with NADPH than in catalyzing the reverse reaction (oxidation of dihydrolipoic acid with NADP+). In contrast, LipDH was only 0.048 times as efficient in the forward reaction as compared to the reverse reaction (using NADH and NAD+). We conclude that all or part of the previously described NADPH-dependent lipoamide dehydrogenase (diaphorase) activities in mammalian systems should be attributed to TR. Our results suggest that in mammalian cells a significant part of the therapeutically important reduction of lipoic acid is catalyzed by thioredoxin reductase.
Content may be subject to copyright.
BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
225, 268274 (1996)
ARTICLE NO
. 1165
Efficient Reduction of Lipoamide and Lipoic Acid
by Mammalian Thioredoxin Reductase
Elias S. J. Arne
´r,
1
Jonas Nordberg, and Arne Holmgren
Medical Nobel Institute for Biochemistry I, Department of Medical Biochemistry and Biophysics,
Karolinska Institutet, S-171 77 Stockholm, Sweden
Received June 27, 1996
Reduction of the antioxidant lipoic acid has been proposed to be catalyzed in vivo by lipoamide dehydroge-
nase (LipDH) or glutathione reductase (GR). We have found that thioredoxin reductase (TR) from calf
thymus, calf liver, human placenta, and rat liver efficiently reduced both lipoic acid and lipoamide with
Michaelis-Menten type kinetics in NADPH-dependent reactions. In contrast to LipDH, lipoic acid was
reduced almost as efficiently as lipoamide. Under equivalent conditions at 20
7
C, pH 8.0, mammalian TR
reduced lipoic acid by NADPH 15 times more efficiently than the corresponding NADH dependent reduction
catalyzed by LipDH (297 min
01
for TR vs. 20.3 min
01
for LipDH). Moreover, TR was 2.5 times faster in
reducing lipoic acid with NADPH than in catalyzing the reverse reaction (oxidation of dihydrolipoic acid
with NADP
/
). In contrast, LipDH was only 0.048 times as efficient in the forward reaction as compared
to the reverse reaction (using NADH and NAD
/
). We conclude that all or part of the previously described
NADPH-dependent lipoamide dehydrogenase (diaphorase) activities in mammalian systems should be attrib-
uted to TR. Our results suggest that in mammalian cells a significant part of the therapeutically important
reduction of lipoic acid is catalyzed by thioredoxin reductase.
q
1996 Academic Press, Inc.
Thioredoxin reductase (TR), lipoamide dehydrogenase (LipDH) and glutathione reductase
(GR) belong to a family of pyridine nucleotide-disulfide oxidoreductases containing FAD and
a redox active disulfide (1). The two latter enzymes are highly similar homo-dimeric enzymes
with 50 kDa subunits conserved between all species. In contrast, the properties of thioredoxin
reductase (EC 1.6.4.5) differs between Escherichia coli and mammalian cells (2). The reaction
catalyzed by TR from all species is:
NADPH
/
H
/
/
Trx-S
2
S
NADP
/
/
Trx-(SH)
2
where Trx-S
2
is oxidized thioredoxin and Trx-(SH)
2
reduced thioredoxin. Thioredoxin (12
kDa) in reduced form is an efficient protein disulfide reductase with numerous functions,
including function as hydrogen donor of ribonucleotide reductase, regulation of chroloplast
enzyme activities, redox control of transcription factors and cytokine effects on normal and
malignant cells. For reviews on thioredoxin, see (3-5).
Thioredoxin reductase from E. coli with a subunit M
r
of 35 kDa has been studied in depth,
with cloning, sequencing and a high resolution X-ray structure of the enzyme (6, 7). The three-
dimensional structure including the amino acid sequence around the redox active disulfide in
1
Corresponding author. Fax: 46-8-31 15 51. E-mail: Elias.Arner@mbb.ki.se.
Abbreviations used are: TR, thioredoxin reductase; LipDH, lipoamide dehydrogenase; GR, glutathione reductase;
Trx, thioredoxin; Trx-S
2
, oxidized Trx; Trx-(SH)
2
, reduced Trx; DTNB, 5,5
*
-dithiobis-(2-nitrobenzoic acid); lipoamide,
DL-6,8-thioctic acid amide; lipoic acid, oxidized DL-6,8-thioctic acid; dihydrolipoic acid, reduced DL-6,8-thioctic
acid; TE buffer, 50 mM Tris-Cl, 2 mM EDTA, pH 8.0.
0006-291X/96 $18.00
Copyright
q
1996 by Academic Press, Inc.
All rights of reproduction in any form reserved.
268
AID BBRC 5144 / 6905$$$861
07-17-96 07:41:18 bbrcas AP: BBRC
Vol. 225, No. 1, 1996 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
E. coli TR is strikingly different from the corresponding consensus structures of GR and
LipDH and it was concluded that the disulfide reductase activities of E coli TR and GR must
have evolved convergently (7). Mammalian TR was originally purified from calf liver and
thymus (8), and to homogeneity from rat liver (9). The rat liver enzyme showed subunits with
M
r
of 58,000 and the human placenta enzyme had similar properties with even higher subunit
M
r
(10). Mammalian TR, in contrast to the E. coli enzyme, shows a broad substrate specificity
and reduces not only thioredoxins from many species (8) but also several low molecular weight
substrates including 5,5
*
-dithiobis(2-nitrobenzoic acid) (DTNB), vitamin K, selenodiglutathi-
one, selenite, lipid hydroperoxides, and alloxan (reviewed in (4). Inhibitors of mammalian TR
include antitumour quinones (11, 12), nitrosoureas (13), 13-cis-retinoic acid (14), and 1-chloro-
2,4-dinitrobenzene (DNCB) (15). DNCB also induces NADPH oxidase activity after alkylation
of mammalian TR (15), which resembles the NADPH oxidase (or diaphorase) activity of both
glutathione reductase (16) and lipoamide dehydrogenase (1).
We have sequenced a large number of internal peptides of bovine TR and cloned and
sequenced the rat enzyme (manuscript submitted) which largely confirms the cDNA sequence
of a putative human TR recently reported by Powis and coworkers (17). The sequences revealed
a close structural relationship between mammalian TR and GR or LipDH. We have therefore
reinvestigated the activity of mammalian TR with the principal substrates for GR and LipDH,
and found that lipoamide and lipoic acid were efficient substrates for the enzyme.
Lipoic acid bound in amide linkage to transacetylase or transsuccinylase is an essential
cofactor in the mitochondrial oxoacid dehydrogenase complexes (1, 18, 19). Recently lipoic
acid has gained interest as an antioxidant, with potential therapeutic applications in a range
of conditions, including ischemia-reperfusion injuries, cataract formation, HIV activation, neu-
rodegeneration and radiation injury (19).
Both lipoic acid and dihydrolipoic acid are present extracellularly and human plasma levels
have been reported to be 1-25 ng/ml and 33-145 ng/ml, respectively (20). Exogenously adminis-
tered lipoic acid is rapidly and to a high extent reduced to dihydrolipoic acid by mammalian
cells (19, 21, 22). LipDH cannot be the sole enzyme catalyzing this reduction, since cells
which lack LipDH, like erythrocytes, still reduce lipoic acid to dihydrolipoic acid by an
NADPH dependent reaction (22). Glutathione reductase is an NADPH dependent lipoic acid
reducing enzyme, but the efficiency in this reaction was very low compared to reduction of
GSSG (22, 23).
MATERIALS AND METHODS
Material. 2
*
,5
*
-ADP-Sepharose and Q Sepharose were from Pharmacia Inc. Recombinant human Trx was prepared
as described (24) while E. coli Trx came from IMCO (Sweden). Porcine LipDH was purchased from Sigma, as was
lipoamide (oxidized DL-6,8-thioctic acid amide), lipoic acid (oxidized DL-6,8-thioctic acid) and dihydrolipoic acid
(reduced DL-6,8-thioctic acid). Dihydrolipoic acid was used within two hours upon opening of the sealed ampule.
All other chemicals used were of analytical grade or better. TR from bovine liver and thymus, rat liver or human
placenta was purified based upon the method described earlier (9). The purifications typically resulted in 2.5 mg of
TR from 1 kg of tissue, homogenous as judged by SDS PAGE and with a specific activity in DTNB reduction (8)
of more than 1000 A
412
/min/mg.
Ion exchange chromatography. Aliquots of TR (6
m
g in 200
m
l TE buffer) were loaded on a Mono Q PC 1.6/5
column (SMART system, Pharmacia Biotech Inc., Sweden) equilibrated with TE buffer. TE buffer was allowed to
run through until UV baselines were steady (approximately 5 min at 100
m
l/min) whereupon protein was eluted with
a linear NaCl gradient 0 -0.3 M in TE buffer in 20 min. Fractions of 100
m
l were continuously collected and stored
at
0
20
7
C until analyzed for enzyme activity. Protein content in each fraction was determined using the SMART
manager software for calculation of the integrated absorbance at 280 nm.
Enzyme activity determination. Enzyme activity was measured spectrophotometrically at 20
7
C in TE buffer as
described earlier (8, 9), with the following modifications. Screening of activity in fractions of the MonoQ chromatogra-
phy (Figure 2) was carried out in 96-well Cel-Cult plates (Sterilin Ltd., England) and consumption of NADPH was
measured through decrease of absorbance at 340 nm determined using the THERMOmax microplate reader (Molecular
269
AID BBRC 5144 / 6905$$$862
07-17-96 07:41:18 bbrcas AP: BBRC
Vol. 225, No. 1, 1996 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
Devices Corp., CA) with the kinetic application of the accompanying SOFTmax software. Samples (10
m
l) were
added to wells containing 150
m
l TE buffer with 200
m
M NADPH and substrate, i.e. either SeO
20
3
(200
m
M), lipoamide
(400
m
M) or insulin (100
m
M), the latter supplemented with either human (5
m
M) or E coli (5
m
M) Trx. In each case,
reference wells contained only TE buffer and 200
m
M NADPH. Reduction of DTNB (500
m
M) in the fractions was
performed in the same manner, but in the presence of 300
m
M NADPH and the reduction of DTNB determined by
increase of absorbance at 405 nm (instead of 412 nm). A well containing TE buffer, 300
m
M NADPH and 500
m
M
DTNB but no enzyme was used as reference.
When lipoamide and lipoic acid as substrates were further analyzed (Tables I and II, Figure 2), enzyme activity
was determined at 20
7
C using a Shimadzu double-beam spectrophotometer with semi-micro cuvettes containing 500
m
l TE buffer (pH 8.0) with 200
m
M pyridine nucleotide, enzyme and lipoamide or lipoic acid (0-2500
m
M), with
everything included except lipoamide or lipoic acid in the reference cuvette. In each case, 5
m
l of pure enzyme was
added, which gave final TR concentrations of 1.2 nM (human placenta TR), 6.9 nM (calf thymus TR), 12.4 nM (rat
liver TR) and 13.8 nM (calf liver TR), as determined in separate experiments by DTNB reduction calculating with
an M
r
for the holoenzyme of 116000 and assuming 1200 A
412
units/min/mg for the pure enzyme (4). Porcine LipDH
from Sigma (10.4 mg/ml) was diluted in TE buffer at the time for experiments and an M
r
of 100 000 was used in
calculations of concentrations (3
m
l of 100 times diluted LipDH in 500
m
l was considered to give 6.24 nM LipDH
in final concentration). Conversion of pyridine nucleotide was determined by decrease (oxidation of NAD(P)H) or
increase (reduction of NAD(P)
/
) of absorbance at 340 nm and using a molar extinction coefficient of 6200 M
01
cm
01
.
Lipoamide and lipoic acid were dissolved in EtOH as 50 mM stock solutions and EtOH was therefore added to an
equal final concentration of 4% in all cuvettes. In each case, enzyme and pyridine nucleotide was added first, pre-
incubated 5 min to reduce the enzyme, whereafter substrates were added and activity was measured.
RESULTS
At the onset of this study, we screened several of our preparations of purified mammalian
TR with the primary substrates of GR and LipDH. GSSG was confirmed not to be a substrate,
in agreement with previous results (9). However, we found a significant NADPH dependent
FIG. 1. Ion exchange chromatography of mammalian TR. Purified calf thymus TR was analyzed on a column of
MonoQ anion exchanger with an isocratic NaCl gradient as shown in A (
l
; protein profile, solid line; salt gradient).
B (inset) shows the enzymatic activity in those fractions containing protein, using DTNB (
l
, dashed line), insulin
(coupled to human Trx,
l
,orE coli Trx,
m
), selenite (
j
), or lipoamide (
l
) as substrates.
270
AID BBRC 5144 / 6905$$$862
07-17-96 07:41:18 bbrcas AP: BBRC
Vol. 225, No. 1, 1996 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
TABLE I
Kinetic Parameters of Mammalian Thioredoxin Reductase in
Reduction of Lipoamide, Reduction of Lipoic Acid,
and Oxidation of Dihydrolipoic Acid
a
K
m
k
cat
k
cat
/K
m
Source of TR (mM) (min
01
) (mM
01
min
01
)
Calf thymus 0.49 503 1035
Calf liver 1.00 918 913
Human placenta 0.51 1102 2182
Rat liver 0.86 1296 1498
Kinetic parameters of rat liver TR for
Substrate lipoic acid and dihydrolipoic acid
Lipoic acid 0.71 368 518
Dihydrolipoic acid
b
0.88 173 196
a
Consumption of pyridine nucleotide was determined by following
the change of absorbance at 340 nm using a molar extinction coefficient
of 6200 M
01
cm
01
. Assays were performed at 20
7
C,pH 8.0, in 50 mM
Tris, 2 mM EDTA, and 200
m
M NADPH with 315 nM TR (depending
on preparation) and 0–2500
m
M lipoamide or lipoic acid, as described
in further detail in Materials and Methods. The kinetic parameters given
in the table are those derived if Michaelis-Menten kinetics are fitted to
the experimental data. The fitted Michaelis-Menten plots in reduction
of lipamide and lipoic acid and oxidation of dihydrolipoic acid by rat
liver TR are shown in Fig. 2.
b
NADP
/
used instead of NADPH.
lipoamide reduction in all TR preparations that we analyzed. To exclude contamination of
lipoamide dehydrogenase, one of the pure calf thymus TR preparations (showing one single
distinct band on SDS gel) was further analyzed with MonoQ ion exchange chromatography.
It was clear that previously known TR-catalyzed reactions (Trx-dependent insulin reduction
as well as reduction of DTNB or selenite) co-eluted in one single peak together with reduction
of lipoamide (Figure 1). The same result was repeated with two additional TR preparations;
one from calf thymus and one from rat liver (data not shown). We also tested TR from E.
coli for lipoamide reduction, but this enzyme did not reduce lipoamide to any detectable level
(data not shown).
Kinetic parameters in the NADPH dependent lipoamide reduction by human, rat and calf
TR were then determined (Table I). For the rat liver TR preparation, that had the highest
specific activity, reduction of lipoic acid as well as the corresponding reverse reaction (NADP
/
coupled dihydrolipoic acid oxidation) was also analyzed (Table I). Also lipoic acid and dihy-
drolipoic acid were good substrates, with K
m
values in the same range as those for lipoamide
(Table I). In none of the reactions (reduction of lipoamide or lipoic acid with NADPH or
oxidation of dihydrolipoic acid with NADP
/
) could any cooperativity be observed and the
experimental data were easily fitted to classic Michaeli-Menten type kinetics (Figure 2).
To compare the activities of mammalian TR with those of mammalian LipDH, both
NADP(H) and NAD(H) were used as pyridine nucleotides under equivalent conditions (20
7
C,
50 mM Tris, 1 mM EDTA, pH 8.0, 200
m
M pyridine nucleotide and 1 mM lipoic acid
derivative) with both LipDH and TR. The results of these assays are given in Table II. The
reduction of lipoamide was ten times more efficient by LipDH using NADH than by TR using
271
AID BBRC 5144 / 6905$$$862
07-17-96 07:41:18 bbrcas AP: BBRC
Vol. 225, No. 1, 1996 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
FIG. 2. Reduction of lipoamide and lipoic acid and oxidation of dihydrolipoic acid by rat liver TR. TR from rat
liver (7.4 nM) was analyzed for reduction of lipoamide (
l
) or lipoic acid (
j
)at20
7
C, pH 8.0 in 50 mM Tris-Cl, 2
mM EDTA, and 200
m
M NADPH or oxidation of dihydrolipoic acid (
h
) using 200
m
M NADP
/
instead of NADPH.
The curves fitted to the data are Michaelis-Menten plots and the derived kinetic parameters are given in Table I.
NADPH (9215 min
01
vs. 889 min
01
). TR, on the other hand, was fifteen times more efficient
in reducing lipoic acid than was LipDH (297 min
01
vs. 20.3 min
01
). It was also clear that TR
was more efficient in the forward reaction, i.e. reducing lipoic acid, as compared to the reverse
reaction, i.e. catalyzing oxidation of dihydrolipoic acid (ratio
Å
2.49). The opposite, however,
was true for LipDH, that was 20 times more efficient catalyzing the reverse reaction (ratio
between forward and reverse reactions
Å
0.048).
DISCUSSION
Our results show that mammalian thioredoxin reductase efficiently catalyzes NADPH-depen-
dent lipoamide and lipoic acid reduction. This reduction of lipoamide and lipoic acid illustrates
the close relationship of the mammalian TR with other enzymes of the pyridine nucleotide
disulfide oxidoreductase family, such as LipDH and GR, which has interesting implications
since TR of the E. coli type appears not to have evolved from a common ancestral protein
(7). It should be noted that we did not see evidence of cooperativity in the reduction of
lipoamide or lipoic acid by TR, which is different from the pronounced cooperativity seen in
the case of LipDH (1).
The efficient reduction of lipoic acid by mammalian TR adds to its known wide substrate
specificity (1-4). The strong 20-fold preference of LipDH for dihydrolipoic acid oxidation
compared to lipoic acid reduction under our assay conditions (Table II) can probably in part
be explained by a lack of NAD
/
in the assay, since NAD
/
is a strong positive effector (1).
TR readily reduced lipoic acid under without NADP
/
, demonstrating differences in enzyme
mechanism between these two enzymes. The K
m
-values of mammalian TR for lipoic acid and
its derivatives of 0.49 - 1.0 mM should be compared with K
m
values of 2.5 - 5
m
M for
mammalian Trx (8, 9), illustrating the higher affinity of the enzyme for its natural substrate
thioredoxin. However, the k
cat
values of 503 - 1296 min
01
for lipoamide demonstrate an
272
AID BBRC 5144 / 6905$$$862
07-17-96 07:41:18 bbrcas AP: BBRC
Vol. 225, No. 1, 1996 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
TABLE II
Activity of Mammalian TR and LipDH in Reduction of Lipoamide and Lipoic Acid
and in the Reverse Reaction Oxidizing Dihydrolipoic Acid,
Using Either NADP or NAD as Pyridine Nucleotides
Enzyme activity (mol pyridine
nucleotide/mol enzyme/min)
a
Pyridine
Substrate nucleotide TR LipDH
Lipoamide NADPH 889
õ
1
NADH 43.4 9215
Lipoic acid NADPH 297
õ
1
NADH
õ
1 20.3
Dihydrolipoic acid NADP
/
119 12.9
NAD
/
23.8 424
Ratio in activity between
Substrates compared substrates compared
Lipoamide/lipoic acid 2.99
b
454
c
Lipoic acid/dihydrolipoic acid 2.49
d
0.048
e
a
All assays were performed at 20
7
C in 50 mM Tris, pH 8.0, and 2 mM EDTA with 1 mM lipoic
acid derivative and 200
m
M pyridine nucleotide. The concentration used of purified rat liver TR
was 7.4 nM and that of porcine heart LipDH was 6.2 nM. Activity was determined as oxidation or
reduction of the pyridine nucleotides by following the change in absorbance at 340 nm using a
molar extinction coefficient of 6200 M
01
cm
01
.
b
With NADPH.
c
With NADH.
d
Using NADPH with lipoic acid and NADP
/
with dihydrolipoic acid.
e
Using NADH with lipoic acid and NAD
/
with dihydrolipoic acid.
efficient reaction, when compared to the corresponding values of 3000 - 4200 min
01
for
different mammalian thioredoxins (9, 24).
Several NADPH-dependent diaphorases or NADPH oxidases, with varying degree of over-
lapping lipoamide dehydrogenase activity, have been described (25-29), many for which the
true nature has not yet been fully characterized. Also, NADPH-dependent diaphorase activity
has been associated with NO synthase expression in the nervous system, but additional neuronal
NADPH-dependent diaphorase activity which is not dependent on NO synthase has been
clearly demonstrated (30, 31). It will be important to determine how TR contributes to these
less characterized diaphorase activities. It is known that the enzyme is present in the nervous
system (32, 33) and the NADPH oxidase activity of TR (15) also readily reduces nitro blue
tetrazolium (NBT), the classic substrate in determination of neuronal NADPH dependent
diaphorase activity (Ane
´r, Nordberg and Holmgren, unpublished observations).
It should be noted that mammalian lipoamide dehydrogenase is a mitochondrial enzyme as
part of several
a
-keto acid dehydrogenase complexes (1, 18) whereas TR is a cytosolic enzyme
with some association with membrane structures (33, 34). The association of TR with the
plasma membrane is worth emphasizing, since lipoamide dehydrogenase activity has also been
detected in plasma membrane fractions (35).
Dihydrolipoic acid and lipoic acid have different antioxidant properties and reduction of
lipoic acid is a necessary prerequisite for several of its antioxidant effects (19). The lipoic acid
reducing activity of mammalian TR should therefore play a significant role in the metabolism of
this compound and for activation of its many antioxidant and redox regulatory qualities.
273
AID BBRC 5144 / 6905$$$862
07-17-96 07:41:18 bbrcas AP: BBRC
Vol. 225, No. 1, 1996 BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS
ACKNOWLEDGMENTS
This study was supported by the Swedish Cancer Society (961), the Swedish Medical Research Council (13X-
3529), Inga-Britt and Arne Lundbergs Stiftelse, Knut and Alice Wallenbergs Stiftelse, Makarna Agnes och Gustaf
Backlunds fond fo
¨r cancerforskning, Stiftelsen Sigurd och Elsa Goljes minne, and the Karolinska Institute.
REFERENCES
1. Williams, C. H., Jr. (1992) in Chemistry and Biochemistry of Flavoenzymes. (Mu
¨ller, F., Ed.) pp. 121–211, CRC
Press, Boca Raton, FL.
2. Holmgren, A. (1989) J. Biol. Chem. 264, 1396313966.
3. Holmgren, A. (1985) Annu. Rev. Biochem. 54, 237271.
4. Holmgren, A., and Bjo
¨rnstedt, M. (1995) Methods Enzymol. 252, 199208.
5. Yodoi, J., and Tursz, T. (1991) Adv. Cancer Res. 57, 381411.
6. Waksman, G., Krishna, T. S., Williams, C. H., Jr., and Kuriyan, J. (1994) J. Mol. Biol. 236, 800816.
7. Kuriyan, J., Krishna, T. S. R., Wong, L., Guenther, B., Pahler, A., Williams, C. H., Jr., and Model, P. (1991)
Nature 352, 172174.
8. Holmgren, A. (1977) J. Biol. Chem. 252, 46004606.
9. Luthman, M., and Holmgren, A. (1982) Biochemistry 21, 66286633.
10. Oblong, J. E., Gasdaska, P. Y., Sherrill, K., and Powis, G. (1993) Biochemistry 32, 72717277.
11. Mau, B.-L., and Powis, G. (1992) Biochem. Pharm. 43, 16131620.
12. Mau, B.-L., and Powis, G. (1992) Biochem. Pharm. 43, 16211627.
13. Schallreuter, K. U., Gleason, F. K., and Wood, J. M. (1990) Biochim. Biophys. Acta 1054, 1420.
14. Schallreuter, K. U., and Wood, J. M. (1989) Biochem. Biophys. Res. Commun. 160, 573–9.
15. Arne
´r, E. S. J., Bjo
¨rnstedt, M., and Holmgren, A. (1995) J. Biol. Chem. 270, 34793482.
16. Carlberg, I., and Mannervik, B. (1980) FEBS Lett. 115, 265268.
17. Gasdaska, P. Y., Gasdaska, J. R., Cochran, S., and Powis, G. (1995) FEBS Lett. 373, 5–9.
18. Perham, R. N. (1991) Biochemistry 30, 85018512.
19. Packer, L., Witt, E. H., and Tritschler, H. J. (1995) Free Rad. Biol. Med. 19, 227250.
20. Teichert, J., and Preiss, R. (1992) Int. J. Clin. Pharmacol. Ther. Toxicol. 30, 511512.
21. Handelman, G. J., Han, D., Tritschler, H., and Packer, L. (1994) Biochem. Pharmacol. 47, 17251730.
22. Constantinescu, A., Pick, U., Handelman, G. J., Haramaki, N., Han, D., Podda, M., Tritschler, H. J., and Packer,
L. (1995) Biochem. Pharmacol. 50, 253261.
23. Pick, U., Harmaki, N., Constantinescu, A., Handelman, G. J., Tritschler, H. J., and Packer, L. (1995) Biochem.
Biophys. Res. Commun. 206, 724730.
24. Ren, X., Bjo
¨rnstedt, M., Shen, B., Ericson, M. L., and Holmgren, A. (1993) Biochemistry 32, 97018.
25. Cross, A. R., Yarchover, J. L., and Curnutte, J. T. (1994) J. Biol. Chem. 269, 2144821454.
26. Gasdaska, P. Y., Fisher, H., and Powis, G. (1995) Cancer Res. 55, 25422547.
27. Carlberg, I., Sahlman, L., and Mannervik, B. (1985) FEBS Lett. 180, 102106.
28. Otsuka-Murakami, H., and Nisimoto, Y. (1995) FEBS Lett. 361, 206210.
29. Sreider, C. M., Grinblat, L., and Stoppani, A. O. M. (1990) Biochem. Pharmacol. 40, 18491857.
30. Afework, M., Tomlinson, A., and Burnstock, G. (1994) Cell Tissue Res. 276, 133141.
31. Hope, B. T., Michael, G. J., Knigge, K. M., and Vincent, S. R. (1991) Proc. Natl. Acad. Sci. U.S.A. 88, 2811
2814.
32. Hansson, H.-A., Holmgren, A., Norstedt, G., and Rozell, B. (1989) Exp. Eye Res. 48, 411420.
33. Rozell, B., Hansson, H. A., Luthman, M., and Holmgren, A. (1985) Eur. J. Cell Biol. 38, 7986.
34. Schallreuter, K. U., Ja
¨nner, M., Mensing, H., Breitbart, E. W., Berger, J., and Wood, J. M. (1991) Int. J. Cancer
48, 15–9.
35. Danson, M. J. (1988) Biochem. Soc. Trans. 16, 8789.
274
AID BBRC 5144 / 6905$$$862
07-17-96 07:41:18 bbrcas AP: BBRC
... Enlightened by the early finding that the five-membered cyclic disulfide compounds lipoamide and lipoic acid could be efficiently opened by mammalian TrxR but not GSH [77], the Fang group developed the first off-on fluorescent probe, called TRFS-green ( Figure 6, probe 1) [78], for mammalian TrxR by linking the fluorophore naphthalimide to an artificial cyclic disulfide 1,2-dithiolane via a carbamate linker. With the reduction by NADPH under the catalysis of TrxR, TRFS-green emits a bright green fluorescence signal at λ = 538 nm when it is excited at λ = 438 nm. ...
Article
Full-text available
The modulation of numerous signaling pathways is orchestrated by redox regulation of cellular environments. Maintaining dynamic redox homeostasis is of utmost importance for human health, given the common occurrence of altered redox status in various pathological conditions. The cardinal component of the thioredoxin system, mammalian thioredoxin reductase (TrxR) plays a vital role in supporting various physiological functions; however, its malfunction, disrupting redox balance, is intimately associated with the pathogenesis of multiple diseases. Accordingly, the dynamic monitoring of TrxR of live organisms represents a powerful direction to facilitate the comprehensive understanding and exploration of the profound significance of redox biology in cellular processes. A number of classic assays have been developed for the determination of TrxR activity in biological samples, yet their application is constrained when exploring the real-time dynamics of TrxR activity in live organisms. Fluorescent probes offer several advantages for in situ imaging and the quantification of biological targets, such as non-destructiveness, real-time analysis, and high spatiotemporal resolution. These benefits facilitate the transition from a poise to a flux understanding of cellular targets, further advancing scientific studies in related fields. This review aims to introduce the progress in the development and application of TrxR fluorescent probes in the past years, and it mainly focuses on analyzing their reaction mechanisms, construction strategies, and potential drawbacks. Finally, this study discusses the critical challenges and issues encountered during the development of selective TrxR probes and proposes future directions for their advancement. We anticipate the comprehensive analysis of the present TrxR probes will offer some glitters of enlightenment, and we also expect that this review may shed light on the design and development of novel TrxR probes.
... Among them are the peroxiredoxins (Prxs) which are peroxidases contributing to H 2 O 2 detoxification, and ribonucleotide reductase providing deoxyribonucleotides for DNA synthesis. Mammalian TrxRs are also capable of reducing substrates beyond Trxs, such as several additional Trx-fold proteins including protein disulfide-isomerase (PDI), TRP14 (TXNDC17) and TRP32 (TXNL1), as well as small molecule substrates including alloxan, menadione, and lipoic acid [9,[47][48][49][50][51]. ...
Article
Human thioredoxin reductase (TrxR) is a selenoprotein with a central role in cellular redox homeostasis, utilizing a highly reactive and solvent exposed selenocysteine (Sec) residue in its active site. Pharmacological modulation of TrxR can be obtained with several classes of small compounds showing different mechanisms of action, but most often dependent upon interactions with its Sec residue. The clinical implications of small compound-mediated TrxR modulation have been studied in diverse diseases, from rheumatoid arthritis and ischemia to cancer and parasitic infections. The involvement of TrxR in these diseases was in some cases serendipitously discovered, by finding that existing clinically used drugs are also TrxR inhibitors. Inhibiting isoforms of human TrxR is however not the only strategy for human disease treatment, as some pathogenic parasites also depend upon Sec-containing TrxR variants, including S. mansoni, B. malayi or O. volvulus. Inhibiting parasite TrxR has been shown to selectively kill parasites and can thus become a promising treatment strategy, especially in the context of quickly emerging resistance towards other drugs. Here we have summarized the basis for the targeting of selenoprotein TrxR variants with small molecules for therapeutic purposes in different human disease contexts. We discuss how Sec engagement appears to be an indispensable part of treatment efficacy and how some therapeutically promising compounds have been evaluated in preclinical or clinical studies. Several research questions remain before a wider application of selenoprotein TrxR inhibition as a first-line treatment strategy might be developed. These include further mechanistic studies of downstream effects that may mediate treatment efficacy, identification of isoform-specific enzyme inhibition patterns for some given therapeutic compounds, and the further elucidation of cell-specific effects in disease contexts such as in the tumor microenvironment or in host-parasite interactions, and which of these effects may be dependent upon the specific targeting of Sec in distinct TrxR isoforms.
... The oxidized forms of EGT and Asc have clear structural similarities, as highlighted in blue and red. Sec-TrxR, a selenoenzyme, is also able to reduce the number of other small molecule substrates, including S-nitrosoglutathione, lipoic acid/lipoamide, lipid hydroperoxides, and ubiquinone [28][29][30][31]. ...
Article
Full-text available
Ergothioneine (EGT) is a sulfur-containing amino acid analog that is biosynthesized in fungi and bacteria, accumulated in plants, and ingested by humans where it is concentrated in tissues under oxidative stress. While the physiological function of EGT is not yet fully understood, EGT is a potent antioxidant in vitro. Here we report that oxidized forms of EGT, EGT-disulfide (ESSE) and 5-oxo-EGT, can be reduced by the selenoenzyme mammalian thioredoxin reductase (Sec-TrxR). ESSE and 5-oxo-EGT are formed upon reaction with biologically relevant reactive oxygen species. We found that glutathione reductase (GR) can reduce ESSE, but only with the aid of glutathione (GSH). The reduction of ESSE by TrxR was found to be selenium dependent, with non-selenium-containing TrxR enzymes having little or no ability to reduce ESSE. In comparing the reduction of ESSE by Sec-TrxR in the presence of thioredoxin to that of GR/GSH, we find that the glutathione system is 10-fold more efficient, but Sec-TrxR has the advantage of being able to reduce both ESSE and 5-oxo-EGT directly. This represents the first discovered direct enzymatic recycling system for oxidized forms of EGT. Based on our in vitro results, the thioredoxin system may be important for EGT redox biology and requires further in vivo investigation.
Article
Three self-assembled nanoaggregates (CPUL1-LA NAs, CPUL1-DA NAs, and CPUL1-AA NAs) were constructed through lipoic acid (LA), dithiodipropionic acid (DA), and adipic acid (AA) decorated TrxR inhibitor (CPUL1), respectively. Measurements of DLS, TEM, UV-vis, fluorescence, 1H NMR, ITC, and MTT assays verified disulfide-containing CPUL1-LA NAs and CPUL1-DA NAs spontaneously assembled carrier-free nanoparticles in aqueous solution, which possessed high drug contents, excellent stability, improved cytotoxicity against HUH7 hepatoma cells, and potential biosafety because of low cytotoxicity against L02 normal cells. In contrast, disulfide-free CPUL1-AA NAs happened to aggregate and precipitate after 48 h, which showed distinct instability in aqueous solution. Thus, disulfide units seemed to be crucial for constructing controllable and stable nanoaggregates. While measuring the reduction of nanoaggregates by TrxR/NADPH and GSH/GR/NADPH, cyclic disulfide of LA and linear disulfide of DA were verified to endow the nanoaggregates with targeting ability to respond specifically to TrxR over GSH. Furthermore, by tests of flow cytometry, fluorescence images, and CLSM, both CPUL1-LA NAs and CPUL1-DA NAs displayed a faster cellular uptake characteristic to be internalized by cancer cells and could generate more abundant ROS to induce cell apoptosis than that of free CPUL1, resulting in significantly improved antitumor efficacy against HUH7 cells in vitro.
Article
Full-text available
In this comprehensive evidence-based analysis of ulcerative colitis (UC), a causal role is identified for colonic epithelial hydrogen peroxide (H2O2) in both the pathogenesis and relapse of this debilitating inflammatory bowel disease. Studies have shown that H2O2 production is significantly increased in the non-inflamed colonic epithelium of individuals with UC. H2O2 is a powerful neutrophilic chemotactic agent that can diffuse through colonic epithelial cell membranes creating an interstitial chemotactic molecular "trail" that attracts adjacent intravascular neutrophils into the colonic epithelium leading to mucosal inflammation and UC. A novel therapy aimed at removing the inappropriate H2O2 mediated chemotactic signal has been highly effective in achieving complete histologic resolution of colitis in patients experiencing refractory disease with at least one (biopsy-proven) histologic remission lasting 14 years to date. The evidence implies that therapeutic intervention to prevent the re-establishment of a pathologic H2O2 mediated chemotactic signaling gradient will indefinitely preclude neutrophilic migration into the colonic epithelium constituting a functional cure for this disease. Cumulative data indicate that individuals with UC have normal immune systems and current treatment guidelines calling for the suppression of the immune response based on the belief that UC is caused by an underlying immune dysfunction are not supported by the evidence and may cause serious adverse effects. It is the aim of this paper to present experimental and clinical evidence that identifies H2O2 produced by the colonic epithelium as the causal agent in the pathogenesis of UC. A detailed explanation of a novel therapeutic intervention to normalize colonic H2O2, its rationale, components, and formulation is also provided.
Article
Significance: Nutraceuticals are ingested for health benefits in addition to their general nutritional value. These dietary supplements have become increasingly popular since the late twentieth century and they are a rapidly expanding global industry approaching a half-trillion US dollars annually. Many nutraceuticals are promulgated as potent antioxidants. Recent advances: Experimental support for the efficacy of nutraceuticals has lagged behind anecdotal exuberance. However, accumulating epidemiological evidence, and recent, well-controlled clinical trials, are beginning to support earlier animal and in vitro studies. Although still somewhat limited, encouraging results have been suggested in essentially all organ systems and against a wide range of pathophysiological conditions. Critical issues: Health benefits of the 'antioxidant' nutraceuticals are largely attributed to their ability to scavenge oxidants. This has been criticized based on several factors including limited bioavailability, short tissue retention time, and the preponderance of endogenous antioxidants. Recent attention has turned to nutraceutical activation of downstream antioxidant systems, especially the Keap 1/Nrf2 axis. The question now becomes, how do nutraceuticals activate this axis? Future directions: Reactive sulfur species (RSS), including hydrogen sulfide (H2S) and its metabolites are potent activators of the Keap 1/Nrf2 axis and avid scavengers of reactive oxygen species (ROS). Evidence is beginning to accumulate that a variety of nutraceuticals increase cellular RSS by directly providing RSS in the diet, or through a number of catalytic mechanisms that increase endogenous RSS production. We propose that nutraceutical-specific targeting of RSS metabolism will lead to the design and development of even more efficacious antioxidant therapeutic strategies.
Article
Background: The thioredoxin system (Trx), comprising of Trx, Thioredoxin reductase (TrxR) and NADPH aids in donating hydrogen group to support Ribonucleotide reductase (RNR) catalysis during de-novo DNA biosynthesis. However, it has been observed that inhibiting TrxR does not affect the viability of cancer cells that are susceptible to pharmacological glutathione (GSH) depletion. This prompted us to study the potential antioxidant redundancies that might prolong RNR activity. Methods: To study the RNR activity assay, the RNR complex was reconstituted by mixing purified mouse recombinant RNR subunits and the conversion of [3 H] CDP into [3 H] dCDP was monitored. In the assay system, either purified Trx and GSH or Lipoamide system was supplemented as reducing agents to support RNR catalysis. Results: Herein, we have found that GSH-dependent Trx reduction supports mammalian class I RNR catalysis in absence of TrxR in the system. Our data also presents the first report that the LAM system is capable of supporting in-vitro RNR activity in the complete absence of either Trx or Grx systems. Conclusions: We conclude that GSH-mediated Trx reduction and LAM systems support basal level RNR activity in vitro; in absence of TrxR and complete redoxin systems respectively and hypothesize that potential redundancy between the various antioxidant systems might synergize in sustaining RNR activity.
Article
Background Thioredoxin reductase (TrxR), one of the representative selenoenzymes, is an important antioxidant enzyme suppressing oxidative stress in living organisms. At the active site of human TrxR, the presence of a Sec•••His•••Glu catalytic triad was previously suggested. Method. In this study, a short selenopeptide mimicking this plausible triad, i.e., H-CUGHGE-OH (1), was designed, synthesized, and evaluated for the TrxR-like catalytic activity. Method In this study, a short selenopeptide mimicking this plausible triad, i.e., H-CUGHGE-OH (1), was designed, synthesized, and evaluated for the TrxR-like catalytic activity Results The molecular simulation in advance by REMC/SAAP3D predicted the preferential formation of Sec•••His•••Glu hydrogen bonding networks in the aqueous solution. Indeed, a significant antioxidant activity was observed for 1 in the activity assay using NADPH as a reductant and H2O2 as a substrate. Tracking the reaction between 1 and GSH by 77Se NMR revealed a reductive cleavage of the selenosulfide (Se-S) bond to generate the diselenide species. The observation suggested that in the transiently formed mixed Se-S intermediate, the NH•••Se hydrogen bond between the Sec and His residues leads a nucleophilic attack of the second thiol molecule not to the intrinsically more electrophilic Se atom but to the less electrophilic S atom of the Se-S bond. Ab initio calculations for the complex between MeSeSMe and an imidazolium ion at the MP2/6-31++G(d,p) level demonstrated that NH•••Se and NH•••S hydrogen bonds are equally favorable as the interaction modes. Thus, importance of the relative spatial arrangement of the Se-S bond with respect to the imidazole ring was suggested for the exertion of the TrxR-like catalytic activity. Conclusion The proposed umpolung effect of NH•••Se hydrogen bond on the reactivity of a Se-S bond will be a useful tool for developing efficient TrxR models with high redox catalytic activity.
Article
Full-text available
Thioredoxin reductase, which catalyzes the reduction of the disulfide bridge in thioredoxin by NADPH, was purified from calf liver and thymus. Preparation methods, involving chromatography on DEAE cellulose, TEAE cellulose, and Sephadex G 200 or G 100 were used to purify the calf liver enzyme 1100 fold and the thymus enzyme 2800 fold. The enzyme was shown to catalyze an NADPH dependent reduction of 5,5' dithiobis(2 nitrobenzoic acid) which could be used to develop a simple and rapid assay in addition to a specific calf liver thioredoxin dependent reduction of disulfide bonds in bovine insulin. The purified enzyme, which was inhibited by 0.1 mM arsenite, showed a wider substrate specificity than the corresponding enzyme from Escherichia coli and rapidly reduced E. coli thioredoxin, yeast thioredoxin, and 5,5' dithiobis(2 nitrobenzoic acid). Phage T4 thioredoxin was slowly reduced. The apparent k(m) values for 5,5' dithiobis(2 nitrobenzoic acid) and calf liver thioredoxin were 1.5 mM and 5 μM, respectively. In vitro oxidized preparations of calf liver thioredoxin were shown to contain high molecular weight mixed disulfide aggregates that were reduced by the enzyme with kinetics which supported a process of autoactivation. This may be of importance as a control mechanism for the activity of the bovine thioredoxin system. Reduction of disulfide bonds in insulin or L cystine by NADPH and thioredoxin reductase was absolutely dependent upon the presence of thioredoxin as intermediate electron carrier. With the coupled system, fast reduction of insulin was obtained with as little as 3 x 10-8 M calf liver thioredoxin. By a number of criteria the bovine thioredoxin system (thioredoxin, thioredoxin reductase, and NADPH) was identical with the enzyme NADPH protein disulfide reductase and is suggested to play a major role in metabolic oxidoreductions of protein disulfides.
Article
Full-text available
NADPH diaphorase histochemistry selectively labels a number of discrete populations of neurons throughout the nervous system. This simple and robust technique has been used in a great many experimental and neuropathological studies; however, the function of this enzyme has remained a matter of speculation. We, therefore, undertook to characterize this enzyme biochemically. With biochemical and immunochemical assays, NADPH diaphorase was purified to apparent homogeneity from rat brain by affinity chromatography and anion-exchange HPLC. Western (immunoblot) transfer and immunostaining with an antibody specific for NADPH diaphorase labeled a single protein of 150 kDa. Nitric oxide synthase was recently shown to be a 150-kDa, NADPH-dependent enzyme in brain. It is responsible for the calcium/calmodulin-dependent synthesis of the guanylyl cyclase activator nitric oxide from L-arginine. We have found that nitric oxide synthase activity and NADPH diaphorase copurify to homogeneity and that both activities could be immunoprecipitated with an antibody recognizing neuronal NADPH diaphorase. Furthermore, nitric oxide synthase was competitively inhibited by the NADPH diaphorase substrate, nitro blue tetrazolium. Thus, neuronal NADPH diaphorase is a nitric oxide synthase, and NADPH diaphorase histochemistry, therefore, provides a specific histochemical marker for neurons producing nitric oxide.
Article
Full-text available
An example of two related enzymes that catalyse similar reactions but possess different active sites is provided by comparing the structure of Escherichia coli thioredoxin reductase with glutathione reductase. Both are dimeric enzymes that catalyse the reduction of disulphides by pyridine nucleotides through an enzyme disulphide and a flavin. Human glutathione reductase contains four structural domains within each molecule: the flavin-adenine dinucleotide (FAD)- and nicotinamide-adenine dinucleotide phosphate (NADPH)-binding domains, the 'central' domain and the C-terminal domain that provides the dimer interface and part of the active site. Although both enzymes share the same catalytic mechanism and similar tertiary structures, their active sites do not resemble each other. We have determined the crystal structure of E. coli thioredoxin reductase at 2 A resolution, and show that thioredoxin reductase lacks the domain that provides the dimer interface in glutathione reductase, and forms a completely different dimeric structure. The catalytically active disulphides are located in different domains on opposite sides of the flavin ring system. This suggests that these enzymes diverged from an ancestral nucleotide-binding protein and acquired their disulphide reductase activities independently.
Article
Lipoic acid has been reported recently to be an effective antioxidant in biological systems. It may act in vivo through reduction to its dithiol form, dihydrolipoic acid. Using a dual Hg/Au electrode, and HPLC with electrochemical detection, a method was developed which allowed simultaneous measurement of lipoic acid and dihydrolipoic acid, at nanomolar levels. (RS)-α-Lipoic acid was added to human cells in tissue culture (Jurkat T-lymphocytes and primary neonatal diploid fibroblasts). Lipoic acid was converted rapidly by the cells to dihydrolipoic acid, which accumulated in the cell pellet. Monitored over a 2-hr interval, dihydrolipoic acid was released, and several-fold more dihydrolipoic acid could be found in the medium than in the pellet.
Article
Quinoids undergo metabolism by a number of flavoenzymes. Reactive species formed during the metabolism of some quinoids might be anticipated to inhibit flavoenzyme activity. Several quinoids have been tested for their ability to inhibit rat liver thioredoxin reductase (TR). The antitumor quinones diaziquone and doxorubicin, and the quinoneimine 2,6-dichloroindophenol, were found to be inhibitors of the reduction of 5,5'-dithiobis-2-nitrobenzoic acid (DTNB) by TR. The inhibition was most marked after incubation of the quinoid with NADPH and the enzyme for 60 min before adding DTNB, with Ki values of 0.5 microM for diaziquone, 0.5 microM for doxorubicin, and 0.07 microM for 2,6-dichloroindophenol. The three quinoids all produced a time-dependent and first order loss of TR activity. There was formation of electron spin resonance-detectable semiquinoid free radicals upon incubation of diaziquone, doxorubicin and 2,6-dichloroindophenol with TR and NADPH under anaerobic conditions. Oxygen radicals formed by redox cycling of the quinoids did not make a major contribution to the inhibition of TR by the quinoids, as shown by the absence of significant reversal of the inhibition by anaerobic incubation conditions and the lack of effect of the oxygen radical scavengers dimethyl sulfoxide, superoxide dismutase and catalase. It was not possible to demonstrate NADPH-dependent covalent binding of radiolabeled diaziquone or doxorubicin to the TR apoprotein. It is possible that the quinoids bind noncovalently to the enzyme apoprotein, or bind to the FAD prosthetic group. The results of the study suggest that some antitumor quinoids are mechanism-based inhibitors of TR showing metabolism- and time-dependent irreversible inhibition of enzyme activity.
Article
Adult T-cell leukemia-derived factor (ADF) plays an important role in the transforming properties of HTLV-I by interfering with the expression of the interleukin-2 receptor (IL-2R) complex. In particular, ADF appears to act as an autocrine growth factor for an Epstein–Barr virus (EBV)-positive B cell line established independently by another group and to synergize with suboptimal doses of other lymphokines, making these cells sensitive to a series of growth factors. The fact that both HTLV-I-transformed T lymphocytes and EBV-immortalized B cells are able to produce the same factor implies that some underlying mechanisms of transformation could be common to the two viruses, which could possibly interact with similar cellular genes. The amino acid sequence of ADF is highly homologous to prokaryotic and mammalian thioredoxins. Thus, ADF appears to be a member of the thioredoxin family of protein cofactors involved in thiol-dependent redox reactions in many cell types and at various steps of cellular metabolism. ADF secreted by the cells infected with HTLV-I or EBV exhibits a very high reducing potential. This enzymatic activity of ADF might, at least partly, explain its role in cellular activities, including IL-2R expression, increased susceptibility to a variety of growth factors, and promotion of cell proliferation.
Article
Plasma membrane-associated thioredoxin reductase activities have been determined on primary melanoma tissues and their surrounding skin in 29 patients. Compared to patient's normal skin, enzyme activities in melanoma were higher in some patients (n = 24) and lower in others (n = 5). Those melanomas with high TR activities yielded low activities in the adjacent epidermis, reaching normal activity 3 to 5 cm away from each primary site (n = 4). Tumors with low activities showed higher than normal activities on the immediate surrounding skin (i.e., 1 cm away from the tumor) compared to the normal skin (n = 3). Earlier it was shown that in both keratinocytes and melanoma cells, calcium regulates thioredoxin reductase activity by an allosteric mechanism. The differences in TR activities within the high and low groups may be caused by a calcium flux between the primary tumor and the surrounding epidermis, and vice versa. A comparison of TR activities to tumor invasiveness (Breslow level) in 28 primary melanomas showed a significant correlation using regression analysis (p = 0.031). A 4-fold difference in TR activity corresponds to a one-unit change in Breslow determination. These preliminary results suggest that TR activity may be another useful and sensitive assay for melanoma spread.