SlideShare a Scribd company logo
1 of 4
Download to read offline
Neuroscience Letters 364 (2004) 164–167




       Endogenous glutathione and catalase protect cultured rat astrocytes
            from the iron-mediated toxicity of hydrogen peroxide
                                 Jeff R. Liddell a , Stephen R. Robinson a , Ralf Dringen a,b,∗
                                         a Department of Psychology, Monash University, Clayton, Vic. 3800, Australia
                             b   Interfakultäres Institut für Biochemie der Universität Tuebingen, D-72076 Tuebingen, Germany

                                  Received 21 March 2004; received in revised form 15 April 2004; accepted 15 April 2004




Abstract

   Primary astrocyte cultures from rat brain were exposed to hydrogen peroxide (H2 O2 ) to investigate peroxide toxicity and clearance by
astrocytes. After bolus application of H2 O2 (100 M), the peroxide was eliminated from the incubation medium following first-order kinetics
with a half-time of approximately 4 min. The rate of peroxide detoxification was significantly slowed by pre-incubating the cells with the
glutathione synthesis inhibitor buthionine sulfoximine (BSO), or the catalase inhibitor 3-amino-1,2,4-triazole (3AT), and was retarded further
when both treatments were combined. H2 O2 application killed a small proportion of cells, as indicated by the levels of the cytosolic enzyme
lactate dehydrogenase in the media 1 and 24 h later. In contrast, cell viability was strongly compromised when the cells were pre-incubated with
3AT and/or BSO before peroxide application. The iron chelator deferoxamine completely prevented this cell loss. These results demonstrate
that chelatable iron is involved in the toxicity of H2 O2 and that both the glutathione system and catalase protect astrocytes from this toxicity.
© 2004 Elsevier Ireland Ltd. All rights reserved.

Keywords: Astrocytes; Catalase; Glutathione; Hydrogen peroxide; Iron; Oxidative stress



Hydrogen peroxide is generated during aerobic meta-                               when acutely applied at concentrations of 100 M [5].
bolism by the action of superoxide dismutases and various                         These studies have also found that chemical inactivation
oxidases [4]. H2 O2 and superoxide, in the presence of iron                       of either GPx or catalase only slightly retards the rate of
ions, can lead to the formation of highly reactive and cyto-                      H2 O2 detoxification, and does not appreciably increase
toxic hydroxyl radicals [3,13]. Such processes contribute to                      the extent of cell loss during the period of H2 O2 exposure
the cell loss associated with reperfusion injury [11], and to                     [5].
the neurodegeneration associated with chronic conditions                             The preceding observations question whether cells are
such as Alzheimer’s and Parkinson’s diseases [12,15]. To                          only vulnerable to H2 O2 when both antioxidant systems
reduce the likelihood of radical formation from peroxides,                        have been inactivated, or whether cell loss occurs but
brain cells use two antioxidant systems that can rapidly                          is delayed beyond the first few hours. It is also unclear
inactivate H2 O2 . In a reaction catalyzed by glutathione per-                    from these observations whether endogenous iron is an
oxidases (GPx), the tripeptide glutathione (GSH) serves as                        important factor in peroxide-mediated toxicity, as has
an electron donor to reduce H2 O2 to water. The product                           been suggested by some other studies [1,16,17]. Answers
of the oxidation of GSH is glutathione disulfide. Besides                          to these questions could assist the design of therapeutic
the glutathione system, the diffusion-controlled catalase                         approaches that are aimed at reducing the extent of neu-
also inactivates H2 O2 , especially if this peroxide is present                   rodegeneration following acute episodes, such as stroke
in high concentrations [2]. Experiments using cultured                            and ischemia. The present study therefore investigates:
cells have established that endogenous levels of GSH and                          (i) the relative contributions of GSH and catalase to the
catalase are sufficient to rapidly inactivate H2 O2 , even                         detoxification of H2 O2 by cultured astrocytes; (ii) the con-
                                                                                  tributions of both systems to the protection of astrocytes
  ∗ Corresponding author. Present address: Interfakultäres Institut für
                                                                                  from H2 O2 -mediated damage in the 24 h period following
Biochemie der Universität Tübingen, Hoppe-Seyler-Str. 4, D-72076 Tübin-
                                                                                  peroxide exposure; and (iii) whether the iron chelator de-
gen, Germany. Tel.: +49-7071-2973334; fax: +49-7071-295360.                       feroxamine (DFX) can play a protective role against H2 O2
   E-mail address: ralf.dringen@uni-tuebingen.de (R. Dringen).                    toxicity.

0304-3940/$ – see front matter © 2004 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.neulet.2004.04.042
J.R. Liddell et al. / Neuroscience Letters 364 (2004) 164–167                                        165

   H2 O2 was applied in a concentration of 100 M to as-                       ability of astrocyte cultures, DFX was applied in a final
trocyte primary cultures in 24-well plates that had been                      concentration of 2 mM either 2 h before the application
prepared from the brains of newborn Wistar rats, as de-                       of H2 O2 (pre-peroxide) and/or during the 24 h incubation
scribed previously [8,9]. To measure the rate of peroxide                     following the H2 O2 exposure (post-peroxide). Exposure of
clearance, cells were washed with 2 ml of incubation buffer                   cultured astrocytes to 1 mM DFX is sufficient to up-regulate
(20 mM HEPES, 145 mM NaCl, 1.8 mM CaCl2 , 5.4 mM                              transferrin receptor expression and down-regulate ferritin
KCl, 1 mM MgCl2 , 0.8 mM Na2 HPO4 , 5 mM glucose, pH                          expression [10]. Such changes are indicative of depleted in-
7.4) and then provided with 500 l of incubation buffer (con-                  tracellular iron levels [3]. Since 2 mM DFX was used in the
taining 100 M H2 O2 ) at 37 ◦ C. During the incubation the                    present experiments, it is likely that most of the chelatable
extracellular concentration of H2 O2 was repeatedly mea-                      iron was effectively bound by the DFX. The protein con-
sured by the colorimetric method described previously [6].                    tent of the cell cultures was determined with the method of
   To assess the relative contributions of GSH and catalase to                Lowry et al. [14] using bovine serum albumin as a standard.
H2 O2 toxicity, astrocyte cultures were pre-incubated either                     When untreated astrocyte cultures (control) were exposed
for 24 h with 1 ml of Dulbecco’s modified Eagles’s medium                      to 100 M H2 O2 , the peroxide was depleted from the incuba-
(DMEM) containing the GSH synthesis inhibitor buthionine                      tion buffer (Fig. 1A) following first-order kinetics (Fig. 1B),
sulfoximine (BSO; 1 mM), or for 2 h in 1 ml DMEM with                         with a half-time of about 4 min (Table 1). This result is con-
the catalase inhibitor 3-amino-1,2,4-triazole (3AT; 10 mM).                   sistent with previous reports concerning the rate of H2 O2
Under these conditions BSO reduces the GSH content of                         detoxification by cultured astrocytes [5,6,9]. In the absence
astrocytes to 14% of controls and 3AT completely inhibits                     of cells, but otherwise under identical conditions, H2 O2 is
catalase activity [5]. For BSO + 3AT conditions, cells were                   stable for at least 1 h [7]. LDH measurements indicated
incubated for 22 h with BSO and for an additional 2 h with                    that control astrocytes which had been treated with 100 M
BSO plus 3AT. The subsequent incubation with H2 O2 was                        H2 O2 did not undergo substantial cell damage in the subse-
carried out in the absence of inhibitors.                                     quent 24 h incubation period (Fig. 2B, control).
   Cell viability was analyzed by determining the extracel-                      Astrocytes that had been pre-incubated with BSO or 3AT
lular activity of lactate dehydrogenase (LDH) in the cultures                 disposed of the H2 O2 more slowly than controls (Fig. 1;
[6]. The LDH activity in the incubation buffer after 60 min                   Table 1). Semi-logarithmic plots of the data obtained dur-
incubation with H2 O2 (or in DMEM 24 h after removal of                       ing the initial 8 min of H2 O2 detoxification showed that un-
the peroxide) was compared to the LDH activity in lysates                     der these conditions the clearance of the peroxide follows
of untreated astrocyte cultures. These lysates were obtained                  first-order kinetics (Fig. 1B). The rate of peroxide disposal
by exposing cultures to 1% Triton X-100 for 30 min [6].                       was normalized to the protein content of the respective wells
Pre-incubation of astrocytes with BSO, DFX or 3AT did                         by calculating the specific H2 O2 detoxification rate constant
not alter the total LDH activity of astrocyte cultures (data                  (D = 1/(half-time in minutes × initial protein content in
not shown). To investigate the effect of DFX on the vi-                       milligrams)). D is proportional to the capacity of cells to




Fig. 1. Disposal of exogenous H2 O2 by cultured astrocytes. The cells were pre-incubated for 24 h in DMEM (control), for 24 h in DMEM containing
1 mM BSO, for 2 h in DMEM containing 3AT (10 mM) or for 24 h with BSO (1 mM) as well as for 2 h with 10 mM 3AT (BSO + 3AT). Subsequently,
astrocytes were incubated with 500 l of incubation buffer containing 100 M H2 O2 . (A) Time course of the concentration of H2 O2 in the incubation
buffer of the cultures. (B) Semi-logarithmic representation of the data obtained. This figure shows a representative experiment performed on a 20 days
old culture. The mean protein content was 146 ± 9 g per well.
166                                           J.R. Liddell et al. / Neuroscience Letters 364 (2004) 164–167

Table 1
Half-times of H2 O2 and specific H2 O2 detoxification rate constant D of astrocyte cultures
Treatment         No DFX                                                             DFX                                                              n

                  Half-time (min)      D (minutes × milligrams of    protein)−1      Half-time (min)     D (minutes × milligrams of   protein)−1

Control            4.0   ±   0.4       1.78   ±   0.19                               4.1   ±   0.6       1.74   ±   0.23                              18
BSO                5.1   ±   0.8       1.35   ±   0.21∗                              5.6   ±   0.6       1.19   ±   0.10∗                              9
3AT                6.1   ±   1.0       1.19   ±   0.23∗                              5.7   ±   0.8       1.26   ±   0.18∗                              9
BSO + 3AT         11.1   ±   2.5       0.64   ±   0.18∗                              9.8   ±   2.6       0.72   ±   0.17∗                             18
The cells were pre-incubated for 24 h in DMEM (control) or for 24 h in DMEM containing 1 mM BSO and/or for 2 h in the presence of 10 mM 3AT
and/or 2 mM DFX. Subsequently, astrocytes were incubated with 100 M H2 O2 for 1 h. The decline in extracellular concentration of the peroxide was
monitored and half-times of the peroxide were calculated from the semi-logarithmic representations of the data. The specific detoxification rate constant
D is defined as 1/(half-time in minutes × initial protein content in milligrams). The data represent mean values ± S.D. of (n) wells derived from three
to six experiments. The significance of the differences of the D values compared to those of the respective controls (control) was calculated by ANOVA
followed by Bonferroni’s post hoc test (∗ P < 0.001). Corresponding data obtained for cells treated with and without DFX were not significantly different,
as calculated by Student’s t-test.

detoxify a peroxide [7]. The higher the D value, the better                       damage than control astrocytes (Fig. 2). 1 h after application
the capacity of the cultured cells to dispose of a peroxide.                      of the peroxide about 20% of total LDH was measured ex-
The specific detoxification rate constant D of astrocytes that                      tracellularly for cells that had been treated with inhibitors,
were treated with BSO or 3AT was significantly reduced                             compared to 13% for controls (Fig. 2A). Cell loss was con-
compared to that in control cultures, indicating that the rate                    siderably greater 24 h later, with extracellular LDH values
of peroxide disposal was reduced by pre-incubating the cells                      reaching up to 53% of the total (Fig. 2B). These findings
with either BSO or 3AT (Table 1). Combined treatment with                         demonstrate that while astrocytes treated with BSO and/or
both inhibitors slowed the rate of peroxide detoxification                         3AT can effectively detoxify H2 O2 , they nonetheless sustain
to a much greater extent than was achieved with either in-                        oxidative injury, which results in their death several hours
hibitor alone. This result provides clear evidence that both                      later. It is noteworthy that BSO-treated cells were signifi-
GSH and catalase participate in the inactivation of H2 O2 by                      cantly more sensitive to peroxide treatment (Fig. 2B) than
astrocytes.                                                                       cells which had been pre-incubated with 3AT (P < 0.001),
   Astrocytes that had a reduced rate of H2 O2 disposal due                       indicating that under the conditions used the GSH system is
to pre-incubation with either BSO, 3AT, or BSO + 3AT                              more effective at preventing peroxide-mediated damage than
(Table 1) underwent significantly higher amounts of cell                           catalase. A reason for this difference could be that catalase
                                                                                  can only inactivate H2 O2 , whereas GSH can also inactivate
                                                                                  radicals [4].
                                                                                     Iron has been implicated in the potentiation of peroxide-
                                                                                  induced cell damage in brain cells [1,16,17]. In order to
                                                                                  examine the contribution of cellular iron to the toxicity
                                                                                  observed after H2 O2 treatment, astrocyte cultures were in-
                                                                                  cubated with DFX before and after H2 O2 treatment. DFX
                                                                                  treatment did not affect the rate of clearance of H2 O2 under
                                                                                  the various conditions investigated, as indicated by identical
                                                                                  D values and half-times for the peroxide (Table 1). These
                                                                                  observations demonstrate that chelatable iron does not con-
                                                                                  tribute to the disappearance of H2 O2 from the incubation
Fig. 2. LDH release from astrocyte cultures after peroxide stress. The            medium and that DFX does not compromise the capacity
cells were pre-incubated for 24 h in DMEM (control) or for 24 h in
DMEM containing 1 mM BSO and/or for 2 h in the presence of 10 mM
                                                                                  of astrocytes to inactivate H2 O2 . Nonetheless, DFX com-
3AT and/or 2 mM DFX. Subsequently, astrocytes were incubated with                 pletely prevented the elevated LDH release that normally
500 l of incubation buffer containing 100 M H2 O2 for 1 h. Cells were             resulted when astrocytes were exposed to H2 O2 following
then incubated for a further 24 h in 1 ml DMEM in the absence or                  pre-incubation with BSO, 3AT, or BSO + 3AT (Fig. 2).
presence of 2 mM DFX. Cell viability was assessed by determining the              This prevention of cell damage by DFX was evident both
activity of extracellular LDH 1 h (A) or 24 h (B) after the cells had been
incubated with hydrogen peroxide. The data represent mean values ± S.D.
                                                                                  1 h (Fig. 2A) and 24 h (Fig. 2B) after H2 O2 treatment.
of 8 or 9 wells of the respective treatments obtained from experiments               To determine when the protective action of DFX is most
performed on three independently prepared cultures. The significance of            critical, DFX application was limited to either the pre- or
the differences in LDH release between peroxide-treated control cells and         the post-peroxide period. It was found that pre-incubation
cells pre-incubated with BSO and/or 3AT was calculated by ANOVA                   with DFX provides protection against the toxicity of H2 O2
followed by Bonferroni’s post hoc test (+ P < 0.001). The significance of
the differences in LDH release obtained from astrocyte cultures treated
                                                                                  whereas post-incubation with DFX provides no significant
with 2 mM DFX to those treated without DFX was calculated by Student’s            protection (Table 2). However, the presence of DFX dur-
t-tests (∗ P < 0.001).                                                            ing both the pre- and post-peroxide incubation periods
J.R. Liddell et al. / Neuroscience Letters 364 (2004) 164–167                                              167

Table 2                                                                          ment of Psychology, Monash University, for financial sup-
Effects of DFX treatment on the LDH release from peroxide-treated                port of the research conducted in this study. Jan Riemer
astrocytes
                                                                                 and Hans-Hermann Hoepken provided valuable technical
Treatment       LDH release (percent of total LDH)                       n       advice.
                No DFX      Pre-          Post-        Pre- and
                            peroxide      peroxide     post-peroxide
Control         13 ± 4       6±3          20 ± 10      13 ± 5            9       References
BSO + 3AT       42 ± 7∗     19 ± 4∗,+     36 ± 5∗      10 ± 4+           9
The cells were pre-incubated for 24 h in DMEM (control) or for 24 h               [1] K. Abe, H. Saito, Characterization of t-butyl hydroperoxide toxicity
in DMEM containing BSO (1 mM) plus for 2 h in the presence of 3AT                     in cultured cortical neurones and astrocytes, Pharmacol. Toxicol. 83
(10 mM). Subsequently, astrocytes were incubated with 100 M H2 O2 for                 (1998) 40–46.
1 h and for further 24 h in DMEM. DFX (2 mM) was present 2 h before               [2] H.E. Aebi, Catalase, in: H.U. Bergmeyer, J. Bergmeyer, M. Grassl
peroxide application (pre-peroxide) and/or during the 24 h incubation af-             (Eds.), Methods of Enzymatic Analysis, vol. 3, Verlag Chemie,
ter removal of the peroxide (post-peroxide). Cell viability was assessed              Weinheim, Germany, 1984, pp. 273–286.
by monitoring the amount of LDH release after the 24 h incubation that            [3] R.R. Crichton, S. Wilmet, R. Legssyer, R.J. Ward, Molecular and
followed the peroxide treatment. The data represent mean values ± S.D.                cellular mechanisms of iron homeostasis and toxicity in mammalian
of (n) wells obtained from experiments performed on three independently               cells, J. Inorg. Biochem. 91 (2002) 9–18.
prepared cultures. The significance of the differences in LDH release ob-          [4] R. Dringen, Metabolism and functions of glutathione in brain, Prog.
tained for control or BSO + 3AT-treated cells was calculated by Student’s             Neurobiol. 62 (2000) 649–671.
t-test (∗ P < 0.001). The significance of the differences between cells that       [5] R. Dringen, B. Hamprecht, Involvement of glutathione peroxidase
were treated in the absence of DFX (no DFX) or with DFX for the given                 and catalase in the disposal of exogenous hydrogen peroxide by
incubation periods was calculated by ANOVA followed by Bonferroni’s                   cultured astroglial cells, Brain Res. 759 (1997) 67–75.
post hoc test (+ P < 0.001).                                                      [6] R. Dringen, L. Kussmaul, B. Hamprecht, Detoxification of exogenous
                                                                                      hydrogen peroxide and organic hydroperoxides by cultured astroglial
                                                                                      cells assessed by a microtiter plate assay, Brain Res. Protoc. 2 (1998)
completely rescued cells from peroxide-mediated damage                                223–228.
and was significantly (P < 0.01) more protective than                              [7] R. Dringen, L. Kussmaul, J.M. Gutterer, J. Hirrlinger, B. Hamprecht,
pre-incubation with DFX (Table 2). These data demon-                                  The glutathione system of peroxide detoxification is less efficient
strate that the presence of chelatable iron is a key factor                           in neurons than in astroglial cells, J. Neurochem. 72 (1999) 2523–
                                                                                      2530.
in the peroxide-mediated damage of astrocytes that have
                                                                                  [8] B. Hamprecht, F. Löffler, Primary glial cultures as a model system
a compromised capacity to clear H2 O2 . Since chelatable                              for studying hormone action, Methods Enzymol. 109 (1985) 341–
iron catalyzes the formation of hydroxyl radicals in the                              345.
presence of superoxide and H2 O2 by the Fenton reaction                           [9] J. Hirrlinger, A. Resch, J.M. Gutterer, R. Dringen, Oligodendroglial
or the Haber–Weiss cycle [3,13], the protection provided                              cells in culture effectively dispose of exogenous hydrogen peroxide:
                                                                                      comparison with cultured neurones, astroglial and microglial cells,
to H2 O2 -treated astrocytes by DFX is probably due to a
                                                                                      J. Neurochem. 82 (2002) 635–644.
reduction in the amount of iron that is available to catalyze                    [10] H.H. Hoepken, T. Korten, S.R. Robinson, R. Dringen, Iron accumu-
the formation of cytotoxic hydroxyl radicals.                                         lation, iron-mediated toxicity and altered levels of ferritin and trans-
   In summary, both GSH and catalase contribute to the                                ferrin receptor in cultured astrocytes during incubation with ferric
clearance of H2 O2 by astrocytes in vitro. Impairment of                              ammonium citrate, J. Neurochem. 88 (2004) 1194–1202.
                                                                                 [11] P.A. Hyslop, Z. Zhang, D.V. Pearson, L.A. Phebus, Measurement of
either antioxidant system increases the vulnerability of as-
                                                                                      striatal H2 O2 by microdialysis following global forebrain ischemia
trocytes to H2 O2 -induced injury, and even small reductions                          and reperfusion in the rat: correlation with the cytotoxic potential of
in the rate of peroxide disposal can lead to substantial cell                         H2 O2 in vitro, Brain Res. 671 (1995) 181–186.
damage over the following 24 h period. It is of particular                       [12] P. Jenner, Oxidative stress in Parkinson’s disease, Ann. Neurol.
interest that peroxide-induced cell loss in astrocyte cultures                        53 (Suppl. 3) (2003) S26–S36.
                                                                                 [13] M. Kruszewski, Labile iron pool: the main determinant of cellular
can be completely prevented with DFX. Viewed in the con-
                                                                                      response to oxidative stress, Mutat. Res. 531 (2003) 81–92.
text of the brain, the present results suggest that chelatable                   [14] O.H. Lowry, N.J. Rosebrough, A.L. Farr, R.J. Randall, Protein mea-
iron, as well as endogenous levels of GSH and catalase in                             surement with the Folin phenol reagent, J. Biol. Chem. 193 (1951)
astrocytes, may represent useful therapeutic targets in the                           265–275.
treatment of certain neurodegenerative conditions.                               [15] N.G. Milton, Role of hydrogen peroxide in the aethiology of
                                                                                      Alzheimer’s disease: implications for treatment, Drugs Aging 21
                                                                                      (2004) 81–100.
                                                                                 [16] S.J. Robb, J.R. Connor, An in vitro model for analysis of oxidative
Acknowledgements                                                                      death in primary mouse astrocytes, Brain Res. 788 (1998) 125–132.
                                                                                 [17] W. Ying, S.K. Han, J.W. Miller, R.A. Swanson, Acidosis potentiates
  R.D. was supported by a Senior Research Fellowship                                  oxidative neuronal death by multiple mechanisms, J. Neurochem. 73
                                                                                      (1999) 1549–1556.
awarded by NeuroSciences Victoria. We thank the Depart-

More Related Content

What's hot

A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND ANTIANDROGENIC E...
A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND  ANTIANDROGENIC E...A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND  ANTIANDROGENIC E...
A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND ANTIANDROGENIC E...Dr. Pradeep mitharwal
 
A review on antioxidant methods
A review on antioxidant methodsA review on antioxidant methods
A review on antioxidant methodsneeluyadav
 
Ameliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation inducedAmeliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation inducedRam Sahu
 
Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008
Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008
Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008joecali26
 
International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)inventionjournals
 
Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...
Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...
Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...Mahendra Kumar Trivedi
 
ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, A PROPHYLACTIC AND THERAPEUTIC MIXTU...
ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, APROPHYLACTIC AND THERAPEUTIC MIXTU...ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, APROPHYLACTIC AND THERAPEUTIC MIXTU...
ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, A PROPHYLACTIC AND THERAPEUTIC MIXTU...Dmitri Popov
 
In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...
In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...
In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...pharmaindexing
 
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complexElectrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complexIOSR Journals
 
SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...
SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...
SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...Tomsk Polytechnic University
 
Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...
Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...
Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...IOSR Journals
 

What's hot (19)

2
22
2
 
I0363044052
I0363044052I0363044052
I0363044052
 
A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND ANTIANDROGENIC E...
A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND  ANTIANDROGENIC E...A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND  ANTIANDROGENIC E...
A STUDY TO EVALUATE THE IN VITRO ANTIMICROBIAL ACTIVITY AND ANTIANDROGENIC E...
 
DPPH assay
DPPH assayDPPH assay
DPPH assay
 
A review on antioxidant methods
A review on antioxidant methodsA review on antioxidant methods
A review on antioxidant methods
 
Ameliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation inducedAmeliorative effect of salicin against gamma irradiation induced
Ameliorative effect of salicin against gamma irradiation induced
 
Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008
Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008
Comp Of Chem&amp;Cell Based Antiox Jafc Sep 2008
 
Viva 01032011
Viva 01032011Viva 01032011
Viva 01032011
 
2007 revised nmr data for incartine an alkaloid from galanthus
2007 revised nmr data for incartine an alkaloid from galanthus2007 revised nmr data for incartine an alkaloid from galanthus
2007 revised nmr data for incartine an alkaloid from galanthus
 
International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)International Journal of Pharmaceutical Science Invention (IJPSI)
International Journal of Pharmaceutical Science Invention (IJPSI)
 
Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...
Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...
Thermal and Physical Properties of Biofield Treated Bile Salt and Proteose Pe...
 
B045004013
B045004013B045004013
B045004013
 
ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, A PROPHYLACTIC AND THERAPEUTIC MIXTU...
ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, APROPHYLACTIC AND THERAPEUTIC MIXTU...ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, APROPHYLACTIC AND THERAPEUTIC MIXTU...
ANTIOXIDANTS AND POTASSIUM FERROCYANIDE, A PROPHYLACTIC AND THERAPEUTIC MIXTU...
 
26248 28893-1-pb
26248 28893-1-pb26248 28893-1-pb
26248 28893-1-pb
 
In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...
In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...
In Vitro Antioxidant Studies of Whole Plant Ethanolic Extract of Blepharisrep...
 
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complexElectrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
Electrochemical, in-vitro in-vivo study of Co (II)-ofloxacin complex
 
In vitro antioxidant-activities_of_ethan (1)
In vitro antioxidant-activities_of_ethan (1)In vitro antioxidant-activities_of_ethan (1)
In vitro antioxidant-activities_of_ethan (1)
 
SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...
SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...
SELF-ASSEMBLY AMPHIPHILIC CHITOSAN NANOCARRIERS FOR ORAL CO-DELIVERY OF HYDRO...
 
Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...
Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...
Synthesis, spectroscopic, magnetic properties and superoxide dismutase (SOD) ...
 

Viewers also liked

Personalidades
PersonalidadesPersonalidades
PersonalidadesLuciane
 
Les Satellites v2 - Le Nouveau Chapitre
Les Satellites v2 - Le Nouveau ChapitreLes Satellites v2 - Le Nouveau Chapitre
Les Satellites v2 - Le Nouveau ChapitreNicolas Bergé
 
Whitepaper: Integrating People and Technology
Whitepaper: Integrating People and TechnologyWhitepaper: Integrating People and Technology
Whitepaper: Integrating People and TechnologyTolero Solutions
 
Présentation du projet Douzi'Action
Présentation du projet Douzi'ActionPrésentation du projet Douzi'Action
Présentation du projet Douzi'ActionPaul Mariuzzo-Raynaud
 
Ao k human and natural sciences - enzyme lab
Ao k   human and natural sciences - enzyme labAo k   human and natural sciences - enzyme lab
Ao k human and natural sciences - enzyme labPreston Fernandez
 
Gsh detoxcell 1250
 Gsh detoxcell 1250  Gsh detoxcell 1250
Gsh detoxcell 1250 markanne
 

Viewers also liked (11)

Personalidades
PersonalidadesPersonalidades
Personalidades
 
C&rsquo;est Skaistkalne
C&rsquo;est SkaistkalneC&rsquo;est Skaistkalne
C&rsquo;est Skaistkalne
 
Les Satellites v2 - Le Nouveau Chapitre
Les Satellites v2 - Le Nouveau ChapitreLes Satellites v2 - Le Nouveau Chapitre
Les Satellites v2 - Le Nouveau Chapitre
 
Présentation réunion 26.06.15
Présentation réunion 26.06.15Présentation réunion 26.06.15
Présentation réunion 26.06.15
 
Whitepaper: Integrating People and Technology
Whitepaper: Integrating People and TechnologyWhitepaper: Integrating People and Technology
Whitepaper: Integrating People and Technology
 
Présentation du projet Douzi'Action
Présentation du projet Douzi'ActionPrésentation du projet Douzi'Action
Présentation du projet Douzi'Action
 
Satalase eun jeong
Satalase eun jeongSatalase eun jeong
Satalase eun jeong
 
Ao k human and natural sciences - enzyme lab
Ao k   human and natural sciences - enzyme labAo k   human and natural sciences - enzyme lab
Ao k human and natural sciences - enzyme lab
 
Enzmes
EnzmesEnzmes
Enzmes
 
BBB Asas Matematik
BBB Asas MatematikBBB Asas Matematik
BBB Asas Matematik
 
Gsh detoxcell 1250
 Gsh detoxcell 1250  Gsh detoxcell 1250
Gsh detoxcell 1250
 

Similar to Ting Endogenous Glutathione And Catalase Protect Cultured Rat Astrocytes From The Iron Mediated Toxicity Of Hydrogen Peroxide

Chan & Roth 2008
Chan & Roth 2008Chan & Roth 2008
Chan & Roth 2008Kin Chan
 
1308-JonathanBakerCa-SummerPoster
1308-JonathanBakerCa-SummerPoster1308-JonathanBakerCa-SummerPoster
1308-JonathanBakerCa-SummerPosterJonathan Baker
 
PhD subject proposal_Devaux
PhD subject proposal_DevauxPhD subject proposal_Devaux
PhD subject proposal_DevauxJules Devaux
 
medicinal_chemistry_of_some_NSAIDs.pptx
medicinal_chemistry_of_some_NSAIDs.pptxmedicinal_chemistry_of_some_NSAIDs.pptx
medicinal_chemistry_of_some_NSAIDs.pptxNajmedinAzizi
 
Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...
Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...
Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...CrimsonPublishersUrologyJournal
 
Radiation Protection : Phospholipase A
Radiation Protection : Phospholipase ARadiation Protection : Phospholipase A
Radiation Protection : Phospholipase ADmitri Popov
 
Nico wanandy unsw mechanism of antioxidant for the skin
Nico wanandy   unsw mechanism of antioxidant for the skinNico wanandy   unsw mechanism of antioxidant for the skin
Nico wanandy unsw mechanism of antioxidant for the skinNico Wanandy
 
Reactive oxygen species
Reactive oxygen speciesReactive oxygen species
Reactive oxygen speciesRupali Patil
 
Response and tolerance strategies of microorganisms to oxidative
Response and tolerance strategies  of microorganisms to oxidativeResponse and tolerance strategies  of microorganisms to oxidative
Response and tolerance strategies of microorganisms to oxidativeKumar Purushotam
 
Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...
Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...
Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...BRNSSPublicationHubI
 
(THPS BIOCIDE BASE ) moniotoring in sea water
(THPS BIOCIDE BASE ) moniotoring in sea water(THPS BIOCIDE BASE ) moniotoring in sea water
(THPS BIOCIDE BASE ) moniotoring in sea waterAlsayed Yakoot
 
Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)
Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)
Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)IOSR Journals
 
Lead Acetate Induced histopathological Changes in Kidney Tissue of Rat
Lead Acetate Induced histopathological Changes in Kidney Tissue of RatLead Acetate Induced histopathological Changes in Kidney Tissue of Rat
Lead Acetate Induced histopathological Changes in Kidney Tissue of RatIOSRJAVS
 
Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...
Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...
Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...Georgi Daskalov
 
OXIDATIVE_STRESS_RESPONSE_mitochondria.ppt
OXIDATIVE_STRESS_RESPONSE_mitochondria.pptOXIDATIVE_STRESS_RESPONSE_mitochondria.ppt
OXIDATIVE_STRESS_RESPONSE_mitochondria.pptskellchansky
 
In Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional FoodIn Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional FoodInstitut Kurz
 

Similar to Ting Endogenous Glutathione And Catalase Protect Cultured Rat Astrocytes From The Iron Mediated Toxicity Of Hydrogen Peroxide (20)

Chan & Roth 2008
Chan & Roth 2008Chan & Roth 2008
Chan & Roth 2008
 
1308-JonathanBakerCa-SummerPoster
1308-JonathanBakerCa-SummerPoster1308-JonathanBakerCa-SummerPoster
1308-JonathanBakerCa-SummerPoster
 
PhD subject proposal_Devaux
PhD subject proposal_DevauxPhD subject proposal_Devaux
PhD subject proposal_Devaux
 
medicinal_chemistry_of_some_NSAIDs.pptx
medicinal_chemistry_of_some_NSAIDs.pptxmedicinal_chemistry_of_some_NSAIDs.pptx
medicinal_chemistry_of_some_NSAIDs.pptx
 
Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...
Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...
Crimson Publishers-Hyperoxaluria Induces Oxidative DNA Damage and Results in ...
 
Radiation Protection : Phospholipase A
Radiation Protection : Phospholipase ARadiation Protection : Phospholipase A
Radiation Protection : Phospholipase A
 
Lesion por oxigeno
Lesion por oxigenoLesion por oxigeno
Lesion por oxigeno
 
Nico wanandy unsw mechanism of antioxidant for the skin
Nico wanandy   unsw mechanism of antioxidant for the skinNico wanandy   unsw mechanism of antioxidant for the skin
Nico wanandy unsw mechanism of antioxidant for the skin
 
Reactive oxygen species
Reactive oxygen speciesReactive oxygen species
Reactive oxygen species
 
Response and tolerance strategies of microorganisms to oxidative
Response and tolerance strategies  of microorganisms to oxidativeResponse and tolerance strategies  of microorganisms to oxidative
Response and tolerance strategies of microorganisms to oxidative
 
Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...
Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...
Hydrogen Sulfide Suppress the Pathological Alterations of Endocrine Glands In...
 
(THPS BIOCIDE BASE ) moniotoring in sea water
(THPS BIOCIDE BASE ) moniotoring in sea water(THPS BIOCIDE BASE ) moniotoring in sea water
(THPS BIOCIDE BASE ) moniotoring in sea water
 
Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)
Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)
Structural and Spectroscopical Studies for a Complex Macromolecule (hGH)
 
Lead Acetate Induced histopathological Changes in Kidney Tissue of Rat
Lead Acetate Induced histopathological Changes in Kidney Tissue of RatLead Acetate Induced histopathological Changes in Kidney Tissue of Rat
Lead Acetate Induced histopathological Changes in Kidney Tissue of Rat
 
Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...
Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...
Some In Vitro/In Vivo Chemically-Induced Experimental Models of Liver Oxidati...
 
Reactive oxygen species
Reactive oxygen speciesReactive oxygen species
Reactive oxygen species
 
OXIDATIVE_STRESS_RESPONSE_mitochondria.ppt
OXIDATIVE_STRESS_RESPONSE_mitochondria.pptOXIDATIVE_STRESS_RESPONSE_mitochondria.ppt
OXIDATIVE_STRESS_RESPONSE_mitochondria.ppt
 
In Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional FoodIn Vitro Cell Tests for Functional Food
In Vitro Cell Tests for Functional Food
 
PROTEOMICS.pptx
PROTEOMICS.pptxPROTEOMICS.pptx
PROTEOMICS.pptx
 
2013 antioxidant properties of xanthones
2013 antioxidant properties of xanthones2013 antioxidant properties of xanthones
2013 antioxidant properties of xanthones
 

Ting Endogenous Glutathione And Catalase Protect Cultured Rat Astrocytes From The Iron Mediated Toxicity Of Hydrogen Peroxide

  • 1. Neuroscience Letters 364 (2004) 164–167 Endogenous glutathione and catalase protect cultured rat astrocytes from the iron-mediated toxicity of hydrogen peroxide Jeff R. Liddell a , Stephen R. Robinson a , Ralf Dringen a,b,∗ a Department of Psychology, Monash University, Clayton, Vic. 3800, Australia b Interfakultäres Institut für Biochemie der Universität Tuebingen, D-72076 Tuebingen, Germany Received 21 March 2004; received in revised form 15 April 2004; accepted 15 April 2004 Abstract Primary astrocyte cultures from rat brain were exposed to hydrogen peroxide (H2 O2 ) to investigate peroxide toxicity and clearance by astrocytes. After bolus application of H2 O2 (100 M), the peroxide was eliminated from the incubation medium following first-order kinetics with a half-time of approximately 4 min. The rate of peroxide detoxification was significantly slowed by pre-incubating the cells with the glutathione synthesis inhibitor buthionine sulfoximine (BSO), or the catalase inhibitor 3-amino-1,2,4-triazole (3AT), and was retarded further when both treatments were combined. H2 O2 application killed a small proportion of cells, as indicated by the levels of the cytosolic enzyme lactate dehydrogenase in the media 1 and 24 h later. In contrast, cell viability was strongly compromised when the cells were pre-incubated with 3AT and/or BSO before peroxide application. The iron chelator deferoxamine completely prevented this cell loss. These results demonstrate that chelatable iron is involved in the toxicity of H2 O2 and that both the glutathione system and catalase protect astrocytes from this toxicity. © 2004 Elsevier Ireland Ltd. All rights reserved. Keywords: Astrocytes; Catalase; Glutathione; Hydrogen peroxide; Iron; Oxidative stress Hydrogen peroxide is generated during aerobic meta- when acutely applied at concentrations of 100 M [5]. bolism by the action of superoxide dismutases and various These studies have also found that chemical inactivation oxidases [4]. H2 O2 and superoxide, in the presence of iron of either GPx or catalase only slightly retards the rate of ions, can lead to the formation of highly reactive and cyto- H2 O2 detoxification, and does not appreciably increase toxic hydroxyl radicals [3,13]. Such processes contribute to the extent of cell loss during the period of H2 O2 exposure the cell loss associated with reperfusion injury [11], and to [5]. the neurodegeneration associated with chronic conditions The preceding observations question whether cells are such as Alzheimer’s and Parkinson’s diseases [12,15]. To only vulnerable to H2 O2 when both antioxidant systems reduce the likelihood of radical formation from peroxides, have been inactivated, or whether cell loss occurs but brain cells use two antioxidant systems that can rapidly is delayed beyond the first few hours. It is also unclear inactivate H2 O2 . In a reaction catalyzed by glutathione per- from these observations whether endogenous iron is an oxidases (GPx), the tripeptide glutathione (GSH) serves as important factor in peroxide-mediated toxicity, as has an electron donor to reduce H2 O2 to water. The product been suggested by some other studies [1,16,17]. Answers of the oxidation of GSH is glutathione disulfide. Besides to these questions could assist the design of therapeutic the glutathione system, the diffusion-controlled catalase approaches that are aimed at reducing the extent of neu- also inactivates H2 O2 , especially if this peroxide is present rodegeneration following acute episodes, such as stroke in high concentrations [2]. Experiments using cultured and ischemia. The present study therefore investigates: cells have established that endogenous levels of GSH and (i) the relative contributions of GSH and catalase to the catalase are sufficient to rapidly inactivate H2 O2 , even detoxification of H2 O2 by cultured astrocytes; (ii) the con- tributions of both systems to the protection of astrocytes ∗ Corresponding author. Present address: Interfakultäres Institut für from H2 O2 -mediated damage in the 24 h period following Biochemie der Universität Tübingen, Hoppe-Seyler-Str. 4, D-72076 Tübin- peroxide exposure; and (iii) whether the iron chelator de- gen, Germany. Tel.: +49-7071-2973334; fax: +49-7071-295360. feroxamine (DFX) can play a protective role against H2 O2 E-mail address: ralf.dringen@uni-tuebingen.de (R. Dringen). toxicity. 0304-3940/$ – see front matter © 2004 Elsevier Ireland Ltd. All rights reserved. doi:10.1016/j.neulet.2004.04.042
  • 2. J.R. Liddell et al. / Neuroscience Letters 364 (2004) 164–167 165 H2 O2 was applied in a concentration of 100 M to as- ability of astrocyte cultures, DFX was applied in a final trocyte primary cultures in 24-well plates that had been concentration of 2 mM either 2 h before the application prepared from the brains of newborn Wistar rats, as de- of H2 O2 (pre-peroxide) and/or during the 24 h incubation scribed previously [8,9]. To measure the rate of peroxide following the H2 O2 exposure (post-peroxide). Exposure of clearance, cells were washed with 2 ml of incubation buffer cultured astrocytes to 1 mM DFX is sufficient to up-regulate (20 mM HEPES, 145 mM NaCl, 1.8 mM CaCl2 , 5.4 mM transferrin receptor expression and down-regulate ferritin KCl, 1 mM MgCl2 , 0.8 mM Na2 HPO4 , 5 mM glucose, pH expression [10]. Such changes are indicative of depleted in- 7.4) and then provided with 500 l of incubation buffer (con- tracellular iron levels [3]. Since 2 mM DFX was used in the taining 100 M H2 O2 ) at 37 ◦ C. During the incubation the present experiments, it is likely that most of the chelatable extracellular concentration of H2 O2 was repeatedly mea- iron was effectively bound by the DFX. The protein con- sured by the colorimetric method described previously [6]. tent of the cell cultures was determined with the method of To assess the relative contributions of GSH and catalase to Lowry et al. [14] using bovine serum albumin as a standard. H2 O2 toxicity, astrocyte cultures were pre-incubated either When untreated astrocyte cultures (control) were exposed for 24 h with 1 ml of Dulbecco’s modified Eagles’s medium to 100 M H2 O2 , the peroxide was depleted from the incuba- (DMEM) containing the GSH synthesis inhibitor buthionine tion buffer (Fig. 1A) following first-order kinetics (Fig. 1B), sulfoximine (BSO; 1 mM), or for 2 h in 1 ml DMEM with with a half-time of about 4 min (Table 1). This result is con- the catalase inhibitor 3-amino-1,2,4-triazole (3AT; 10 mM). sistent with previous reports concerning the rate of H2 O2 Under these conditions BSO reduces the GSH content of detoxification by cultured astrocytes [5,6,9]. In the absence astrocytes to 14% of controls and 3AT completely inhibits of cells, but otherwise under identical conditions, H2 O2 is catalase activity [5]. For BSO + 3AT conditions, cells were stable for at least 1 h [7]. LDH measurements indicated incubated for 22 h with BSO and for an additional 2 h with that control astrocytes which had been treated with 100 M BSO plus 3AT. The subsequent incubation with H2 O2 was H2 O2 did not undergo substantial cell damage in the subse- carried out in the absence of inhibitors. quent 24 h incubation period (Fig. 2B, control). Cell viability was analyzed by determining the extracel- Astrocytes that had been pre-incubated with BSO or 3AT lular activity of lactate dehydrogenase (LDH) in the cultures disposed of the H2 O2 more slowly than controls (Fig. 1; [6]. The LDH activity in the incubation buffer after 60 min Table 1). Semi-logarithmic plots of the data obtained dur- incubation with H2 O2 (or in DMEM 24 h after removal of ing the initial 8 min of H2 O2 detoxification showed that un- the peroxide) was compared to the LDH activity in lysates der these conditions the clearance of the peroxide follows of untreated astrocyte cultures. These lysates were obtained first-order kinetics (Fig. 1B). The rate of peroxide disposal by exposing cultures to 1% Triton X-100 for 30 min [6]. was normalized to the protein content of the respective wells Pre-incubation of astrocytes with BSO, DFX or 3AT did by calculating the specific H2 O2 detoxification rate constant not alter the total LDH activity of astrocyte cultures (data (D = 1/(half-time in minutes × initial protein content in not shown). To investigate the effect of DFX on the vi- milligrams)). D is proportional to the capacity of cells to Fig. 1. Disposal of exogenous H2 O2 by cultured astrocytes. The cells were pre-incubated for 24 h in DMEM (control), for 24 h in DMEM containing 1 mM BSO, for 2 h in DMEM containing 3AT (10 mM) or for 24 h with BSO (1 mM) as well as for 2 h with 10 mM 3AT (BSO + 3AT). Subsequently, astrocytes were incubated with 500 l of incubation buffer containing 100 M H2 O2 . (A) Time course of the concentration of H2 O2 in the incubation buffer of the cultures. (B) Semi-logarithmic representation of the data obtained. This figure shows a representative experiment performed on a 20 days old culture. The mean protein content was 146 ± 9 g per well.
  • 3. 166 J.R. Liddell et al. / Neuroscience Letters 364 (2004) 164–167 Table 1 Half-times of H2 O2 and specific H2 O2 detoxification rate constant D of astrocyte cultures Treatment No DFX DFX n Half-time (min) D (minutes × milligrams of protein)−1 Half-time (min) D (minutes × milligrams of protein)−1 Control 4.0 ± 0.4 1.78 ± 0.19 4.1 ± 0.6 1.74 ± 0.23 18 BSO 5.1 ± 0.8 1.35 ± 0.21∗ 5.6 ± 0.6 1.19 ± 0.10∗ 9 3AT 6.1 ± 1.0 1.19 ± 0.23∗ 5.7 ± 0.8 1.26 ± 0.18∗ 9 BSO + 3AT 11.1 ± 2.5 0.64 ± 0.18∗ 9.8 ± 2.6 0.72 ± 0.17∗ 18 The cells were pre-incubated for 24 h in DMEM (control) or for 24 h in DMEM containing 1 mM BSO and/or for 2 h in the presence of 10 mM 3AT and/or 2 mM DFX. Subsequently, astrocytes were incubated with 100 M H2 O2 for 1 h. The decline in extracellular concentration of the peroxide was monitored and half-times of the peroxide were calculated from the semi-logarithmic representations of the data. The specific detoxification rate constant D is defined as 1/(half-time in minutes × initial protein content in milligrams). The data represent mean values ± S.D. of (n) wells derived from three to six experiments. The significance of the differences of the D values compared to those of the respective controls (control) was calculated by ANOVA followed by Bonferroni’s post hoc test (∗ P < 0.001). Corresponding data obtained for cells treated with and without DFX were not significantly different, as calculated by Student’s t-test. detoxify a peroxide [7]. The higher the D value, the better damage than control astrocytes (Fig. 2). 1 h after application the capacity of the cultured cells to dispose of a peroxide. of the peroxide about 20% of total LDH was measured ex- The specific detoxification rate constant D of astrocytes that tracellularly for cells that had been treated with inhibitors, were treated with BSO or 3AT was significantly reduced compared to 13% for controls (Fig. 2A). Cell loss was con- compared to that in control cultures, indicating that the rate siderably greater 24 h later, with extracellular LDH values of peroxide disposal was reduced by pre-incubating the cells reaching up to 53% of the total (Fig. 2B). These findings with either BSO or 3AT (Table 1). Combined treatment with demonstrate that while astrocytes treated with BSO and/or both inhibitors slowed the rate of peroxide detoxification 3AT can effectively detoxify H2 O2 , they nonetheless sustain to a much greater extent than was achieved with either in- oxidative injury, which results in their death several hours hibitor alone. This result provides clear evidence that both later. It is noteworthy that BSO-treated cells were signifi- GSH and catalase participate in the inactivation of H2 O2 by cantly more sensitive to peroxide treatment (Fig. 2B) than astrocytes. cells which had been pre-incubated with 3AT (P < 0.001), Astrocytes that had a reduced rate of H2 O2 disposal due indicating that under the conditions used the GSH system is to pre-incubation with either BSO, 3AT, or BSO + 3AT more effective at preventing peroxide-mediated damage than (Table 1) underwent significantly higher amounts of cell catalase. A reason for this difference could be that catalase can only inactivate H2 O2 , whereas GSH can also inactivate radicals [4]. Iron has been implicated in the potentiation of peroxide- induced cell damage in brain cells [1,16,17]. In order to examine the contribution of cellular iron to the toxicity observed after H2 O2 treatment, astrocyte cultures were in- cubated with DFX before and after H2 O2 treatment. DFX treatment did not affect the rate of clearance of H2 O2 under the various conditions investigated, as indicated by identical D values and half-times for the peroxide (Table 1). These observations demonstrate that chelatable iron does not con- tribute to the disappearance of H2 O2 from the incubation Fig. 2. LDH release from astrocyte cultures after peroxide stress. The medium and that DFX does not compromise the capacity cells were pre-incubated for 24 h in DMEM (control) or for 24 h in DMEM containing 1 mM BSO and/or for 2 h in the presence of 10 mM of astrocytes to inactivate H2 O2 . Nonetheless, DFX com- 3AT and/or 2 mM DFX. Subsequently, astrocytes were incubated with pletely prevented the elevated LDH release that normally 500 l of incubation buffer containing 100 M H2 O2 for 1 h. Cells were resulted when astrocytes were exposed to H2 O2 following then incubated for a further 24 h in 1 ml DMEM in the absence or pre-incubation with BSO, 3AT, or BSO + 3AT (Fig. 2). presence of 2 mM DFX. Cell viability was assessed by determining the This prevention of cell damage by DFX was evident both activity of extracellular LDH 1 h (A) or 24 h (B) after the cells had been incubated with hydrogen peroxide. The data represent mean values ± S.D. 1 h (Fig. 2A) and 24 h (Fig. 2B) after H2 O2 treatment. of 8 or 9 wells of the respective treatments obtained from experiments To determine when the protective action of DFX is most performed on three independently prepared cultures. The significance of critical, DFX application was limited to either the pre- or the differences in LDH release between peroxide-treated control cells and the post-peroxide period. It was found that pre-incubation cells pre-incubated with BSO and/or 3AT was calculated by ANOVA with DFX provides protection against the toxicity of H2 O2 followed by Bonferroni’s post hoc test (+ P < 0.001). The significance of the differences in LDH release obtained from astrocyte cultures treated whereas post-incubation with DFX provides no significant with 2 mM DFX to those treated without DFX was calculated by Student’s protection (Table 2). However, the presence of DFX dur- t-tests (∗ P < 0.001). ing both the pre- and post-peroxide incubation periods
  • 4. J.R. Liddell et al. / Neuroscience Letters 364 (2004) 164–167 167 Table 2 ment of Psychology, Monash University, for financial sup- Effects of DFX treatment on the LDH release from peroxide-treated port of the research conducted in this study. Jan Riemer astrocytes and Hans-Hermann Hoepken provided valuable technical Treatment LDH release (percent of total LDH) n advice. No DFX Pre- Post- Pre- and peroxide peroxide post-peroxide Control 13 ± 4 6±3 20 ± 10 13 ± 5 9 References BSO + 3AT 42 ± 7∗ 19 ± 4∗,+ 36 ± 5∗ 10 ± 4+ 9 The cells were pre-incubated for 24 h in DMEM (control) or for 24 h [1] K. Abe, H. Saito, Characterization of t-butyl hydroperoxide toxicity in DMEM containing BSO (1 mM) plus for 2 h in the presence of 3AT in cultured cortical neurones and astrocytes, Pharmacol. Toxicol. 83 (10 mM). Subsequently, astrocytes were incubated with 100 M H2 O2 for (1998) 40–46. 1 h and for further 24 h in DMEM. DFX (2 mM) was present 2 h before [2] H.E. Aebi, Catalase, in: H.U. Bergmeyer, J. Bergmeyer, M. Grassl peroxide application (pre-peroxide) and/or during the 24 h incubation af- (Eds.), Methods of Enzymatic Analysis, vol. 3, Verlag Chemie, ter removal of the peroxide (post-peroxide). Cell viability was assessed Weinheim, Germany, 1984, pp. 273–286. by monitoring the amount of LDH release after the 24 h incubation that [3] R.R. Crichton, S. Wilmet, R. Legssyer, R.J. Ward, Molecular and followed the peroxide treatment. The data represent mean values ± S.D. cellular mechanisms of iron homeostasis and toxicity in mammalian of (n) wells obtained from experiments performed on three independently cells, J. Inorg. Biochem. 91 (2002) 9–18. prepared cultures. The significance of the differences in LDH release ob- [4] R. Dringen, Metabolism and functions of glutathione in brain, Prog. tained for control or BSO + 3AT-treated cells was calculated by Student’s Neurobiol. 62 (2000) 649–671. t-test (∗ P < 0.001). The significance of the differences between cells that [5] R. Dringen, B. Hamprecht, Involvement of glutathione peroxidase were treated in the absence of DFX (no DFX) or with DFX for the given and catalase in the disposal of exogenous hydrogen peroxide by incubation periods was calculated by ANOVA followed by Bonferroni’s cultured astroglial cells, Brain Res. 759 (1997) 67–75. post hoc test (+ P < 0.001). [6] R. Dringen, L. Kussmaul, B. Hamprecht, Detoxification of exogenous hydrogen peroxide and organic hydroperoxides by cultured astroglial cells assessed by a microtiter plate assay, Brain Res. Protoc. 2 (1998) completely rescued cells from peroxide-mediated damage 223–228. and was significantly (P < 0.01) more protective than [7] R. Dringen, L. Kussmaul, J.M. Gutterer, J. Hirrlinger, B. Hamprecht, pre-incubation with DFX (Table 2). These data demon- The glutathione system of peroxide detoxification is less efficient strate that the presence of chelatable iron is a key factor in neurons than in astroglial cells, J. Neurochem. 72 (1999) 2523– 2530. in the peroxide-mediated damage of astrocytes that have [8] B. Hamprecht, F. Löffler, Primary glial cultures as a model system a compromised capacity to clear H2 O2 . Since chelatable for studying hormone action, Methods Enzymol. 109 (1985) 341– iron catalyzes the formation of hydroxyl radicals in the 345. presence of superoxide and H2 O2 by the Fenton reaction [9] J. Hirrlinger, A. Resch, J.M. Gutterer, R. Dringen, Oligodendroglial or the Haber–Weiss cycle [3,13], the protection provided cells in culture effectively dispose of exogenous hydrogen peroxide: comparison with cultured neurones, astroglial and microglial cells, to H2 O2 -treated astrocytes by DFX is probably due to a J. Neurochem. 82 (2002) 635–644. reduction in the amount of iron that is available to catalyze [10] H.H. Hoepken, T. Korten, S.R. Robinson, R. Dringen, Iron accumu- the formation of cytotoxic hydroxyl radicals. lation, iron-mediated toxicity and altered levels of ferritin and trans- In summary, both GSH and catalase contribute to the ferrin receptor in cultured astrocytes during incubation with ferric clearance of H2 O2 by astrocytes in vitro. Impairment of ammonium citrate, J. Neurochem. 88 (2004) 1194–1202. [11] P.A. Hyslop, Z. Zhang, D.V. Pearson, L.A. Phebus, Measurement of either antioxidant system increases the vulnerability of as- striatal H2 O2 by microdialysis following global forebrain ischemia trocytes to H2 O2 -induced injury, and even small reductions and reperfusion in the rat: correlation with the cytotoxic potential of in the rate of peroxide disposal can lead to substantial cell H2 O2 in vitro, Brain Res. 671 (1995) 181–186. damage over the following 24 h period. It is of particular [12] P. Jenner, Oxidative stress in Parkinson’s disease, Ann. Neurol. interest that peroxide-induced cell loss in astrocyte cultures 53 (Suppl. 3) (2003) S26–S36. [13] M. Kruszewski, Labile iron pool: the main determinant of cellular can be completely prevented with DFX. Viewed in the con- response to oxidative stress, Mutat. Res. 531 (2003) 81–92. text of the brain, the present results suggest that chelatable [14] O.H. Lowry, N.J. Rosebrough, A.L. Farr, R.J. Randall, Protein mea- iron, as well as endogenous levels of GSH and catalase in surement with the Folin phenol reagent, J. Biol. Chem. 193 (1951) astrocytes, may represent useful therapeutic targets in the 265–275. treatment of certain neurodegenerative conditions. [15] N.G. Milton, Role of hydrogen peroxide in the aethiology of Alzheimer’s disease: implications for treatment, Drugs Aging 21 (2004) 81–100. [16] S.J. Robb, J.R. Connor, An in vitro model for analysis of oxidative Acknowledgements death in primary mouse astrocytes, Brain Res. 788 (1998) 125–132. [17] W. Ying, S.K. Han, J.W. Miller, R.A. Swanson, Acidosis potentiates R.D. was supported by a Senior Research Fellowship oxidative neuronal death by multiple mechanisms, J. Neurochem. 73 (1999) 1549–1556. awarded by NeuroSciences Victoria. We thank the Depart-