Attenuation of Rotenone-Induced Mitochondrial Oxidative Damage and Neurotoxicty in Drosophila Melanogaster Supplemented With Creatine

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Neurochem Res (2010) 35:1402–1412

DOI 10.1007/s11064-010-0198-z

ORIGINAL PAPER

Attenuation of Rotenone-Induced Mitochondrial Oxidative


Damage and Neurotoxicty in Drosophila melanogaster
Supplemented with Creatine
Ravikumar Hosamani • Saraf R. Ramesh •

Muralidhara

Accepted: 15 May 2010 / Published online: 1 June 2010


Ó Springer Science+Business Media, LLC 2010

Abstract Creatine (Cr), an ergogenic nutritional supple- antioxidant activity and ability to abrogate rotenone
ment is demonstrated to possess bioenergetic, anti- induced mitochondrial oxidative stress.
excitotoxic and antioxidant properties. This study investigated
the neuroprotective effects of Cr against rotenone induced Keywords Creatine  Drosophila  Oxidative stress 
oxidative stress, mortality and neurotoxicty in Drosophila Mitochondria  Dopamine  Neurotoxicty
melanogaster. We found significant diminution in the
endogenous levels of oxidative markers in whole body
homogenates of flies exposed to Cr (2–10 mM). Cr sup- Introduction
plementation resulted in reduced mortality in flies
exposed to rotenone (500 lM) and better performance in Creatine (Cr), a natural substance in the human body, is
a negative geotaxis assay. Further Cr (10 mM) markedly partly synthesized endogenously as well as ingested
offset rotenone induced mitochondrial oxidative stress, exogenously from food especially meat and fish. It plays a
completely restored the GSH levels, nitric oxide levels, vital role in providing rapid energy during muscle con-
activity of Mn-SOD and dopamine depletion. In an traction which involves the transfer of N-phosphoryl group
oxidative stress bioassay, flies given Cr prophylaxis from phosphorylcreatine (PCr) to ADP to regenerate ATP
exhibited marked resistance to paraquat exposure. These through a reversible reaction catalyzed by phosphorylcre-
data allow us to hypothesize that the neuroprotective atine kinase (PCK) [1]. Cr chiefly functions to transfer
action of Cr in Drosophila may be related to its direct energy from mitochondria to cytosol. The PCr-PCK system
serves as an energy buffer by connecting the mitochondrial
sites of energy production with cytosolic sites of energy
consumption in tissues with high energy demand such as
brain and muscle [2]. Of the various physiological func-
tions assigned to the PCr-PCk system in vivo, prevention of
oxidative stress via direct and indirect antioxidant action is
Presented in part during the international conference: 12th European
Drosophila Neurobiology Conference, NEUROFLY 2008’’
one of the major roles [3].
September 6th–10th 2008 Wurzburg, Germany (abstract published in Currently Cr is widely used as an ergogenic nutri-
Journal of Neurogenetics special issue, 2009; vol, 23(1): S1–S102. tional supplement regularly by athletes to improve
muscular performance [4]. Several studies have con-
R. Hosamani  Muralidhara (&)
firmed the beneficial effects of Cr supplementation in
Department of Biochemistry and Nutrition, Central Food
Technological Research Institute, Council of Scientific and patients suffering from atrophy, muscle weakness, and
Industrial Research, Mysore 570020, India metabolic dysfunctions. Recent investigations have
e-mail: mura16@yahoo.com revealed the potential therapeutic effects of Cr supple-
mentation such as—improved brain function among
S. R. Ramesh
Department of Studies in Zoology, Manasagangotri, University young subjects and elderly people [5], attenuation of
of Mysore, Mysore 570020, India stress-induced cognitive impairment and reduction of

123
Neurochem Res (2010) 35:1402–1412 1403

mental fatigue [6]. Several evidences indicate specific Experimental Procedure


protective effects of Cr in various animal models of
neurodegenerative diseases or ischemic stroke [7–9]. The Materials
neuroprotective effects of Cr has been ascribed to the
buffering capacity of cellular ATP levels coupled with Creatine monohydrate, Rotenone, Paraquat, Dopamine,
mitochondrial targeted antioxidant properties in cell and Thiobarbituric acid (TBA), 1,1,3,3-tetramethoxypropane,
mammalian models [10–12]. 2,7, dichloro-fluoroscein (DCF), 2,7, dichlorofluorescein
Non-mammalian model such as Drosophila melano- diacetate (DCF-DA), and Griess reagent were procured
gaster has been extensively exploited as a powerful from Sigma Chemical Co. St Louis, USA. All other
genetic tool to understand complex biological problems. chemicals used were of analytical grade.
In recent times, numerous researchers have utilized this
model to understand various neurodegenerative diseases Drosophila Culture and Husbandry
[13, 14]. Proteomic analyses have revealed that greater
than 70% of the disease-related loci in humans have a Synchronized Drosophila melanogaster, wild (Oregon K),
clear orthologs in Drosophila [13]. The high degree of adult male flies (8–10d old) were obtained from the
conservation revealed through proteomic analyses, the Drosophila stock culture facility at Manasagangothri,
presence of a complex nervous system in an intact University of Mysore, Karnataka. The flies were main-
organism amenable to genetic manipulation, and the rel- tained at 25 ± 1°C and 70–80% relative humidity and fed
atively short lifespan of the flies make Drosophila ideal on a standard wheat flour-agar diet with yeast granules.
for studying progressive neurological conditions including Diet was prepared according to a standard protocol [20].
Parkinson’s disease (PD). Familial forms of PD have led Male adult flies were isolated by following a standardized
to the discovery of affected genes and important mecha- protocol. In brief, a given number of flies are exposed to
nistic insight into the disease. However, the majorities of few drops of diethyl ether in a small airtight glass container
cases are sporadic of unknown etiology or thought to be for 1 min. In case of over anesthetization, flies will be
the result of exposure to environmental toxins such as found dead with their wings stretched perpendicular to the
rotenone, paraquat etc. [15, 16]. body axis.
Rotenone, a high affinity specific inhibitor of mito-
chondrial complex I is capable of causing mitochondrial Preliminary Study
dysfunctions that phenocopies PD [17–19]. Chronic rote-
none treatment has been used in Drosophila to create a Preliminary studies were conducted with small fly numbers
pharmacological model of PD and induce both dopami- with three concentrations of Cr viz., 2, 5 and 10 mM to
nergic cell loss and locomotor impairments [13, 17]. determine the effect of Cr alone on the survival of the flies
Although Drosophila is employed to elucidate mechanistic during the experimental period. However, for the deter-
basis of various neurological disorders, it is less employed minative studies such as mortality and climbing assay, only
for evaluating potential therapeutic interventions. Since two concentrations viz., 5 and 10 mM per unit of medium
Drosophila allows rapid screening of putative neurothera- were chosen. Further, for co-exposure study only one
peutic agents, we have employed this model to obtain concentration of Cr viz., 10 mM was employed. The
mechanistic insights of specific neuroprotective com- selection of 10 mM concentration was based on recent
pounds under oxidative stress mediated neurotoxicty [20]. findings in cell models [41] and our own observation of
In this study, we examined the neuroprotective action of maximum reduction of endogenous MDA levels in flies.
creatine against rotenone induced mortality, mitochondrial
oxidative stress and neurotoxicty in Drosophila. We found Rotenone (Rot) Exposure
that Cr supplementation in flies not only reduced the
incidence of rotenone induced mortality, but also signifi- In a preliminary study, flies were exposed to rotenone at
cantly abrogated mitochondrial oxidative stress, restored concentrations of 250, 500 and 1000 lM for 7 days to
the depleted dopamine levels and attenuated rotenone- determine lethality. However, only one concentration of
induced neurotoxicity as determined in a negative geotaxis rotenone (500 lM) was employed to assess the neuropro-
assay. Further, flies given Cr prophylaxis exhibited resis- tective effect of Cr. For these studies, rotenone exposed
tance against paraquat intoxication. Based on our results flies were provided with Cr (10 mM) in the diet and the
we hypothesize that the neuroprotective action of creatine modulatory effect of Cr on rotenone-induced lethality,
in Drosophila may be largely ascribed to the combined locomotor dysfunctions, dopamine levels, oxidative
action as an antioxidant and its ability to mitigate mito- impairments in both cytosol and mitochondria were
chondrial oxidative stress. determined. For mortality studies, a minimum of 50 adult

123
1404 Neurochem Res (2010) 35:1402–1412

flies per replicate (three replicates) were exposed to Cr or were homogenized in sodium-phosphate buffer (0.1 M, pH
rotenone or combination of Cr and rotenone for 7 days. 7.4), followed by centrifugation at 25009g for 10 min at
Lethality profile of the flies were recorded every 24 h till 4°C. Supernatant was used directly to estimate the dopa-
the end of the experiment and expressed in terms of percent mine level by HPLC method [21].
mortality. Similarly, behavioral and biochemical investi-
gations were also carried out. Assay for Lipid Peroxidation and Hydroperoxide
Levels
Biochemical Measurements
Lipid peroxidation was measured by employing thiobar-
Following various treatments, flies were mildly anesthe- baturic acid (TBA). Briefly, the reaction mixture contained
tized using diethyl ether in a small airtight glass container 500 ll fly homogenate, 1.5 ml acetic acid (pH 3.5, 20%),
for 1 min. Quantification of oxidative markers viz., Mal- 1.5 ml of TBA (0.8% w/v), 200 ll sodium lauryl sulphate
ondialdehyde (MDA), hydroperoxide (HP) levels, reduced (SDS) (8% w/v). The mixture was heated in a boiling water
glutathione (GSH) content, total thiols (TSH) and activities bath for 45 min and adducts formed were extracted into
of antioxidant enzymes viz., catalase and superoxide dis- 3 ml of 1-butanol. The absorbance was measured at
mutase (SOD) were determined only in whole body 532 nm and quantified as malondialdehyde equivalents
homogenates. However, the dopamine levels were deter- using 1,1,3,3-tetramethoxypropane as the standard [22].
mined in head and body region of flies. Further, mito- Hydroperoxide generation was determined according to the
chondrial oxidative impairments were assessed in terms of method of Wolff [23]. An aliquot of homogenate (100 lg
reactive oxygen species (ROS) generation, nitric oxide protein) was added to 1 ml of FOX reagent (250 lM Fer-
(NO) levels and activities of complexes I–III and complex rous ammonium sulphate; 100 mM sorbitol; 25 mM
II–III were determined. In addition, cholinergic functions H2SO4; 100 lM xylenol orange), and incubated for 30 min
were measured in terms of the activities of acetylcholin- at room temperature. The absorbance was taken at 560 nm
esterase (AChE) and butyrylcholinesterase (BChE) in and expressed as nmoles hydroperoxides/mg protein.
whole body homogenates.
Determination of Reduced Glutathione and Total Thiols
Isolation of Cytosol and Mitochondria from Flies
Reduced glutathione (GSH) content was estimated
Whole body homogenate/cytosol was prepared by using according to the fluorimetric method employing o-phthal-
sodium-phosphate buffer, (0.1 M, pH 7.4). Following aldehyde (OPT). An aliquot of whole body homogenate
homogenization, samples were centrifuged at 25009g for was added to 0.1 M formic acid and spun at 52009g for
10 min at 4°C, supernatant filtered through nylon mesh 10 min to precipitate protein, then the supernatant was
(pore size, 10 lm) and used as cytosol for further bio- allowed to react with OPT (1 mg/ml in methanol) at room
chemical assays. Mitochondria from whole body of flies temperature for 30 min and fluorescence measured at
were prepared by differential centrifugation. Briefly, fly excitation of 345 nm and emission at 425 nm [24]. To
homogenate (4%) was prepared in ice-cold Tris–sucrose estimate total thiols, 200 ll of homogenate (400 lg pro-
buffer (Tris buffer, 2 mM and Sucrose, 250 mM) of tein) was added to 300 ll of tris–buffer (2 mM, pH 8.2),
0.25 M, pH 7.4 using a glass-Teflon grinder, and filtered 25ul of 5, 50 -dithiobis 2-nitrobenzoic acid (DTNB) (10 mM
through 10 lM tissue sieve. The filtrate was centrifuged in methanol) and 1.975 ll of methanol and allowed to
(25009g for 10 min), pellet was discarded and supernatant stand for 30 min for room temperature with occasional
further subjected to centrifugation at 78009g for 10 min to shaking. Following centrifugation at 30009g for 15 min,
obtain the nuclear pellet. Mitochondria were obtained by the supernatant was read at 412 nm against distilled water
centrifuging the post-nuclear supernatant at 10,0009g for blank and calculated using molar extinction coefficient
10 min. The pellet was washed mannitol-sucrose-HEPES (MEC) 13.6 mM-1 cm-1 [25].
buffer (Mannitol, 200 mM; Sucrose, 70 mM; EDTA,
0.1 mM and HEPES, 10 mM) and resuspended in the Enzyme Activities: Catalase and Superoxide Dismutase
buffer, stored at -20°C until future use.
Catalase activity was measured following the method of
Preparation of Homogenate from Head and Body Aebi [26]. To 1 ml reaction mixture containing 8.8 mM
Regions for Dopamine Estimation H2O2 (3%), 0.1 M sodium phosphate buffer, pH 7.0. The
reaction was initiated by adding an aliquot (equivalent to
Known numbers of fly heads were separated from rest of 10 lg protein). The decrease in H2O2 was monitored for
the body using a sharp blade. 20 heads and body regions 3 min at 240 nm and expressed as lmol of H2O2

123
Neurochem Res (2010) 35:1402–1412 1405

decomposed/min/mg protein (MEC-H2O2 44.1 mM-1 10 mM) pH 7.4, 0.1 ml homogenate (100 lg protein) and
cm-1). SOD activity was determined by monitoring inhi- 10 ll of DCFH-DA (5 lM) was incubated for 15 min at
bition of quercetin auto oxidation. Total volume of 1 ml room temperature to allow the DCFH-DA to be incorpo-
reaction mixture containing 3–5 lg protein, 0.016 M rated into any membrane bound vesicles and diacetate
sodium phosphate buffer, pH 7.8; N,N,N,N tetramethyl group to be cleaved by esterases. After 15 min of further
ethylenediamine (TEMED), 8 mM and ethylenediamine- incubation, the conversion of DCFH-DA to the fluorescent
teraacetic acid (EDTA, 0.08 mM) and reaction was started product DCF was measured in a spectrofluorometer with
by adding 0.15% quercetin dissolved in dimethyl form- excitation wavelength of 484 nm and emission at 530 nm.
amide (DMF). Reaction was monitored for 3 min at Background fluorescence was corrected by the inclusion of
406 nm, expressed as amount of protein required to inhibit parallel blanks. ROS formation was quantified from a DCF
50% of quercetin auto oxidation [27]. standard curve and data are expressed as pmoles DCF
formed/min/mg protein [30].
Nitric oxide levels were measured by using commer-
Activities of Acetylcholinesterase (AChE)
cially available Griess reagent (1.5% Sulphanilamide and
and Butyrylcholinesterase (BChE)
0.15% N-1-naphthyl-ethylene diamine in 1 N HCl) pro-
cured from M/s Sigma Chemicals (St. Louis, MO, USA).
AChE and BChE activities were determined according to
The principle of assay is based on the enzymatic conver-
the method of Ellmann [28]. To the reaction mixture
sion of nitrate to nitrite by nitrate reductase. The reaction is
containing 1 ml phosphate buffer (0.1 M, pH 8.0), 5,5-
based on two step diazotization reaction in which acidified
dithiobis 2-nitrobenzoic acid (DTNB,10 mM), sample
NO2 produces a nitrosating agent which reacts with sul-
(cytosol) and acetylthiocholineiodide (78 mM)/butyrylthi-
phanilic acid to produce the diazonium ion which is cou-
ocholine iodide were added and the change in absorbance
pled with NED, N-(1-napthylethylenediamine to form the
was monitored at 412 nm for 3 min. The enzyme activities
chromophoric azo derivative which absorbs light at
were expressed as nmoles of substrate hydrolyzed/min/mg
540 nm. Nitric oxide levels were quantified from a sodium
protein.
nitrate standard curve [31].
Assays in Mitochondria: Activities of NADH-
Cytochrome C Reductase (Complex I–III) Determination of Dopamine Levels by HPLC
and Succinate-Cytochrome C Reductase (Complex II–III)
Head/rest of the body were homogenized in 100/500 ll of
Mitochondria isolated from whole body of adult flies ice cold 0.1 M phosphate buffer (pH, 7.4) containing
(50 lg) were added to phosphate buffer (0.1 M, pH 7.4) 1 mM EDTA. Following centrifugation at 25009g for
containing NADH (0.2 mM) and KCN (1 mM). The reac- 10 min, the supernatant was filtered through sieve and
tion was initiated by addition of 0.1 mM cytochrome C and injected directly into HPLC column (C-18 column reverse
decrease in absorbance was monitored for 3 min at 550 nm. phase, Linchrospher 100RD-18, 14.5 cm, 5 lm, E. Merck)
The activity was expressed as lmol cytochrome C reduced/ equipped with ultraviolet detector set at 280 nm. Mobile
min/mg protein (molar extinction coefficient (MEC) phase consisted of 0.2% aqueous trifluoroacetic acid and
-19.6 mM/cm) [29]. An aliquot of mitochondria from methanol (70:30 v/v) and the flow rate was maintained at
whole body (50 lg) were added to phosphate buffer (0.1 M, 1 ml/min [20, 21].
PH 7.4, 2 mM EDTA) containing succinate (20 mM) and
KCN (1 mM). The reaction was initiated by addition of
Negative Geotaxis Assay (Climbing Assay)
0.1 mM cytochrome C and decrease in absorbance was
monitored for 3 min at 550 nm. The activity was expressed
Test flies were anesthetized and placed in a vertical glass
as lmol cytochrome C reduced/min/mg protein (molar
column (standard length, 25 cm; diameter, 1.5 cm). After a
extinction coefficient (MEC) -19.6 mM/cm) [29].
brief recovery period, flies were gently tapped to the bot-
tom of the column. Following 1 min, flies that reached the
Reactive Oxygen Species (ROS) and Nitric Oxide (NO) top of the column and flies that remained at the bottom
Levels were counted separately. Data was expressed as percent
flies escaped beyond minimum distance of 6 cm in 60 s of
ROS was assayed using dihydro dichlorofluorescein diac- interval [32]. Twenty adults per replication were used for
etate. Briefly, the reaction mixture (2 ml) containing assay. The assays were repeated three times and the score
Locke’s buffer (Nacl, 154 mM; Kcl, 5.6 mM; NaHCo3, for each replication was an average of three such trials for
3.6 mM; HEPES, 5 mM; CaCl2, 20 mM; D-Glucose, each group of insects including control.

123
1406 Neurochem Res (2010) 35:1402–1412

Paraquat Resistance Test between 4–7 days and terminally the cumulative percent
lethality was: 250 lM-14%; 500 lM-55%; 1000 lM-78%.
Paraquat is a well known in vivo free radical generator. In order to examine the modulatory effect of creatine, we
The ‘paraquat resistance’ test has been employed to mea- used only one concentration of rotenone (500 lM) and two
sure acquired resistance to oxidative stress. Flies were concentrations of creatine. Interestingly, co-exposure of
exposed to paraquat for 48 h (20 and 40 mM, in 5% flies with creatine (5 and 10 mM) resulted in lower inci-
sucrose solution) as described previously [20, 33]. The rate dence of mortality among rotenone exposed flies. The
of survival of flies among untreated controls and Cr treated degree of protection was robust (74 and 52%) (Fig. 1b)
group was recorded during 72 h of observation period. indicating its ability to protect against rotenone induced
lethality.
Determination of Protein
Creatine Protects Rotenone-Induced Locomotor Deficits
Protein concentrations in the cytosol and mitochondria of
whole body flies were determined by Lowry’s method [34] Data obtained in the negative geotaxis assay in flies
using bovine serum albumin as the standard. exposed previously to rotenone for 7 days revealed con-
centration dependent locomotor dysfunctions (Fig. 2a). At
Statistical Analysis higher concentrations of rotenone, more number of flies
had a tendency to stay at the bottom of vial. Among
Results are represented as the group means ± standard untreated controls, more than 94% flies were able to reach
error (SE) for each experimental group. The data was at the top of the vial within a minute, while rotenone
analyzed using one way ANOVA followed by post hoc exposed flies exhibited significant decrease in climbing
‘Tukey’ test and P value less than 0.05 was set as the ability with increasing concentration of rotenone (250 lM,
minimum level of significance. 44%; 500 lM, 19% and 1000 lM, 15%) clearly suggesting
the induction of locomotor deficits. In a parallel experi-
ment, we also measured the modulatory effect of Cr on
Results rotenone induced locomotor deficits in a similar paradigm.
Rotenone treated flies exhibited severe locomotor impair-
Effect of Creatine (Cr) on Endogenous Markers ments, while co-exposure with creatine significantly
of Oxidative Stress and Cholinergic Function improved (35 and 45%) the performances of flies (Fig. 2b)
in the negative geotaxis test. In general, co-exposed flies
A concentration related diminution in both MDA and appeared to be more active than rotenone alone treated
hydroperoxide levels was observed in whole body flies. Further, Cr at both the tested concentration alone had
homogenates of adult flies fed with Cr supplemented diet no significant effect on locomotor behavior among flies.
for 7 days (Table 1). The reduction in MDA levels were
more marked (17–44%) compared to the reduction in HP Effect on Rotenone-Induced Depletion of Dopamine (DA)
levels (17–21%). However, Cr did not have any effect on Levels
GSH and total thiols content in whole body homogenates.
Likewise, the activities of antioxidant enzymes viz., Cat- Cr alone treatment did not significantly affect the DA
alase and Superoxide dismutase also showed no significant levels measured either in head or body homogenates of
alterations. The activities of both acetylcholinesterase flies (Fig. 3). In contrast, rotenone caused robust DA
and butyrylcholinesterase enzymes among flies fed with Cr depletion in both the regions (head, 41%; body region,
for 7 days remained unchanged at all Cr concentrations 70%). Interestingly, the degree of rotenone induced DA
tested (Table 1). depletion was less robust among flies co-exposed to Cr.
Restoration of DA levels affected by Cr treatment was 50%
Protective Effect of Creatine Against Rotenone- in head and 28% in rest of the body region of flies.
Induced Neurotoxicty
Effect of Creatine Against Rotenone-Induced
Modulatory Effect of Creatine against Rotenone induced Mitochondrial Oxidative Stress
lethality
Effect on ROS Generation and GSH Levels
Exposure of adult flies to Rotenone resulted in a concen-
tration dependent lethality over a 7-day experimental per- Flies fed with Cr alone for 7d showed marginal (14%)
iod (Fig. 1a). Neurotoxicant induced deaths occurred decrease in endogenous ROS levels. On exposure to

123
Neurochem Res (2010) 35:1402–1412 1407

Table 1 Effect of Creatine (Cr) supplementation for 7 days on the endogenous markers of oxidative stress, activities of antioxidant enzymes and
cholinergic function in whole body homogenates of adult Drosophila melanogaster
Parameters Creatine (mM)
0 2 5 10

Malondialdehydea 3.51 ± 0.32 2.91 ± 0.23 2.26 ± 0.11* 1.98 ± 0.36*


Hydroperoxideb 0.52 ± 0.08 0.42 ± 0.009 0.43 ± 0.019* 0.41 ± 0.062*
Glutathionec 31.33 ± 0.89 34.34 ± 2.14 36.09 ± 1.02* 32.30 ± 2.27
Total thiolsd 0.07 ± 0.005 0.064 ± 0.004 0.076 ± 0.003 0.067 ± 0.006
SODe 204.2 ± 1.73 244.1 ± 18.2 253.2 ± 38 238.2 ± 28.3
f
Catalase 0.174 ± 0.01 0.178 ± 0.003 0.171 ± 0.014 0.136 ± 0.005
Acetylcholinesteraseg 0.080 ± 0.001 0.084 ± 0.005 0.083 ± 0.0034 0.076 ± 0.009
Butyrylcholinesteraseg 0.041 ± 0.002 0.041 ± 0.002 0.043 ± 0.002 0.038 ± 0.004
Adult flies were provided with creatine in feed at concentrations of 2, 5 and 10 mM for 7 days. All biochemical parameters were quantified in
whole body homogenates
Values are mean ± SE (Three sets of flies). Data analyzed by one way ANOVA followed by post hoc ‘Tukey’ test (*P \ 0.05)
a
gmol MDA/mg protein; bgmol hydroperoxides/mg protein; clg GSH/mg protein; dmmol DTNB/min/mg protein; eUnits/min/mg protein; flmol
H2O2 hydr/min/mg protein; gqmol DTNB/min/mg protein
For more details, see ‘‘Material and Methods’’

A 100 rotenone, mitochondria obtained from whole body


Rotenone 250µM Rotenone 500µM
homogenate of flies revealed marked (43%) increase in
Rotenone 1000µM
80 ROS levels. Interestingly, co-exposure with Cr completely
Mortality (%)

abrogated the rotenone induced ROS generation as


60
revealed by near normal levels of ROS (nearly 90% pro-
tection) (Fig. 4a). While rotenone only treated flies showed
40
significant (31%) depletion of GSH levels, flies co exposed
to creatine exhibited normal GSH levels (Fig. 4b). Further,
20
GSH levels among Cr alone treated flies remained
0 unaltered.
96 120 144 168
Exposure time (hrs) Effect on the Activity of Mn-SOD and Nitric Oxide (NO)
Levels
B 100

80 Cr treatment alone did not alter the activity levels of


Mortality (%)

Mn-SOD enzyme measured in mitochondria. However,


60
rotenone alone treated flies showed elevated (by 19%) SOD
40 activity. Surprisingly, co-exposed (rotenone and creatine)
20
flies showed normalized activity compared to rotenone
treated group (Fig. 5a). Flies exposed to Cr (10 mM) alone
0 showed significantly decreased endogenous nitric oxide
M

ot

ot
M
l
tro

0m

levels (by 33%). However, rotenone exposure resulted in



m

R
on

r5

50

+
r1

M
C

significant increase in nitric oxide levels (29%). Interest-


C

ot

0m
r5
R

r1
C

ingly, Cr supplementation resulted in complete restoration


C

of NO to normal levels (Fig. 5b).


Fig. 1 Rotenone (Rot) induced mortality response in Drosophila
melanogaster and its modulation by creatine (Cr) supplementation.
a Mortality pattern among adult Drosophila exposed to different Effect on the Activities of ETC Enzymes
concentrations of Rotenone in feed (250, 500 and 1000 lM).
Rotenone exposure caused a concentration dependent lethality. Cr alone fed flies showed no change in the complex I–III
b Modulation of Rotenone induced mortality among flies co-exposed
with creatine. Cr supplementation significantly reduced the incidence
activity compared to control. However, significant inhibi-
of Rotenone induced mortality tion (29%) was evident among rotenone only treated flies.

123
1408 Neurochem Res (2010) 35:1402–1412

A 100 Head Rest of body

µg dopamine/mg protein
15
* *
80
% Flies escaped/min

12

9
60
6
* *
40 3

M
20

ot

ot
ol

ol
M

M


0m

0m
tr

tr
+R

+R
on

on
50

50
M

M
r1

r1
C

C
0m

0m
ot

ot
C

C
R

R
r1

r1
0

C
0 250 500 1000
Fig. 3 Modulatory effect of creatine (Cr) supplementation on
Rotenone (µM) Rotenone (Rot)-induced dopamine depletion among Drosophila
melanogaster determined in homogenates prepared from head and
B body region separately. Cr supplementation significantly restored
100
Rotenone induced dopamine depletion in both head and body regions.
% Flies escaped/min

Values are mean ± SE (in triplicates), data was analyzed by one way
80
ANOVA followed by post hoc Tukey test (*P \ 0.05). Significances
were determined by making comparisons between control v/s Cr, Rot;
60
Rot v/s Rot ? Cr. (n = 50 flies per replicate, three such replication
used for assay)
40

20 supplemented flies were able to survive for longer time


compared to rotenone alone treatment at both observation
0
times (Fig. 6). The percent protection at 48 h was—33 and
ol

ot

ot
M
M

52% and at 72 h; 49 and 47% at both 20 and 40 mM of PQ,


tr

R
0m
on

r5

+
50
r1

M
C

respectively.
M
C

0m
ot

m
C

r5

r1
C

Fig. 2 Rotenone (Rot) induced locomotor deficits (expressed as


percent flies escaped) and its modulation by creatine (Cr) supple-
Discussion
mentation determined in a negative geotaxis assay. a Rotenone
exposure resulted in a concentration dependent locomotor impairment Although there is lack of certainty on the pathophysiology
in adult Drosophila melanogaster. b Modulation of Rotenone induced of neurodegenerative mechanisms, it is well accepted that
locomotor deficits among flies given Cr supplementation. (n = 50
flies per replicate, three such replication used for assay). Cr
energy depletion, oxidative stress and mitochondrial dys-
supplemtation markedly improved the performance of Rotenone functions are vital factors associated with most of these
treated flies in the negative geotaxis assay disorders [35, 36]. Drosophila is widely used as a model to
understand the pathophysiolgy of several neurodegenera-
Interestingly, the activity levels of complex I–III were tive diseases (NDD). In recent times it is also effectively
completely restored among Cr co-exposed flies (Table 2). serving as an easy platform to screen several phyto-
Further, Cr alone fed flies showed no change in the activity chemicals for their neuroprotective properties and is
level of complex II–III enzyme. Furthermore, rotenone anticipated to be highly valuable for first-step drug screen.
alone and co-treatment with creatine also failed to modu- Previously, using Drosophila we demonstrated the pro-
late the activity level of complex II–III. tective effects of Bacopa monnieri against rotenone
induced neurotoxicty [20]. In this study we tested the
Creatine Prophylaxis Confers Resistance to Paraquat hypothesis that creatine supplementation can significantly
Exposure offset rotenone induced mitochondrial oxidative stress in
Drosophila and rescue the flies from the neurotoxic
Initially, we determined the LC50 concentration of paraquat consequences.
among adult flies. At 24 h in both the tested concentration Creatine has been widely used as an ergogenic aid to
no significant mortality was recorded. However, significant improve exercise performance in humans and also to
mortality was evident at 48 and 72 h. The incidence of ameliorate oxidative stress mediated diseases. However,
mortality was: 15, 42% at 48 h and 39 & 95% at 72 h the underlying mechanism/s of its action are less well
at both the concentration, respectively. Interestingly, Cr understood in vivo [37–39]. In the present study, flies

123
Neurochem Res (2010) 35:1402–1412 1409

* * A 600
* *
A

Units/min/mg protein
0.20
picomole dichloro flouroscein

Mn-SOD activity
formed/mg protein/min

0.15 400

0.10
200

0.05

0
0.00 Control Cr 10mM Rot 500µM Cr +Rot
Control Cr 10mM Rot 500µM Cr +Rot

B 0.8
* *
B 100 *

Absorbance/mg protein
*
µg GSH/mg protein

0.6

Nitrite formed
75

0.4
50

0.2
25

0
0 Control Cr 10mM Rot 500µM Cr +Rot
Control Cr 10mM Rot 500µM Cr +Rot
Fig. 5 Modulatory effect of creatine (Cr) supplementation on
Fig. 4 Modulatory effect of creatine (Cr) supplementation on mitochondrial Superoxide dismutase activity (Mn-SOD) (a) and
Rotenone (Rot)-induced oxidative stress measured as reactive oxygen nitric oxide (NO) levels (b) in whole body mitochondrial fractions of
species (ROS) (a) and reduced glutathione (GSH) (b) levels in whole Drosophila melanogaster exposed to Rotenone. Mitochondrial SOD
body mitochondrial fraction of Drosophila melanogaster. Cr supple- activity and NO levels were markedly restored to near normalcy
mentation significantly protected against Rotenone induced ROS among flies given Cr Supplementation. Values are mean ± SE (in
generation and GSH depletion. Values are mean ± SE. Data was triplicates), data was analyzed by one way ANOVA followed by post
analyzed by one way ANOVA followed by post hoc Tukey test hoc Tukey test (*P \ 0.05). Significances were determined by
(*P \ 0.05). Significance was determined by making comparisons making comparisons between control v/s Cr, Rot; Rot v/s Rot ? Cr.
between control v/s Cr, Rot; Rot v/s Rot ? Cr. (n = 50 flies per (n = 50 flies per replicate, three such replication used for assay). For
replicate, three such replication used for assay). For more details, see more details, see ‘‘Material and Methods’’
‘‘Material and Methods’’
ABTS?, superoxide anions and peroxynitrite radicals were
provided with Cr supplements exhibited significant reduc- demonstrated in vitro [40]. Studies in cell models also
tion in the endogenous levels of oxidative markers such as demonstrated the direct antioxidant activity of Cr via a
malondialdehyde and hydroperoxides. While the lowest scavenging mechanism in oxidatively injured mammalian
concentration of Cr had no appreciable effect, at higher cells and its DNA protective action against oxidative attack
concentrations significant diminitution of MDA and [41, 42]. Further long term Cr supplementation in aged
hydroperoxide levels in whole body homogenates sug- mice was reported to diminish the ROS levels, decrease the
gested its potential to modulate endogenous levels of oxi- accumulation of lipofuscin pigments in brain and extend
dative markers. These findings of diminished levels of longevity [43]. In a recent study, Cr supplementation was
oxidative markers associated with enhanced GSH levels in shown to reduce endogenous lipid peroxidation biomarkers
whole body homogenates clearly suggest the antioxidative in rats suggesting a protective role against oxidative dam-
effects of Cr. Owing to higher reduction in MDA levels age [44].
observed with 10 mM Cr, we employed this concentration Rotenone, a respiratory chain complex I inhibitor is
for our further experimentation. The observed effects in hypothesized to be a vital environmental toxin (along with
flies are consistent with previous reports on the direct other chemicals such as MPTP and Paraquat) contributing
antioxidant properties of Cr both under in vitro and in vivo significantly towards the occurrence of sporadic PD in
conditions. Previously the potential of Cr to scavenge humans [45]. Chronic exposure of Drosophila to sub lethal

123
1410 Neurochem Res (2010) 35:1402–1412

Table 2 Effect of dietary supplementation of Creatine (Cr) (10 mM) Earlier findings in Drosophila viz., prevention of rotenone
against rotenone (500 lM) induced mitochondrial dysfunctions induced locomotor deficits and DA neuronal loss by the
measured in whole body mitochondrial fractions of adult Drosophila
antioxidant melatonin clearly suggest the involvement of
melanogaster
oxidative stress mechanisms [47]. Interestingly, similar
Groups Enzyme activity signs of oxidative damages have been reported frequently
Complex I–IIIa Complex II–IIIb in dopaminergic neurons from PD patients, suggesting
implication of oxidative stress in this disease [48, 49]. In
Control 0.059 ± 0.008 0.048 ± 0.003
the present study, rotenone induced significant oxidative
Creatine, 10 mM 0.049 ± 0.009 0.040 ± 0.003
markers as evidenced by enhanced MDA and hydroper-
Rotenone, 500 lM 0.042 ± 0.005* 0.043 ± 0.001
oxide levels, elevated activities of antioxidant enzymes
Creatine ? rotenone 0.064 ± 0.004* 0.037 ± 0.001 (Catalase and SOD). These findings are consistent with our
Adult flies were exposed to rotenone (500 lM) with or without cre- own findings in Drosophila [20] and previous reports in
atine (10 mM) for 7 days and mitochondria isolated from whole body rats exposed to rotenone [50, 51]. Severe depletion in
homogenates were assayed for the activities of ETC enzymes; Values
cellular GSH levels upon rotenone exposure in Drosophila
are mean ± SE (Three sets of flies). Data analyzed by one way
ANOVA followed by post hoc ‘Tukey’ test (*P \ 0.05) adds further evidence that a state of oxidative stress exists
a, b
lmol cytochrome c reduced/min/mg protein in vivo which may lead to mitochondrial damage and
For more details, see ‘‘Material and Methods’’ increase in free radical generation.
Cr supplementation restored the depleted GSH pool and
normalized the ROS levels in rotenone treated flies. This
* * protective action may be chiefly attributable to the direct
100 quenching of free radicals or alternatively due to the up
regulation of antioxidative defenses. While the primary
80 48 hrs 72 hrs effect of Cr supplementation on GSH synthesis in flies
needs further investigations, it may be the result of de novo
Mortality (%)

60
synthesis of GSH and phase II detoxification in response to
* *
* * rotenone induced altered redox status. The lower incidence
of mortality among Cr supplemented flies clearly suggests
40
the protective action of Cr. Furthermore, rotenone induced
* neurotoxicty was clearly evidenced by high rate of loco-
20 motor deficits measured in the negative geotaxis assay.
Flies with locomotor deficits have tendency to stay at the
0 bottom of vial and do not appear to coordinate their legs in
normal fashion. The development of this phenotypic
PQ mM

PQ mM
M

M
ol
M

M
ol

PQ mM

PQ M

M
M
m

m
0m

0m
tr
tr

m
m

on
on

40
40
20

20
20

20

40
40
r1

r1

expression of among flies exposed to rotenone is speculated


C

C
PQ

PQ
PQ

PQ
C

C
+

to be due to the deficits in high energy levels of ambulatory


+

M
M

M
M

0m
0m

0m
0m

and flight muscles which are rich in mitochondria. Hence it


r1

r1

r1

r1
C

is likely that uncoupled mitochondrial machinery may be


Fig. 6 Paraquat (PQ) induced mortality response in Drosophila responsible of the induced locomotor dysfunctions as evi-
melanogaster and its modulation by creatine (Cr) prophylaxis. Time
course lethality response expressed as percent mortality among flies
denced by severe complex I inhibition. Another possible
exposed to various concentrations of PQ and its modulation by Cr reason might be due to the differential and significant
prophylaxis (n = 50 flies per replicate, three such replication used for depletion of dopamine pool as observed in both head and
assay). For more details, see ‘‘Material and Methods’’ body homogenate. Interestingly Cr supplementation was
able to rescue the flies significantly from deteriorating
doses of rotenone is well demonstrated to recapitulate the locomotor dysfunctions indicating its potential to restore
main symptomatic features of PD viz., a selective loss of dopamine pool and protect the mitochondrial function. This
dopaminergic neurons including locomotor defects [17, thinking is consistent with earlier findings which suggest
46]. In the present model, rotenone induced a concentration significant correlation between locomotor dysfunction and
related mortality and locomotor deficits in adult flies, data dopamine deficiency [13]. Although speculative, the better
which is consistent with previous reports [13, 46]. While performance resulting from Cr supplements among rote-
the exact mechanism of rotenone action leading to neuro- none exposed flies could be partly related to its well known
toxicity is not well understood, in the insect model, rote- physiological role in the maintenance of ATP/ADP ratio
none action is attributed to specific sensitivity of resulting in enhanced mitochondrial respiration [1, 52].
dopaminergic neurons to ROS and oxidative damage [45]. However further investigations are necessary to delineate

123
Neurochem Res (2010) 35:1402–1412 1411

the direct effects of Cr on flight muscles among flies 5. Watanabe A, Kato N, Kato T (2002) Effects of creatine on mental
exposed to rotenone. fatigue and cerebral haemoglobin oxygenation. Neurosci Res
42:279–285
Employing a prophylactic approach, we obtained further 6. Mc Moris T, Harris RC, Howard AN et al (2007) Creatine sup-
evidence that Cr has the potential to confer protection plementation, sleep deprivation, cortisol, melatonin and behav-
against paraquat induced oxidative insult. Paraquat, a well iour. Physiol Behav 90:21–28
known herbicide is demonstrated to cause selective 7. Baker SK, Tornopolsky MA (2003) Targeting cellular energy
production in neurological disorders. Expert Opin Investig Drugs
degeneration of dopaminergic neurons in substantia nigra 12:1655–1679
pars compacta in animal models [53]. It is known to induce 8. Bender A, Koch W, Elstner M et al (2006) Creatine supple-
oxidative stress in mammals by participating in redox mentation in Parkinson disease: a placebo-controlled randomized
cycling with cellular enzymes [54, 55] and in Drosophila pilot trial. Neurology 67:1262–1264
9. Bender A, Samtleben W, Elstner M et al (2008) Long-term cre-
[56]. Data obtained in our bioassay revealed significant atine supplemetaion is safe in aged patients with Parkinson’s
reduction in the degree of paraquat-induced lethality disease. Nutr Res 28:172–178
among flies given Cr prophylaxis clearly indicating that 10. Andres RH, Huber W, Pérez-bouza O et al (2005) Effects of
flies were able to successfully mitigate the PQ induced creatine treatment on the survival of dopaminergic neurons
in cultured fetal ventral mesencephalic tissue. J Neurosci
oxidative stress resulting in better survival rate. Further 133:701–713
investigations on the mechanism by which Cr protects flies 11. Lensman M, Korzhe Vskii DE, Mourovets VO et al (2006)
against PQ induced oxidative stress and neurotoxicty is Intracerebroventricular administration of creatine protects
underway. against damage by global cerebral ischemia in rat. Brain Res
1114:187–194
In conclusion, we have demonstrated the neuroprotec- 12. Sestili P, Martinelli C, Bravi G et al (2006) Creatine supple-
tive efficacy of creatine supplementation in Drosophila as mentation affords cytoprotection in oxidatively injured cultured
evidenced by its ability to modulate endogenous oxidative mammalian cells via direct antioxidant activity. Free Radic Biol
markers and its propensity to mitigate rotenone induced Med 40:837–849
13. Celotto AM, Palladino MJ (2005) Drosophila, a model system to
mitochondrial oxidative stress, restore dopamine levels and study neurodegeneration. Molecular Interventions 5:292–303
neurotoxicty. Additional evidence of its neuroprotective 14. Reiter RJ, Acuna-Castrviej OD, Tan DX et al (2001) Free radical
action was exemplified by the enhanced resistance to mediated molecular damage, mechanisms for the protective
Paraquat. While the precise mechanisms by which creatine actions of melatonin in the central nervous system. Ann NY Acad
Sci 939:200–215
offers neuroprotection in this model needs further investi- 15. Francesca C, Drouin-Ouellet J, Robert EG (2009) Environmental
gation, we hypothesize that it may be due to the combined toxins and Parkinson’s disease: what have we learned from pes-
effects of its antioxidative action as well as direct physio- ticide-induced animal models? Trends Pharmacol Sci 30:475–483
logical effects on mitochondria. More importantly, these 16. Dinis-Oliveira RJ, Remia F, Carmo H et al (2005) Paraquat
exposure as an etiological factor of Parkinson’s disease. Neuro-
results confirm the utility of Drosophila as a primary tool Toxicology 27:1110–1122
to rapidly screen compounds suspected to possess neuro- 17. Coulom H, Birman S (2004) Chronic exposure to rotenone
pharmacolgical properties prior to their testing in mam- models sporadic Parkinson’s disease in Drosophila melanogaster.
malian models and further therapeutic use in humans. J Neurosci 24:10993–10998
18. Sherer TB, Betarbet R, Stout AK et al (2002) An in vitro model
of Parkinson’s disease: linking mitochondrial impairment to
Acknowledgments We wish to thank the Director, CFTRI for his altered a-synuclein metabolism and oxidative damage. J Neurosci
encouragement in this study. The first author (RKH) thanks the 22:7006–7015
University Grant Commission (UGC), New Delhi, India for the award 19. Betarbet R, Sherer TB, Mackenzie G et al (2000) Chronic sys-
of Junior/Senior Research Fellowships. temic pesticide exposure reproduces features of Parkinson’s dis-
ease. Nat Neurosci 3:1301–1306
Declaration of Interest The authors report no conflicts of interest. 20. Ravikumar H, Muralidhara (2009) Neuroprotective efficacy of
The authors alone are responsible for the content and writing of the Bacopa monnieri against rotenone induced oxidative stress and
manuscript. neurotoxicty in Drosophila melanogaster. NeuroToxicology
30:977–985
21. Dalpiaz A, Filosa K, de Caprariis P et al (2007) Molecular
mechanism involved in the transport of a prodrug dopamine
References glycosyl conjugate. Intl J Pharm 336:133–139
22. Ohakawa H, Ohishi U, Yagi K (1979) Assay of lipid peroxidation in
1. Gualano B, Artioli GG, Poortmans JR et al (2010) Exploring ther- rat tissues by thiobarbituric reaction. Anal Biochem 95:145–149
apeutic role of creatine supplementation. Amino acids 38:31–44 23. Wolf SP (1994) Ferrous ion oxidation in presence of ferric ion
2. Wyss M, Kaddurah-Daouk R (2000) Creatine and creatine indicator xylenol orange for measurement of hydroperoxide.
metabolism. Physiol Rev 80:1107–1213 Methods Enzymol 233:182–189
3. Greenhalf PL (2001) The creatine phospho creatine system: 24. Hissin PJ, Hilf R (1976) A flourimetric method for determination
there’s more than one song in its repertoire. J Physiol 537:657 of oxidized and reduced glutathione in tissues. Anal Biochem
4. Froiland K, Koszewski W, Hingst J et al (2004) Nutritional 74:214–216
supplement use among college athletes and their sources of 25. Ellman A (1959) Tissue sulfhydral groups. Arch Biochem Bio-
information. Intl J Sport Nutr Exerc Metab 14:104–120 phys 82:70–77

123
1412 Neurochem Res (2010) 35:1402–1412

26. Aebi H (1984) Catalase in vitro. Methods Enzymol 05:121–125 43. Bender A, Beckers J, Schneider I et al (2008) Creatine improves
27. Vladimir A, Kostyuk A, Potapovich AI (1989) Superoxide driven health and survival of mice. Neurobiol Aging 29:1404–1411
oxidation of quercetin and a simple sensitive assay for determi- 44. Deminice R, Portari GV, Vannuchi H et al. (2009) Effects of
nation of superoxide dismutase. Biochem Intl 19:117–1124 creatine supplementation on Homocysteine levels and lipid per-
28. Ellmann GE, Courtney KD, Anderson V et al (1961) A new oxidation in rats. British J Nutr 102:110–116
calorimetric determination of acetyl cholinesterase activity. 45. Sherer TB, Betarbet R, Testa CM et al (2003) Mechanism of
Biochem Pharmacol 7:88–95 toxicity in rotenone models of Parkinson’s disease. J Neurosci
29. Navarro A, Sanchez Del Pino MJ, Gomez C et al (2002) 23:10756–10764
Behavioral dysfunction, brain oxidative stress and impaired 46. Chowdhary A, Bowling K, Funderburk C et al (2007) Interaction
mitochondrial electron transfer in aging mice. Am J Physiol of genetic and environmental factors in a Drosophila Parkin-
Regul Integ Comp Physiol 282:985–992 sonism model. J Neurosci 27:2457–2467
30. Driver AS, Kodavanti PS, Mundy WR (2000) Age related 47. Tan DX, Reiter RJ, Manchester LC (2002) Chemical physical
changes in reactive oxygen species production in rat brain properties and potential mechanisms: melatonin as a broad-
homogenates. Neurotoxicol Teratol 22:175–181 spectrum antioxidant and free radical scavenger. Curr Top Med
31. Choi BM, Pae HO, Jang SI et al (2002) Nitric oxide as a proa- Chem 2:181–198
poptotic as well as antiapoptotic modulator. J Biochem Mol Biol 48. Coto-montes A, Hardeland R (1999) Antioxidant effects of
35:116–126 melatonin in Drosophila melanogaster: antagonization of damage
32. Feany MB, Bender WW (2000) A Drosophila model of Parkin- induced by the inhibition of catalase. J Pineal Res 27:154–158
son’s disease. Nature 404:394–398 49. Beal MF (2003) Mitochondria, oxidative damage and inflam-
33. Lin A (1998) Extended lifespan and stress resistance in the mation in Parkinson’s disease. Ann NY Acad Sci 991:120–131
Drosophila mutant Methuselah. Science 282:943–946 50. Jennar P (2003) Oxidative stress in Parkinson’s disease. Ann
34. Lowry OH, Rosebrough NJ, Farr AL et al (1951) Protein mea- Neurol 53:S26–S38
surement using Folin phenol reagent. J Biol Chem 193:265–275 51. Bove J, Prou D, Perier C et al (2005) Toxin-induced models of
35. Dawson TM, Dawson VL (2003) Molecular pathways of neuro- Parkinson’s disease. J Am Exptl Therpeutic 2:484–494
degeneration in Parkinson’s disease. Science 302:819–822 52. Mccling JM, Gand GA, Davis JM et al (2003) Effect of creatine
36. Lin MT, Beal MF (2006) Mitochondrial dysfunction and oxida- supplementation on cardiac muscle of exercise stress rats. Europ J
tive stress in neurodegenerative diseases. Nature 443:787–795 Appl Physiol 89:26–33
37. Wyss M, Schulze A (2002) Health implications of creatine: can 53. Dupuis L, Oudart H, Rene E et al (2004) Evidence for defective
oral creatine supplementation protect against neurological and energy homeostasis in amyotrophic lateral sclerosis; benefit of
atherosclerotic disease? J Neurosci 112:243–260 high energy diet in transgenic mouse model. Proc Natl Acad Sci
38. Persky AM, Brazeau GA (2001) Critical pharmacology of the US 101:11159–11164
dietary supplement creatine monohydrate. Pharmacol Rev 54. Peng J, Peng L, Stevenson FF et al (2007) Iron and paraquat as
53:161–176 synergistic environmental risk factors in sporadic Parkinson’s
39. Andres RH, Ducry AD, Schlattner U et al (2008) Functions and disease accelerates age-related neurodegeneration. J Neurosci
effects of creatine the central nervous system. Brain Res Bull 27:6914–6922
76:329–343 55. Cocheme HM, Murphy MP (2008) Complex I is the major site of
40. Lawler JM, Barnes WS, Wu G et al (2002) Direct antioxidant mitochondrial superoxide production by paraquat. J Biol Chem
properties of creatine. Biochem Biophysic Res Commun 290:47–52 283:1786–1798
41. Guidi C, Potenza L, Sestili P et al (2008) Differential effect of 56. Ravikumar H, Muralidhara (2010) Paraquat induced oxidative
creatine on oxidatively injured mitochondrial and nuclear DNA. perturbations and lethality in Drosophila melanogaster are miti-
Biochem Biophys Acta 1780:16–26 gated by prophylactic treatment with Bacopa monnieri. Indian J
42. Azzi A, Davies KJ, Kelly F (2004) Free radical biology-termi- Biochem Biophys 47:75–82
nology and critical thinking. FEBS Lett 558:3–6

123

You might also like