Download Ecstasy-induced toxicity in rat liver

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Glucose wikipedia , lookup

Glycolysis wikipedia , lookup

Ketosis wikipedia , lookup

Pharmacometabolomics wikipedia , lookup

Glyceroneogenesis wikipedia , lookup

Wilson's disease wikipedia , lookup

Transcript
Z ooo
t',,,,,,.,;,,,1>,.
........
_ 4//,.,_,h_,
,,.,,.,,,.,;
Ecstasy-induced
_
_[iver
toxicity in rat liver
Beitia G, Cobreros A. Sainz L, Cenarruzabeitia
ity in rat liver.
Liver 2000: 20: 8-15. C: Munksgaard, 2(/00
E. Ecstasy-induced
toxic-
Abstract: BackgroundAims." The aim of the present study was to examine
the effects of single and repeated administration
of 3.4-methylenedioxymethamphetamine (MDMA, "ecstasy") on rat liver. Mefllods." Animals were
given an acute (20 mg/kg) and repeated (20 mg/kg, b.i.d., for 4 consecutive day's) intraperitoneal dose of MDMA. and at various times after administration the hepatic and serum determinations were made. Resuhs. The
effect of acute MDMA administration
included increased triglyceride and
cholesterol levels and an increase in all enzyme activities 6 h post administration. The toxic effect of MDMA was also observed in other hepatic
processes. Gly'cogen content showed a marked decrease, which was accompanied by' a decrease in serum glucose levels. No significant changes
in lipid peroxidation and hepatic GSH content were observed. In contrast, multiple MDMA administration produced some evidence of oxidative stress, namely, increased MDA content and decreased GSH content, a
small decrease in liver glycogen at 3 h recovering 6 h post dose. no effect
on blood glucose and increased AST and ALP activities but no effects
' on ALl" activity. Seven days after the last MDMA injection a tendency
towards recovery was shown. Com'lusiom Our results show that the liver
. toxicity' caused by' MDMA administration
involves several mechanisms,
3,4-methylenedioxymethamphetamine
(MDMA)
commonly' known as "ecstasy" is a synthetic amphetamine derivative that rapidly became a widely
abused drug (1). Patented in 1914 by the E. Merck
Company as an appetite suppressant,
it was never
marketed, but in the 1970s and early 1980s the
drug gained popularity
as an adjunct to psychotherapy (2). Since MDMA's
classification
in 1985
as a Schedule I drug under the Controlled
Substances Act (3), there has been an increase in clandestine synthesis and illicit use which has been accompanied by an equivalent increase in the number
of
MDMA
toxicity
reports.
MDMA
intoxication
is predominantly
associated
with the
ability of the drug to destroy serotonergic
nerve
terminals
(4-7), but there are some reported cases
of severe toxicity resulting from recreational misuse of small amounts of MDMA in which the pattern of toxicity included
cardiac
arrhythmias,
fulminate hyperthermia,
convulsions,
disseminated
intravascular
coagulation,
rhabdomyolysis,
and
acute renal failure (8-10). Hepatotoxicity
effects
have only recently been noted. However, there is
increasing evidence that "ecstasy"
is hepatotoxic,
and liver changes have been reported following bi8
be ,'4--',
o.- !
G. Beitia, A. Cobreros,L. Sainzand
E. Cenarruzabeitia
Department
of Pharmacology,
University
of
Navarra. 31008 Pamplona, Spain
Key words: 3,4-methylenedioxymethamphetamine(MDMA)- liver - lipid per0xidation -
reducedglutathi0ne(GSH)- glycogen
Dr.E.Cenarruzabeitia.
University of Navarra,
SchoolofPharmacy,
c/Irunlarrea
1,
31008Pamplona,
Spain
Received
23 October1998.acceptedfor
publication5 August1999
opsy, which varied from foci of individual cell necrosis to centrilobular
necrosis, hepatitis and extensive fibrosis (1 1-13). The principal evidence of
hepatic lesions, resulting from MDMA in humans,
is an unexplained jaundice or hepatomegaly
(14,
15). Acute hepatitis has also been associated with
repeated use of ecstasy (16, 17).
The aim of the present study was to obtain evidence for the mechanism involved in MDMA-induced hepatotoxicity
in rats. For this purpose, several parameters
of rat liver function were determined.
Materials and methods
Drugs used
The MDMA was obtained from the Audiencia
Provincial de Navarra. All other reagents were of
the highest grade commercially available and were
obtained
from E. Merck (Darmstadt,
Germany)
and Sigma Chemical Company
(St. Louis, MO,
U.S.A.). Catalase, glutathione reductase, glutathione peroxidase,
glutathione-S-transferase
and superoxide dismutase were purchased
from Randox
Laboratories
Ltd. (Crumlin, UK).
Ecstasy-induced injury
Animalsand treatments
Measurementof hepatic reducedglutathione(GSH)levels
Male Wistar rats (200-220 g) were housed in plastic cages in a room with a controlled temperature
(22°C) and maintained on a 12 h light-dark cycle
with free access to food and water. Two different
schedules of treatment with MDMA were used in
this study. Acute treatment consisted of a single
dose of MDMA (20 mg/kg, i.p.), the rats being
sacrificed 3 h and 6 h later. Repeated treatment
consisted of the same dose of MDMA (20 mg/kg,
i.p.) which was given b.i.d, for 4 consecutive days,
the rats being sacrificed 3 h, 6 h and 7 days later,
The dose of MDMA (20 mg/kg, i.p.) was chosen
because it is in a similar range of doses (10-20 rag/
kg) that have been shown to produce neurotoxicity
in rats (17). MDMA dose refers to the hydrochloride. The time points (3 h, 6 h and 7 days) were
selected because of the reported neurotoxic effects
(18).
Hepatic GSH content was estimated by a colorimetric method as previously described by Ellman
(21). Samples of liver (0.3 g)were homogenised in 6
ml of 10% Trychloroacetic acid, containing 5 mM
sodium ethylenediamine tetraacetic acid (EDTA),
and later centrifuged at 3000×g for 15 min. The
volume of the supernatants was fixed in 10 ml by
addition of phosphate buffer (pH 7.4). Aliquots of
1.0 ml were taken and diluted to 10 ml with phosphate buffer. Then, 3.750 ml of these dilutions was
mixed with a solution of 5,5'-dithiobis-(2-nitrobenzoic acid) (DTNB) (0.08mg/ml in phosphate buffer) and incubated in a bath at 37°C for 30 rain.
After cooling, the absorbance of each sample was
determined at 412 nm, and the GSH concentration
of these samples was calculated with a standard
curve using a dissolution of GSH (0.12 mg/ml) as
standard.
Measurement of lipid peroxidation
Measurement of antioxidant enzymatic activities:
superoxide dismutase (SOD), glutathione peroxidase (GPx),
The extent of lipid peroxidation,
which can be
measured by the formation of malondialdehyde
(MDA) after the breakdown of polyunsaturated
fatty acids, was measured by assay for thiobarbituric acid reactive substances (TBARS) at 535 rim,
as described by Buegue and Aust (19). Briefly, liver
samples were homogenised with 50 mM Tris buffer, pH 7.5. Then, 2 ml of TBA reagent (0.375%
w/v thiobarbituric acid, 15°/,, w/v trichloroacetic
acid in 0.25 N HC1) was added to a fraction from
the homogenate samples. The mixture was heated
for 10 min in a boiling water bath, allowed to cool
and then centrifuged at 1000Xg for 10 rain.
TBARS are presented as malondialdehyde equivalents, calculated using an extinction coefficient of
1.56×105 M-_/cm -_
Liver protein content was evaluated by the
method of Bradford using bovine serum albumin
(BSA) as standard (20).
glutathione-S-transferase (GST), glutathi0ne reductase
(GR)and catalase (CAT)
Animals were sacrificed by decapitation and their
livers quickly removed and placed in homogenisation medium (250 mM mannitol, 70 mM sucrose,
1 mM EDTA adjusted with Tris to pH 7.4). One
part of the liver was immediately homogenised in a
Potter-Elvehjem glass homogeniser (1 g of tissue
plus 10 ml homogenisation medium). Unbroken
cells, cell debris, and nuclei were sedimented at
800×g for 10 min, and the supernatant was used as
homogenate. This was kept on ice while being sonicated at 300 w for 30 secondsin three 10 secondsintervals and centrifuged at 20000x g for 10 min. Aliquots ( 1.0 ml) of the resulting supernatant were pipetted into plastic tubes, stoppered, and stored at
-70°C until assayed. The enzyme activities of SOD,
GR, GPx, GST and CAT were measured using a
Cobas MIRA clinical analyser (Roche Diagnostic
Systems, Inc., Basel, Switzerland). Analyses, and
calculations were performed automatically accord-
Measurement of hepatic triglyceridesand cholesterol levels
ing to the programmed instructions.
Animals were sacrificed by decapitation and their
livers immediately
removed and placed in homogenisation medium (250 mM mannitol, 70 mM sucrose,
1 mM EDTA adjusted with Tris to pH 7.4). One
part of the liver was immediately homogenised in a
Potter-Elvehjem glass homogeniser (1 g of tissue
plus 10 ml homogenisation medium). Cholesterol
and triglycerides from this homogenate were measured by automated assays using Hitachi kits for
each determination(HITACHl
704analyser).
Assay of serum enzymatic activities and glucose serum
levels
In order to determine serum transaminase and alkaline phosphatase activities and glucose serum
levels, animals were anaesthetised using sodium
pentobarbital,
50-60 mg/kg body weight, and
blood samples were removed from the retro-orbital
sinus. The enzymatic activities and glucose levels
were measured by automated enzymatic assays
9
deitia et ai.
ElControl
r_MDMA3 h
NMDMA6 h
o .'-',×
< ._
7"
DControl
I_MDMA3h
t:1MDMA6h
NMDMA7 d
2]
1.5'
L
"r
Acute treatment
(20 mg/kg,
-
.J.*
i,.ll._
111.4
_....
Results
Effectof MDMA
onlipidperoxidation
///,,
II1"1
/,1./,'
h'll
tffJ
_5_
555.I
I//l
"1""
_//'/_
Repeatedtreatment
1
,_
<
_ 0.5
_" _
_
Fie, 1. Effect of single
test (ANOVA), followed b\ the Scheffc test. The
minimuna number of animals used to determine
mean values and statistical differences was eight
for each treatment.
i.p.) and repeated
A single dose of MDMA (20 m_/kg, i.p.) did not
cause any significant change in lipid peroxidation.
However, MDMA (20 mg/kg, i.p., b.i.d., for 4 consecutive days) produced an increase in lipid peroxidation, the eflect being significant 3 h (18%).and 6
h (26%) after last injection. Seven days later, MDA
levels returned to control values (Fig. 1).
(20 mg/kg.
i.p., b.i.d.. 4 consecutive
days) administration
of MDMA
on
lipid peroxidation
in rat liver. Animals were sacrificed 3 h, 6 h
and 7 days after the last MDMA administration.
Data are the
Effect of MDMA on hepatic triglycerides and cholesterol
mean__SEM
A series of experiments
was performed
to evaluate
hepatic levels of triglycerides
and cholesterol after
group
of 8-10
(one-way
rats.
analysis
*p<0,05,
**p<0.01
of the variance
vs. to control
followed
by Scheffe
test),
using Technicon-Ba,xer kits for each enzyme (Technicon R.A.1000 autoanalyser),
Measurement
of hepatic glycogen content
Hepatic glycogen content was determined according to the method of McGarry and Kawajima (22).
Hepatic samples (0.3 g) were digested with 5 M
KOH and were precipitated with ethanol. Later,
samples were centrifuged at 1000Xg for 15 min.
An aliquot of the precipitate was mixed with 5 m]
of anthrone reagent (0.05% w/v anthrone, 1% w/v
thiourea in sulfuric acid __7"_'"'_,,
v/v). The colour was
developed by' placing the tubes in a boiling bath
for 10 min. The absorbance of each sample was
determined at 620 nm and the glycogenconcentration of these samples was calculated with a standard curve using a glycogen kit (E. Merck, Darmstadt, Germany).
levels
acuteand repeatedMDMAadministration.Acute
MDMA treatment caused a significant increase in
hepatic triglycerides and cholesterol contenL the
effect being significant 6 h after drug injection. Repealed administration of MDMA produced a significant increase in cholesterol levels, both 3 h and
6 h after drug administration, which returned to
control values seven days later. However, no significant change in hepatic triglycerides content was
observed after repeated MDMA treatment at any'
time of sacrifice (Table 1).
Effect of MDMA on reduced glutathione (GSH) hepatic
content
A single dose of MDMA did not cause any significant change in hepatic GSH content. The effect
!
Table 1. Effect of single and repeated administration of MDMAon choiester0i
and triglycerides
content
inratliver
Survival
Treatment
Histology
Statistical analysis
Data
were reported
as mean_ + SD and
were
ann-
lysed using the one-way analysis of the variance
10
Cholesterol
Triglycerides
(mg/g liver)
(mg/g liver)
Control
0.93±009
8.81-_-0,48
MDMA
MDMA
Control
MDMA
MDMA
MDMA
1.06+_0.10 9.91=0.41
1.4±0.10" 11,12:0,50"
0.78_*0.09 943+-0.29
1.11+_0.05"*10.24±0.64
1.40_.0.05"**8.78=0.88
0.85*_0.04
7.19+_0.26
time
Single
Immediately after the sacrifice of the animals, liver
sections were excised and immersed in 10% buffered neutral formalin for 24 h. Fixed portions were
then processed, embedded in paraffin blocks, sectioned at about 5 #m, mounted on glass slides, and
stained with hematoxylin-eosin
and Masson stains.
3h
6h
Repeated
3h
7 6h
days
_,
Animalsreceivedsaline (control group), MDMA(20 mg/kg i.p.) or MDMA(20
mg/kg i.p., b.i.d for 4 days) for single or repeated treatment respectively.
Rats were sacrificed 3 h, 6 h and 7 days after the Last MDMA injection.
Valuesare mean±SEM from 8-10 rats. *p<0.05. **p<0.01 ***p<0.001
vs control group using one-way analysis of the variance lollowed by Schefie
test.
'
Ecstasy-induced injury
FlControl
[3Control
EIMDMA
mMDMA
3h
WMDMA6h
A
1.6.
1-.
'g" 1.2
"-_
g
tathione reductase in liver were determined in rats
alter single or repeated MDMA administration.
Liver enzymes behaved differently. With the exceptionof GPx, the catalyticactivitiesof the antioxidant enzymes in tile liver did not show any significant change between different groups (data not
shown). However, MDMA (20 mg/kg, i.p.) caused
a slightdecreasein glutathioneperoxidase(GPx)
activity,the effectbeingsignificant7 daysafter repeated MDMA treatment (Fig. 2B).
3 h
t':IMDMA
6h
WMDMA 7d
T
i
0.8
,//,,r ....
0.4-
Effect of MDMAon serum transaminase and alkaline
phosphatase activities
Acute treatment
B
Repeated treatment
Y
1.5.
r-J :/:r i:i
, :::_:
: :::::.
_ ::::
:::::
::::
:ii
0
_:,
.....
Acutetreatment
!
0.5'
Fig. 2. Effect
" " "•
•:
of single (20 m_kg,
on serum ALT, AST and
ALP activitiesis shownin Table2. AST activity
showedan increasingtendencyafter acuteMDMA
treatment, but the significant increase became evi-
2-
,:,
The effect of MDMA
_/,-_
,
dent 6 h after drug administration.
Repeated
MDMA treatment, causeda significantincreasein
(/Z,
,///
""
,,-,-,
_,
[] Control
EtMDMA 3 h
EMDMA 6 h
¢//.
Ill*
A
rnControl
E_MDMA3 h
t':1MDMA6 h
IIMDMA 7 d
5
Repeatedtreatment
i.p.) and repeated
""
4
(20 m_kg,
i.p., b.i.d., 4 consecutive days) administration
of MDMA on
GSH content (A) and on glutathione peroxidase activiD (B) in
rat liver. Animals were sacrificed
3 h, 6 h and 7 days
- after the
Z
r'n N
Last MDMA administration.
Data are the mean+_SEM
rats. *p<0.05. **p<0.01 xs. to control group lone-way
of the _ariance follo_ed b._ Scheffe test).
_
_
_
"_
of 8 10
analysis
3
_0
2
*
1
0
of repeated MDMA treatment was more marked.
A significant reduction of approximately 22% and
27% in GSH hepatic content was found 3 h and 6
h respectively after the last injection. Seven days
later, hepatic glutathione levels returned to control
values
(Fig.2A).
i
Acute treatment
B
Repeatedtreatment
240
180
-
_a
r,_ ,..-,
Effectof MDMAon antioxidanthepaticenzymeactivities
To establish the mechanism of toxicity as oxygen
radical-mediated, there are a number of direct and
indirect methods that can be employed. Direct
methods include the measurementof superoxide
hydrogen peroxide, or hydroxyl radical. These species are very reactive and their quantitation can be
difficult. Therefore, indirect methods of study are
often used. One such method is the measurement
of changes ill endogenous antioxidant enzyme activitv.
, In this sense, the five principal antioxidant
enzymes superoxide dismutase, catatase, glutathione
peroxidase, glutathione-S-transferase
and glu-
{
"I"
8
"1- .
"l-
"_ 120
_
60
:
0
Acute treatment
(20 m_kg.
Fig. 3. Effect of single
i.p., b.i.d.,
hepatic
4 consecutive
gl?,cogen content
days)
Repeatedtreatment
i.p.)and
repeated
administration
(A) and on glucose
(20 mjkg.
of MDMA
serum
on
lcxels IB)
in
rat. Animals were sacrificed
3 h.the
6 hmean_-SEMof
and 7 days after8 the
last
MDMAadmmistration.
Data are
10rats.
*/,<0.05.
**p<0.01
the _ariance
follov,,ed
vs. to control
b? Scheffe
group
(one-way
analssis
of
test).
11
I',eitia el al.
Table 2 Effect of single ar)d repeated adminisl)at)orl
Treatment
ol MDMA on serum ALT AST and ALP activilies
Time
S)ngle
ALTiU/L)
in rat
AST (U/L)
ALP {ULI
3h
6h
Control
MDMA
MDMA
2868 - 2.20
29.33 " 151
4434-537"
8392 : 5.89
86.70:443
133 971478""
251 67 : 3 67
36117 : 11.76'
384.50-35.59""
3h
6h
Control
MDMA
MDMA
3200 _125
34.80:204
3100- 321
79.83- 1.45
10200-291"**
98.00-2.55*"
25043 - 10.05
33738-76.99*
336.86' 291"
7days
MDMA
33831.28
101 43-3.67"**
38529-32.14"*
Repeated
Animals received saline ieontrol group). MDMA (20 mg/kg ip ) or MDMA (20 mg/kg i.p bi.d for 4 days) for single or repeatedtreatment respectively.Blood
samples were taken 3 h. 6 h and 7 days after the last MDMAinjection Valuesare mean-SEM from 8-I0 rats. "p. 0.05 **p<0.01 "**p- 0.001 vs control
group using one-way analysis o1 the variance followed by Scheffe test
AST activity. Seven days later AST activit\ remained significantly increased.
A single injection of MDMA significantly increased ALl- activity 6 h after drug administration.
Repeated MDMA treatment did not elicit an\ significant change in ALT activity at any time of sacrifice,
MDMA caused a significant increase in ALP acti\it\ at both. 3 h and 6 h after single or repeated
drug administration.
Furthermore.
seven days
acute treatment.
Seven days after repeated
MDMA treatment, glscogen hepatic levels returned to control values (Fig. 3A).
Serum glucose levels after single or multiple administration of MDMA are shown in Fig. 3B.
MDMA caused a substantial reduction in the glucose levels6 h after a singleinjection.However,
no significant effect on glucose serum levels after
repeated MDMA treatment was observed.
later.
creased. ALP
Histological examination
activit\
remained
significantl)
in-
Figure 4A shows the liver section of a control rat
Effectof MDMAon glycogenhepaticlevelsand glucose
serumlevels
MDMA caused a significant decrease in glycogen
hepatic content, the effect being more marked after
showing, in the centre off the photograph, the portal tract. Striking changes were identified in the
liver.Acute MDMA treatmentcausedcell necrosis
particularly in portal areas with inflammatory infiltrate consisting in lymphocytes and macro-
/-7,,,.4 L.i\cr histology sho\_ ing the portnl lracl in the centre of the photograph.
Hepatic necrosis with the portal area being infiltrated
by I__mphoc._tcs and macrophagcs
following administration
off NII)M a, &nimal received saline (control group) (A) or MDMA (20
mg/kg, i.p.) (B) being sacriiiccd 6 hours laler.
12
Ecstasy-induced injury
Fi%. 5. Liver histology showing the hepatic xein in the centre of the photograph. Hepatic necrosis with inflammatory infihrate
around hepatic vein following administration or MDMA. Animal receixed saline (control group) IA) or MDMA {20 mg/kg, i.p..
b.i.d., l\)r 4 days) (B) being sacrificed 6 hours htter.
phages denser in portal tracts, 6 h post administration (Fig. 4B). Figure 5A shows the liver section of
a control rat showing, in the centre of the photograph, the hepatic vein. Repeated rejection produced cell necrosis and inflammatory infiltrate
around the hepatic _ein. These effects were more
marked 6 h after the last administration (Fig. 5B)
bi,tt seven days later no changes were observed,
Discussion
Hepatotoxicity is one of the medical consequences
of MDMA consumption (14). Jaundice, hepatomegaly, centrilobular necrosis and hepatitis are
some of the MDMA-induced liver conditions (11.
14). The most reported form of liver injury is hepatitis (15-17, 23).
The mechanism of MDMA-induced hepatic injury is unclear but a spectrum of severity seems to
exist as assessed by histological changes varying
from mild to moderate lobular hepatitis and to
features of massive hepatic parenchymal collapse
with areas of nodular regeneration. The severity of
liver damage does not seem to correlate with either
the amount or frequency of MDMA ingested suggesting an idiosyncratic type of reaction (13).
In the present study, the major
biochemical
events indicative of liver injur,<'after acute and repeated MDMA administration to rats. wereinxestigated. The attack of reactive oxygen species on
polyunsaturated fatt\ acids, essential constituents
of biological membranes, has been sho_vn to result
in peroxidative damage of these lipids (24). Malondialdehyde, the reaction product of lipid peroxidation usually determined in experiments, exerts
several biological effects including cross-linking of
proteins and nucleic acids, and is presumably mutagenic and carcinogenic. Lipid peroxidation and
subsequent cellular damage are regarded as an inaportant mechanism underlying the toxicity of several xenobiotics (25t.
Living organisms contain various free radical
scavenging systems for the clarification of the
pathologic role of free radicals, it is therefore essential to estimate the changes in both the generation and the scavenging of free radicals. The reduced form of glutathione (GSH), the major intracellular thiol, neutralises many kinds of radicals,
either directly or in association witl'lglutathione
peroxidase (26).
Depletion of GSH, especially in the liver, can
cause irreversible damage and cell death. GSH
plays an important role in protecting cells against
reactive 02 intermediates and free radicals. Glutathione deficiency worsens free radical-induced
toxicity, while free radical production depletes glutathione. Decreased ghitathione is an early marker
of free radical insult and is evident before overt cell
death occtlrs (27).
A single dose of MDMA did not change glutathione levels, and free radical production
from
MDMA was not quantitati,,ely significant enough
to deplete glutathione and allo,<vuncontrolled reaction with ,,ital cclh.llar components.
In contrast, re-
13
Beitia et al,
peated
MDMA
injection produccd
a sufficicnl
quantity of free radicals to deplete glutathione,
Furthermore, glutathione
peroxidase activity showed a
tendency to decrease after MDMA injection. In addition, not only GSH itself but also tile GSH redox
cycle, catalvsed by the enzyme glutathione peroxidase, could contribute to radical scavenging,
Numerous
studies have documented
that decreases in glutathione precede lethal cell injury from
free radical generators (28). The leakage of intracellular enzymes, suggests irreversible damage. In
this sense, ALl- and AST serum activities as indi-
suggested
that MDMA
could be regarded
as a
chemical stressor as M DMA-induced
neurochcn>
teal, behavioural
and endocrine aherations closely
resemble those elicited by exposure to acute stress
(,__). McGuinness
et al. established that during
chronic stress hormone infusion the rise in epinephrine exerts potent stimulatorv
effects on glucose
production
principally by' enhancing hepatic glycogenolysis
(33). Thus, these results SUgGest that
MDMA-induced
stimulation
of glycogenolysis
could be associated with the sympathomimetic
elfects observed after acute MDMA administration
cators of liver injury were determined. Three hours
after a single dose of MDMA,
no significant
changes were observed. In contrast, a significant increase in ALT and AST activities, 6 h after drug administration
were obtained.
Repeated
MDMA
treatment produced a significant increase ill AST
activity but no effect on ALT activity was observed,
In all cases, MDMA produced a disproportionate
increase in AST activity compared with ALT activitv. This effect is frequently all index of important
cell necrosis. Furthermore,
ALP activity as indicatire of cholestasis was determined.
MDMA caused
an increase in ALP activity, the effect being more
marked after a single dose. Seven days after repeated
MDMA
treatment.
ALP activity remained increased. These effects resemble those seen in
humans in which increases in AST, ALT and ALP
activities have been reported (13. 15.29, 301. These
findings were corroborated
with histological examination
which,
according
to the post-mortem
changes was reported in deaths associated
with
MDMA
intake (12-17). Hepatic necrosis and illflammatory
infiltrate were evident and overall findings were consistent with a drug related hepatitis.
There appears to be a relationship
between glutathione
deficiency
and glycogen
metabolism.
Recently: it has been reported
that glutathione
depletion caused by various
drugs stimulates glycoten
breakdown
(tl).
Our
results
show
that
(34).
Taken together with the present results, the liver
toxicity caused by acute MDMA
treatment
involves several mechanisms
which cause increased
lipid levels, increased liver enzymes and impaired
gluconeogenesis
leading to a fall ill blood glucose
and depletion
of liver glycogen. In contrast repeated treatment produced some evidence of oxidative stress, namely, increased MDA content and
decreased GSH content, but no evidence of hepatocyte damage. As far as we know. these results
could be the first major step for research in liver
injury'. Furthermore.
according to the sympathomimetic effects reported after acute MDMA mjeclion (34), these results could be explained by the
acute effect of the last dose rather than any longterrn chronic effects.
-
repeated
administration
of MDMA
(20 mg/kg,
i.p.) causes a significant decrease in hepatic glycogen content which is not accompanied by a modification in serum glucose levels. In this sense, the
effect on hepatic glycogen content
could be explained by the significant GSH depletion observed
after repeated MDMA treatment.
On tile other hand, 3 h after a single dose off
MDMA a 60" ,, reduction of liver glycogen content,
witllout additional
was
tent
crease
effect
on
seruna
glucose levels
observed. A larger reduction
of glycogen COlaI)/
(approximately
83,,t
and
a signiticant
deof plasma glucose levels were found
6 h after
MDMA.
B\ contrast
glutathione
levels did not change
after acute MDMA treatment.
It has been recently
14
Acknowledgements
We thank Caju Pamplona
i\_r a fello_ship
1o one of us (G.B.B.j.
We also _ish to thank Mrs. Mariu Luz Muro for her excellent
contributions to this work.
References
1. BRONSO>,
M E. BARRIOS-Z_MI_F,A?VO
k. JIANG_i eta]. Behavioural
and
de\elopmemal
effects
of t,_o 3.4-moth\len-
edioxymethamphetamine
(MDMA)
derixatixes.
Drug AIcoholDepend1994:
36:161 6.
2 GREER G. TOLBERI R. Subjeclixe
reporls of Ihe effects of
MDMA
in a clinical setting..1
Psychoactixe
Drugs
1986:
is: 319-27.
3. Law>, J (2. Federal Register
1988:53: 5156.
4. SPRA(iL'[ J E. EVIiR\IAN S L, NICHOLS D E. _n integrated
hypothesis
for the serotonergic
axonal loss induced by 3.4methvlenedioxymethamphe(amine.
Neurotoxicology
1998:
19:427 42.
5. A(;tlRm
N. B.,_RlOXt'l\()M.
L-XSH_R..XS
B. Dtl Rio J. The
role of dopaminergic
systems in the pcrinatal
sensiti\it 3
to 3.4-mcthylencdiox)methamphclaminc-induccd
neurotoxicit\ in rats..1 Pharmacol
ExpThcr
1998: 2,";6:1159 65.
6. S..xl_()L K E. kl<\v R. Rl(u.,xm)s .1 B. et al. Meth._lencdiox._methamphetaminc-ihduced
serotonin
deficits
urc follo_ed
b, partial reco\er\
oxer u 52-x_eek period. Part I: synaptosomal uptake and tissue concentrations.
J Pharmacol
Exp
Ther 199(_: 27(x 846 54.
7. Gt:_)_!l.s_
G A. t.a_l.._,_u)r() B K. NAsI_ .l E Pmentiation
of
3.4-methylenedioxymethamphetaminc-induced
dop-
Ecstasy-induced injury
8.
amine
release
ceptor
agonists.
CtlADWICK
and
[ S,
serotonin
neurotoxicity
Eur J Pharmacol
LINSLtY
A,
I994:
FREI{\It)NT
by 5-HT(2)
264:325
A
J.
DORAN
re-
21. ELLMAN G L. Tissue
30.
ophys
B.
Ec-
22.
sulph,,dryl
groups.
Arch
Biochcnl
Bi-
1959: 82:70-7.
_,I('GARRY
I D,
N.,XW-_.JIM.\
M.
From dietary
glucose
to li,_er
stasy. 3.4-methylenedioxynlethan_phetamme
(MDMA).
a
tatality associated ,aith coagulopath,,
and hyperthcrmia..1
R Soc Med 1991 84: 371.
FxltAI. 1 H. S.XLLO,_n D E Y..',(,_oof_M. BtII. G M. Acute
renal failure after ecstas}. BriI Med J 992 305: 29.
R.XNDALL T. Ecstas>-fuelcd
"'ruNe'" parties become dunces
of death for English }ouths. JA.MA 1992: 268:1505
6.
MILROY C M. CI.ARK J C. F()RREST A i \M Patholog}
of
deaths associated with "'ecstas\" and 'e;e" misuse. J Clin
Pathol 1996: 49:149
53.
KHAKOO S I. COLES C J, ARMsrRoN(; J S. BARRY R E.
gl}cogen, the full c_cle round. Annu Rex Nutr 1'-)87: 7:51
73.
23. FU)Lli_: H. DHIIA.(}N A. GtRTNIR
D. Bt r_{}t _;HS A.
Chronic
ecstasy
3.4-nleth_lencdioxxmethanlp
letl n _c}
abuse: it recurrent
and unpredictable
c:m_,e o1 sc,,cre acute
hepatitis. J Hcpatol
1996: 25:563
6.
24. H.',I.LIWELL B, CtlIRI('() S. Lipid peroxidation:
its mechanism. measurement
and significance.
\m J (lin Nutr 1_)93:
57: 715S 25.
25. Kt.l,,OSl! l. HIGtT(HI H. K.\lo S. et _.11.Ethanol-reduced
oxidative stress in the liner. Alcohol Clin Exp Res 1996: 20:
Hepatotoxicity
and acceleTated fibrosis following
3.4meth>lenedioxymethamphetamine
tecstasy")
usage. J Clin
Gastroenterol [995: 20: 244-7.
13. Et.LIS A J. WENDON J A, PORTM.X.NNB. VV'ILLIAMSR. Acute
77A 85.
26. KoB..xY.XSul H. NOXAMI T. KUROK.',W_ T. et al. Changes in
the glutathione redox s,_stcm during ischemia and reperfusion in rat liver. Scand J Gastroenterol
1992: 27: 71l 6.
liver damage and ecstas_ ingestioq.
Gut !996: 38:454
8.
I4. HI!NRY J A, JEFFRtCfS K J. DAWt.IN(; S. Toxicity and deaths
27. MITCHELL J B. Russo A. The role _1 glutathione
ation and drug induced cEtotoxicit.,,. Br J Cancer
from 3.4-methylenedioxymethumphetamine.
Lancet 1992:
340: 384_7.
15. DYKHUIZEN R S, BRUNT P W. :_TKINSON P. et al. Ecstas,_
(suppl 8): 105- 12.
28. P{}wls O. Free radical formation
Free Rudic Biol Med 1989: 6:63
induced hepatitis mimicking
viral hepatitis.
Gut 1995: 36:
939_,I.
t6. SHEARMAN J D. CHAPMAN R W G. SATSANGI J, RYI]_2¢ N
29. BROWN C. OSTERLOH J. Multiple sexere complications
from
recreational
ingestion of MDMA (ecstasx)[Letter].
JAMA
1987: 258:780
1.
G. Misuse of ecstas,, [Letter].
Brit Med J 1992: 305: 309.
17. GORARI) D A. DAVIES S E. CLARK N'I k. Misuse of ecstas._
30. IBRAHIM H E SALLOMI D E YAOOOB M. BELL G M. Acute
renal failure after ecstas}.
Brit Med J 1002: 305: 29.
[Letter].
Brit Med J 1992: 305: 309.
18. AGUIRRE N. GAt.BFTE J L, LASHERASB, DEL Rio J. Methyl-
31. BRAUN L. CSAL.X M. Poussw A. ctal. Glutathione
depletion
induces glycogenolysis
dependent
ascorbate
s.,,nthesis in
cnedioxymethamphetamine
induces opposite
changes in
central pre-and postsynaptic
5-HTt \ receptors in rats. Eur
J Pharmacol 1995: 281: 10l 5.
19. BUEGWEJ A. AUNT S D. Microsomal
lipid peroxidation,
isolated murine hepatocytes.
FEBS Lett 1996: 388:173 6.
32. (/ONNOR T J, N|CNAMARA N/[ G. FINN D, et al. ,-',cute 3.4methvlenediox.vmethamphetamine
(MDMA) administration produces a rapid and sustained sappression of immune
Methods
Enz>mol 1978: 52:
20. BRADFORD M M. A rapid
q uantitation
of microgram
the principle of protein-dye
72:..48 _4.
function in the rat. Immunopharmacology
1998: 38: "__ 60
33. MCGUINNESS O P, SHAW V. Bt.:NSON E M. et al. Am J Physiol
1997: _ _ 8674 81.
34. GREER G. SIRASSMAN R J. Information
on "'Ecstasy". Am
J Psychiatry1985:142:1391.
9.
10.
1l.
I2.
_0_-,.
and sensitive method for the
quantities of protein utilizing
binding. Anal Biochem
1976:
b\ antitumor
101.
m radi1987:55
quinones.
15