Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Aquatic Toxicology 82 (2007) 204–213

Ammonia toxicity and tolerance in the brain of the


African sharptooth catfish, Clarias gariepinus
Nicklaus L.J. Wee a , Yvonne Y.M. Tng a , Hin T. Cheng a , Serene M.L. Lee a ,
Shit F. Chew b , Yuen K. Ip a,∗
a Department of Biological Sciences, National University of Singapore, Kent Ridge, Singapore 117543, Republic of Singapore
b Natural Sciences and Science Education, National Institute of Education, Nanyang Technological University,

1 Nanyang Walk, Singapore 637616, Republic of Singapore


Received 7 December 2006; received in revised form 9 February 2007; accepted 22 February 2007

Abstract
The African sharptooth catfish Clarias gariepinus lives in freshwater, is an obligatory air-breather, and can survive on land during drought. The
objective of this study was to elucidate the mechanism of acute ammonia toxicity in C. gariepinus, and to examine whether methionine sulfoximine
[MSO; an inhibitor of glutamine synthetase (GS)] or MK801 [an antagonist of N-methyl d-aspartate type glutamate (NMDA) receptors] had
protective effects against acute ammonia toxicity in this fish. After 48 h of exposure to a sublethal concentration (75 mmol l−1 ) of environmental
ammonia, the brain glutamine and ammonia contents in C. gariepinus increased to 15 ␮mol g−1 and 4 ␮mol g−1 , respectively. Thus, C. gariepinus
detoxified ammonia to glutamine and could tolerate high levels of glutamine in its brain. After C. gariepinus was injected intraperitoneally with a
sublethal dose of ammonium acetate (CH3 COONH4 ; 8 ␮mol g−1 fish) followed with emersion, brain ammonia and glutamine contents increased
continuously during the subsequent 24-h period, reaching 7 and 18 ␮mol g−1 , respectively, at hour 24. These results suggest that when confronted
with acute ammonia toxicity, the survival of C. gariepinus was crucially determined by its high tolerance of ammonia and high capacity to detoxify
ammonia to glutamine in the brain. For fish injected with a sublethal dose of CH3 COONH4 (10 ␮mol g−1 fish) followed with immersion, there
were transient but significant increases in brain ammonia and glutamine contents, which peaked at hour 2 (4 ␮mol g−1 ) and hour 6 (6 ␮mol g−1 ),
respectively. From these results, it can be deduced that C. gariepinus accumulated glutamine in preference to ammonia in its brain. By contrast, for
fish injected with a lethal dose (20 ␮mol g−1 fish) of CH3 COONH4 followed with immersion, the brain ammonia content increased drastically to
10 ␮mol g−1 after 30 min, while the brain glutamine content remained relatively low at 5 ␮mol g−1 . Therefore, it can be concluded that increased
synthesis and accumulation of glutamine in the brain was not the major cause of death in C. gariepinus confronted with acute ammonia toxicity. The
determining factor of acute ammonia toxicity appeared to be the rate of ammonia build-up in the brain. MK801 (2 ␮g g−1 fish) had no protective
effect on C. gariepinus injected with a lethal dose of CH3 COONH4 (20 ␮mol g−1 fish) indicating that activation of NMDA receptors might not
be involved. By contrast, the prior administration of MSO (100 ␮g g−1 fish) reduced the mortality rate from 100% to 80% and at the same time
prolonged the time of death significantly from 27 min to 48 min. However, the protective effect of MSO was apparently unrelated to the inhibition
of glutamine synthetase and prevention of glutamine accumulation in the brain. Instead, MSO affected activities of glutamate dehydrogenase and
alanine aminotransferase and suppressed the rate of ammonia build up in the brain of fish injected with a lethal dose of CH3 COONH4 .
© 2007 Elsevier B.V. All rights reserved.

Keywords: Ammonia; Ammonia toxicity; Brain; Clarias gariepinus; Glutamine; Glutamine synthetase; MSO; MK801; Nitrogen metabolism

1. Introduction Randall, 1979; McKenzie et al., 1993), hyper-excitability, coma,


convulsions and finally death. It also affects the ionic balance
Ammonia is toxic to fish because of many reasons. At the in fish, because NH4 + can substitute for K+ in Na+ /K+ -ATPase
organismal level, ammonia causes hyperventilation (Hillaby and and in Na+ /K+ /2Cl− co-transporter (see Wilkie, 1997, 2002 for
reviews; Person Le Ruyet et al., 1997). NH4 + can substitute
for H+ in Na+ /H+ exchanger (NHE), probably NHE2 and/or
∗ Corresponding author. Tel.: +65 6516 2702; fax: +65 6779 2486. NHE3 (Randall et al., 1999). At the cellular level, ammonia can
E-mail address: dbsipyk@nus.edu.sg (Y.K. Ip). interfere with energy metabolism through impairment of the tri-

0166-445X/$ – see front matter © 2007 Elsevier B.V. All rights reserved.
doi:10.1016/j.aquatox.2007.02.015
N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213 205

carboxylic acid cycle in fish (Arillo et al., 1981). In addition, phosphate synthetase III activity can be detected from its liver,
NH4 + can substitute for K+ (Binstock and Lecar, 1969) affecting not even after exposure to 100 mmol l−1 NH4 Cl at pH 7.0 for 5
the membrane potential. Smart (1978) suggested that the mech- days (Ip et al., 2004c). Ip et al. (2004c) demonstrated that the
anism of ammonia toxicity in fish might be similar to that in ability of C. gariepinus to survive in high concentrations of envi-
mammals during hepatic encephalopathy. Several theories, i.e. ronmental ammonia was partially related to its ability to actively
glutamatergic dysfunction, glutamine accumulation leading to excrete ammonia, presumably NH4 + . Furthermore, Ip et al.
astrocyte swelling, and/or activation of N-methyl-d-aspartate- (2005a) reported that four days of emersion did not induce ureo-
type glutamate (NMDA) receptors, have been proposed as genesis or ureotely in this catfish, and ammonia was not excreted
mechanisms involved in chronic and/or acute ammonia toxicity as NH3 through volatilization during emersion. In addition,
in mammalian brains (Felipo et al., 1994; Margulies et al., 1999; there were no changes in levels of alanine in the muscle, liver,
Hermenegildo et al., 2000; Desjardins et al., 2001; Brusilow, and plasma; neither were there any changes in the glutamine
2002; Felipo and Butterworth, 2002; Rose, 2002). However, it levels in these tissues. More importantly, there were extraordi-
has been proposed recently that mechanisms of and defence narily high levels of ammonia in the muscle (14 ␮mol g−1 ), liver
against ammonia toxicity in brains of fishes with high ammonia (18 ␮mol g−1 ) and brain (11 ␮mol g−1 ) of fish exposed to terres-
tolerance could be different from those in mammals (see Ip et trial conditions for 4 days. Thus, Ip et al. (2005a) concluded that
al., 2004a,b; Chew et al., 2006 for reviews). C. gariepinus could tolerate high levels of ammonia at the cel-
In mammals, ammonia toxicity involves the activation lular and tissue levels, especially in the brain, during emersion.
of NMDA receptors (Fan and Szerb, 1993; Hermenegildo That being the case, the mechanisms of ammonia toxicity in the
et al., 2000), and the administration of a NMDA receptor brain of C. gariepinus remains enigmatic because encephalopa-
antagonist, (5R, 10S)-(+)-methyl-10, 11-dihydro-5H-dibenzo[a, thy would develop in mammalian brains with 1–3 ␮mol g−1
d]cyclohepten-5,10-imine hydrogen maleate (MK801), at a of ammonia (Cooper and Plum, 1987). Therefore, the present
dosage of 2 ␮g g−1 animal delays or even eliminates the fatal study was undertaken to elucidate the mechanisms of ammonia
effects of ammonia toxicity in rats (Warren and Schenker, 1964; toxicity and tolerance in the brain of C. gariepinus.
Takahashi et al., 1991; Willard-Mack et al., 1996; Brusilow, Five sets of experiments were performed in this study. First,
2002; Marcaida et al., 1992; Hermenegildo et al., 1996). While experiments were undertaken to establish the levels of ammonia,
a similar dosage of MK801 appears to have a protective effect glutamine, glutamate, and alanine in the brains of fish immersed,
on the weatherloach (Tsui et al., 2004), such an effect cannot but with free access to air, in freshwater or water containing
be detected in two mudskippers (Periophthalmodon schlosseri sublethal concentrations (15 or 75 mmol l−1 ) of environmen-
and Boleophthalmus boddarti) confronted with acute ammo- tal ammonia for 48 h. The objectives of this set of experiments
nia toxicity (Ip et al., 2005b). Furthermore, ammonia toxicity were to determine how high the brain ammonia and glutamine
in mammals could be a result of increased glutamine pro- levels could be and to examine whether the brain glutamate
duction and accumulation, which cause increased astrocyte and alanine contents would decrease as a result of increased
swelling, leading to cellular dysfunction, brain edema and glutamine synthesis during chronic exposure to environmental
death (Brusilow, 2002). Indeed, l-methionine S-sulfoximine ammonia. Second, experiments were undertaken to determine
(MSO) inhibits glutamine synthetase (GS), reduces edema, contents of ammonia, glutamine, glutamate, and alanine in the
attenuates ammonia-induced increases in brain extracellular brains collected at various time points from fish that had been
K+ , and ameliorates ammonia toxicity when administered at a injected intraperitoneally with a sublethal dose (8 ␮mol g−1
dosage of 100 ␮g g−1 to patients suffering from acute hepatic fish) of ammonium acetate (CH3 COONH4 ) followed by 24 h of
encephalopathy (Brusilow, 2002). However, increases in glu- emersion. During emersion, ammonia could not be effectively
tamine synthesis and accumulation occur in the brain of the Gulf excreted and consequently the experimental fish had to defend
toadfish, Opsanus beta, without causing brain edema (Veauvy against the toxicity of almost all of the exogenous ammonia
et al., 2005); and, MSO does not have any significant protective injected into them. The objective of this set of experiments was
effects on mudskippers confronted with acute ammonia toxicity to evaluate the upper limits of ammonia and glutamine con-
(Ip et al., 2005b). tents in the brain of this fish. Third, fish were euthanized at
Many freshwater catfishes (members of families Clariidae specific time points during 24 h of immersion after being injected
and Heteropneustidae) in the tropics are obligatory air-breathers; intraperitoneally with a sublethal dose (10 ␮mol g−1 fish) of
they can survive on land for many days and exhibit high tolerance CH3 COONH4 . In addition to examining contents of ammonia
of environmental ammonia. The African sharptooth catfish Clar- and glutamine in the brains of these fish, we aimed to determine
ias gariepinus (Burchell) has, in addition to degenerate gills, whether C. gariepinus, despite having degenerate gills, could up-
accessory breathing organs for air-breathing (Graham, 1997). It regulate ammonia excretion in water effectively to ameliorate the
is ammonotelic in water as urea-N accounts for only ∼10% of the toxicity of ammonia. Fourth, fish were euthanized for tissue sam-
total waste-N (Eddy et al., 1980; Ip et al., 2004c, 2005a). Despite pling 30 min after the intraperitoneal injection with a lethal dose
being able to tolerate high concentrations of environmental (20 ␮mol g−1 fish) of CH3 COONH4 followed by immersion. It
ammonia (Eddy et al., 1980; Ip et al., 2004c), C. gariepinus was hoped that results obtained from this set of experiments
does not detoxify ammonia to urea or amino acids (including would shed light on whether glutamine and/or ammonia accu-
glutamine) during ammonia exposure (Ip et al., 2004c). It does mulation could be the cause of death in C. gariepinus confronted
not possess a functional ornithine urea cycle, and no carbamoyl with acute ammonia toxicity. Fifth, experiments were performed
206 N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213

to evaluate whether MSO or MK801 had protective effects 2.2.3. Intraperitoneal injection with a sublethal dose
on C. gariepinus injected intraperitoneally with a lethal dose (8 μmol g−1 fish) of CH3 COONH4 followed by 24 h of
(20 ␮mol g−1 fish) of CH3 COONH4 followed by immersion. emersion
Fish were anaesthetized and injected intraperitoneally with
2. Materials and methods 10 ␮mol g−1 fish of CH3 COONH4 or CH3 COONa (control).
Fish were subjected to emersion in plastic tanks (L 28 cm × W
2.1. Experimental animals 19 cm × H 18 cm) containing only 200 ml of freshwater (pH
7.0). Under such terrestrial conditions, only the ventral surface
Clarias gariepinus (80–200 g body mass) were purchased of the fish was in contact with a thin film of water. After 0.5,
from the China Town wet market, Singapore. They were main- 2, 6, 12 or 24 h, fish were euthanized and brains were collected
tained in plastic aquaria filled with dechlorinated tap water at and kept at −80 ◦ C until further analysis.
25 ◦ C in the laboratory, and water was changed daily. No attempt
was made to separate the sexes. The fish were acclimated to lab- 2.2.4. Intraperitoneal injection with a sublethal dose
oratory conditions for at least three weeks. During the adaptation (10 μmol g−1 fish) of CH3 COONH4 followed by 24 h of
period, C. gariepinus was fed an artificial diet. Food was with- immersion
drawn 48 h prior to experiments, which gave sufficient time for Fish were anaesthetized and injected intraperitoneally with
the gut to be emptied of all food and waste. 10 ␮mol g−1 fish of CH3 COONH4 or CH3 COONa (control).
Fish were immersed individually in 2.5 l of freshwater (pH.
2.2. Experimental conditions 7.0) with free access to air in plastic containers (L 28 cm × W
19 cm × H 18 cm). After 0.5, 2, 6, 12 or 24 h, fish were eutha-
2.2.1. Immersion of fish in water containing 15 or nized and brains were collected and kept at −80 ◦ C until further
75 mmol l−1 NH4 Cl for 48 h analysis.
Experimental fish were immersed individually in 2 l of
dechlorinated water containing a sublethal dose (15 or 2.2.5. Intraperitoneal injection with a lethal dose
75 mmol l−1 ) of NH4 Cl in plastic tanks (L 28 cm × W 19 cm × H (20 μmol g−1 fish) of CH3 COONH4 and euthanized after
18 cm) for 48 h, because Ip et al. (2004) reported that C. gariepi- 30 min of immersion
nus could survive in 100 mmol l−1 of NH4 Cl. There was no Fish were anaesthetized and injected intraperitoneally with
need to aerate the water because C. gariepinus is an air-breather. 20 ␮mol g−1 fish of CH3 COONH4 or CH3 COONa (control).
While immersed in water, fish had free access to the water sur- Fish were immersed individually in 2.5 l of freshwater, and euth-
face to breathe air. Control fish were immersed in dechlorinated anized after 30 min. Brain samples were collected and kept at
water for the same period. At the end of 48 h, fish were eutha- −80 ◦ C until further analysis.
nized and the brains were excised and freeze-clamped in liquid
nitrogen. Brain samples were kept at −80 ◦ C until the analysis 2.3. Determination of ammonia, glutamine, glutamate and
of ammonia, glutamine, glutamate and alanine. alanine contents

2.2.2. Determination of sublethal and lethal dose of The frozen brain samples were weighed and ground to a
CH3 COONH4 injected into the peritoneal cavity powder in liquid nitrogen. Five volumes (w/v) of ice-cold 6%
To determine the 24 h sublethal and lethal dose of trichloroacetic acid (TCA) were added and the mixture was
CH3 COONH4 to be used in this study, 3 groups of 10 fish homogenized three times for 20 s each (with 10 s off intervals)
each (a total of 30 fish) were anaesthetized for approximately with an Ultra-Turrax homogenizer at 24,000 revs min−1 . The
10 min in 0.1% phenoxyethanol (Sigma Chemical Co., St. Louis, samples were then centrifuged for 20 min at 10,000 × g at 4 ◦ C
MO, USA) and injected intraperitoneally (0.5 ml 100 g−1 fish) to obtain the supernatant.
with 10, 15 or 20 ␮mol g−1 fish of CH3 COONH4 . Prior to the For ammonia analysis, the pH of the supernatant was adjusted
experiment, all equipment were sterilized to prevent infection. to between 6.0 and 6.5 with 2 mol l−1 KHCO3 . The ammo-
Control fish (N = 10) were injected with CH3 COONa followed nia content was determined using the enzymatic method of
by immersion. Fish were then immersed in 2.5 l of strongly aer- Bergmeyer and Beutler (1985). The change in absorbance at
ated freshwater, and they usually recovered and responded to 25 ◦ C and 340 nm was monitored using a Shimadzu UV-160A
mechanical stimulation within 7–10 min. For control fish, no spectrophotometer. Freshly prepared NH4 Cl solution was used
mortality was recorded at hour 24. For experimental fish, mor- as the standard for comparison.
tality and time to death were recorded during the subsequent 24-h For glutamine, glutamate and alanine analyses, the super-
period. Fish was regarded as dead when there was no observable natant was diluted with an equal volume of 0.2 mol l−1
respiratory activity and no reaction to mechanical stimulation. lithium citrate buffer and adjusted to pH 2.2 with 4 mol l−1
The sublethal and lethal doses of CH3 COONH4 for immersed LiOH. The free amino acid contents in the tissues were
fish were 10 and 20 ␮mol g−1 fish, respectively. A similar set of determined using a Shimadzu LC-6A Amino Acid Analy-
experiment was performed with fish exposed to 24 h of emersion sis System (Kyoto, Japan) with a Shim-pack ISC-07/S1504
after the injection with CH3 COONH4 . The sublethal dose was Li-type column. Despite complete FAA analyses being per-
determined to be 8 ␮mol CH3 COONH4 g−1 fish. formed on every sample, only contents (␮mol g−1 brain tissue)
N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213 207

of glutamine, glutamate and alanine were presented in this times in 5 volumes (w/v) of ice-cold extraction buffer con-
report. taining 50 mmol l−1 imidazole-HCl (pH 7.0), 50 mmol l−1 NaF,
3 mmol l−1 EGTA, 3 mmol l−1 EDTA at 24,000 revs min−1 for
2.4. To evaluate the possible protective effects of MK801 20 s each with a 10 s off interval. The homogenate was cen-
and MSO on acute ammonia toxicity trifuged at 10,000 × g at 4 ◦ C for 20 min. The supernatant
obtained was passed through a 5 ml Econo-Pac 10DG desalt-
2.4.1. Protective effect of MK801 ing column (Bio-Rad Laboratories Inc., CA, USA) equilibrated
The possible protective effect of MK801 (Sigma–Aldrich with the same buffer solution. The resulting eluent was used for
Chemical Co., St. Louis, MI, USA) was evaluated by inject- enzyme assays. Protein concentrations of the extract were mon-
ing 2 ␮g g−1 fish of MK80l into the peritoneal cavity of fish itored before and after filtration to determine the dilution factor
(N = 10 each) 15 min prior to the injection with a lethal dose involved. Protein was determined according to the method of
of CH3 COONH4 . Preliminary results indicated that the injec- Bradford (1976).
tion of 2 ␮g MK801 g−1 fish alone had no observable effects on Glutamine synthetase (GS, EC 6.3.1.2) was determined as
C. gariepinus. Controls (N = 10 each) were injected with 0.9% transferase activity by the method of Shankar and Anderson
NaCl solution in place of MK801 prior to the injection with (1985) with some modifications. The reaction mixture consisted
CH3 COONH4 . of 67 mmol l−1 imidazole (pH 7.0), 25 mmol l−1 hydroxyla-
mine, 30 mmol−1 arsenate, 100 mmol l−1 glutamine, 1 mmol l−1
2.4.2. Protective effect of MSO ADP, 1.7 mmol l−1 MnCl2 and 0.1 ml sample in a final volume
To evaluate if MSO had a protective effect against acute of 0.8 ml. After incubation at 25 ◦ C for 10 min, the reaction was
ammonia toxicity on C. gariepinus, fish (N = 10) were injected terminated with 0.4 ml of ferric chloride reagent (0.37 mmol l−1
intraperitoneally with 100 ␮g g−1 of MSO (pH 7.0) 15 min FeCl3 , 0.20 mmol l−1 TCA and 0.67 mmol l−1 HCl). The solu-
prior to the injection with a lethal dose of CH3 COONH4 tion was centrifuged at 10,000 × g at 4 ◦ C for 5 min to remove
(20 ␮mol g−1 fish). Preliminary results indicated that injection precipitated proteins. Reaction terminated at time 0 was used
of 100 ␮g MSO g−1 fish alone had no observable effects on as blank. GS activity was measured at 500 nm and expressed
C. gariepinus. Fishes (N = 10 each) injected with 0.9% NaCl as ␮mol ␥-glutamyl hydroxamate formed min−1 mg−1 protein.
solution in place of MSO served as controls for comparison. Freshly prepared glutamic acid ␥-monohydroxamate solution
The mortality rate and time of death for succumbed fish were was used as a standard for comparison.
recorded during the subsequent 24 h. Glutamic dehydrogenase (GDH, E.C. 1.4.1.3) was assayed
in the amination direction according to Ip et al. (1993) with
2.5. Effects of MSO on brain ammonia, glutamine and some modifications. The assay was performed in a total vol-
glutamate contents and plasma ammonia concentration ume of 1.5 ml containing 200 mmol l−1 imidazole (pH 7.8),
10 mmol l−1 ␣- ketoglutarate, 1 mmol l−1 ADP, 0.17 mmol l−1
Fish (N = 5 each) were injected intraperitoneally with NADH and 0.1 ml sample. The reaction was initiated by the
100 ␮g g−1 of MSO or 0.9% NaCl (control) 15 min prior to addition of 0.2 ml of ammonium acetate at a final concentra-
the injection with a lethal dose of CH3 COONH4 (20 ␮mol g−1 tion of 25 mmol l−1 . Change in absorbance was monitored at
fish), followed with immersion in 2 l of freshwater. Fish were 340 nm. Enzyme activity was expressed as ␮mol NADH oxi-
killed 30 min later. Brains were excised and kept at −80 ◦ C until dised min−1 mg−1 protein.
analysis. Contents of ammonia, glutamine and glutamate in the Alanine aminotransferase (ALT, E.C. 2.6.1.2) in the direction
brain were determined as described above. Blood samples were of alanine degradation was determined following the method of
collected from the severed caudal peduncle into heparinized Peng et al. (1994) in a reaction mixture of 1.2 ml containing
Eppendorf tubes and centrifuged at 5,000 × g at 4 ◦ C for 5 min to 50 mmol l−1 imidazole (pH 7.4), 10 mmol l−1 ␣-ketoglutarate,
obtain the plasma. The plasma was deproteinized by adding one 0.025 mmol l−1 pyridoxal phosphate, 0.15 mmol l−1 NADH,
volume (v/v) of ice-cold 6% TCA and centrifuged at 10,000 × g 1 i.u. LDH and 0.05 ml of sample. The reaction was initiated by
at 4 ◦ C for 15 min. The resulting supernatant was kept at −20 ◦ C the addition of 0.2 ml of 0.2 mol l−1 l-alanine and the change
until ammonia analysis following the method described above. in absorbance was monitored at 340 nm. Enzyme activity was
expressed as ␮mol NADH oxidised min−1 mg−1 protein.
2.6. Effects of MSO injected into the peritoneal cavity on
activities of some enzymes from the brain samples 2.7. Effects of MSO in vitro on activities of some enzymes
from the brain samples
Fish (N = 5 each) were injected intraperitoneally with
100 ␮g g−1 fish of MSO or 0.9% NaCl 15 min prior to the injec- Brain samples were collected from control fish immersed
tion with a lethal dose of 20 ␮mol g−1 fish CH3 COONH4 . Fish in fresh water, and enzyme extracts were prepared as described
were killed 30 min after immersion in 2.5 l of freshwater. The above. To determine the in vitro effects of MSO, activities of GS,
brain was quickly excised, freeze-clamped in liquid nitrogen, GDH, and ALT were assayed as described above with or without
and stored at −80 ◦ C until analysis. (blank) 30 min pre-incubation of the enzyme extract with 0.1 ml
The frozen brain, liver and muscle samples were weighed of 2 mmol l−1 MSO (pH 7.0) in the assay medium prior to the
and homogenized, using an Ultra-Turrax homogenizer, three initiation of reactions.
208 N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213

2.8. Statistics Table 1


Contents of ammonia, glutamine, glutamate and alanine in the brain (␮mol g−1
tissue) of Clarias gariepinus immersed for 48 h in freshwater (control) or water
Results were presented as means ± standard errors of mean containing a sublethal concentration (15 mmol l−1 or 75 mmol l−1 ) of NH4 Cl
(S.E.M.). Student’s t-test and one-way analysis of variance fol-
lowed by Bonferroni’s multiple range test were used to evaluate Control 15 mmol l−1 NH4 Cl 75 mmol l−1 NH4 Cl
differences between means where applicable. Differences with Ammonia 1.72 ± 0.12a 1.70 ± 0.16a 4.41 ± 0.68b
P < 0.05 were regarded as statistically significant. Glutamine 1.79 ± 0.16a 5.46 ± 0.87b 15.4 ± 1.5c
Glutamate 4.21 ± 0.69 4.84 ± 0.37 5.41 ± 0.45
Alanine 0.35 ± 0.07 0.31 ± 0.04 0.40 ± 0.02
3. Results
Results represent means ± S.E.M. (N = 4). Means not sharing the same letter are
significantly different, P < 0.05.
3.1. Contents of ammonia, glutamine, glutamate and
alanine

3.1.1. Fish exposed to environmental ammonia (15 or 3.1.3. Fish injected intraperitoneally with a sublethal dose
75 mmol l−1 NH4 Cl) for 48 h (10 μmol g−1 fish) of CH3 COONH4 followed by 24 h of
There was no mortality in fish exposed to 15 or 75 mmol l−1 immersion
NH4 Cl for 48 h. The ammonia content in the brain of fish The injection with 10 ␮mol g−1 fish of CH3 COONH4 fol-
exposed to 15 mmol l−1 NH4 Cl for 48 h remained unchanged lowed by 24 h of immersion resulted in no mortality among the
but that of fish exposed to 75 mmol l−1 NH4 Cl increased sig- experimental fish. However, it led to significant increases in the
nificantly (2.6-fold) to 4.41 ± 0.68 ␮mol g−1 tissue (Table 1). contents of ammonia (4-fold) and glutamine (3.3-fold) in the
The brain glutamine content of fish exposed to 15 and brain within the first 2 h (Table 3). The brain glutamine content
75 mmol l−1 NH4 Cl increased significantly to 5.46 ± 0.87 (3- continued to increase and peaked (6.18 ± 0.38 ␮mol g−1 tissue)
fold) and 15.4 ± 1.5 (9-fold) ␮mol g−1 tissue, respectively at hour 6, even though the brain ammonia content returned
(Table 1). Despite a significant increase in glutamine content, to a level comparable to that of the control. By contrast, the
there were no significant changes in contents of glutamate brain glutamate content decreased significantly at hour 2, but
and alanine in the brain of fish exposed to 15 or 75 mmol l−1 returned to control level at hour 6. There was no significant
NH4 Cl. change in the alanine content in the brain throughout the 24-h
period.
3.1.2. Fish injected intraperitoneally with a sublethal dose The peak levels of ammonia and glutamine in the brain of fish
(8 μmol g−1 fish) of CH3 COONH4 followed by 24 h of injected with 10 ␮mol g−1 fish of CH3 COONH4 followed by
emersion 24 h of immersion (Table 3) were significantly lower (P < 0.05)
No mortality was observed in fish injected intraperitoneally than those of fish injected with 8 ␮mol g−1 fish of CH3 COONH4
with 8 ␮mol g−1 fish of CH3 COONH4 followed by 24 h of followed by 24 h of emersion (Table 2).
emersion. There were significant increases in ammonia and glu-
tamine contents in the brain throughout the 24-h period, reaching 3.1.4. Fish injected intraperitoneally with a lethal dose
7.07 ± 0.82 and 17.72 ± 1.35 ␮mol g−1 tissue, respectively, in (20 μmol g−1 fish) of CH3 COONH4 and euthanized after
the brains of the experimental fish at hour 24 (Table 2). No 30 min of immersion
significant changes in brain glutamate and alanine levels were For C. gariepinus injected with a lethal dose of CH3 COONH4
observed throughout the 24-h period in the experimental fish as and euthanized after 30 min of immersion, the ammonia and
compared with the control. glutamine levels were 9.63 ± 0.30 and 5.36 ± 0.32 ␮mol g−1

Table 2
Contents of ammonia, glutamine, glutamate and alanine in the brain (␮mol g−1 brain) of Clarias gariepinus after the intraperitoneal injection with 8 ␮mol g−1 fish
of CH3 COONa (control) or a sublethal dose (8 ␮mol g−1 fish) of CH3 COONH4 followed by 0.5, 2, 6, 12 or 24 h of emersion
Condition 0.5 h 2h 6h 12 h 24 h

Ammonia Control 1.32 ± 0.16 1.81 ± 0.09 2.31 ± 0.23 2.16 ± 0.12 2.07 ± 0.31
CH3 COONH4 4.10 ± 0.54a,* 6.23 ± 0.52b,* 5.26 ± 0.23a,* 7.36 ± 0.33b,* 7.07 ± 0.82b,*
Glutamine Control 1.93 ± 0.23a 2.87 ± 0.51a 3.58 ± 0.79a 5.99 ± 0.54b 7.53 ± 1.85b
CH3 COONH4 1.79 ± 0.32a 4.23 ± 1.11a 13.60 ± 0.62b,* 16.53 ± 1.32c,* 17.72 ± 1.35c,*
Glutamate Control 5.48 ± 0.27 4.86 ± 0.73 5.67 ± 1.11 5.34 ± 0.58 6.00 ± 0.85
CH3 COONH4 4.48 ± 0.40a 5.04 ± 0.47a 6.53 ± 0.46b 6.58 ± 0.34b 6.47 ± 0.52b
Alanine Control 0.40 ± 0.03 0.53 ± 0.12 0.45 ± 0.05 0.55 ± 0.09 0.47 ± 0.14
CH3 COONH4 0.53 ± 0.07 0.52 ± 0.04 0.39 ± 0.10 0.65 ± 0.10 0.67 ± 0.12

Results represent means ± S.E.M. (N = 4). (*) Significantly different from the corresponding control value, P < 0.05. Means not sharing the same letter are significantly
different, P < 0.05.
N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213 209

Table 3
Contents of ammonia, glutamine, glutamate and alanine in the brain (␮mol g−1 brain) of Clarias gariepinus after the intraperitoneal injection with 10 ␮mol g−1 fish
of CH3 COONa (control) or a sublethal dose (10 ␮mol g−1 fish) of CH3 COONH4 followed by 0.5, 2, 6, 12 or 24 h of immersion
Condition 0.5 h 2h 6h 12 h 24 h

Ammonia Control 1.10 ± 0.17 1.00 ± 0.10 1.21 ± 0.31 1.16 ± 0.09 0.98 ± 0.08
CH3 COONH4 3.50 ± 0.50b,* 3.91 ± 0.38b,* 1.09 ± 0.04a 1.04 ± 0.09a 1.15 ± 0.08a
Glutamine Control 2.03 ± 0.24 1.66 ± 0.08 1.62 ± 0.14 1.85 ± 0.03 2.26 ± 0.26
CH3 COONH4 3.33 ± 0.27a 5.42 ± 0.13b,* 6.18 ± 0.38b,* 3.28 ± 0.22a,* 2.65 ± 0.18a
Glutamate Control 6.15 ± 0.11 6.10 ± 0.42 5.73 ± 0.23 6.34 ± 0.45 5.61 ± 0.18
CH3 COONH4 5.21 ± 0.36 4.42 ± 0.11* 5.53 ± 0.22 5.21 ± 0.24 4.80 ± 0.13
Alanine Control 0.30 ± 0.09 0.22 ± 0.03 0.18 ± 0.02 0.22 ± 0.04 0.21 ± 0.02
CH3 COONH4 0.32 ± 0.06 0.20 ± 0.02 0.13 ± 0.01 0.17 ± 0.02 0.16 ± 0.03

Results represent means ± S.E.M. (N = 4). (*) Significantly different from the corresponding control value, P < 0.05. Means not sharing the same letter are significantly
different, P < 0.05.

Table 4
Contents of ammonia, glutamate and glutamine in the brain (␮mol g−1 tissue) and the concentration of ammonia in the plasma (␮mol ml−1 ) of Clarias gariepinus
30 min after the intraperitoneal injection with a lethal dose of 20 ␮mol CH3 COONH4 g−1 fish with the prior injection (15 min) of saline solution (0.9% NaCl; control)
or methionine sulfoximine (MSO; 100 ␮g g−1 fish)
Saline Saline + CH3 COONH4 (20 ␮mol g−1 fish) MSO + CH3 COONH4 (20 ␮mol g−1 fish)

Brain
Ammonia 1.04 ± 0.28a 9.63 ± 0.3c 5.95 ± 0.32b
Glutamine 2.29 ± 0.08a 5.36 ± 0.32b 6.16 ± 0.16b
Glutamate 6.49 ± 0.26a 6.44 ± 0.24a 7.39 ± 0.24b
Plasma
Ammonia 0.37 ± 0.02a 19.73 ± 1.65b 17.99 ± 0.78b

Results represent means ± S.E.M. (N = 4). Means not sharing the same letter are significantly different, P < 0.05.

tissue, respectively, which were significantly higher than 3.3.2. Effects of MSO on brain ammonia, glutamine and
the corresponding values of the control (1.04 ± 0.28 and glutamate contents and plasma ammonia concentration
2.29 ± 0.08 ␮mol g−1 tissue, respectively). Despite the sharp The ammonia content in the brain of fish injected
increase in the brain ammonia level, the glutamate level in the with MSO followed by a lethal dose of CH3 COONH4
brain remained unaltered (Table 4).The brain ammonia con- (5.95 ± 0.32 ␮mol g−1 tissue) was significantly lower than that
tent of the experimental fish injected with a lethal dose of (9.63 ± 0.30 ␮mol g−1 tissue) of fish injected with a lethal dose
CH3 COONH4 followed by immersion (Table 4) was higher than of CH3 COONH4 only (Table 4). However, the concentrations
the peak levels of ammonia in the brain of fish injected with a of ammonia in the plasma of these two groups of fish were com-
sublethal dose (8 ␮mol g−1 fish) of CH3 COONH4 followed by parable (Table 4). Surprisingly, MSO had no significant effect
24 h of emersion (Table 2) and fish injected with 10 ␮mol g−1 on the brain glutamine content of fish injected with a lethal dose
fish of CH3 COONH4 followed by 24 h of immersion (Table 3). of CH3 COONH4 . However, there was a slight but significant
However, an opposite phenomenon was observed for the brain increase in the glutamate content in the brain of fish injected
glutamine content. with MSO and a lethal dose of CH3 COONH4 as compared with

3.2. Protective effect of MK801


Table 5
MK801 (2 ␮g g−1
fish) did not exert any protective effect Mortality (%) and time to death (min) of Clarias gariepinus injected intraperi-
toneally with a lethal dose (20 ␮mol g−1 fish) of CH3 COONH4 with the prior
on C. gariepinus injected with a lethal dose of CH3 COONH4 injection of saline solution (0.9% NaCl; control), MK801 (2 ␮g g−1 fish), or
(20 ␮mol g−1 fish) (Table 5). methionine sulfoximine (MSO; 100 ␮g g−1 fish)
Conditions Mortality (%) Time to death
3.3. Protective effect of MSO (min)

Control (Saline + CH3 COONH4 ) 100 26.7 ± 3.3a


3.3.1. Effects of MSO on mortality and time to death MK801 + CH3 COONH4 100 27.0 ± 2.7a
MSO (100 ␮g g−1 fish) reduced the mortality of fish injected MSO + CH3 COONH4 80 47.8 ± 6.8b
with 20 ␮mol CH3 COONH4 g−1 fish from 100% to 80%. Results of time to death represent means ± S.E.M. (N = 10), except for prior
In addition, it delayed the time to death significantly from injection of MSO (N = 8). Means not sharing the same letter are significantly
26.7 ± 3.3 min to 47.8 ± 6.8 min (Table 5). different, P < 0.05.
210 N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213

Table 6 synthesis of glutamine following glutamate formation might


Specific activities (␮mol min−1 mg−1 protein) of glutamine synthetase (GS), be the reason for the reduction of cerebral ATP as observed in
glutamate dehydrogenase (GDH) in the amination direction and alanine amino-
transferase (ALT) from the brain sample of Clarias gariepinus 30 min after
ammonia-exposed rainbow trout (Arillo et al., 1981). However,
the intraperitoneal injection with a lethal dose of 20 ␮mol CH3 COONH4 g−1 in recent years, glutamatergic dysfunction, astrocyte swelling
fish with the prior injection (15 min) of saline solution (0.9% NaCl; control) or and brain herniation, and activation of N-methyl-d-aspartate-
methionine sulfoximine (MSO; 100 ␮g g−1 fish) type glutamate receptors related to ammonium ion induced
Enzyme Control MSO (Intraperitoneal Percentage membrane depolarization have been proposed as mechanisms of
injection) change acute ammonia toxicity in mammalian brains (Brusilow, 2002;
GS 0.31 ± 0.01 0.24 ± 0.01* −21.5%
Felipo and Butterworth, 2002; Rose, 2002).
GDH 0.21 ± 0.01 0.18 ± 0.02 NA The increased synthesis of glutamine (Meijer et al., 1990)
ALT 0.0179 ± 0.0014 0.0177 ± 0.0012 NA and its accumulation leading to astrocyte swelling is known to
Results represent means ± S.E.M. (N = 5). (*) Significantly different from the
be one of the reasons behind hepatic encephalopathy in mam-
corresponding control value, P < 0.05. NA = not applicable. mals (Brusilow, 2002). Fish brains are also known to detoxify
ammonia to glutamine (Peng et al., 1998; Ip et al., 2004a,b;
Table 7 Chew et al., 2006). By exposing the Gulf toadfish, O. beta, to
15 NH Cl, Rodicio et al. (2003) demonstrated that the glutamine
Specific activities (␮mol min−1 mg−1 protein) of glutamine synthetase (GS), 4
glutamate dehydrogenase (GDH) in the amination direction and alanine amino- in the brain became 15 N-labelled. Indeed, after 48 h of expo-
transferase (ALT) from the brain sample of Clarias gariepinus determined with sure to a sublethal concentration (75 mmol l−1 ) of environmental
or without 30 min of pre-incubation with methionine sulfoximine (MSO) in vitro
ammonia, the brain glutamine content in C. gariepinus increased
Enzyme Control MSO (in vitro) Percentage inhibition (%) to an exceptionally high level of 15.4 ␮mol g−1 . In compari-
GS 0.299 ± 0.013 0.166 ± 0.014* −44.9 son, the increase in the brain ammonia content (4.4 ␮mol g−1 )
GDH 0.188 ± 0.012 0.087 ± 0.005* −52.8 was much less drastic. Thus, C. gariepinus could up-regulate
ALT 0.016 ± 0.001 0.010 ± 0.001* −37.3 glutamine synthesis and tolerate high levels of brain glu-
Results represent means ± S.E.M. (N = 5). (*) Significantly different from the tamine with no mortality when exposed to 75 mmol l−1 NH4 Cl.
corresponding control value, P < 0.05. Similarly, Ip et al. (2005b) demonstrated that brains of mud-
skippers, P. schlosseri and B. boddarti, accumulated high levels
of glutamine (14–18 ␮mol g−1 ) after exposure to a sub-lethal
that of fish injected with saline and a lethal dose of CH3 COONH4
concentration (100 and 8 mmol l−1 , respectively, at pH 7.0)
(Table 4).
of environmental ammonia. Taken together, these results indi-
cate that increased glutamine synthesis and accumulation are
3.3.3. Effects of MSO on activities of enzymes from the not the major mechanisms of ammonia toxicity in these fishes.
brain samples In addition, using magnetic resonance imaging, Veauvy et al.
The specific activity of GS from the brain of fish injected (2005) demonstrated a lack of association between increase in
with MSO followed by a lethal dose of CH3 COONH4 was sig- brain glutamine level and brain hydration status in O. beta,
nificantly lower than that from the brain of fish injected with and concluded that increased glutamine synthesis and accu-
a lethal dose of CH3 COONH4 only (Table 6). However, MSO mulation did not cause cerebral swelling in this species. Even
apparently had no significant effects in vivo on the specific activ- with swelling, the presence of excess cranial space in fishes
ities of GDH and ALT from the brain of fish injected with a lethal in general would prevent the build up of pressure therein (Ip
dose of CH3 COONH4 (Table 6). et al., 2005b). It is important to note that increased glutamine
synthesis in C. gariepinus exposed to environmental ammonia
3.3.4. In vitro effects of MSO on activities of enzymes from did not result in a decrease in glutamate or alanine content,
the brain samples which suggests that there must be an increase in the synthe-
The specific activities of GS, GDH and ALT from brain sis of glutamate. If indeed that was the case, C. gariepinus
samples of C. gariepinus were reduced significantly when deter- probably could resolve problems associated with redox balance
mined in the presence of MSO in vitro, with 44.9%, 52.8%, and and the depletion of ␣-ketoglutarate during the increased detox-
37.3% of inhibition, respectively (Table 7). ification of ammonia to glutamate in its brain. Furthermore,
the lack of increase in alanine content suggests that increased
4. Discussion glutamate synthesis proceeded in tandem with increased glu-
tamine synthesis and was separated from other transamination
4.1. The brain of C. gariepinus could tolerate high levels of reactions.
glutamine and ammonia when exposed to a sublethal
concentration of environmental ammonia 4.2. Increased glutamine synthesis and accumulation was
not the major cause of death in fish confronted with acute
It was previously thought that ammonia toxicity was due to ammonia toxicity
the disruption of redox and depletion of ␣-ketoglutarate in the
brain when ammonia was converted to glutamate to remove In addition to high tolerance of environmental ammonia, C.
ammonia. Furthermore, the increase in ATP demand for the gariepinus could tolerate high dose of CH3 COONH4 injected
N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213 211

into its peritoneal cavity followed by emersion or immersion. in ammonia level happened to be gradual (e.g. through a 4-day
With immersion, it had a 24 h LC50 value of 15 ␮mol g−1 fish period) as in the case of emersion.
(N. L. J. Wee and Y.K. Ip, unpublished results), which is com- In addition, it has been well established in mammals that
parable to that (15.6 ␮mol g−1 fish) of the giant mudskipper, the normal brain ammonia level is approximately 2 times that
P. schlosseri (Ip et al., 2005b). C. gariepinus could continue of the blood because of the pH gradient involved (Cooper and
to excrete excess ammonia injected into its peritoneal cavity Plum, 1987). In contrast, for C. gariepinus injected intraperi-
during subsequent immersion, but ammonia excretion would toneally with a lethal dose of CH3 COONH4 , the brain ammonia
become relatively ineffective during emersion (Ip et al., 2005a). content (9.6 ␮mol g−1 ) was only half of the plasma ammonia
Indeed, with emersion, it had a 24 h LC50 value of 12 ␮mol g−1 concentration (19.7 ␮mol ml−1 ). How the brain of C. gariepinus
fish (Y.K. Ip, unpublished results). After fish were injected confronted with acute ammonia toxicity maintains its ammo-
with a sublethal dose of CH3 COONH4 (8 ␮mol g−1 fish) nia content to be lower than that of the plasma awaits future
followed by emersion, the ammonia and glutamine contents studies.
in the brain increased continuously during the subsequent
24-h period, reaching 7 and 18 ␮mol g−1 , respectively, at hour 4.3. MK801 did not exhibit a protective effect against acute
24. By contrast, mammals would succumb to encephalopathy ammonia toxicity
at a brain ammonia content of 1–3 ␮mol g−1 (Cooper and
Plum, 1987; Felipo and Butterworth, 2002). These results Mortality in mammals, rats in particular, confronted with
suggest that when confronted with acute ammonia toxicity, acute ammonia toxicity could be prevented by the administration
the survival of C. gariepinus was crucially dependent on its of a wide range of NMDA receptor antagonists (Marcaida et al.,
high tolerance of ammonia and high capacity to detoxify 1992; Hermenegildo et al., 1996). Studies using in vivo cerebral
ammonia to glutamine in the brain, and corroborate in essence microdialysis show that acute ammonia exposure results in the
the proposition that increased glutamine synthesis and accu- activation of NMDA receptor-coupled nitric oxide-cyclic GMP
mulation are not the major mechanism of ammonia toxicity in signal transduction pathway in the rat brain (Hermenegildo et
C. gariepinus. al., 2000). An increase in intracellular NH4 + can also lead to a
After C. gariepinus was injected with a sublethal dose of depolarization of membrane potential (Sugden and Newsholme,
CH3 COONH4 (10 ␮mol g−1 fish) followed with immersion, 1975), and depolarization can result in the reversal of glutamate
there was a transient but significant increase in ammonia level transport resulting in an increase in the extracellular gluta-
in the brain between hours 0.5 and 2, reaching 3.9 ␮mol g−1 . mate concentration (Szatkowski et al., 1990). Besides increasing
The transient nature of ammonia accumulation could be a result extracellular glutamate concentration, changes in membrane
of the continual excretion of ammonia during immersion. There potential can lead to the removal of Mg2+ block on NMDA
was also a significant increase in the brain glutamine content receptors and hence its activation (Fan and Szerb, 1993). The
between hours 2 and 12, with a peak of 6.2 ␮mol g−1 at hour 6. deleterious effects of ammonia toxicity in mouse could be
From these results, it can be deduced that C. gariepinus accu- ameliorated through the administration of MK801, a NMDA
mulated glutamine in preference to ammonia in its brain, and receptor antagonist. At a dosage of 2 ␮g g−1 , MK801 provides
the content of the latter was maintained lower than that of the significant protection in animals injected intraperitoneally with
former whenever possible. At hour 2, there was a transient but a lethal dose of ammonia, and there is a significant improvement
significant decrease in glutamate content in the brain, which indi- in clinical grading with lower electroencephalogram activity
rectly implies that the rate of glutamine synthesis momentarily (Marcaida et al., 1992).
outpaced the rate of glutamate production. By contrast, MK801 (2 ␮g g−1 fish) had no protective effect
By contrast, for fish injected with a lethal dose (20 ␮mol g−1 on C. gariepinus injected with a lethal dose of CH3 COONH4
fish) of CH3 COONH4 followed with immersion, the brain (20 ␮mol g−1 fish). Because of the negative result, it became
ammonia content increased drastically to 9.6 ␮mol g−1 after essential to verify that the same batch of MK801 exhibited a pro-
30 min, while the brain glutamine content remained relatively tective effect on another animal species. Indeed, when the same
low at 5.4 ␮mol g−1 . Therefore, it can be concluded that batch of MK801 was tested at the same dosage on the Chinese
increased synthesis and accumulation of glutamine in the brain soft-shelled turtle, Pelodiscus sinensis, there was a significant
was not the major cause of death in C. gariepinus confronted delay in the time to death for turtle injected intraperitoneally
with acute ammonia toxicity. The brain ammonia level of with a lethal dose of CH3 COONH4 (S. M. L. Lee and Y. K. Ip,
9.6 ␮mol g−1 in fish injected with a lethal dose of ammonia was unpublished results). In view of the relatively high brain ammo-
actually lower than that (11 ␮mol g−1 ) reported for fish exposed nia content in C. gariepinus confronted with acute ammonia
to 4 days of emersion (Ip et al., 2005a). However, it is impor- toxicity and the lack of a protective effect from MK801, it is log-
tant to consider the fact that the ammonia level was reached ical to conclude that either membrane depolarization occurred
within a 30-min period in the former fish injected with a lethal but did not lead to activation of NMDA receptors, or membrane
dose of ammonia. Taken together, it can be deduced that one of potentials were resilient to NH4 + interference due to the pres-
the determining factors of acute ammonia toxicity was the rate ence of K+ channels and Na+ /K+ -ATPase with high substrate
at which ammonia built up in the brain. There could be some specificity for K+ , in the brain of this catfish. A similar conclu-
degree of plasticity because C. gariepinus was able to accom- sion was drawn by Ip et al. (2005b) on mudskippers, P. schlosseri
modate a higher level of ammonia in its brain when the increase and B. boddarti.
212 N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213

4.4. MSO had a slight protective effect against acute 4.5. MSO exhibited inhibitory effects on GDH and ALT
ammonia toxicity, which appeared unrelated to the activities in vitro
inhibition of GS
It was reported previously that the administration of MSO
MSO is an irreversible inhibitor (Folbergrova, 1964) of GS alone induced an increase in GDH activity, in the amination
in the mammalian system. Administration of MSO could ame- direction, and a decrease in ALT activity in astrocytes of rat brain
liorate effects of ammonia toxicity in patients suffering from (Subbalakshmi and Murthy, 1983). Subbalakshmi and Murthy
hyperammonemic encephalopathy, by reducing the levels of (1983) hypothesized that the GDH reaction might become an
glutamine synthesized and hence preventing death via the pre- important ammonia detoxification mechanism when the activity
vention of brain edema (Brusilow, 2002; Butterworth, 2002). In of GS was inhibited by MSO. The glutamate, so formed, would
the case of rats injected intraperitoneally with CH3 COONH4 , accumulate instead of being converted to glutamine as observed
the protective effects of MSO can also be a result of the pre- by Folbergrova et al. (1969).
vention of glutamate release and the prevention of activation While the brain of C. gariepinus injected with MSO + CH3
of NMDA receptors (Kosenko et al., 1994). In the case of COONH4 had higher content of glutamate, unlike rat brain
C. gariepinus injected with a lethal dose of CH3 COONH4 astrocytes, the specific activity of GDH, in the amination
(20 ␮mol g−1 fish), the administration of MSO (100 ␮g g−1 fish) direction, was comparable with that of fish injected with
reduced the mortality rate from 100% to 80% and at the same saline + CH3 COONH4 . However, when tested in vitro, MSO
time prolonged the time of death significantly from 26.7 min to exhibited quite a prominent inhibitory effect (53%) in vitro on
47.8 min. Thus, MSO apparently offered some forms of protec- the activity of GDH from the brain of C. gariepinus. There-
tion to C. gariepinus confronted with acute ammonia toxicity. fore, unlike GS, the inhibitory effect of MSO on GDH from
Superficially, these results appear to contradict the earlier con- C. gariepinus might be reversible because the GDH activ-
clusion that increased synthesis and accumulation of glutamine ity from brains of fish injected with MSO + CH3 COONH4
was not the major cause of death in C. gariepinus injected was not significantly different from that of fish injected with
intraperitoneally with a lethal dose of CH3 COONH4 . How- saline + CH3 COONH4 . Superficially, these observations appear
ever, a detailed analysis of the results reveals that MSO did to contradict the fact that MSO lead to an increase in the brain
not reduce the glutamine level in the brain of the experimental glutamate content when injected into the fish prior to the injec-
fish. This phenomenon occurred in spite of the facts that MSO tion with a lethal dose of CH3 COONH4 . However, it can be
inhibited (by 45%) GS activity in vitro from the brain of C. reasoned that MSO would simultaneously exert an inhibitory
gariepinus, and the prior administration of MSO resulted in a effect on the deaminating activity of GDH from the brain of
significant decrease (by 21.5%) in GS activity in the brain of fish this fish. Taken altogether, these results indicate that MSO sup-
injected intraperitoneally with a lethal dose of CH3 COONH4 . pressed the turnover of glutamate upon its production or release
In the latter fish, the negative effects of a decrease in GS activ- from protein degradation, resulting in its accumulation in the
ity on glutamine formation could have been compensated for brain despite its being siphoned off for glutamine synthesis. In
by an increase in glutamate concentration which shifted the addition, when tested in vitro, MSO also inhibited ATL activ-
equilibrium in favour of glutamine formation (see discussion ity (in the direction of alanine degradation) from the brain of C.
below). gariepinus. Thus, the decrease in rate of ammonia accumulation
More importantly, a prior injection with MSO resulted unex- in fish injected with MSO + CH3 COONH4 could also result in
pectedly in a significant decrease (by 38%) in the ammonia part from a decrease in the production of endogenous ammonia
content in the brain of fish injected with a lethal dosage through transdeamination. These results suggest that C. gariepi-
(20 ␮mol g−1 fish) of CH3 COONH4 . Thus, the protective effects nus could regulate the rate of ammonia production in the brain
of MSO on the mortality rate and the time of death of these when confronted with acute ammonia toxicity, and therefore
experimental fish were unrelated to the inhibition of glutamine efforts should be made in the future to elucidate the regulatory
synthesis, but resulted from a slower build up of ammonia in mechanisms involved. In addition, the effects of MSO alone on
the brain. This observation is novel, and has not been reported nitrogen metabolism in fish brain should be examined in greater
before for other fish or animal species. How did MSO slow down detail in future studies.
the build up of ammonia in the brain of C. garienpinus? This
could not be a result of the brains of the experimental fish being References
exposed to a lower level of ammonia, because the plasma ammo-
nia concentrations in fish injected with saline + CH3 COONH4 Arillo, A., Margiocco, C., Melodia, F., Mensi, P., Schenone, G., 1981. Ammonia
toxicity mechanisms in fish: studies on rainbow trout (Salmo gairdneri Rich).
and those injected with MSO + CH3 COONH4 happened to be Ecotoxicol. Environ. Saf. 5, 316–325.
comparable. An original and important observation made in this Bergmeyer, H.U., Beutler, H.O., 1985. Ammonia. In: Bergmeyer, H.U.,
study is that the glutamate content in the brain of fish with a Bergmeyer, J., Grapi, M. (Eds.), Methods of Enzymatic Analysis, vol.VIII.
prior injection of MSO was significantly higher than that of Academic Press, New York, pp. 454–461.
Binstock, L., Lecar, H., 1969. Ammonium ion currents in the squid giant axon.
the control fish with a prior injection of saline. These results
J. Gen. Physiol. 53, 342–361.
give indication for the first time that MSO might have an effect Bradford, M.M., 1976. Rapid and sensitive method for quantitation of microgram
on glutamate metabolism, and hence activities of GDH, in fish quantities of protein utilizing the principle of protein–dye binding. Anal.
brains. Biochem. 72, 248–254.
N.L.J. Wee et al. / Aquatic Toxicology 82 (2007) 204–213 213

Brusilow, S.W., 2002. Hyperammoniemic encephalopathy. Medicine 81, Margulies, J.E., Thompson, R.C., Demetriou, A.A., 1999. Aquaporin-4 water
240–249. channel is up-regulated in the brain in fulminant hepatic failure. Hepatology
Butterworth, R.F., 2002. Pathophysiology of hepatic encephalopathy. A new 30, 395A.
look at ammonia. Metab. Brain Dis. 17, 221–226. McKenzie, D.J., Randall, D.J., Lin, H., Aota, S., 1993. Effects of changes in
Chew, S.F., Wilson, J.M., Ip, Y.K., Randall, D.J., 2006. Nitrogenous excretion plasma pH, CO2 and ammonia on ventilation in trout. Fish Physiol. Biochem.
and defense against ammonia toxicity. In: Val, A., Almedia-Val, V., Randall, 10, 507–515.
D.J. (Eds.), Fish Physiology. The Physiology of Tropical Fishes, vol. 21. Meijer, A.J., Lamers, W.H., Chauleau, A.F.M., 1990. Nitrogen metabolism and
Academic Press, New York, pp. 307–395. ornithine cycle function. Physiol. Rev. 70, 701–748.
Cooper, J.L., Plum, F., 1987. Biochemistry and physiology of brain ammonia. Peng, K.W., Chew, S.F., Ip, Y.K., 1994. Free amino acids and cell volume
Physiol. Rev. 67, 440–519. regulation in the Sipunculid Phascolosoma arcuatum. Physiol. Zool. 67,
Desjardins, P., Michalak, A., Therrien, G., Chatauret, N., Butterworth, R.F., 580–597.
2001. Increased expression of the astrocytic/endothelial cell glucose trans- Peng, K.W., Chew, S.F., Lim, C.B., Kuah, S.S.L., Kok, W.K., Ip, Y.K., 1998.
porter (and water channel) protein GLUT-1 in relation to brain glucose The mudskippers Periophthalmodon schlosseri and Boleophthalmus bod-
metabolism and edema in acute liver failure. Hepatology 34, 253 (A237). daerti can tolerate environmental NH3 concentrations of 446 and 36 ␮M,
Eddy, F.B., Bamford, O.S., Maloiy, G.M.O., 1980. Sodium and chloride balance respectively. Fish Physiol. Biochem. 19, 59–69.
in the African catfish Clarias mossambicus. Comp. Biochem. Physiol. 66A, Person Le Ruyet, J., Galland, R., Le Roux, A., Chartois, H., 1997. Chronic
637–641. ammonia toxicity in juvenile turbot (Scophthalmus maximus). Aquaculture
Fan, P., Szerb, J.C., 1993. Effects of ammonium ions on synaptic transmission 154, 155–171.
and on responses to quisqualate and N-methyl-d-aspartate in hippocampal Randall, D.J., Wilson, J.M., Peng, K.W., Kok, T.W.K, Kuah, S.S.L., Chew, S.F.,
CA1 pyramidal neurons in vitro. Brain Res. 632, 225–231. Lam, T.J., Ip, Y.K., 1999. The mudskipper, Periophthalmodon schlosseri,
Felipo, V., Butterworth, R.F., 2002. Neurobiology of ammonia. Progr. Neurobiol. actively transports NH4 + against a concentration gradient. Am. J. Physiol.
67, 259–279. 277, R1562–R1567.
Felipo, V., Kosenko, E., Minana, M.D., Marcaida, G., Grisolia, S., 1994. Molecu- Rodicio, L.P., Sternberg, L.S., Walsh, P.J., 2003. Metabolic fate of exogenous
lar mechanism of acute ammonia toxicity and of its prevention by l-carnitine. 15 NH Cl in the gulf toadfish (Opsanus beta). Comp. Biochem. Physiol. C.
4
In: Felipo, V., Grisola, S. (Eds.), Hepatic Encephalopathy, Hyperammonemia 136, 157–164.
and Ammonia Toxicity. Plenum Press, New York, pp. 65–77. Rose, C., 2002. Increased extracellular brain glutamate in acute liver fail-
Folbergrova, J., 1964. Free glutamine level in the rat brain in vivo after methio- ure: decreased uptake or increased release? Metab. Brain Dis. 17, 251–
nine sulfoximine administration. Physiol. Bohemoslov. 13, 21–26. 261.
Folbergrova, J., Passonneau, J.V., Lowry, O.H., Schulz, D.W., 1969. Glycogen, Shankar, R.A., Anderson, P.M., 1985. Purification and properties of glutamine
ammonia and related metabolities in the brain during seizures evoked by synthetase from the liver of Squalus acanthias. Arch. Biochem. Biophys.
methionine sulphoximine. J. Neurochem. 16, 191–203. 239, 248–259.
Graham, J.B., 1997. Air-breathing fishes. Academic Press, San Diego, p. 299. Smart, G.R., 1978. Investigations of the toxic mechanisms of ammonia to fish-
Hermenegildo, C., Marcaida, G., Montoliu, C., Grisolia, S., Minana, M., Felipo, gas exchange in rainbow trout (Salmo gairdneri) exposed to acutely lethal
V., 1996. NMDA receptor antagonists prevent acute ammonia toxicity in conditions. J. Fish Biol. 12, 93–104.
mice. Neurochem. Res. 21, 1237–1244. Subbalakshmi, G.Y.C.V., Murthy, Ch.R.K., 1983. Effects of methionine sulfox-
Hermenegildo, C., Monfor, C.P., Felipo, V., 2000. Activation of N-methyl-d- imine on the enzymes of glutamate metabolism in isolated astrocytes of rat
aspartate receptors in rat brain in vivo following acute ammonia intoxication: brain. Biochem. Pharmacol. 22, 3695–3700.
characterization by in vivo brain microdialysis. Hepatology 31, 709–715. Sugden, P.H., Newsholme, E.A., 1975. The effects of ammonium, inorganic
Hillaby, B.A., Randall, D.J., 1979. Acute ammonia toxicity and ammonia excre- phosphate and potassium ions on the activity of phosphofructokinases from
tion in rainbow trout (Salmo gairdneri). J. Fish. Res. Board Can. 36, 621–629. muscle and nervous tissues of vertebrates and invertebrates. J. Biochem. 150,
Ip, Y.K., Lee, C.Y., Chew, S.F., Low, W.P., Peng, K.W., 1993. Differences in the 113–122.
responses of two mudskippers to terrestrial exposure. Zool. Sci. 10, 511–519. Szatkowski, M., Barbour, B., Attwell, D., 1990. Nonvesicular release of gluta-
Ip, Y.K., Chew, S.F., Randall, D.J., 2004a. Five tropical fishes, six different mate from glial cells by reversed electrogenic glutamate uptake. Nature 384,
strategies to defend against ammonia toxicity on land. Physiol. Biochem. 443–446.
Zool. 77, 768–782. Takahashi, H., Koehler, R.C., Brusilow, S.W., Traystman, R.J., 1991. Inhibition
Ip, Y.K., Chew, S.F., Wilson, J.M., Randall, D.J., 2004b. Defences against ammo- of brain glutamine accumulation prevents cerebral edema in hyperammone-
nia toxicity in tropical air-breathing fishes exposed to high concentrations mic rats. Am. J. Physiol. 261, H285–H829.
of environmental ammonia: a review. J. Comp. Physiol. 174, 565–575. Tsui, T.K.N., Randall, D.J., Hanson, L., Farrell, A.P., Chew, S.F., Ip, Y.K., 2004.
Ip, Y.K., Subaidah, R.M., Liew, P.C., Loong, A.M., Hiong, K.C., Wong, W.P., Dogmas and controversies in the handling of nitrogenous wastes: ammonia
Chew, S.F., 2004c. African Sharptooth catfish Clarias gariepinus does not tolerance in the oriental weatherloach Misgurnus anguillicaudatus. J. Exp.
detoxify ammonia to urea or amino acids but actively excretes ammonia Biol. 207, 1977–1983.
during exposure to environmental ammonia. Physiol. Biochem. Zool. 77, Veauvy, C.M., McDonald, M.D., Audekerke, J.V., Vanhoutte, G., Camp, N.V.,
242–254. Van der Linden, A., Walsh, P.J., 2005. Ammonia affects brain nitrogen
Ip, Y.K., Lau, I.Y., Wong, W.P., Lee, S.L., Chew, S.F., 2005a. The African sharp- metabolism but not hydration status in the Gulf toadfish (Opsanus beta).
tooth catfish Clarias gariepinus can tolerate high levels of ammonia in its Aquat. Toxicol. 74, 32–46.
tissues and organs during four days of aerial exposure. Physiol. Biochem. Warren, K.S., Schenker, S., 1964. Effect of an inhibitor of glutamine synthesis
Zool. 78, 630–640. (methionine sulfoximine) on ammonia toxicity and metabolism. J. Lab. Clin.
Ip, Y.K., Leong, M.W.F., Sim, M.Y., Goh, G.S., Chew, S.F., 2005b. Chronic Med. 64, 442–449.
and acute ammonia toxicity in mudskippers, Periophthalmodon schlosseri Wilkie, M.P., 1997. Mechanisms of ammonia excretion across fish gills. Comp.
and Boleophthalmus boddaerti: brain ammonia and glutamine contents, and Biochem. Physiol. 118, 39–50.
effects of methionine sulfoximine and MK801. J. Exp. Biol. 208, 1993–2004. Wilkie, M.P., 2002. Ammonia excretion and urea handling by fish gills: present
Kosenko, E., Kaminsky, Y., Grau, E., Minana, M.D., Marcaida, G., Grisolia, understanding and future research challenges. J. Exp. Zool. 293, 284–
S., Felipo, V., 1994. Brain ATP depletion induced by acute ammonia intox- 301.
ication in rats is mediated by activation of the NMDA receptor and Na+ , Willard-Mack, C.L., Koehler, R.C., Hirata, T., Cork, L.C., Takahashi, H.,
K+ -ATPase. J. Neurochem. 63, 2172–2178. Traystman, R.J., Brusilow, S.W., 1996. Inhibition of glutamine synthetase
Marcaida, G., Felipo, V., Hermenegildo, C., Minana, M.D., Grisolia, S., 1992. reduces ammonia-induced astrocyte swelling in rat. Neurosci. 71, 589–
Acute ammonia toxicity is mediated by NMDA type of glutamate receptors. 599.
FEBS Lett. 296, 67–68.

You might also like