You are on page 1of 10

e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227

Available online at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/etap

Sub-chronic exposure to chlorpyrifos induces


hematological, metabolic disorders and oxidative stress in
rat: Attenuation by glutathione

Eman E. Elsharkawy a , Doha Yahia a , Neveen A. El-Nisr b


a Department of Forensic Medicine and Toxicology Faculty of Veterinary Medicine, Assuit University, Egypt
b Animal Health Institute of Research, Egypt

a r t i c l e i n f o a b s t r a c t

Article history: The current work aimed to investigate the different toxic effects of chlorpyrifos (CPF) in
Received 15 June 2012 subchronic exposure. Two groups of Sprague-Dawley male rats were exposed to CPF alone
Received in revised form in a dose of 30 mg/kg body weight, or CPF dose as previous plus glutathione (GSH) in a
8 December 2012 dose of 100 mg/kg body weight, for 90 days, twice weekly, orally. Another two groups of rat
Accepted 19 December 2012 were given corn oil (control) or GSH. There is a significant decrease in hemoglobin concen-
Available online 27 December 2012 tration, haematocrit percentage, thrombocytic indices, total protein and albumin levels in
CPF-exposed group. CPF induced hyperglycemia and significant increase in total cholesterol,
Keywords: but a significant decrease in triglyceride levels was obtained. A significant increase in the
Chlorpyrifos levels of lipid peroxidation was obtained while a significant decrease of the total antioxidant
Glutathione was recorded. The decrease in glycogen content and some histopathological changes were
Hematological observed in liver after CPF exposure. Furthermore, co-administration of GSH can restore
Oxidative stress some of these alterations.
Metabolic © 2012 Elsevier B.V. All rights reserved.
Histochemical

pesticides used extensively throughout the world, including


1. Introduction Egypt. CPF is classified as a moderately hazardous, Class II
insecticide by the WHO (1997). Its acute toxicity varies accord-
Occupational exposure to pesticides is becoming a com- ing to the species and route of exposure. For example, acute
mon and increasingly alarming world-wide phenomenon. The oral LD50 for male rats is estimated to be 80 mg/kg bodyweight
World Health Organization estimates that the incidence of (Karanth et al., 2004).
pesticide poisoning in developing countries doubled between The chief action mechanism of OP pesticides occurs by
1987 and 1997 (WHO, 1997). Organophosphate (OP) pesti- inhibiting neuronal cholinesterase activity, a key enzyme
cides are among the most widely used synthetic chemicals involved in neurotransmission (Richardson et al., 1993).
for controlling a wide variety of pests. Residual amounts of CPF elicits a number of other effects, including: hepatic
(OP) pesticides have been detected in the soil, water bod- dysfunction, genotoxicity, embryotoxicity, teratogenicity, neu-
ies, vegetables, grains, and other food products (IARC, 1983; robehavioral, and neurochemical changes (Yasmashita et al.,
Poet et al., 2004). Chlorpyrifos (O,O -dithyl-O-3,5,6-trichloro-2- 1997; Goel et al., 2000). Acute and chronic exposure to CPF
pyrydyl phosphorothionate, or CPF) is among the leading OP has resulted in considerable liver damage, as evidenced by

E-mail address: medicine1971@yahoo.com (E.E. Elsharkawy).


1382-6689/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.etap.2012.12.009
e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227 219

changes in several liver enzymes (Goel et al., 2000). Several


2. Materials and methods
studies reported that hyperglycaemia is one of the side effects
of poisoning by OP in acute treatment and in sub-chronic
2.1. Chemicals
exposure via glycogenolysis and gluconeogenesis. There are
reports of incidents of transient glycosuria and hyperglycemia
Chlorpyriphos (CPF) of purity 96.5% (Technical Grade) and
in humans following acute exposure to OPs (Zadik et al., 1983;
glutathione 100% (pure powdered form) were purchased
Shobha and Prakash, 2000).
from Sigma Chemicals Co. (St. Louis, USA). Total antioxidant
The liver plays a major role in blood-glucose homeosta-
capacity and lipid peroxide malondialdehyde (MDA) and 4-
sis by maintaining a balance between the uptake and storage
hydroxyalkenals (HAE) were measured using commercial test
of glucose via glycogenesis and the release of glucose via
kits supplied Bio-diagnostics (Bio-diagnostics, Cairo, Egypt).
glycogenolysis and gluconeogenesis (Hers, 1990; Nordlie et al.,
Glucose, total cholesterol, triglycerides, total proteins were
1999; Abdollahi et al., 2003a,b). These toxic effects proba-
determined using colorimetric kits purchased from Stanbio
bly occur through the generation of reactive oxygen species
laboratories, USA. Histochemical determination of glycogen
(ROS) causing damage to the various membranous compo-
content was done using a Periodic Acid-Schiff Stain kit (PAS)
nents of the cells. ROS had been implicated in important
supplied from Dia-Technology, Egypt. All other chemicals were
pathologies such as cardiovascular, pulmonary and autoim-
of the highest grade available commercially.
mune diseases, inherited metabolic disorders, cancer, and
aging (Halliwell and Gutteridge, 1999). Pesticides are known
to produce oxidative stress, and extensive data suggest that
2.2. Animals and treatments
oxygen free-radical formation can be a major contributor to
the toxicity of pesticides (Lodovici et al., 1994; Bagchi et al.,
Sprague-Dawley rats, weighing between 100 and 150 g,
2002). Several studies point to the production of ROS as a sec-
obtained from the Animal Laboratory House of Assiut Uni-
ondary means of toxicity (Bebe and Panemangalore, 2003).
versity, Assiut, Egypt were used for the study. The animals
These include hydroxyl, peroxyl radicals and hydrogen per-
were housed in plastic cages and allowed to adjust to the new
oxide that target and inactivate biological macromolecules
environment for a week before starting the experiment. Rats
eventually damaging membranes and other tissues (Meister,
were fed on standard food pellets and tap water ad libitum. The
1998). Cells succumb to oxidative damage when the indige-
rats were housed at 24–25 ◦ C and in a daily dark/light cycle.
nous store of antioxidants is used up by the oxidant exposure.
The design of the study was in accordance with the ethical
The antioxidant machinery is composed of enzymes and non-
guidelines prescribed by the Institution. Rats were free from
enzymatic components.
infection.
The enzymatic component is made of free radical scaven-
Animals were randomly divided into four groups of seven
gers like catalase (CAT) and superoxide dismutase (SOD) as
animals each. Animals in CPF-treated group: were given
well as the glutathione-dependent enzymes like glutathione
twice/weekly a dose of CPF alone (30 mg/kg body weight,
peroxidase (GPx), glutathione reductase (GR) and glutathione
oral) (1/45 LD50 ) for 90 days. The acute oral LD50 of CPF was
S-transferase (GST) (Gutteridge and Halliwell, 2000). The non-
136 mg/kg body weight for rats (Suleiman et al., 2010). Animals
enzymatic component is primarily composed of thiols and
in CPF plus GSH-treated group: were simultaneously given glu-
glutathione (GSH). In vitro, exposure to CPF has been shown
tathione as an aqueous solution in a dose of (100 mg/kg body
to affect activities of all these enzymes (Gultekin et al.,
weight, oral); twice/weekly, during the 90 days and exposed
2000).
to the CPF at the previous dose. The GSH dose was used
Glutathione is a major water-soluble antioxidant, low
according to Sharma and Mishra (2006). Animals in GSH-
molecular-weight thiol compound of living cells and plays
treated group: were given aqueous solution of glutathione
an important role in the detoxification of potentially toxic
alone; twice/weekly for 90 days. Control group: were given only
compounds by its conjugation with xenobiotic or their
standard pellet diet and corn oil.
metabolites (Selvam, 2002). Its net negative charge and over-
all hydrophilic nature greatly increases the aqueous solubility
of the lipophilic moieties with which it becomes conjugated
(Rana et al., 2002). GSH may bind to free-radicals with their 2.3. Necropsy
sulfhydryl groups resulting in an activation of the brain antiox-
idant enzymes and thus limiting the free-radical action more At the end of the study period, rats were fasted overnight;
extensively. Therapy with this agent was found to normalize anesthetized by inhalation of diethyl ether and blood sam-
the cellular antioxidant system, enzyme scavengers, inter- ple was drawn by puncturing the orbital sinus of the animals
rupted membrane LPO reaction and ATPase inactivation in rats under light anesthesia. Blood samples were put immediately
(Rana et al., 2002). The aim of this study was to determine the into silicon disposable glass tubes with ethylenediamine tetra
effect of a subchronic 90-days exposure to CPF on the hema- acetic acid (EDTA) as the anticoagulant to determine the dif-
tological, metabolic parameters and oxidative status of rats. ferent hematological parameters and in a clean glass test tube
And also was to evaluate the role of water soluble antioxidant and allowed to stand for 30 min at room temperature to clot
glutathione, to attenuate these changes following subchronic and centrifuged at 4000 × g for 10 min for serum separation.
exposure to CPF. This is the first time to our knowledge that the Rats were sacrificed after anesthetizing with chloroform, liv-
antioxidant glutathione has been employed to protect against ers were immediately excised and fixed in 10% neutral buffer
CPF’s toxic effects. formalin and were processed for light microscopy.
220 e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227

2.4. Hematological parameters in paraffin wax. Paraffin sections (5 ␮m thick) were stained for
routine histological study using hematoxylin and eosin (H&E).
Hematological parameters [red blood cell count (RBC),
hemoglobin (HB), haematocrite (HCT), red Blood cell dis- 2.9. Histochemical determination of glycogen content
tribution width (RDW), mean corpuscular volume (MCV),
mean corpuscular hemoglobin (MCH), mean corpuscular Paraffin sections were stained by Periodic acid Schiff’s reagent
hemoglobin concentration (MCHC), white blood cell count technique (PAS) for demonstrating glycogen content (Pears,
(WBC) and differential leucocytic count]. Thrombocytic 1985).
indices [total platelet count (PLT), mean platelet volume (MPV),
total platelet crit (PCT) and platelet distribution width (PDW)] 2.10. Statistical analysis
were analyzed by the automated parameter called Electronic
Blood Cell Counter (Medonic vet. CA620, Sweden). Data were analyzed using one-way analysis of variance
(ANOVA) followed by Duncan’s multiple range test. All data
2.5. Metabolic parameters were expressed as mean ± SD for all experimental and con-
trol animals. P < 0.05 was considered significant compared to
2.5.1. Glucose assay control.
Glucose was measured by the glucose oxidase and peroxi-
dase using quinoneimine as a chromogen. The amount of
plasma glucose is related to the amount of quinoneimine, 3. Results
which is measured spectrophotometrically at 505 nm accord-
ing to Bergmeyer et al. (1974). 3.1. Changes in hematological parameters

2.5.2. Protein assay When the GSH-treated animals were compared with the con-
Total protein (TP) and albumin (Alb) were determined in serum trol animals at the end of the 90 days, they did not differ
according to Doumas, 1971. significantly in any terms of the hematological parameters.
When the CPF-treated group and the CPF plus GSH-treated
2.5.3. Total cholesterol and triglycerides assay group were compared with the control group, they did not
Total cholesterol and triglycerides were determined in serum differ significantly in RBC counts but significantly lower in
according to Tietz, 1994 and Carlson, 1963, respectively. terms of HB, HCT, MCV, MCH, MCHC and RDW values (Table 1).
CPF-treated group had significantly higher WBC, lymphocyte
2.6. Determination of lipid peroxidation and monocyte counts but significantly lower in granulocyte
counts than the control animals. CPF plus GSH-treated group
Measurements of MDA and HAE have been used as an indi- was significantly more lymphocytes and significantly fewer
cator of lipid peroxidation. The colorimetric kit was used to granulocytes than the control animals (Table 2). CPF-treated
determine the levels of oxidized lipid according to Buege and group and the CPF plus GSH-treated group were significantly
Aust, 1978. This assay is based on the reaction of chromogenic decreased in terms of PLT, MPV, PCT and PDW values than
reagent with MDA and HAE at 45 ◦ C to yield a stable chro- the control. However, the CPF plus GSH-treated group was
mophore with maximum absorbance at 586 nm. The rate of significantly differed in most previous parameters than the
lipid peroxidation was expressed as nmol of reactive sub- CPF-treated group (Tables 1–3).
stance formed/min/mg protein.
3.2. Changes in glucose assay
2.7. Determination of total antioxidant
The CPF-treated group and the CPF plus GSH-treated group
Total antioxidant status was assessed by using commercial had significantly higher glucose levels than the control group.
test kit and using spectrophotometer, according to the meth- There is a significant difference between the CPF and CPF plus
ods described by Koracevic et al. (2001). The basis of latter GSH-treated group (Table 4).
method is a change of 2,2 azino-di (3-ethylbenzthiazoline) sul-
fonate (ABTS), incubated with peroxidase (metmyoglobin) and 3.3. Changes in protein assay
H2 O2 into its cation form. This leads to a change in color of
the sample examined proportional to the concentration. After In comparison with the control group, the CPF-treated group
addition of 1 mL of ABTS incubated with metmyoglobin to and the CPF plus GSH-treated group had significantly lower
20 mL of the supernatant, the sample was centrifuged and the total protein and albumin levels, when the CPF plus GSH-
absorbance was read at a wavelength of 600 nm. Then, 200 mL treated group was compared with the CPF-treated group;
of H2 O2 was added, and another reading was obtained after they had significantly higher total protein and albumin levels
3 min. The data are presented as mmol/L. (Table 4).

2.8. Histopathological study 3.4. Changes in total cholesterol and triglycerides

Liver samples were dissected and fixed in 10% neutral forma- The CPF-treated group and the CPF plus GSH-treated group
lin, dehydrated in ascending grades of alcohol and embedded both had significantly higher total cholesterol levels and
e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227 221

Table 1 – Effect of sub-chronic exposure to chlorpyrifos on the hematological parameters and the protective effect of
glutathione.
RBCs (×106 /mm3 ) HB (g/dl) HCT (%) MCV (␮m) MCH (pg) MCHC (g/dl) RDW (%)
CPF 8.40 ± 0.35 14.22 ± 0.57*bc 40.55 ± 1.80*c 48.27 ± 1.21*c 16.97 ± 0.33*c 35.20 ± 0.21*c 35.60 ± 1.11*c
CPF + GSH 8.77 ± 0.38 15.20 ± 0.47a 42.30 ± 1.41*c 48.23 ± 0.82*c 17.36 ± 0.23*c 36.00 ± 0.20*c 35.80 ± 1.26*c
GSH 8.34 ± 0.15 15.78 ± 0.31a 48.65 ± 1.35ab 49.41 ± 0.56ab 18.25 ± 0.01ab 37.65 ± 0.22ab 36.83 ± 1.24ab
Control 8.64 ± 0.19 15.58 ± 0.31 47.65 ± 1.35 50.50 ± 0.46 18.15 ± 0.02 37.05 ± 0.25 36.63 ± 1.54
Data are expressed as means ± S.D. of seven animals per group.*denotes P < 0.05 as compared to control group, a denotes P < 0.05 as compared
to CPF-group. b denotes P < 0.05 as compared to CPF + GSH-group. C denotes P < 0.05 as compared to GSH-group (One-way ANOVA/Duncan).

Table 2 – Effect of sub-chronic exposure to chlorpyrifos on the total and differential leucocytes counts (103 mm−3 ) and the
protective effect of glutathione.
WBCs Lymphocytes Granulocytes Moncytes
CPF 11.05 ± 2.18*bc 6.92 ± 1.01*bc 3.10 ± 0.98*bc 1.02 ± 0.19*bc
CPF + GSH 9. 93 ± 0.90ac 5.10 ± 0.60*ac 2.35 ± 0.27*ac 0.60 ± 0.34a
GSH 10.55 ± 0.24ab 4.23 ± 0.80ab 5.98 ± 0.80ab 0.88 ± 0.19a
Control 10.53 ± 0.34 4.13 ± 0.12 5.50 ± 0.25 0.80 ± 0.05
Data are expressed as means ± S.D. of seven animals per group.*denotes P < 0.05 as compared to control group, a denotes P < 0.05 as compared
to CPF-group. b denotes P < 0.05 as compared to CPF + GSH-group. C denotes P < 0.05 as compared to GSH-group (One-way ANOVA/Duncan).

Table 3 – Effect of sub-chronic exposure to chlorpyrifos on the thrombocytic indices and the protective effect of
glutathione.
PLT (103 /ml) MPV (␮m) PDW (␮m) PCT (%)
CPF 468.6 ± 62.4*c 5.45 ± 0.25 c 8.35 ± 0.41*c 0.24 ± 0.02*bc
CPF + GSH 497.4 ± 41.7*c 5.50 ± 0.11*c 8.45 ± 0.17*c 0.27 ± 0.04*ac
GSH 698.6 ± 15.8 ab 6.48 ± 0.38ab 9.29 ± 0.15ab 0.33 ± 0.01ab
Control 696.3 ± 15.8 6.06 ± 0.08 9.16 ± 0.17 0.30 ± 0.01
Data are expressed as means ± S.D. of seven animals per group.*denotes P < 0.05 as compared to control group, a denotes P < 0.05 as compared
to CPF-group. b denotes P < 0.05 as compared to CPF + GSH-group. C denotes P < 0.05 as compared to GSH-group (One-way ANOVA/Duncan).

Table 4 – Effect of sub-chronic exposure to chlorpyrifos on the biochemical parameters and the protective effect of
glutathione.
TP (g\dl) Alb (g\dl) Total cholesterol (mg/dl) Triglycerides (mg/dl) Glucose (mg/dl)
CPF 5.98 ± 0.1*bc 4.33 ± 0.45*bc 115.00 ± 4.25*bc 80.65 ± 5.6*bc 81.5 ± 4.8*bc
CPF + GSH 7.40 ± 0.9*ac 4.82 ± 0.91*ac 93.72 ± 3.11*ac 105.08 ± 5.2*ac 72.6 ± 5.5*ac
GSH 7.86 ± 0.27ab 5.47 ± 0.56ab 79.42 ± 3.51ab 125.20 ± 6.8 ab 60.0 ± 4.2ab
Control 7.88 ± 0.17 5.67 ± 0.56 81.07 ± 3.23 123.93 ± 7.15 63.8 ± 5.2
Data are expressed as means ± S.D. of seven animals per group.*denotes P < 0.05 as compared to control group, a denotes P < 0.05 as compared
to CPF-group. b denotes P < 0.05 as compared to CPF + GSH-group. C denotes P < 0.05 as compared to GSH-group (One-way ANOVA/Duncan).

significantly lower triglyceride than the control group. When


Table 5 – Effect of sub-chronic exposure to chlorpyrifos
the CPF plus GSH-treated groups were compared to the CPF- on the oxidative status and the protective effect of
treated group, they had significantly lower total cholesterol glutathione.
levels and significantly higher triglyceride’s levels (Table 4).
Total antioxidant MDA&HAE
(mM/L) (nmol/mg protein)
3.5. Lipid peroxidation and total antioxidant CPF 0.36 ± 0.02*bc 20.58 ± 0.51*bc
CPF + GSH 0.45 ± 0.01*ac 14.53 ± 0.44*ac
Table 5 shows that the CPF-treated group and the CPF GSH 0.1 ± 0.12ab 13.80 ± 0.21ab
plus GSH-treated group in rats caused significantly marked Control 0.98 ± 0.14 13.50 ± 0.31
increase in the levels of MDA and 4HNE in blood when com- Data are expressed as means ± S.D. of seven animals per
pared with control. While the total antioxidant status was group.*denotes. P < 0.05 as compared to control group, a denotes
statistically significantly lower than the control group. In P < 0.05 as compared to CPF-group. b denotes P < 0.05 as compared
to CPF + GSH-group. C denotes P < 0.05 as compared to GSH-group
contrast, treatment with GSH resulted in a significant ame-
(One-way ANOVA/Duncan).
lioration of the activities of both the levels of MDA and 4HNE
and the total antioxidant, when compared with CPF-treated
group and the CPF plus GSH-treated group.
222 e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227

Fig. 1 – Livers from the control (a) exhibited normal arrangement and structure of the hepatocytes’ H&E X40. Liver sections
of CPF-treated rat showed (b) sever vacuolar degeneration H&E X40, (c) disarrangement of the hepatic cord H&E X25, (d)
congestion of the portal blood vessels which, surrounded with thick connective tissue and monocytic cellular infiltration
H&E X10, (e) the portal area blockaded with fibrous strands associated with kupffer cell infiltration H&E X40. Liver of rat of
CPF plus GSH-group showed (f) more or less normal hepatocytes associated with regular architecture’s H&E X25.

3.6. Histopathological study CPF plus GSH-treated animals exhibited histopathological


changes: The livers of the CPF-treated animals displayed hem-
Light microscopic examination of the livers revealed that orrhage, vacuolar degeneration, dilation of sinusoids (Fig. 1b).
those of the control exhibited normal arrangement of the Hepatocytes lost their radial arrangement (Fig. 1c) with mono-
hepatocytes and there were visible sinusoids in most places. nuclear cell infiltration, vascular congestion, and necrosis
The nuclei of the hepatocytes had a normal vesicular struc- (Fig. 1d–e), while the livers of the CPF plus GSH-treated animals
ture, and the cytoplasm appeared uniform and regular (Fig. 1a). exhibited regular arrangement of the hepatocytes with little
In contrast, the livers of the CPF-treated animals and the mononuclear cell infiltration and slight vascular congestion
e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227 223

Fig. 2 – Glycogen in paraffin sections of rat liver stained with PAS. Original magnification is ×400. (a) Control liver showed
the homogenous feature of glycogen that was arranged in regular lines in all hepatocytes. (b) Liver from CPF-treated groups
showed marked disturbance of glycogen content that was polling in one side of most the hepatocytes. (c) The livers of the
CPF plus GSH-treated animals showed less polling, decrease and disturbance of glycogen content than CPF-treated animals.

(Fig. 1f). However, the latter livers did not display the vacuolar residues, and this might be a possible physiological reason for
degeneration, calcification and necrosis that were detected in the inverse proportion to the result obtained. They also sug-
the livers of CPF-treated animals. gested that the poisoning by pesticide residue induce anemia
due to interference of hemoglobin biosynthesis and short-
3.7. Histochemical study ening of the life span of circulating erythrocytes. Therefore,
the obtained decrease in the hemoglobin concentration might
Glycogen in paraffin sections of rat liver stained with PAS is be due to the effect of pesticide on erythropoietic tissue and
illustrated in (Fig. 2) containing three figures (a, b and c liver). haem biosynthesis in rats. Our findings suggested that the
Control liver showed the homogenous feature of glycogen exposure of rats to CPF result in microcytic hypochromic ane-
that was arranged in regular lines in all hepatocytes (Fig. 2a). mia, which may be due to the inhibition of erythropoiesis and
While sections from animals exposed to CPF and CPF plus GSH haemosynthesis.
showed disturbance of glycogen content that was polling in Another study has shown that some OP insecticides can
one side of most the hepatocytes (Fig. 2b). However, the livers increase WBC counts (Celik et al., 2009). These findings are
of the CPF-treated animals showed more obvious disturbance consistent with our results, which revealed that CPF in sub-
and pooling of glycogen content than CPF plus GSH-treated chronic exposure elevated WBC, lymphocyte and monocyte
animals (Fig. 2c). counts. It has been shown that leukocytosis may be due to
increased leukocyte mobilization and can be directly pro-
portional to the severity of the causative stress condition
4. Discussion (Celik et al., 2009). Thus, the leukocytosis seen in the CPF-
treated rats may reflect the response of the immune system
Several studies have shown that OP insecticides can alter to the stress imposed by CPF. It is possible that the toxic
hematological parameters in experimental animals (Kalender effects of CPF weaken the constitution of the rats, which
et al., 2006; Yehia et al., 2007; Celik and Suzek, 2008) and in then acquire infections that provoke an immune response
clinical cases (Patil and Govindwar, 2003). The present results (Celik and Suzek, 2008). In addition, OP insecticides have been
revealed that exposure to CPF was associated with significant shown to induce hemorrhage, inflammatory cell infiltration
decreases in HB, HCT, MCV, MCHC and RDW values but no sig- (Morowati, 1998; Elhalwagy et al., 2008), tissue damage, and
nificant change was observed in RBC counts. A similar trend necrosis (Kalender et al., 2006), which could provoke increased
has been observed in mice treated with Nuvacron and Furadan WBC and lymphocyte counts. This possibility is supported
(Gupta et al., 1982), and in rats treated with CPF (Shalby, 1998). by our histopathological analysis of the CPF-treated rats in
In this study, no significant change was observed in RBC counts this study, which revealed the occurrence of hepatic hem-
but HB content was decreased in CPF-treated rats. Because orrhage, inflammatory cell infiltration, edema and necrosis.
of this a significant decease was observed in both MCH and The results obtained from this study revealed that the CPF-
MCHC contents. As MCH shows hemoglobin content in eryth- treated group exhibited decreases in PLT, MPV, PCT and PDW
rocytes, and MCHC means hemoglobin rate to RBC (Nussey values compared to the control. Similarly, Kalender et al.
et al., 1995), the reduction in HB content was suggested to be (2006) reported that a significant decrease was observed in
due to an adverse effect of OP on the bone marrow (Seiverd, thrombocyte counts at the end of 1st and 4th weeks of diazi-
1972). MCV has been reported to provide information about non exposure. And also, a significant decrease was observed
the size and status of erythrocytes (Nussey et al., 1995). Thus, in platelets counts indices in diazinon sub-acute exposure
the observed decrease in the mean of MCV and RDW in rats (Hariria et al., 2010 and Hariria et al., 2011). In contrast,
exposed to CPF was a manifestation of atrophied erythrocytes Kalender et al. (2009) reported that malathion acute exposure
and microcytic anemia. Patil and Govindwar, 2003) stated that induced increase in thrombocyte’s count. Our results indi-
many steps in the heme biosynthesis are inhibited by pesticide cated that CPF induced thrombocytopenia in exposed rats. It
224 e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227

should be noted that it is the first time we know a study discuss after malathion treatment (Gupta, 1974). It is well estab-
the effect of CPF on thrombocytic indices. The thrombocy- lished that the increase in the hepatic glycogen rate is due to
topenia may develop due to antibody production against viral the stimulation of the key enzyme of glycogenesis (glycogen
antigens attached to platelet surfaces or to nonspecific binding synthetase) (Rezg et al., 2006). One possible explanation for
of antigen-antibody complexes to platelet surfaces (Cheung these results could be the turnover of glucose by its release
et al., 2005). Furthermore, drug and xenobiotic-induced throm- through a succession of glycogenolysis and gluconeogenesis
bocytopenia has been reported in dogs, cats, and horses. One (Pournourmohammadi et al., 2004), which involves abnormal
mechanism is marrow suppression of megakaryocytes or pos- hyperglycemia and its storage via glycogenesis (Rezg et al.,
sibly even generalized marrow stem cell suppression after 2006). This is proven by marked changes to the overall histo-
administration of xenobiotics (Gibbins and Mahaut-Smith, architecture of the liver (Gupta, 1974). Hepatic atrophy has
2004). Another mechanism is increased platelet destruction also been observed when repeated doses of CPF are given. In
and consumption resulting in impair of platelet function. addition, Radiger et al. (2003) stated that the decline of glyco-
Numerous xenobiotics were reported to block platelet receptor gen content may reflect a disruption in enzyme pathways of
binding or to change platelet membrane charge or permeabil- glycolysis.
ity (Page et al., 2002). Quantitative platelet disorders have been Pesticides are known to produce oxidative stress, and
reported in liver disease with or without coagulation protein extensive data suggest that oxygen free-radical formation can
deficiencies (Tkaczyk and Baj, 2002). be a major contributor to the toxicity of pesticides (Lodovici
In this study, the CPF-treated animals exhibited signifi- et al., 1994; Bagchi et al., 2002). It has been previously reported
cantly lower total protein and albumin levels than the control that acute exposure to OP pesticides may induce oxidative
animals. Albumin, which is the most abundant blood plasma stress in rats (Koner et al., 1997). Our result strengthens the
protein, is produced by the liver and several studies have hypothesis and suggests that induction of oxidative stress
shown that its production can be decreased by OP such as CPF is perhaps the central mechanism by which OP pesticides
(Kalender et al., 2005; Yousef et al., 2006; Ogutcu et al., 2008). exert their cellular action. Oxidative damage primarily occurs
Since reductions in albumin levels are generally suggestive through production of reactive oxygen species, including
of liver disease, it is possible that OP alters protein and free hydroxyl radicals and hydrogen peroxide that subsequently
amino acid metabolism and their synthesis within the liver react with biological molecules as well as causing damage
(Ncibi et al., 2008). to membranes and other tissues (Banerjee et al., 1999). A
OP insecticides generally increase the total cholesterol and significant decrease in the total antioxidant level and a con-
total lipid levels (Kalender et al., 2005; Ogutcu et al., 2008; comitant increase in LPO level following administration of the
Lasram et al., 2009) and indeed, in the present study, CPF ele- pesticide dose were observed in the present study. Suppres-
vated the total cholesterol levels of the rats. This increase sion in the total antioxidant capacity is due to the decrease
in serum cholesterol can be attributed to the effect of pes- in biologic antioxidant enzymes. The results are in agree-
ticides on the permeability of the liver cell membrane (Yousef ment with previous studies by Vidyasagar et al. (2004), where
et al., 2006). In addition, the increase in serum total choles- alterations in glutathione, total thiols, lipid peroxidation and
terol levels may be due to the blockage of the liver bile ducts, changes in the activity of SOD and catalase in erythrocytes
which reduces or stops cholesterol secretion into the duode- have been noted following exposure to OP pesticides. Deple-
num (Zaahkouk et al., 2000; Ogutcu et al., 2008). Increased tion in biologic antioxidant enzymes is responsible for the
serum cholesterol levels may be a sign of liver damage. Some most cytotoxic effects of OP (John et al., 2001). CPF induced
pesticides also decrease the triglyceride levels (Kalender et al., oxidative stress (Verma et al., 2007), and effected on thio-
2005; Ogutcu et al., 2008). It has been shown that parenchymal barbituric acid reactive substances, scavenging enzymes and
liver diseases are associated with decreases in triglyceride lev- glutathione in rat tissues (Verma and Srivastava, 2003). The
els (Kalender et al., 2005). In the present study, CPF exposure basis of OP toxicity in the production of oxidative stress may
reduced the triglyceride levels. These observations are consis- be due either to: (a) their “redox-cycling” activity, where they
tent in the presence of liver cell damage that was seen in the readily accept an electron to form free radicals and then trans-
CPF-exposed rats by light microscopy. fer them to oxygen to generate superoxide anions and hence
Additionally, several studies indicate that OP exposure hydrogen peroxide through disputation reactions, or (b) to ROS
causes hyperglycemia, as an immediate consequence of generation via changes in normal antioxidant homeostasis
malathion administration (Gupta, 1974; Rodrigues et al., 1987), resulting in antioxidant depletion, if the requirement of con-
which was explained by stimulation of glycogenolysis in sev- tinuous antioxidants is not maintained (Banerjee et al., 1999;
eral organs (Pournourmohammadi et al., 2004; Abdollahi et al., Kovacic, 2003; Vidyasagar et al., 2004).
2004) and gluconeogenesis in liver (Abdollahi et al., 2004), OP insecticides are known to induce various histopatho-
provoking the release of glucose into the blood. Treatment logical changes in the liver tissues (Goel et al., 2005; Gokcimen
of rats with acute diazinon also resulted in hyperglycemia, et al., 2007; Sayım, 2007; Yehia et al., 2007) and indeed,
increased activity of the hepatic gluconeogenic enzyme and we found that CPF-induced inflammatory cell infiltration,
phosphoenolpyruvate carboxykinase (PEPCK) (Matin et al., hemorrhage, calcification, vacuolar degeneration, dilation of
1990). In the present study, CPF induced hyperglycemia sinusoids, vascular congestion and necrosis in the rat liver.
and a significant disturbance in hepatic glycogen distri- These changes are entirely consistent with the changes in var-
bution rate after 90 days of sub-chronic exposure. It has ious biochemical parameters that were also observed. Such
been shown that acute exposure to malathion leads to an liver damage may arise from the toxic effects of CPF, which dis-
increase of glycogen deposition in the liver during 6–24 h turbs the detoxification mechanisms of the liver. In addition,
e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227 225

it is possible that CPF, like several other insecticides, adversely cAMP levels, glycogenolysis, gluconeogenesis and
affects the cytochrome P450 system or the mitochondrial susceptibility to a mitochondrial toxin. Mol. Cell. Biochem.
membrane transport system of hepatocytes. Exposure to CPF 252, 205–211.
Abdollahi, M., Soleimani, F., Kangarlou, S., 2003b. A review on
may lead to membrane damage in liver cells and eventual
blood glucose variations and affecting parameters. Middle
loss of membrane integrity (Gokcimen et al., 2007). It has East Pharm. 11, 6–10.
been reported previously that during liver damage, there is an Abdollahi, M., Donyavi, M., Pournourmohammadi, S., Saadat, M.,
observed decrease in antioxidant defenses in the liver (Seven 2004. Hyperglycemia associated with increased hepatic
et al., 2004). Glycogen is the major storage form of carbo- glycogen phosphorylase and phosphoenolpyruvate
hydrates in the animal liver (Murray et al., 2000). A marked carboxykinase in rats following subchronic exposure to
decrease of hepatocyte’s glycogen was noticed after CPF expo- malathion. Comp. Biochem. Physiol. C 137 (4),
343–347.
sure, focally to the central vein region indicating the increase
Bagchi, D., Balmoori, J., Bagchi, M., Ye, X., Williams, C.B., Stohs,
of vacuolated cells that may affect hepatocytes function. How- S.J., 2002. Comparative effects of TCDD, endrin, naphthalene
ever, marked increases in glycogen deposits in one side of and chromium (IV) on oxidative stress and tissue damage in
some hepatocytes may be due to the detoxification by hepato- the liver and brain tissues of mice. Toxicology 175,
cytes. These findings are in good agreement with the results of 73–82.
a previous study by Radiger et al. (2003). They stated that, the Banerjee, B.D., Seth, V., Bhattacharya, A., Pasha, S.T., Chakraborty,
A.K., 1999. Biochemical effects of some pesticides on lipid
decline of glycogen content may reflect a disruption in enzyme
peroxidation and free-radical scavengers. Toxicol. Lett. 107,
pathways of glycolysis.
33–47.
Antioxidants have a number of biological activities, includ- Bebe, F.N., Panemangalore, M., 2003. Exposure to low doses of
ing immune stimulation and alteration of the metabolic endosulfan and chlorpyriphos modifies endogenous
activities of carcinogens. These vitamins can also prevent antioxidants in tissues of rats. J. Environ. Sci. Health 38,
genetic changes by inhibiting the DNA damage induced by 349–363.
reactive oxygen metabolites (Verma et al., 2007). Glutathione, Bergmeyer, H.U., Bernt, E., Schmidt, F., Stork H, 1974. d-glucose:
determination with hexokinase and glucose-6-phosphate
the major water-soluble antioxidant, is known to protect cel-
dehydrogenase. In: Bergmeyer, H.U. (Ed.), Methods of
lular membranes and lipoproteins from peroxidation (Rana Enzymatic Analysis Vol. 3. Academic Press, New York, pp.
et al., 2002), may minimize lipid peroxidation in biological 1196–1201.
systems (Selvam, 2002) and acts to protect both cytosol and Buege, J.A., Aust, S.D., 1978. Microsomal lipid peroxidation.
membranes against free-radical attack Hogg, 1998. Its high Methods Enzymol. 52, 306–310.
electron donating capacity combined with its intensive intra- Carlson, L.A., 1963. Determination of serum triglycerides. J.
cellular concentration endows GSH with great reducing power, Atheroscler. Res. 3, 334–336.
Celik, I., Suzek, H., 2008. The hematological effects of methyl
which is used to regulate a complex thiol exchange system
parathion in rats. J. Hazard. Mater. 153, 1117–1121.
(Meister, 1994). Celik, I., Yilmaz, Z., Turkoglu, V., 2009. Hematotoxic and
The hematological, metabolic disorders and the changes hepatotoxic effects of dichlorvos at sublethal dosages in rats.
of the oxidative status that were induced by CPF exposure Environ. Toxicol. 24, 128–132.
were at least partially normalized when the GSH antioxidant Cheung, R., McAuley, R., Pollard, J., 2005. High mortality rate in
was given with CPF. Moreover, our light microscopic anal- patients with advanced liver disease independent of exposure
to general anesthesia. J. Clin. Anesth. 17 (3), 172–176.
yses revealed that the antioxidant-treated animals did not
Doumas, B., 1971. Biochemical determination of albumin
exhibit the hepatic calcification, vacuolar degeneration and
concentration. Clin. Chem. Acta, 31–87.
necrosis seen in the livers of the CPF-treated group. Further- Elhalwagy, M.E.A., Darwish, N.S., Zaher, E.M., 2008. Prophylactic
more, while both groups exhibited hepatic inflammatory cell effect of green tea polyphenols against liver and kidney injury
infiltration, hemorrhage, dilation of sinusoids, and vascular induced by fenitrothion insecticide. Pestic. Biochem. Physiol.
congestion, these changes were less severe in the CPF plus 91, 81–89.
GSH-treated group. Thus, GSH may be could ameliorate the Gibbins, M.J., Mahaut-Smith, P.M., 2004. Platelets and
Megakaryocytes: Perspectives and Techniques. Humana Press
liver damage induced by CPF exposure. In conclusion, our
Inc., New York city, ISBN: 1588291014.
results demonstrate that sub-chronic exposure to CPF leads Goel., A., Chauhan, D.P., Dhawan, D.K., 2000. Protective effects of
to significant hematological, metabolic disorders and oxida- zinc in chlorpyriphos induced hepatotoxicity: a biochemical
tive damage. Administration of non-enzymatic water soluble and trace elemental study. Biol. Trace Elem. Res. 74,
antioxidant GSH exerts a significant protective role against the 171–183.
CPF toxic effect in rat. Goel, A., Dani, V., Dhawan, D.K., 2005. Protective effects of zinc on
lipid peroxidation, antioxidant enzymes and hepatic
histoarchitecture in chlorpyriphos-induced toxicity. Chem.
Biol. Interact. 156, 131–140.
Conflicts of interest
Gokcimen, A., Gulle, K., Demirin, H., Bayram, D., Kocak, A.,
Altuntas, I., 2007. Effects of diazinon at different doses on rat
None declared. liver and pancreas tissues. Pestic. Biochem. Physiol. 87,
103–108.
Gultekin, F., Ozturk, M., Akdogan, M., 2000. The effect of
references
organophosphate insecticide chlorpyrifos-ethyl on lipid
peroxidation and antioxidant enzymes in vitro. Arch. Toxicol.
74, 533–538.
Abdollahi, M., Chan, T.S., Subrahmanyam, V., O’Brien, P.J., 2003a. Gupta, P.K., 1974. Malathion induced biochemical changes in rats.
Effects of phosphodiesterase 3,4,5 inhibitors on hepatocyte Acta Pharmacol. Toxicol. 35 (3), 191–194.
226 e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227

Gupta, M., Bagchi, G., Bandyopadhyay, S., Sasmal, D., Chatterjee, Meister, A., 1998. Glutathione metabolism and its selective
T., Dey, S.N., 1982. Hematological changes produced in mice modification. J. Biol. Chem. 263, 17205–17208.
by Nuvacron or Furadan. Toxicology 25, 255–260. Morowati, M., 1998. Inhalation toxicity studies of thimet
Gutteridge, J.M., Halliwell, B., 2000. Free radicals and antioxidants (phorate) in the male swiss albino mouse, Mus musculus: II.
in the year 2000: a historical look to the future. Ann. N. Y. Lung histopathology, pseudocholinesterase level and
Acad. Sci. 899, 136–147. haematological studies. Environ. Pollut. 103,
Halliwell, B., Gutteridge, J.M.C., 1999. Free Radicals in Biology and 309–315.
Medicine. Oxford Science Publications, Oxford. Murray, R., Granner, D., Maysr, P., Rodwell, V.W., 2000.
Hariria, A.T., Moallemb, S.A., Mahmoudic, M., Memard, B., Biochemistry Harper’s, 32nd ed. Leton and Long.
Hosseinzadeh, H., 2010. Sub-acute effects of diazinon on Ncibi, S., Othman, M.B., Akacha, A., Krifi, M.N., Zourgi, L., 2008.
biochemical indices and specific biomarkers in rats: Opuntia ficus indica extract protects against
protective effects of crocin and safranal. Food Chem. Toxicol. chlorpyrifos-induced damage on mice liver. Food Chem.
48 (10), 2803–2808. Toxicol. 46, 797–802.
Hariria, A.T., Moallemb, S.A., Mahmoudic, M., Memard, B., Nordlie, R.C., Foster, J.D., Lange, A.J., 1999. Regulation of glucose
Hosseinzadeh, H., 2011. The effect of crocin and safranal, production by the liver. Annu. Rev. Nutr. 19,
constituents of saffron, against subacute effect of diazinon on 379–406.
hematological and genotoxicity indices in rats. Nussey, G.J., Van Vuren, J.H.J., du Preez, H.H., 1995. Effect of
Phytomedicine 18 (6), 499–504. copper on haematology and osmoregulation of the
Hers, H.G., 1990. Mechanisms of blood glucose homeostasis. J. Mozambique tilapia, Oreochromis mossambicus (Cichlidae).
Inherit. Metab. Dis. 13, 395–410. Comp. Biochem. Physiol. 111, 369–380.
Hogg, N., 1998. Free radicals in disease. Semin. Reprod. Ogutcu, A., Suludere, Z., Kalender, Y., 2008. Dichlorvos-induced
Endocrinol. 16, 241–248. hepatotoxicity in rats and the protective effects of vitamins C
IARC International Agency for Research on Cancer, 1983. and E. Environ. Toxicol. Pharmacol. 26, 355–361.
Monograph on the Evaluation of Carcinogenic Risk of Page, C., Fuster, V., Gresele, P., Vermylen, J., 2002. Platelets in
Chemicals to Man. Miscellaneous Pesticides, Vol. 30. Lyon, Thrombotic and Non-Thrombotic Disorders: Pathophysiology.
France. In: Pharmacology and Therapeutics. Cambridge University
John, S., Kale, M., Rathore, N., Bhatnagar, D., 2001. Protective Press, Cambridge city, ISBN: 052180261X.
effect of Vitamin E in dimethoate and malathion induced Patil, A.J., Govindwar, S.P., 2003. Biochemical effects of various
oxidative stress in rat erythrocytes. J. Nutr. Biochem. 12, pesticides on sprayers of grape gardens. Ind. J. Clin. Biochem.
500–504. 18, 16–22.
Kalender, S., Ogutcu, A., Uzunhisarcikli, M., Acikgoz, F., Durak, D., Pears, A.G.E., 1985. Histochemistry “Theoretical and Applied”,
Ulusoy, Y., Kalender, Y., 2005. Diazinon-induced hepatotoxicity Vol. 2., third ed. Churchill Livingstone, Edinburgh, London,
and protective effect of vitamin E on some biochemical Melbourne, New York, p. 740.
indices and ultrastructural changes. Toxicology 211, Poet, T.S., Kousba, A.A., Dennison, S.L., Timchalk, C., 2004.
197–206. Physiologically based. pharmacokinetic/pharmodynamic
Kalender, Y., Uzunhisarcikli, M., Ogutcu, A., Acikgoz, F., Kalender, model for the organophosporus pesticide diazinon.
S., 2006. Effects of diazinon on pseudocholinesterase activity Neurotoxicology 25, 1013–1030.
and haematological indices in rats: the protective role of Pournourmohammadi, S., Farzami, B., Ostad, S.N., Azizi, E.,
vitamin E. Environ. Toxicol. Pharmacol. 22, 46–51. Abdollahi, M., 2004. Effects of malathion subchronic exposure
Kalender, S., Uzun, F.G., Durak, D., Demir, F., Kalender, Y., 2009. on rat skeletal muscle glucose metabolism. Environ. Toxicol.
Malathion-induced hepatotoxicity in rats: the effects of Pharmacol. 19 (1), 169–191.
vitamins C and E. Food Chem. Toxicol. 47, 1903–1908. Radiger, H.A., Gaf, R., Cleaven, P.A., 2003. Liver ischemia
Karanth, S., Liu, J., Oliver, K., Pope, C., 2004. Interactive toxicity of apoptosis as a central mechanism of injury. J. Invest. Surg. 16,
the organophosphorus insecticides chlorpyriphos and methyl 149–159.
parathion in rats. Toxicol. Appl. Pharmacol. 196, 183–190. Rana, S.V.S., Allen, T., Singh, R., 2002. Inevitable glutathione, then
Koner, B.C., Banerjee, B.D., Ray, A., 1997. Organochlorine pesticide and now. Ind. J. Exp. Biol. 40, 706–716.
induced oxidative stress and immune suppression in rats. Rezg, R., Mornagui, B., El-Arbi, M., Kamoun, A., El-Fazaa, S.,
Indian J. Exp. Biol. 35, 1132. Gharbi, N., 2006. Effect of subchronic exposure to malathion
Koracevic, D., Koracevic, G., Djordjevic, V., Andrejevic, S., Cosic, V., on glycogen phosphorylase and hexokinase activities in rat
2001. Method for the measurement of antioxidant activity in liver using native PAGE. Toxicology 223, 9–14.
human fluids. Clin. Pathol. 54, 356–361. Richardson, R.J., Moore, T.B., Kayyali, U.S., Fowke, J.H., Randall,
Kovacic, P., 2003. Mechanism of organophosphates (nerve gases J.C., 1993. Inhibition of hen brain acetylcholinesterase and
and pesticides) and antidotes: electron transfer and oxidative neurotoxic esterase by chlorpyriphos in vivo and kinetics of
stress. Curr. Med. Chem. 10, 2705–2709. inhibition by chlorpyriphos oxon in vitro: application to
Lasram, M.M., Annabi, A.B., Elj, N., Selmi, S., Kamoun, A., assessment of neuropathic risk. Fundam. Appl. Toxicol. 20,
El-Fazaa, S., Gharbi, N., 2009. Metabolic disorders of acute 273–279.
exposure to malathion in adult Wistar rats. J. Hazard. Mater. Rodrigues, M.R., Puga, F.R., Chenker, E., Mazanti, M.T., 1987. Short
163, 1052–1055. term effect of malathion on rats’ blood glucose and on
Lodovici, M., Aiolli, S., Monsenat, C., Dolara, P., Medica, A., Di glucose utilization by mammalian cells in vitro. Ecotoxicol.
Simplicio, P., 1994. Effect of mixture of 15 commonly used Environ. Saf. 12 (2), 110–113.
pesticides on DNA levels of 8-hydroxy-2-deoxyguanosine and Sayım, F., 2007. Dimethoate-induced biochemical and
xenobiotic metabolizing enzymes in rat liver. J. Environ. histopathological changes in the liver of rats. Exp. Toxicol.
Pathol. Toxicol. Oncol. 13, 163–168. Pathol. 59, 237–243.
Matin, M.A., Sattar, S., Husain, K., 1990. The role of adrenals in Seiverd, C.E., 1972. Hematology for Technologists, 4th ed. Lea and
diazinoninduced changes in carbohydrate metabolism in rats. Febiger, Philadelphia, pp. 300–360.
Arch. Ind. Hyg. Toxicol. 41, 347–356. Selvam, R., 2002. Calcium oxalate stone disease: role of lipid
Meister, A., 1994. Glutathione-ascorbic acid antioxidant system peroxidation and antioxidants. Urol. Res. 30,
in animals. J. Biol. Chem. 269 (13), 9397–9400. 35–47.
e n v i r o n m e n t a l t o x i c o l o g y a n d p h a r m a c o l o g y 3 5 ( 2 0 1 3 ) 218–227 227

Seven, A., Güzel, S., Seymen, O., Civelek, S., Bolayirh, M., Uncu, Verma, R.S., Mehta, A., Srivastava, N., 2007. In vivo chlorpyrifos
M., BurÇak, G., 2004. Effects of vitamin E supplementation on induced oxidative stress: attenuation by antioxidant vitamins.
oxidative stress in streptozotocin induced diabetic rats: Pestic. Biochem. Physiol. 88, 191–196.
investigation of liver and plasma. Yonsei Med. J. 45, 703–710. Vidyasagar, J., Karunakar, N., Reddy, M.S., Rajnarayana, K.,
Shalby, A.A., 1998. Toxicological studies on experimental rats. Surender, T., Krishna, D.R., 2004. Oxidative stress and
MSc Thesis, Faculty of Agriculture, University of Mansoura. antioxidant status in acute organophosphorous insecticide
Sharma, P., Mishra, K.P., 2006. Aluminum-induced maternal and poisoning. Indian J. Pharmacol. 36 (2), 76–79.
developmental toxicity and oxidative stress in rat brain: WHO, (1997). The WHO Recommended Classification of
response to combined administration of tiron and Pesticides by Hazard 1996–1997, International Programme on
glutathione. Reprod. Toxicol. 21 (3), 313–321. Chemical Safety, WHO/IPCS/96.3.
Shobha, T.R., Prakash, O., 2000. Glycosuria in organophosphate Yasmashita, M., Tanaka, J., Ando, Y., 1997. Human mortality in
and carbamate poisoning. J. Assoc. Physicians India 48, organophosphate poisonings. Vet. Hum. Toxicol. 39,
1197–1199. 84–85.
Suleiman, F.A., Dayo, O.A., Mufta, S., Abdul Ganiyu, G., Olushola, Yehia, M.A.H., El-Banna, S.G., Okab, A.B., 2007. Diazinon toxicity
O.O., Joseph, O.A., 2010. Chlorpyrifos-induced clinical, affects histophysiological and biochemical parameters in
hematological and biochemical changes in Swiss albino rabbits. Exp. Toxicol. Pathol. 59, 215–225.
mice-mitigating effect by co-administration of vitamins C and Yousef, M.I., Awad, T.I., Mohamed, E.H., 2006.
E. Life Sci. J. 7 (3), 37–44. Deltamethrin-induced oxidative damage and biochemical
Tietz, N.W., 1994. Textbook of Clinical Chemistry, 2nd ed. W.B. alterations in rat and its attenuation by vitamin E. Toxicology
Sounder Co, Philadelphia, PA, p. 851. 227, 240–247.
Tkaczyk, M., Baj, Z., 2002. Surface markers of platelet function in Zaahkouk, S.A.M., Helal, E.G.E., Abd-Rabo, T.E.I., Rashed, S.Z.A.,
idiopathic nephrotic syndrome in children. Pediatr. Nephrol. 2000. Carbamate toxicity and protective effect of vit. A and vit.
17, 673–677. E on some biochemical aspects of male albino rats. Egypt J.
Verma, R.S., Srivastava, N., 2003. Effect of chlorpyrifos on Hosp. Med. 1, 60–77.
thiobarbituric acid reactive substances, scavenging enzymes Zadik, Z., Blachar, Y., Barak, Y., Levin, S., 1983. Organophosphate
and glutathione in rat tissues. Indian J. Biochem. Biophys. 40, poisoning presenting as diabetic ketoacidosis. J. Toxicol. Clin.
423–428. Toxicol. 20, 381–385.

You might also like