Sie sind auf Seite 1von 24

Abdel-Moneim & Ghafeer

75

THE POTENTIAL PROTECTIVE EFFECT OF NATURAL HONEY AGAINST


CADMIUM-INDUCED HEPATOTOXICITY AND NEPHROTOXICITY

BY

Wafaa M. Abdel-Moneim* and Hemmat H. Ghafeer


Departments of Forensic Medicine and Clinical Toxicology* and
Histology, Faculty of Medicine, Assiut University.

ABSTRACT

The therapeutic properties of natural honey once considered a form of folk or preventive medicine. It
is important for the treatment of acute and chronic free radical mediated diseases and toxicity. Oxidative
stress can play a key role in cadmium-induced dysfunction. The aim of this work was to study the effect of
natural honey on cadmium-induced liver and kidney damage. A total of 30 adult male rats were divided
into three groups. Group I animals served as control were injected daily I.P. by 1 ml saline. Group II an-
imals were injected daily I.P. with 0.5mg/kg cadmium chloride dissolved in 1 ml saline for 4 weeks. Ani-
mals of group III were treated with 0.5 mg /kg cadmium chloride I.P. and 0.05ml of natural honey mixed
with water orally concurrently for 4 weeks. Liver function (SGOT),(SGPT), (ALP) and kidney function
(creatinine and urea nitrogen) tests were measured. In addition lipid peroxidation,reduced glutathione
(GSH) and glutathione peroxidase (GPx) were estimated in liver and kidney tissues samples. Light and
transmission electron microscopic examination were used for histological changes. The results revealed
that treatment with Cd caused marked elevation in the level of free radicals (lipid peroxidation) and kid-
ney and liver enzymes, and a decline in GPx activity and GSH level. Administration of honey with Cd in-
duced improvement in all examined parameters. On the other hand, light microscopic examination of kid-
ney cortex of Cd treated group revealed swelling of the cells lining the convoluted tubules and vaculation
of their cytoplasm. Variable degrees of glomerular degeneration were present. The liver showed different
degrees of cell degeneration, necrosis, dilatation and congestion of blood vessels. Results obtained by
EM examination revealed that there were affection of mitochondria and partial loss of microvilli of some
kidney tubules. Furthermore, electron dense mitochondrea, depletion of glycogen granules in a rarified
vaculated cytoplasm were seen in the hepatocytes. It is noticed that concurrent administration of honey
with cadmium improved histological changes in both kidney and liver by light and electron microscope. It
could be concluded that honey via its antioxidant activity has the ability to protect against cadmium-
induced hepatotoxicity and nephrotoxicity.

INTRODUCTlON ment that occurs naturally in ores together


with zinc, lead and copper or is emitted
Cadmium (Cd) is a relatively rare ele- into the air through the process of volcanic

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
76

emission. It became commercial in the terial, bacteriostatic, anti-inflammatory,


20th century due to agricultural and in- wound and sunburn healing activities. Re-
dustrial applications (WHO, 2000 and Jar- cent views propose honey not only as a
up, 2003). health promoting dietary supplement, but
shed light on antioxidant, non-peroxide
Occupational exposure to cadmium, dependent properties. This makes honey
such as working with cadmium con- more than just a nourishment of high val-
taining pigments, plastic, glass, metal al- ue but a valuable dietary source of antioxi-
loys and electrode material in nickel - dants (Beretta et al., 2005).
cadmium batteries, and non occupation-
al exposure, such as food, water and In recent years there has been an in-
cigarette smoke induces uptake of Cd creased interest in the application of anti-
from the environment into the body oxidants to medical treatment as informa-
through pulmonary and enteral pathways tion is available linking the development
(Waisberg et al., 2003). Cadmium ab- of human diseases to oxidative stress (Al-
sorbed and accumulates mainly in the kid- Jadi and Kamaruddin, 2004).
ney and liver, then it is bound to the apo-
protein metallothionein (Morales et al., Honey is a natural antioxidant which
2006). may contain flavinoids, ascorbic acid, to-
copherols, catalase, and phenolic com-
The intracellular release of cadmium is pounds all of which work together to pro-
responsible for the generation of reactive vide a synergistic antioxidant effect,
oxygen species, glutathione depletion, lip- scavenging and eliminating free radicals
id peroxidation, protein cross-liking, DNA (Johnston et al., 2005).
damage, culminating ultimately in oxi-
dant-induced cell death (Brennan, 1996; Little information is available on protec-
Shaikh et al., 1999; Jurezuk et al., 2004 and tive effect of honey against cadmium in-
Babu et al., 2006). duced hepatotoxicity and nephrotoxicity.
In the present study an experimental mod-
Honey has been an ingredient of tradi- el of rats treated with Cd during four
tional medicine on account of its dietary weeks as a model of Cd induced hepato-
and curative properties since ancient toxicity and nephrotoxicity were used. In
times. Starting in the early 1970 research- this model the protective effect of concur-
ers from different scientific fields have in- rent administration of honey on Cd-
vestigated the chemical and biological induced hepatic and renal damage were
properties of honey, including antibac- assessed and if the protective effect of

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
77

honey is based on its antioxidant proper- - Group III (Cd + honey) rats were con-
ties. current by administred of 0.5 ml/kg
CdCl2 I.P. and 0.05 ml natural honey
MATERIAL AND METHODS (Paget and Barnes, 1964) dissolved in
water orally by gastric tube daily for
Chemicals: All chemicals and reagents four weeks.
used were of analytical grades. Cadmium
chloride (CdCl2) was purchased from ICN Blood collection :
pharmaceutical company (USA). Reduced 1- Animals were anesthetized and
glutathione and Ellman's reagent 5,5 Di- blood samples were collected from occular
thiobis-2 nitrotenzoic acid (DTNB) were vascular bed using capillary tubes.
obtained from ICN Biomedica. Inc. (USA).
2- Blood samples were collected into
Animals: Thirty male albino wister rats dry clean tubes on EDTA then centrifuged
weighing 150-200 gm each obtained from for 10 min to isolate plasma and stored at -
the Animal House, Faculty of Medicine, 70oC.
Assiut University were used. Rats were
housed in stainless steel cages at a con- Tissue samples : animals were killed by
stant temperature 25 + 2oC with alternat- cervical decapitation. The liver and kid-
ing 12 hours light and dark cycles and al- neys were taken and each organ was di-
lowed water and food (laboratory chow) vided into two parts.
ad libitum. The research was conducted in
accordance with the internationally ac- The 1st specimen was homogenized in
cepted guidelines for laboratory animal ice cold 100 mM phosphate buffer (pH
use and care. The experiment was ap- 7.4). Homogenates were centrifuged and
proved by the Institutional Ethics Com- the resulting supernatant was storted at -
mittee. 70oC for biochemical analysis.

Treatment: Rats were divided into The 2nd specimen was preserved in
three groups 10 animals each. 10% formaline for histological examina-
- Group I (control) animals were inject- tion.
ed intraperitoneally (I.P.) with 1 ml
saline daily for four weeks. Biochemical measurements:
- Group II animals were injected IP dai- A) The plasma samples were taken for
ly with 0.5 mg/kg CdCl2 for four determination of :
weeks (Satarug and Moore, 2004). 1- Liver function tests: glutamyl - oxal-

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
78

oacetic acid transaminase (SGOT), glu- and glutathione reductase. The en-
tamyl pyruvic transaminase (SGPT) zyme activity is expressed as U/mg
and alkaline phosphatase (ALP). protein.

2- Kidney function tests: creatinine and 3- Reduced glutathione (GSH): An equal


urea nitrogen. volume of 10% metaphosphoric acid
The two tests were measured spectro- was added to a part of the homogen-
photometrically using standardized ates and mixed by vortexing. The mix-
commercially available Diamond Kits ture was allowed to stand for 5 min at
(Modern Laboratory, Egypt). room temperature. After centrifuga-
tion for 5min, the supernatant was col-
3- Oxidative stress indices: lipid peroxi- lected carefully without disturbing the
dation level was estimated by the precipitate. The GSH contents of the
measurement of malondialdehyde neutralized supernatant were assayed
(MDA), an end product of lipid perox- using Ellman's reagent (5, 5'-dithiobis-
idation level were determined using 2-nitrobenzoic acid (DTND solution)
spectrophotometre employing thiobar- according to the method of Griffith
bituric acid reactive substances de- (1980). A standard reference curve was
scribed by Thayer, (1984). Levels were prepared for each assay.
expressed as nmol/ml plasma.
4- Protein contents were determined us-
B) Liver and kidney homogenates: were ing the method of Lowry et al., (1951).
utilized for determination of
1- Lipid peroxidation was measured as Preparation of histological sections:
described by Thayer (1984), levels Specimens of the liver and kidney were
were expressed as nmol/mg protein. taken and fixed in formalin and processed
for light microscope. The specimens were
2- Glutathion peroxidase (GPx): Activity embedded in paraffin and sectioned at a
was measured by the method of Tap- thickness of 7 microns. The sections were
ple (1978). The method is based on oxi- stained with haematoxyline and eosine
dation of glutathione by cumen hydro- stain according to Drury and Wallington
peroxide via glutathione peroxidase. (1980). Other small pieces were fixed in
The reaction in the form of decrease in glutraldehyde and processed for electron
absorbance at 340nm was followed microscope examination. Semithin sec-
when oxidized glutathione converted tions were cut at 1/2-1 micron and stained
to reduced glutathione via NADPH with toluidine blue. Selected sections were

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
79

contrasted and electron micrographs were Table (3) shows the levels of lipid per-
taken with Jeol transmission electron mi- oxidation in plasma, and tissues of liver
croscope (TEM). and kidney. In Cd-group the level of LPO
in plasma, tissues of liver and kidney was
Statistical analysis: significantly higher than controls.In group
* The data were presented as means + received Cd + honey the level of LPO was
standard errors. significantly reduced to near control levels
* For the comparison of statistical sig- compared with the Cd - group.
nificance between two groups student
Newman-keuls t-test was used. The effects of Cd and Cd + honey on
* Values were accepted as being statisti- glutathione peroxidase activity (GPx) in
cally significant if a P value was less tissues of liver and kidney are shown in
than 0.01. table (4). The GPx activity level was signif-
icantly lower in tissues of liver and kidney
RESULTS in Cd-group compared with the control
group.In comparison with Cd group there
Biochemical Results : was significant increase in the level of GPx
Table (1) displays the effects of Cd activity in Cd + honey group in tissues of
alone and Cd with honey on plasma liver liver and kidney. Table (5) shows that re-
enzymes. Levels of SGOT, SGPT and ALP duced glutathione level in tissues of liver
were significantly increased in the cadmi- and kidney of cadmium treated group
um treated group in comparison to the was significantly decreased in comparison
control group. Animals of group III re- to control group.In group received Cd +
ceived Cd + honey showed a significant honey the level of reduced glutathione
reduction in SGOT, SGPT, ALP level com- showed significant increase in tissues of
pared with Cd group. liver and kidney in comparison to Cd
group.
The effects of Cd alone and Cd + honey
on plasma level of kidney function test are Histological Results :
shown in table (2). The plasma levels of I- Liver :
urea and creatinine were significantly A) Light microscope results (Hx&E stain)
higher in Cd-group than controls. Levels Group I: control group :
of urea and creatinine in plasma exhib- The control livers show normal lobular
ited significant reduction in group re- architecture with central vein and radiat-
ceived Cd + honey in comparison with Cd ing cords of hepatocytes, separated by
group. blood sinusoids. Hepatocytes are large

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
80

and polyhedral in shape with slightly formed of rough endoplasmic reticulum


acidophilic granular cytoplasm. They have and mitochondria. These cytoplasmic or-
large, rounded, vesicular nuclei with ganelles were separated by large areas of
prominent nucleoli (Fig. 1) . rarified cytoplasm. The cisternae of the
rough endoplasmic reticulum showed
Group II : animals that received CdCl2 fragmentation in the form of short seg-
only : ments and loss of their attached ribosomes
The liver cells of group II animals (Fig. 5). Whorly appeared structures were
showed obvious histological changes, in demonstrated in the rarified cytoplasm.
the form of distortion in the hepatic organ- Mitochondriae were polymorphic con-
ization, dilatation and congestion of the tained electron-dense granular matrix.
blood sinusoids and central vein. Some Some of them were swollen, lost their cris-
hepatocytes showed signs of degeneration tae and contained vacuoles (Fig. 6).
in the form of hypertrophy with highly
vacuolated cytoplasm and deeply stained Group III : combined treatment with
nuclei. Other hepatocytes exhibited hyali- CdCl2 and honey led to improvement in
nized cytoplasm with pale nuclei and the ultrastructure of hepatocytes which
prominent nucleoli (Fig.2). appeared nearly similar to the control.
Most of the mitochondriae were intact.
Group III: CdCl2 + honey : The cisternae of rough endoplasmic reticu-
The liver cells appeared more or less lum acquired their normal appearance.
similar to those of the control apart from The nuclei of some hepatocytes appeared
few hepatocytes appeared with vacuolat- large, rounded and euchromatic with
ed cytoplasm and pyknotic nuclei (Fig.3). prominent nucleoli (Fig. 7).

B) Electron microscope results: II- Kidney :


Group I : the hepatocytes of control ani- A) Light microscope results (Hx & E
mal exhibit a large rounded euchromatic stain).
nucleus. The cytoplasm contains numer- Group I : the cortex of control kidney is
ous cisternae of rough endoplasmic reticu- mostly occupied by renal corpuscles and
lum, numerous well-preserved mitochon- surrounding proximal and distal convo-
dria and glycogen rosettes (Fig. 4). luted tubules. The renal corpuscle is
formed of glomerular tuft of blood capil-
Group II : after treatment with CdCl2 laries surrounded by capsular space and
the hepatocytes showed clumping of the Bowman's capsule.
cellular organelles which were mainly

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
81

The proximal convoluted tubules are tes are situated between glomerular capil-
lined with large cuboidal cells with deeply laries, and their secondary foot processes
stained acidophilic cytoplasm with apical were extended and rest on the basement
brush border and rounded vesicular nu- membrane. Numerous filtration slit-
clei. The boundaries between the adjacent membranes were observed between the
cells are indistinct. The distal convoluted processes of glomerular capillaries
tubules are lined by low cuboidal cells (Fig.11).
with distinct cell boundaries and less acid-
ophilic cytoplasm (Fig.8). Regarding the cells of the proximal con-
voluted tubule: The apical cell membrane
Group II : the glomerular capillaries of is occupied by numerous microvilli. The
some renal corpuscles of animals showed cytoplasm contains numerous mitochon-
congestion and dilatation. There was dria of various sizes. Few cisternal profiles
swelling of some cells of the proximal con- of rough endoplasmic reticulum and ribo-
voluted tubules leading to diminution or somes were detected in the cytoplasm.
even obliteration of the tubular lumina. The nuclei is rounded euchromatic with
Cytoplasmic vacuolation and deeply prominent nucleolus The basement mem-
stained nuclei were observed compared to brane exhibited basal infoldings with elon-
the control group. Destruction of the gated mitochondria between them
brush borders of the proximal convoluted (Fig.13).
tubules was also detected. The distal con-
voluted tubules showed degenerative Group II : in cadmium treated animals,
changes in the form of pyknotic nuclei and most of the renal corpuscles were highly
vacuolated cytoplasm (Fig.9). affected. There was dilatation and swell-
ing of the glomerular capillaries accompa-
Group III : combined treatment with nied with narrowing of the urinary spaces.
honey and CdCl2 led to obvious improve- The glomerular basement membranes
ment in the histological structures of the showed obvious thickening in most of re-
kidney compared to group II. The renal nal glomeruli. Most of the secondary foot
corpuscles appeared nearly similar to the processes of the podocytes appeared flat-
control. Most of kidney tubules exhibited tened and amulgumated with each other
acidophilic cytoplasm and rounded vesic- with complete disappearance of their slit
ular nuclei (Fig.10). membranes. Some of these processes
contained dense granules and dense irreg-
B) Electron microscope results: ular bodies. The lumina of the capillaries
Group I : ultrastructurally the podocy- contained few exudates and degenerated

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
82

endothelial cells. There was apparent in- and genetic information (Patrick 2003 and
crease in the amorphous secretion of me- DeBurbure et al., 2006). Therefore, some
sangial matrix (Fig. 12). authors have postulated that antioxidants
should be one of the important compo-
Regarding the cells of the proximal nents of an effective treatment of cadmi-
convoluted tubules the apical brush bor- um poisoning (El-Demerdash et al., 2004).
der exhibited partial loss or erosion of
their microvilli. The cytoplasm showed The present study concentrates on the
many vacuoles. The mitochondriae were possible protective effect of honey on oxi-
markedly affected. They appeared small dative damage generated by cadmium in-
with dense matrix and distructed cristae duced hepatotoxicity and nephrotoxicity.
in some of them. The tubular basement Liver and kidney function tests were done
membrane displayed obvious demolish- for different studied groups to assess their
ing of their basal infoldings (Fig.14). status. Biochemical analysis was done for
oxidative stress indices such as lipid per-
Group III : combined treatment with oxidase level. The activity of antioxidants
Cd and honey. The glomerular capillaries was measured e.g. glutathione peroxidase
and the podocytes appeared more or less (GPx) and reduced glutathione (GSH), be-
similar to the control group. The cell of the cause these antioxidants are the common-
proximal convoluted tubule exhibited nor- est to be affected by cadmium toxicity
mal cellular organelles and intact microvil- (Jurezuk et al., 2004). Histological changes
lous border. The apical cytoplasm of some of kidney and liver were examined by
renal tubules was rarified with some light and electron microscopes.
dense bodies (Fig.15).
In this work the liver enzymes SGOT,
DISCUSSION SGPT and ALP in the Cd treated group
were significantly elevated compared
Available literature indicates that no with the control group, denoting the
previous studies have been done to evalu- presence of liver dysfunction (Shimada
ate the antioxidant capacity of honey and et al., 2004). In concurrent administration
its protective effect against cadmium in- of honey and Cd, the levels of liver en-
toxication. zymes activity were significantly reduced
as compared with the Cd-group. This
The mechanisms of cadmium-induced finding indicates the protective effects of
damage include the production of free honey in ameliorating the hepatotoxic ef-
radicals that alter mitochondrial activity fect of Cd.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
83

The plasma level of creatinine and urea cadmium is thought to induce lipid perox-
was significantly increased after cadmium idation and this has often been considered
treatment compared to the control group to be the main cause of its deleterious in-
indicating the impairment in the kidney fluence on membrane-dependent function.
function. Similar observation was ob-
tained by Novelli et al., (1998). In fact, In the present study the elevation in the
urea is the first acute renal marker which free radicals (LPO) induced by cadmium
increases when the kidney suffers any alone was very high significantly de-
kind of injury. Otherwise, creatinine is the creased in the presence of honey. This
most trustable renal marker and increase means that honey minimized the toxic ef-
only when the majority of renal function is fect of cadmium via its antioxidant activi-
lost (Borge et al., 2005). The changes in ty. These results are in line with the view
urea and creatinine level in the present held by Beretta et al., (2005) who con-
study concluded the severe injured effect firmed the role of honey as an antioxidant
of CdCl2 on kidney. Moreover, in the agent in blood. Also Manuela et al., (2007)
present study honey co-administration found that there was a direct link between
with cadmium significantly ameliorated the honey consumption and the level of
the increased plasma levels of creatinine polyphenolic antioxidants in the plasma.
and urea.
In agreement with a previous study, the
In the current study, lipid peroxidation level of GSH was significantly decreased
level was significantly elevated in plasma, in the liver and kidney extracts of cadmi-
liver and kidney tissues of rats treated um treated group compared to the control
with cadmium compared to control group group. This decrease in GSH levels may be
thus suggesting increased oxidative stress. due to its consumption in the prevention
These results were supported by Manca et of free radical-mediated lipid peroxidation
al., (1991) who reported that LPO is an (Demopoulos, 1973; Koyuturk et al., 2006).
early and sensitive consequence of Cd ex- Also, GSH may be consumed in the detox-
posure. Also, Hassoun and Stohs (1996) ification of heavy metals (Kim et al., 1998
demonstrated that oxidative stress was in- and Thevenod, 2003). Furthermore, it has
duced following oral administration of been suggested that the decrease in GSH
cadmium chloride to rats. A similar data levels upon cadmium exposure might im-
had been reported by Jurezuk et al., pair the degradation of lipid peroxides,
(2004). In addition, Elizabeth et al., (2003); thereby leading to its accumulation in the
Jahangir et al., (2005); Eybl et al., (2006) target organs (Sarkars et al., 1997). In con-
and Kawamoto et al., (2007) reported that troversy to the current results, Kamiyama

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
84

et al., (1995) reported an increase in GSH ultrafiltered cadmium is taken up, largely
level in liver and kidney tissues after Cd by the proximal tubules of the kidney and
injection which could be explained as a is accumulated mainly in kidney cortex
protective mechanism. leading to proximal tubule lesions (Lars,
2002). These findings are in agreement
The co-administration of honey with with the current study.
cadmium increased the level of the antiox-
idant (GSH), and approximated to the nor- The nephrotoxicity of Cd has been ex-
mal values of the control group. Al-Waili, tensively studied in various experimental
(2003) confirmed that honey increased the models (Mitsumori et al., 1998; Ohta et al.,
levels of antioxidants and this effect might 2000). Recent papers show that tubular
be attributed to the composition of honey, damage may appear at lower levels of Cd
which contains many nutrient elements exposure than previously anticipated (Jar-
and antioxidants as vitamin C, which is a up et al., 2000; Noonan et al., 2002).
potent antioxidant agent.
The Cd concentrations applied in this
The activities of GPx was significantly study was in the range of previous studies
decreased in the liver and kidney extracts (Brzoska et al., 2003; Choi and Rhee, 2003)
of cadmium treated rats. The decline in according to average human intake data,
the level of GPx activity resulted from Cd soil Cd concentrations from contaminated
toxicity was previously demonstrated in sites (Blanusa et al., 2002; Satarug and
liver tissues (Sidhu et al., 1993; Ossola and Moore, 2004) and Cd levels from animals
Tomara, 1995; Sarkars et al., 1997) and in caught in polluted areas (Damek-
kidney tissues (Bragadin et al., 2004). In Proprawa and Sawicka-Kapusta, 2003).
the present study reduced glutathione per-
oxidase activity was recovered after co- In the present study administration of
administration of honey with cadmium in Cd 0.5 mg/kg I.P. daily for four weeks in-
liver and kidney tissues, which in turn de- duces significant damage in function and
structs the lipid peroxidase (Eaton et al., structure of kidney assessed by increased
1980). This confirm that honey increase an- creatinine and urea concentrations in plas-
tioxidant activity and able to protect from ma and existence of atrophy of and swell-
oxidative stress. ing of some glomerular capillaries as well
as proximal tubular necrosis and apopto-
Epidemiological studies have revealed sis (Damek-Proprawa and Sawicka - Ka-
that cadmium is one of the most toxic of pusta, 2004). The distal convoluted tu-
the heavy metals to humans; 70% of the bules showed degenerative changes with

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
85

pyknotic nuclei, and cytoplasmic vacuola- (1996) and Shaikh et al., (1999) have re-
tion. Ultrastructrually the glomerular ported the mechanism of Cd-induced re-
basement membranes showed obvious nal damage include increased oxidative
thickening in most of the renal gomeruli. stress. Whereas increased oxygen free rad-
The filteration slits were markedly de- icals production seems to be induced by
creased in number. Tubular lesions con- the interaction of Cd and mitochondrial
sisted in partial lost of the brush border structure (Tang and Shaikh, 2001).
microvilli, altered mitochondrial struc-
ture some of them appeared small with In the current study treatment with
dense matrix and districted cristae. This CdCl2 caused liver damage demonstrated
is in contrast with the study of Sandy et functionally by increasing the activity of
al., (2007) who revealed that the toxic ef- SGOT, SGPT and ALP, and histological al-
fects of Cd in the kidney are confined terations. These alterations were in the
to proximal tubular cells. With no signs of form of dilatartion in the hepatic organiza-
glomerular and mitochondrial damage tion, dilatation and congestion of central
were detected. One possible explanation veins and blood sinusoids. Hypertrophy
is that Cd was administrated via the or degeneration of some hepatocytes with
drinking water not by injection as in the either hyalinized and vacuolated cyto-
current study. plasm with pale nuclei and prominent nu-
cleoli. These results coincide with results
These results reveal that honey has a of Borges et al., (2005). These alterations
marked protective effect on renal tubular were observed with electron microscopy.
toxicity and showed decreased lipid per- Most of the hepatocytes showed clumping
oxidation and increased tissue levels of of the cellular organelles. The cisternae of
GSH and GPx activity a potent endoge- the rough endoplasmic reticulum showed
nous antioxidant (Meister and Anderson, fragmentation in the form of short seg-
1983). This protective effect of honey was ments and loss of their attached ribo-
confirmed when renal tissues were ob- somes. Mitochondriae were polymorphic
served by electron microscopy. Most of contained electron-dense granular matrix.
kidney tubules exhibited acidophilic cyto- Some of them were swollen, lost their cris-
plasm and rounded vesicular nuclei. tae and contained vacuoles. These results
Moreover, the observation that Cd+honey are in agreement with study of Mousa,
treated animals did not have mitochondri- (2004). Co-administration of honey with
al damage might be seen as supporting cadmium markedly ameliorated these
the anti-oxidant properties of honey. In histopathological changes induced by Cd.
agreement with this suggestion, Brennan, Most of mitochondrie were intact. The

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
86

cisternae of rough endoplasmic reticulum ing free radicals (Johnston et al., 2005).
acquired their normal appearance.
In conclusion: The present study dem-
In the present study CdCl2 exposure in- onstrated that honey administered in
creased LPO levels in plasma and tissues combination with cadmium minimized its
with alterations in antioxidant defenses hazards. Honey can protect against oxida-
(GPx and GSH). Thus it may be possible tive stress induced by cadmium, by lower-
that oxidative stress and disturbance in ing the free radicals and increased the lev-
antioxidant defenses were the causes for els of antioxidants. Further studies are
liver and kidney damage induced by required to recommend the use of honey
CdCl2 in this experimental model. and it’s therapeutic potential in human.
Several antioxidant assays were utilized in
On hypothesis to explain the benefi- order to evaluate the biological and chemi-
cial effects of honey in ameliorating bi- cal properties of honey. In addition, the
ochemical parameters and histological exposure to cadmium should be reduced
changes is that honey may contains flav- and attention paid to sources of cadmium
onoids, ascorbic acid, tocopherols, catalase in food, water and personal-care products.
and phenolic compounds. All of which Furthermore using diets rich in honey
work together to provide a synergistic an- could be beneficial in alleviating cadmium
tioxidant effect, scavenging and eliminat- toxicity.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
87

Table (1): The effect of cadmium alone and with honey on the liver enzymes.

Items SGOT SGPT ALP


Groups U/L U/L U/L
Group I (Control) 12.08 ± 0.74 10.12 ± 0.56 142.21 ± 9.63
Group II (Cd) 58.20 ± 4.98** 65.21 ± 3.42** 1278.40±4.92**
Group III (Cd+Honey) 15.11 ± 1.1*** 12.34 ± 1.20*** ± 6.63***
150.28±
** P< 0.01 group II vs control. *** P< 0.01 group III vs group II.

Table (2): The effect of cadmium alone and with honey on kidney
function tests (creatinine and urea).

Items Urea (mg/dl) Creatinine (mg/dl)


Groups
Group I 32.24 ± 2.13 0.92 ± 0.04
Group II ± 6.42**
86.5± 3.01 ± 0.3**
Group III ± 2.26***
35.47± 0.95 ± 0.06***
** P< 0.01 group II vs control. *** P< 0.01 group III vs group II.

Table (3): The effect of cadmium alone and with honey on lipid peroxidation level.

Groups GroupI GroupII GroupIII


Organs Control Cd Cd + honey
Plasma nmol/ml 3.56 ± 0.12 9.10 ± 0.53** 4.82 ± 0.11***
Liver nmol/mg 0.79 ± 0.08 2.76 ± 0.38** 0.8 ± 0.04***
Kidney nmol/mg 1.28 ± 0.09 2.67 ± 0.10** 1.58 ± 0.03***
** P< 0.01 group II vs control. *** P< 0.01 group III vs group II.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
88

Table (4): The effect of cadmium alone and with honey on glutathione
peroxidase activity (GPx).

Groups GroupI GroupII GroupIII


Organs Control Cd Cd + honey
Liver U/mg protein 328.50 ± 5.18 ±15.38**
231.28± ±10.36***
331.92±

Kidney U/mg protein 333.06 ± 4.80 224.20 ± 16.63** ± 10.18***


331.20±
** P< 0.01 group II vs control. *** P< 0.01 group III vs group II.

Table (5): The effect of cadmium alone and with honey on reduced glutathione
(GSH) level.

Groups Group I Group II Group III


Organs Control Cd Cd + honey
Liver nmol/mg 6.64 ± 0.35 2.65 ± 0.16** 6.22 ± 0.43***

Kidney nmol/mg 2.96 ± 0.21 1.28 ± 0.09** 2.66 ± 0.29***


** P< 0.01 group II vs control. *** P< 0.01 group III vs group II.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
89

Fig. (1) : A photomicrograph of a section in the Fig. (3): A photomicrograph of a section in the
liver of control adult male albino rat liver of rat of (group III) showing; hep-
(group I) showing; central vein (C) and atocytes appeared more or less similar
hepatocytes with vesicular nuclei () to control apart from few cells with
separated by blood sinusoids(S). (H&E vacuolated cytoplasm ()
X200). (H&E X400).

Fig.(2): A photomicrogaph of a section in the Fig.(4): An electron micrograph of control liver


liver of rat of (group II) showing; dis- cells showing; large nucleus with euch-
organization of the hepatocytes with romatin (N). The cytoplasm contain
vacuolated cytoplasm(V) and deeply numerous mitochondriae(M), strands
stained nuclei. Some of them are hy- of rER and fine glycogen granules (g)
pertrophied with deep acidophilic cyto- (X5000).
plasm (). Note: dilated and congested
central vein (C). (H&E X400).

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
90

Fig.(5): An electron micrograph of liver cell of Fig.(7): An electron-micrograph of the hepato-


(group II) animals showing : clumping cyte of (group III) showing : more or
of cellular organelles, whorly appeared less normal ultrastructure with large
structures () in a rarified cytoplasm vesicular nucleus (N), strands of rER
(X4000). and numerous mitochondria (M).

Fig.(6): A magnified part of a hepatocyte ob- Fig.(8): A photomicrograph of a section of con-


tained from the previous group. Show- trol renal cortex (group I) of adult male
ing : polymorphic mitochondria with albino rat showing : renal corpuscle
electron dense matrix. Note: vacuole (), Proximal (P) and distal (D) con-
(v) and dense bodies (D) inside the de- voluted tubules. (H&E X200).
generated mitochondriae (X8000).

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
91

Fig.(9):A photomicrograph of a section of renal Fig.(11): An electron micrograph of a renal cor-


cortex of (group II) showing : conges- puscle of (group I) showing : podocy-
tion of the glomerular capillaries (G), tes (P) with their minor or secondary
cytoplasmic vacuolation (v) and pyk- foot processes (P2) that rest on normal
notic nuclei (p) in most of the tubules. basement membrane () of capillary
Note: some tubular cells with highly endothelium. (X 6700).
acidophilic cytoplasm ()(H&E X200).

Fig.(10): A photomicrograph of a section of re- Fig.(12): An electron micrograph of a renal cor-


nal cortex of (group III) showing : puscle of (group II) showing : parts of
more or less normal structure podocytes with fused minor processes
(H&E X200). () rest on a thick basement mem-
brane (B). (X6700).

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
92

Fig.(13): An electron-micrograph of proximal Fig.(14): An electron-micrograph of proximal


convoluted tubule of (group I) show- convoluted tubule of (group II) show-
ing : long microvillous border (MV), ing : loss of the microvilli () small
many elongated mitochondria (M), ba- dense mitochondria (M) and cytoplas-
sal spherical nucleus with euchromatin mic vacuoles (V). Note: loss of basal
(N). Note: normal basal infoldings. infolding (X5000).

Fig.(15): An electron-micrograph of proximal


convoluted tubules of (group III) show-
ing : intact microvillous border (MV),
intact mitochondria. Note: rarifaction
of apical cytoplasm with some dense
particles (X5000).

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
93

REFERENCES V.; Nogueira, C. W.; Rocha, J. B. and Zeni


G. (2005) : “Protective effect of diphenyl-
Aljadi A. and Kamaruddin M. (2004) : diselenide on acute liver damage induced
“Evaluation of the phenolic contents and by 2- Nitropropane in rats”. Toxicology.
antioxidant capacities of two Malaysian 210: 1-8.
floral honeys”. Food Chemistry, 85: 513-
518. Bragadin, M.; Bragadin, G.; Scutari, A.;
Fold, A.; Bindoli, M. and Rigobello,
Al-Waili N. (2003) : “Effects of daily (2004) : “Effects of metal complexes on
consumption of honey solution on hemat- thoredoxin reductase and the regulation of
ological indices and blood levels of miner- mitochondria permeability conditions”.
als and enzymes in normal individuals”. J. Ann. N.Y. Acad. Sci. 1030 : 348-354.
Medical Food, 6(2): 894-897.
Brennan R. J. (1996) : “Cadmium is an
Babu, K. R.; H. R. Rajmohan; B. K. Ra- inducer of oxidative stress in yeast”. Mu-
jan and K. M. Kumar, (2006) : “Plasma tat. Res. 356 : 171-178.
lipid peroxidation and erythrocyte antioxi-
dant enzymes status in workers exposed Brzoska, M. M.; Kaminski, M.; Supe-
to cadmium”. Toxicol. Ind. Health., 22: mak-Bobko D., Zwierz K. and Moniusz-
329-35. ko-Jakoniuk J. (2003) : “Changes in the
structure and function of the kidney of
Beretta, G.; Granata, P.; Ferrero, M.; rats chronically exposed to cadmium. I. Bi-
Orioli, M. and Facino, R. (2005) : “Stan- ochemical and histopathological studies”.
dardization of antioxidant properties of Arch. Toxicol., 77 : 344-352.
honey by a combination of spectrophoto-
metric / fluorimetric assays and chemo- Choi H. and Rhee J. H. (2003) : “Effects
metrics”. Analytica Chimica Acta, 533 : of Vitamin E. on renal dysfunction in
185-191. chronic cadmium-poisoned rats”. J. Med.
Food 6 : 209-215.
Blanusa, M.; Mrkovic-Millic, R. and
Durbesic, P. (2002) : “Lead and cadmi- Damek-Proprawa and Sawicka-
um in soil and isopoda woodlice in Kapusta (2003) : “Damage to the liver, kid-
Croatia”. Ecotoxicol. Environ. Saf., 52 : ney and testis with reference to burden of
198-202. heavy metals in yellow-necked mice from
areas around steelworks and zinc smelters
Borges, L. P.; Borges, V. C.; Moro, A. in Poland”. Toxicology, 186 : 1-10.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
94

Damek-Proprawa and Sawicka- Kedwany, F. S. and Baghdadi, H. H.


Kapusta K. (2004) : “Histopathological (2004): “Cadmium-induced changes in lip-
changes in the liver, kidneys, and testes of id peroxidation, blood hematology, bio-
bank voles environmentally exposed to chemical parameters and semen quality of
heavy metal emissions from the steel- male rats: protective role of vitamin E and
works and zinc smelter in Poland, Envi- B-carotene”. Food. Chem. Toxic, 42 : 1563-
ron”. Res. 6:72-78. 1571.

De Burbure, C.; J. B. Buchet; A. Leroy- Elizabeth, A. M.; Rosalyn, D. M.; Jen-


er; C. Nisse; J. M. Haguenoer; A. Mutti; Z. nifer, A. M.; Rebecca, R. W. and Beth A.
Smerhovsky; M. Cikrt; M. Trzcinka- A. (2003) : “Environmental cadmium lev-
Ochocka; G. Razniewska; M. Jakubowski els increase phytochelatin and glutathione
and A. Bernard (2006) : “Renal and neuro- in lettuce grown in a chelator-buffered nu-
logic effects of cadmium, lead, mercury trient solution”. Environ. Qual, 32:1356-
and arsenic in children: evidence of early 1364.
effects and multiple interactions at envi-
ronmental exposure levels. Environ. Eybl, V.; Kotyzova, D.; Leseticky, L.;
Health”. Perspect., 114: 584-590. Blucovska, M. and Koutensky, J. (2006):
“The influence of curcumin and mana-
Demopoulos H. (1973) : “Control of gense complex of curcumin on cadmium-
free radicals in biological system”. Fed. induced oxidative damage and trace ele-
Proc. 32:1903-1908. ments status in tissues of mice”. J. Appl.
Toxicol. 26(3):207-212.
Drury R. A. and Wallington E. A.
(1980) : Carlton's Histological Technique Griffith, O. W. (1980) : “Determina-
5th ed. New York, Oxford University, tion of glutathione and glutathione dis-
Press, P.P. 235-241. ulfide using glutathione reductase and
2. Vinylpyridine”. Anal. Biochem. 106:207-
Eaton, D. L.; Stacey N. H.; Wong, K. L. 212.
and Klaassan, C. D. (1980) : “Dose-
response effects of various metals on rat Hassoun, E. A. and Stohs, S. J. (1996):
liver metallothionein, glutathione, heme “Cadmium-induced production of super-
oxygenase, and cytochrome p. 450. Toxi- oxide anion and nitric oxide, DNA single
col”. Appl. Pharmacol. 55: 393-402. strands breaks and lactate dehydrogenase
leakage in J. 774A. 1 cell cultures”. Toxi-
El-Demerdash, F. M.; Yousef, M. I.; cology, 112:219-226.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
95

Jahangir, T.; Khan, T. H.; Prasad, L. mium exposure in livers and kidneys and
and Sultana, S. (2005) : “Allevation of their relation to glutathione levels”. Res.
free radical mediated oxidative and geno- Commun. Mol. Pathol. Pharmacol. 88 (2):
toxic effects of cadmium by farnesol in 177-186.
Swiss albino mice”. Redox Rep. 10 (6) :
303-310. Kawamoto, S.; Kawamura, T.; Miyaza-
ki, Y. and Hosoya, T. (2007) : “Effects of
Jarup, L.; Hellstrom, L.; Alfven, T.; atorvastatin on hyperlipidemia in kidney
Carlsson, M. D.; Grubb, A.; Persson, B.; disease patients”. Nippon Jinzo Gakkaishi,
Pettersson, C.; Spang, G.; Schutz, A. and 49 (1): 41-48.
Elinder, C. G. (2000) : “Low level expo-
sure to cadmium and early kidney dam- Kim, C. Y., Lee M.J., Lee S.M., Lee
age: the OSCAR study”. Occup. Environ. W.C. and Kim J.S. (1998): “Effect of melat-
Med., 57: 668-672. onin on cadmium induced hepatotoxicity
in male rats”. Tohoku J. Exp. Med. 186:
Jarup, L. (2003) : “Hazards of heavy 205-213.
metal contamination”. Br. Med. Bull., 68:
167-182. Koyuturk, M.; Yanardag, R.; Bulken, S.
and Tunali, S. (2006) : “Influence of com-
Johnston, J.; Sepe, H.; Miano, C.; Bran- bined antioxidants against cadmium in-
nan, R. and Alderton, A. (2005) : “Honey duced testicular damage”. Environ. Toxi-
inhibits lipid oxidation in ready-to-eat col, Pharmacol., 21:235-240.
ground beef patties”. Meat Science, 70 :
627-631. Lars, J. (2002) : “Cadmium overload
and toxicity”. Nephrol. Dial. Transplant,
Jurezuk, M.; Brzoska, J.; Moniuszko- 17: 35-39.
Jakoniuk, M.; Galazyn-Sidorczuk and
Kulikowska-Karpinska, E. (2004) : “Anti- Lowry, O. H.; Rosebrough, N. J.; Farr,
oxidant enzymes activity and lipid peroxi- A. L. and Randal R. T. (1951) : “Protein
dation in liver and kidney of rats exposed measurement with the folin phenol rea-
to cadmium and ethanol”. Food. Chem. gent.” J. Biol. Chem., 193: 265-275.
Toxicol, 42: 429-438.
Manca, D.; Ricard, A. C.; Trottier, B.
Kamiyama, T.; Miyakawa, H.; Li, J. P.; and Chevalier, G. (1991) : “Studies on lip-
Akiba, T.; Liu, J. H.; Marumo, F. and id peroxidation in rat tissues following ad-
Sato, C. (1995) : “Effects of one year cad- ministration of low and moderate doses of

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
96

cadmium chloride”. Toxicology, 67 (3): Noonan, C. W.; Sarasua, S. M.; Cam-


303-323. pagna, D.; Katgman, S. J.; Lybarger, J. A.,
and Mueller, P. W. (2002) : “Effects of ex-
Manuela, B.; Manila, C.; Augusto, A.; posure to low levels of environmental cad-
Maria, P.; Piacentini and Elena, Piatti mium on renal biomarkers”. Environ.
(2007) : “Honey flavonoids as protective Health Perspect., 110: 151-155.
agents against oxidative damage to hu-
man red blood cells”. Food Chemistry, Novelli, E.; Vieira, E., Rodrigues N.
104, (4): 1635-1640. and Ribas B. (1998): “Risk assessment of
cadmium toxicity on hepatic and renal tis-
Meister, A. and Anderson, M. E. sues of rats”. Environ. Res. 79 (2): 102-105.
(1983) : “Glutathione”. Annu. Rev. Bio-
chem., 52:711-760. Ohta, H.; Yamauchi, Y.; Nakakita, M.;
Tanaka, H.; Asami, S.; Seki. Y. and Yoshi-
Mitsumori, K.; Shibutani, M.; Sato, S.; kawa, H. (2000) : “Relationship between
Onodera, H.; Nakagawa, J.; Hayashi, Y. renal dysfunction and bone metabolism
and Ando, M. (1998) : “Relationship be- disorder in male rats after long-term oral
tween the development of hepato-renal quantitative cadmium administration”.
toxicity and cadmium accumulation in Ind. Health, 38: 339-355.
rats given minimum to large amounts of
cadmium chloride in the long-term: Pre- Ossola, J. O. and Tomara, M. L. (1995):
liminary study”. Arch. Toxcicol., 72:545- “Heme oxygenase induction by cadmium
552. chloride evidence for oxidative stress in-
volvement”. Toxicology, 104 (1-3):141-147.
Morales, A.; Vicente, C.; Santiag, J.;
Egido, J.; Mayoral, P.; Arevalo, M.; Fer- Paget, G. E. and Barnes, J. M. (1964) :
nandez, M.; Lopez-Novoa, J. and Perez, F. Toxicity tests. In : Evaluation of Drug Ac-
(2006) : “Protective effect of quercetin on tivities : Pharmacokinetics. (Eds.), Laur-
experimental chronic cadmium nephro- ence, D.R. and Bacharach, A.L. Academic,
toxicity in rats is based on its antioxidant Press, London and New York. Chapter (6)
properties”. Food and Chemical Toxicolo- P.P. 135-166.
gy, 44: 2092-2100.
Partrick, L. (2003) : “Toxic metals and
Mousa, S. A. (2004) : “Expression of ad- antioxidants: Part II. The role of antioxi-
hesion molecules during cadmium hepato- dants in arsenic and cadmium toxicity. Al-
toxicity”. Life Sci. 75, 93-105. tern”. Med. Rev., 8: 106-128.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
97

Sandy, T.; John, M.; Christel, F.; Ivo, L. Yohesh, C.; Awasthi, M. and Ravindra,
and E mmy, V., (2007) : “Chronic expo- N. (1993) : “Effect of chronic cadmium ex-
sure of mice to environmentally relevant, posure on glutathione S-transferase and
low doses of cadmium leads to early glutathione peroxidase activities in Rhesus
renal damage, not predicted by blood monkey: the role of selenium”. Toxciolo-
and urine cadmium levels”. Toxicology. gy, 83: 203-213.
229:145-156.
Tang, W. and Shaikh, Z. A. (2001) :
Sarkars, S.; Poonam, Y. and Bhatnagar, “Renal cortical mitochondrial dysfunction
D. (1997) : “Cadmium-induced lipid per- upon cadmium metallothionein adminis-
oxidation and antioxidant enzymes in rat tration to Sprague-Dawley rats”. J. Toxi-
tissues: role of vitamin E and selenium”. col. Environ. Health, 63: 221-235.
Trace Elem. Electro., 14 (1): 41-45.
Tapple, A. L. (1978) : “Glutathione per-
Satarug, S. and Moore, M. R. (2004): oxidase and hydroperoxidase”. Meth. En-
“Adverse health effects of chronic expo- zymol., 52: 506-513.
sure to low-level cadmium in foodstuffs
and cigarette smoke”. Environ. Health Thayer, W. S. (1984) : “Lipid peroxides
Perspect., 112: 1099-1103. in rats treated chronically with adriamy-
cin”. Biochem. Pharmacol., 33 (14): 2259-
Shaikh, Z. A.; Vu, T. and Zaman, K. 2263.
(1999) : “Oxidative stress as a mechanism
of chronic cadmium heptotoxicity and Thevenod F. (2003) : “Nephrotoxicity
nephrotoxicity and protection by antioxi- and the proximal tubule. Insights
dants”. Toxico. Appl. Pharmacol. 154: 256- from cadmium”. Nephron Physiol., 93 :
263. 87-93.

Shimada, H.; Takamure, Y.; Shimada, Waisberg, M.; Joseph, P.; Hale, B.; Bey-
A.; Yasutake, A.; Waalkes, M. P. and Ima- ersmann, D. (2003) : “Molecular and cellu-
mura, Y. (2004) : “Strain differences of cad- lar mechanisms of cadmium carcinogene-
mium-induced hepatotoxicity in Wister- sis”. Toxicology, 192: 195-117.
irnamichi and Fischer 344 rats: involve-
ment of cadmium accumulation”. Toxicol- World Health organization (WHO)
ogy, 203 (1-3): 189-197. (2000) : Cadmium air quality quide lines
(second ed) WHO, regional office for Eu-
Sidhu, M.; Mridula, S.; Mandeep, B.; rope, Copenhagen Denmark.

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007
Abdel-Moneim & Ghafeer
98

ÈuKJ «Ë ÈbJ « rL « b{ vFOD « qFK qL;« v«u « dOQ «

ÂuOœUJ « s "UM «

Y « v ÊudA*«

Èbd dOH Èbd XL Æœ Ë *bL


 rFM*«b bL
 ¡U Ë Æœ

vÇuuN« r ≠ *WOJOMOKù« ÂuL«Ë v dA« VD« r

◊uO‡‡ √ W‡‡‡F U ≠ V‡‡D« W‡‡‡OK

Ác Xd?√ ¨¡U?C?O?« Ê«d?H?« v ÂuO? œUJU? ÈuKJ«Ë Èb?J« rL??« vK vF?O?D« q?FK v«u« d?OQ?« W? «—b Y ?« «c ¡«d?≈ -

X b???? ≈ YO?  U?? u?L??? Àö v≈ Ê«d???H« rO??I? - r ≤∞∞ ≠ ±µ∞ s t“Ë j u??? ÕË«d? mU i?O√ —Q? ≥∞ œb? vK? W? «—b«

ÂuO? œUJ« b—uKJ XMI ÆW?OU« W? uL?:« ¨vÇuuO?H« `K*« ‰uK 0 vu?Ëd« nu?« q«œ XMIË WDU?{ W u?LL? vË_« W u?L:«

ÆÎUO u rH« od! s dOKOK ∞—∞µ q  q ´ rØr ∞—µ ÂuO œUJ« b—uKJ XMI ∫ WU« W uL:« Ær Ø r ∞—µ

¡«d?ù vK?Ë b??Ë Âœ  UMO?? c?√ - ¨…b*« WU??N v Ê«d??H« `?d?A - ¨lOU?? √ W?F—√ …b* ÎU?O?? u vud?« ¡U??A?G« od! s? sI?(« -

W? «—b WOzU?O?L?OJ« qOU ?« Xd?√ ¨vËd?Jù«Ë vzu?C« »uJ ËdJO*U Uö?)« v W?OÇuu??N«  «dO?G?« W «—b?Ë WOzU?O?LOJ« q?OU ?«

vKJ«Ë bJ?« W√ v b?R*« ‚u Êu?OUuK'«Ë ‰e?<« ÊuOUuK'« q …b?_«  «œU?C  Uu? ”UO? - UL vKJ?«Ë bJ« nzU%Ë

ÆvKJ«Ë bJ« W√ vË U “ö« v ¨…bR*« ‚u Êub« Èu q WbO U_« ◊uGC« ÀËb  ôôœ Èu ”UO -Ë

◊u?G?C« ŸU?H—≈ œu?Ë l jI? Âu?O? œUJU? Wu?I?;« W?OU?« W? u?L?:« v vKJ«Ë b?J« nzU%Ë v? —u?B œu?Ë v?K ZzU?M« Xœ b?Ë

b?Ë ¨…b?_«  «œUC? Èu? v ÷U?H«Ë b?J«Ë vKJ« W?√Ë Âb« qB? s q v …b?R*« ‚u? Êub« W? ŸUH—≈ Wb?O U?_«

Uö?)« iF “öu?O v  «u??Ë WuK*« VOUú? WMD*« Uö)« v r?C vKJ« Uö?) WOU?« W? uL?LK W?OÇuu??N« W «—b«  d?N%√

v ÊU?I?«Ë ŸU?≈ dN% U?L? ¨Uö)« v q?K « iF d?N% b?I b?J« Uö?) W?MU U? √ ÆWuKJ«  UdJ?« WO? Ë√ v qK ?« d«u% iFË

ÆWed*« …œ—Ë_«

Èu??? v h?IË W? Ë—b*« Uö?)« v …b??_«  «œU?C??  Uu??? v …œU“ v≈ Èœ√ vF?O??D« q?F« «b??? ≈ Ê√ ZzU??M« Xœ b?Ë

«b?? ≈ Ê√ ZM p– s? Ë vFO?D« qJA« ÎU?dI vKJ«Ë b?J« Uö?  œUF? ≈ Xu« fH vË  Wb?O? U_« ◊u?GC« …d?(« ozUI?A«

ÆWOÇuuN«Ë WOzUOLOJ« WOUM« s vKJ«Ë bJ« Uö v ÂuO œUJU WOL  «dOQ ÀËb vK v«Ë dOQ t qF«

Mansoura J. Forensic Med. Clin. Toxicol. Vol. XV, No. 2, July 2007

Das könnte Ihnen auch gefallen