ArticlePDF Available

Hepatoprotective, Antioxidant, and Ameliorative Effects of Ginger ( Zingiber officinale Roscoe ) and Vitamin E in Acetaminophen Treated Rats

Authors:
  • National Research Centre, Egypt

Abstract and Figures

ABSTRACT Ginger is a remedy known to possess a number of pharmacological properties. This study investigated efficacy of ginger pretreatment in alleviating acetaminophen-induced acute hepatotoxicity in rats. Rats were divided into six groups; negative control, acetaminophen (APAP) (600 mg/kg single intraperitoneal injection); vitamin E (75 mg/kg), ginger (100 mg/kg), vitamin E + APAP, and ginger + APAP. Administration of APAP elicited significant liver injury that was manifested by remarkable increase in plasma alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), arginase activities, and total bilirubin concentration. Meanwhile, APAP significantly decreased plasma total proteins and albumin levels. APAP administration resulted in substantial increase in each of plasma triacylglycerols (TAGs), malondialdhyde (MDA) levels, and total antioxidant capacity (TAC). However, ginger or vitamin E treatment prior to APAP showed significant hepatoprotective effect by lowering the hepatic marker enzymes (AST, ALT, ALP, and arginase) and total bilirubin in plasma. In addition, they remarkably ameliorated the APAP-induced oxidative stress by inhibiting lipid peroxidation (MDA). Pretreatment by ginger or vitamin E significantly restored TAGs, and total protein levels. Histopathological examination of APAP treated rats showed alterations in normal hepatic histoarchitecture, with necrosis and vacuolization of cells. These alterations were substantially decreased by ginger or vitamin E. Our results demonstrated that ginger can prevent hepatic injuries, alleviating oxidative stress in a manner comparable to that of vitamin E. Combination therapy of ginger and APAP is recommended especially in cases with hepatic disorders or when high doses of APAP are required.
Content may be subject to copyright.
Journal of Dietary Supplements, 10(3):195–209, 2013
C
2013 by Informa Healthcare USA, Inc.
Available online at www.informahealthcare.com/jds
DOI: 10.3109/19390211.2013.822450
Hepatoprotective, Antioxidant, and Ameliorative
Effects of Ginger (Zingiber ofcinale Roscoe)and
Vitamin E in Acetaminophen Treated Rats
Amal S Abdel-Azeem1, Amany M Hegazy1, Khadiga S Ibrahim2,
Abdel-Razik H. Farrag3, & Eman M. El-Sayed1
1Department of Food Science & Nutrition, National Research Centre, Dokki, Cairo,
Egypt, 2Department of Environmental & Occupational Medicine, National Research
Centre, Dokki, Cairo, Egypt, 3Department of Pathology, National Research Centre,
Dokki, Cairo, Egypt
ABSTRACT. Ginger is a remedy known to possess a number of pharmacologi-
cal properties. This study investigated efcacy of ginger pretreatment in alleviating
acetaminophen-induced acute hepatotoxicity in rats. Rats were divided into six groups;
negative control, acetaminophen (APAP) (600 mg/kg single intraperitoneal injection);
vitamin E (75 mg/kg), ginger (100 mg/kg), vitamin E +APAP, and ginger +APAP.
Administration of APAP elicited signicant liver injury that was manifested by remark-
able increase in plasma alanine aminotransferase (ALT), aspartate aminotransferase
(AST), alkaline phosphatase (ALP), arginase activities, and total bilirubin concentra-
tion. Meanwhile, APAP signicantly decreased plasma total proteins and albumin lev-
els. APAP administration resulted in substantial increase in each of plasma triacylglyc-
erols (TAGs), malondialdhyde (MDA) levels, and total antioxidant capacity (TAC).
However, ginger or vitamin E treatment prior to APAP showed signicant hepatopro-
tective effect by lowering the hepatic marker enzymes (AST, ALT, ALP, and arginase)
and total bilirubin in plasma. In addition, they remarkably ameliorated the APAP-
induced oxidative stress by inhibiting lipid peroxidation (MDA). Pretreatment by ginger
or vitamin E signicantly restored TAGs, and total protein levels. Histopathological ex-
amination of APAP treated rats showed alterations in normal hepatic histoarchitecture,
with necrosis and vacuolization of cells. These alterations were substantially decreased
by ginger or vitamin E. Our results demonstrated that ginger can prevent hepatic in-
juries, alleviating oxidative stress in a manner comparable to that of vitamin E. Com-
bination therapy of ginger and APAP is recommended especially in cases with hepatic
disorders or when high doses of APAP are required.
KEYWORDS. acetaminophen, ginger, liver, oxidative stress, proteins, triacylglyc-
erols, vitamin E
Address correspondence to: Khadiga S. Ibrahim, Department of Environmental & Occupational Medicine,
National Research Centre, El-Behoos St. (Tahrir St.Prev.) Dokki, Cairo 12311, Egypt (E-mail khadigasalah@
yahooo).
(Received 29 September 2012; revised 6 March 2013; accepted 3 June 2013)
195
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
196 Abdel-Azeem et al.
INTRODUCTION
The liver plays a pivotal role in regulating various physiological processes in the
body such as metabolism, secretion, and storage. It has a great capacity to detox-
icate toxic substances and synthesize useful ones. Drug metabolism and forma-
tion of reactive toxic metabolites by hepatic microsomal enzyme system plays a
role in the hepatotoxic mechanism (Gonzalez, 1992). Xenobiotics are usually me-
tabolized to inert metabolites that are excreted but unfortunately some of which
are metabolized to more reactive compounds that are more toxic than the par-
ent compound. Acetaminophen {N-acetyl-para-aminophenol (APAP)}is an ex-
tensively prescribed analgesic and antipyretic drug. Although safe when used at
therapeutic doses (Hazai, Monostory, Bakos, Zacher, & Vereczkey, 2001), inten-
tional or unintentional overdose of APAP (>4 g/day) has become the most fre-
quent cause of acute liver failure (Jaeschke, McGill, & Ramachundran, 2012). At
normal doses (<4 g/day), APAP is metabolized by cytochrome P450 monooxyge-
nases (CYPs) to form the highly reactive intermediate, N-acetyl-p-benzoquinone
imine (NAPQI). Metabolizing enzyme; glutathione-S-transferase (GST) catalyzes,
the sequestration of this reactive metabolite through conjugation with glutathione
(GSH). High doses of APAP saturate detoxication pathways, leading to excessive
production of NAPQI which causes glutathione depletion. It freely binds to cellu-
lar molecules producing massive cellular damage and tissue necrosis (Hinson, Reif,
McCullough, & James, 2004; Laine, Auriola, Pasanen, & Juvonen, 2009).
Reactive oxygen species are causally related to oxidative stress that has been
implicated in a number of disease processes, including heart disease, diabetes, can-
cer, and liver injury (Jaeschke, 2000). The balance between ROS and antioxidants
is, therefore, crucial and could be an important mechanism for preventing damage
caused by oxidative stress. This balance has been suggested to have an important
role in preventing APAP toxicity (Jaeschke, Knight, & Bajt, 2003; Lee et al., 2012).
Herbs and spices are generally considered safe and proved to be effective
against various human ailments. Zingiber ofcinale Roscoe commonly known as
ginger, belongs to the family Zigiberaceae. It is a familiar dietary spice with sev-
eral medicinal properties. It has a long history of use in ailments such as nausea,
respiratory, cardiovascular, and rheumatic disorders (Tapsell et al., 2006). It also
has immunomodulatory properties and is reported to inhibit various inammatory
mediators such as prostaglandins and proinammatory cytokines (Grzanna, Lind-
mark, & Frondoza, 2005). Ginger use has been associated with insignicant side
effects and with no known drug or herb interactions, so it has been considered as
a safe herbal medicine (Ali, Blunden, Tanira, & Nemmar, 2008). The constituents
of ginger are numerous and include volatile oils and pungent phenol compounds
known as gingerols, sesquiterpenoids and shogaols. It also has anthocyanin and
tannin in its root bark (Jolad, Lantz, Chen, Bates, & Timmermann, 2005). Sharma
and Singh (2012) demonstrated that ginger is endowed with strong in vitro and in
vivo antioxidant properties. Ginger has been known for its protective role against
a number of toxic agents such as cisplatin (Ajith, Nivitha, & Usha, 2007) and bro-
mobenzene (El-Sharaky, Newairy, Kamel, & Eweda, 2009).This is attributed to its
antioxidant action. A lot had been reported on the chemical characterization of
phytoconstituents and the ethnopharmacological properties of ginger. However,
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
Hepatoprotective Effects of Ginger in Acetaminophen Rats 197
there is still limited information on its potential hepatic protective role against drugs
that induce liver injury when administered at high doses. Hence, the objectives of
the current study were to evaluate the possible hepatoprotective and antioxidant
activities of ginger aqueous suspension against hepatic injury in rats induced by
high dose of acetaminophen. Also, we aimed to determine the effects of ginger ad-
ministration on some nutritional parameters in those rats such as their food intake,
weights and food efciency ratio and to compare the obtained data with those of
vitamin E (Asuku, Atawodi, & Onyike, 2012). Vitamin E has been documented to
protect the liver from several toxins (Ming, Fan, Yang, & Lautt, 2006; Kalender,
Uzun, Ourak, Demir, & Kalender, 2010) by being a standard antioxidant. Thus, it
was used in the current investigation as a reference.
MATERIAL AND METHODS
Drug and Dietary Supplements
Acetaminophen powder was provided by the Arab Drug Company Cairo, Egypt.
Vitamin E was obtained from Pharco Pharmaceuticals, Alexandria, Egypt. Ginger
powder was purchased from MEPACO, Medifood Co. Egypt.
Animals
Adult male Sprague-Dawely rats weighing 120–150 gm were purchased from Ani-
mal House of the National Research Centre, Dokki, Giza, Egypt. They were kept
individually in stainless steel wire bottomed cages at room temperature (25 ±2C)
under 12 hr dark-light cycle. Animals were fed standard balanced diet. Rats had
free access to food and water and they were used after acclimatization period of
one week. Animal experiments were conducted according to the guidelines of An-
imal Care and Ethics Committee of the National Research Centre, Egypt.
EXPERIMENTAL DESIGN
Animals were randomly assigned to six groups each of seven rats as follows; neg-
ative control was orally given distilled water, positive control was injected with a
single dose of acetaminophen 600 mg/kg intraperitoneally (I.P.) (after an overnight
fasting (Hwang et al, 2011). Vitamin E group was orally treated with vitamin E
75 mg/kg/day (third group). The fourth group was orally treated with ginger aque-
ous suspension 100 mg/kg/day (Manju & Nalini, 2010), the fth group (APAP +
vitamin E) and the sixth group (APAP +ginger). Rats were weighed three times
per week and their body weights were recorded. In addition, their food intake was
monitored throughout the experimental period for the calculation of food efciency
ratio (body weight variation/total food intake).
Rats were pretreated daily with vitamin E (groups 3, 5) or with ginger (groups
4, 6) for 14 consecutive days. One hour before the nal treatments on day 14, acute
liver injury was induced in APAP groups (positive control, groups 5 and 6 only) by
APAP (I.P.) injection. Rats were starved overnight and then blood samples were
collected on heparin under light ether anesthesia from retroorbital vein. One part
of blood was left for hemoglobin analysis and the other part was centrifuged at
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
198 Abdel-Azeem et al.
4,000 rpm for 20 min under cooling and plasma was separated and stored at –80C
for subsequent determinations.
Livers were excised, washed quickly with saline and placed immediately in 10%
formalin—Saline buffer for the histopathological examination.
Biochemical Assays
Plasma aspartate and alanine aminotransferases activities were determined accord-
ing to Reitman and Frankel (1957). Alkaline phosphatase activity (ALP) in plasma
was determined according to the principle of Beleld and Goldberg (1971). Plasma
arginase activity was evaluated by using the standard method proposed by March,
Fingerhut, and Miller (1965). Total bilirubin reacted with diazotized sulfanilic acid
in the presence of dimethylsulfoxide to produce azobilirubin which was detected
spectrophotometrically (Walter & Gerard, 1970). Plasma total proteins formed a
blue complex in the presence of copper salt in alkaline solution (Bradford, 1976).
Plasma albumin was quantitatively determined by reacting with bromocresol green
(Doumas, Waston, & Biggs, 1971). Triacylglycerol level (TAGs) was determined en-
zymatically by using the procedure of Fossati and Prencipe (1982). Very low density
lipoprotein concentrations were calculated. Blood hemoglobin was assessed fol-
lowing the standard method proposed by Reuge (1968). Malondialdhyde level, as
a lipid peroxidation end product, was determined by using the thiobarbituric acid
reactive substance (TBARS) assay (Satoh, 1978). Finally, plasma total antioxidant
capacity was assayed calorimetrically (Koracevic, Koracevic, Djordjevic, Andreje-
vic, & Cosic, 2001).
Histopathological Examination
Livers were separated anatomically after anesthetizing the rats by diethylether,
rapidly washed in saline solution to remove the blood. Liver specimens were
removed rapidly from saline, xed in 10% neutral buffered formalin for 24 hr, then
processed up in parafn blocks, sections of 5-mm thick were prepared and stained
with hematoxylin and eosin (Drury & Wallington, 1980) for histopathological
studies.
Statistical Analysis
Data were expressed as mean ±standard error. Statistical differences were assessed
by using F-test, one way of analysis of variance (ANOVA) using SPSS version 14
program. Post-hoc analysis of signicance was made using least-signicant differ-
ence (LSD) test. Differences were considered signicant at p<.05.
RESULTS
APAP single administration resulted in acute liver injury manifested by the signi-
cant increases of plasma AST, ALT, ALP, and arginase activities and total bilirubin
level (p<.01) (Table 1). Pretreatment with ginger signicantly reduced plasma
levels of ALP, ALT, AST, and arginase by 34.9%, 22.2%, 20%, 17.5%, and 42.3%,
respectively, as compared to APAP group (positive control). The recovery from
APAP-induced toxicity by ginger pretreatment was nearly similar to that seen with
vitamin E. As the percentages of reduction produced by vitamin E were 18.6%,
22.2%, 26.7%, 13.1%, and 39.4% respectively. No signicant differences were
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
Hepatoprotective Effects of Ginger in Acetaminophen Rats 199
TABLE 1. Changes in Plasma Hepatic Enzyme Activities and Total Bilirubin of
Concentration Various Experimental Groups
Groups
Negative Positive Vitamin Vitamin Ginger +
Control Control E Ginger E +APAP APAP
Parameters
ALP (U/L) 138 ±8.25 241a∗∗ ±17.1 133 ±5.67 148 ±5.17 196b∗∗ ±1.87 157b∗∗ ±9.43
ALT (u/ml) 23.0 ±2.98 43.7a∗∗ ±3.82 26.7 ±2.03 22.7 ±1.42 34b±3.06 34.0b∗±1.23
AST (u/ml) 48.9 ±3.94 92.9a∗∗ ±3.21 50 ±3.50 54.9 =4.22 68.1b∗∗ ±3.36 74.3b∗∗ ±3.26
Arginase (u/L) 110 ±1.67 137a∗∗ ±1.37 105 ±4.51 116 ±3.92 119b∗∗ ±5.14 113b∗∗ ±1.98
Total bilirubin
(mg/dl)
0.70 ±0.08 1.70a∗∗ ±0.14 0.96 ±0.15 0.71 ±0.04 1.03b∗∗ ±0.15 0.98b∗∗ ±0.16
Values are expressed as mean ±S.E.
avalues significantly differ from normal control.
bvalues significantly differ from APAP.
p<.05; ∗∗p<.01.
observed in enzyme activities or total bilirubin level of healthy rats treated by gin-
ger or vitamin E when compared with those of normal rats.
APAP-administration induced marked oxidative stress (Table 2). Where, it sig-
nicantly increased MDA, a marker for lipid peroxidation. The MDA content in
APAP group was 3 fold that of the negative control. Pretreatment by ginger
signicantly reduced MDA level (p<.05) as compared to that of APAP group
(positive control). Meanwhile, TAC was signicantly higher than that of normal
rats. However, ginger treatment prior to APAP administration showed non signi-
cant changes in TAC. Blood hemoglobin (considered as antioxidant through tran-
sitional iron compartmentalization mechanism) showed nonsignicant change in
positive control as compared to negative control. Overall, ginger administration
produced comparable results as those of vitamin E when their effects were com-
pared to APAP inuences. No signicant differences were observed in MDA or
TAC of normal rats treated by either ginger or vitamin E.
The changes in plasma proteins and lipids of different groups are illustrated in
Table 3. APAP treatment markedly reduced both total proteins and albumin
TABLE 2. Plasma Malondialdehyde Level, Total Antioxidant Capacity and Blood
Hemoglobin Level of Various Experimental Groups
Groups
Negative Positive Vitamin Vitamin Ginger +
Control Control E Ginger E +APAP APAP
Parameters
Malondialdehyde
(nmol/ml)
2.38 ±0.21 7.28a∗∗ ±0.59 3.13 ±0.15 2.61 ±0.48 4.49b∗∗ ±0.42 6.09b±0.07
Total antioxidant
capacity (mol/L)
1.2 6 ±0.12 1.82a∗∗ ±0.027 1.36 ±0.07 1.42 ±0.09 1.86 ±0.02 1.88 ±0.02
Hemoglobin gm/L 15.5 ±0.22 15.0 ±0.25 13.9a∗∗ ±0.22 14.7 ±0.23 14.5 ±0.65 14.3 ±0.29
Values are expressed as mean ±S.E.
avalues significantly differ from normal control.
bvalues significantly differ from APAP.
p<.05; ∗∗p<.01.
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
200 Abdel-Azeem et al.
TABLE 3. Plasma total Proteins, Albumin, Triacylglycerols and Very low Density
Lipoproteins of Different Experimental Groups
Groups
Negative Positive Vitamin Vitamin Ginger +
Control Control E Ginger E +APAP APAP
Parameters
Total proteins
(gm/dl)
6.13 ±0.10 5.74a∗∗ ±0.06 6.75 ±0.16 7.07 ±0.25 6.50b±0.23 6.50b∗∗ ±0.16
Albumin (gm/dl) 4.07 ±0.11 3.89a∗∗ ±0.22 3.73 ±0.17 3.18a∗∗ ±0.05 3.76 ±0.11 3.87 ±0.15
Triacylglycerols
(mg/dl)
45.1 ±3.61 93.7a∗∗ ±5.66 47.2 ±2.49 52.1 ±1.94 78.3b±4.15 59.7b±5.13
VLDL (mg/dl) 9.02 ±0.72 18.7a∗∗ ±1.13 9.26 ±0.52 10.4 ±0.38 15.6b∗∗ ±0.811 11.9b±1.02
Values are expressed as mean ±S.E.
avalues significantly differ from normal control.
bvalues significantly differ from APAP.
p<.05; ∗∗p<.01.
concentrations (p<.01) as compared to those of normal rats. APAP treatment
signicantly (p<.01) increased TAGs and VLDL levels as compared to the cor-
responding values of the negative control. Total proteins, TAGs and VLDL levels
were extremely ameliorated by ginger, where total proteins were signicantly in-
creased, TAGs and VLDL levels were signicantly decreased as compared to pos-
itive control. Ginger effects were similar to those of vitamin E.
No signicant differences were observed in nutritional parameters between
APAP treated and normal rats, Also, food efciency ratio showed insignicant al-
terations throughout various experimental groups (Figure 1).
Histopathological examinations of liver sections from control, rats treated with
vitamin E, and rats treated with ginger showed normal lobular architecture and
normal hepatic cells with a well preserved cytoplasm and well dened nucleus
and nucleoli (Figures 2 A–C). Liver from paracetamol treated animals showed
vacuolization of hepatocytes, sinusoidal dilation, inltration Kupper cells and fatty
degeneration (Figures 2 D–E). The liver of rats treated with vitamin E or ginger
and APAP showed comparatively normal architecture of the liver, having restored
to a large extent, the hepatic lesions produced by the toxins, almost comparable to
the normal (Figures 2 F–I).
FIGURE 1. Food efficiency ratio in various expermintal groups.
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
Hepatoprotective Effects of Ginger in Acetaminophen Rats 201
FIGURE 2. Section of liver of rats (A: negative control, B: vitamin E and C: ginger). Sections
of liver of rats treated with paracetamol (D, E). Sections of liver of rats treated with parac-
etamol and vitamin E (F, G). Sections of liver of rats treated with paracetamol and ginger
(H, I).
DISCUSSION
Acetaminophen is a well-known nonsteroidal antipyretic and analgesic agent,
which is safe in therapeutic doses. However, overdoses of APAP can produce
fatal hepatic necrosis and apoptosis in experimental animals and humans (Amar
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
202 Abdel-Azeem et al.
& Schiff, 2007). The ability of humans to metabolize and clear xenobiotics such
as drugs is a natural process carried out by enzymes called drug metabolizing en-
zymes. APAP subjected to oxidation by CYP2E1 then conjugated by sulfotrans-
ferases (SULT), UDP-glucuronosyltransferases (UGT), and GST (Gonzalez &
Tukey, 2006). The activity of these metabolizing systems is affected by nutrients
(Hwang et al., 2011). Among followed strategies to attenuate APAP toxicity are
dose optimization and the use of combined therapy with antioxidants. Cornas ofc-
inalis fruit extract was reported to protect against APAP hepatic injury (Lee et al.,
2012).
Our data demonstrated that rats treated with APAP developed signicant hep-
atic damage which was observed by a substantial increase in the activities of plasma
enzymes; AST, ALT, ALP, and arginase. Also, plasma total bilirubin level exhibited
a signicant increase. Our results coincided with those of Murayama et al. (2008),
Yassin et al. (2010), Hwang et al. (2011), Sabina et al. (2011), and Lee et al. (2012).
The superiority of arginase to ALT and AST has been demonstrated in toxicant-
induced acute and chronic hepatotoxicity in rats (Murayama, Ikemoto, Fukuda,
Tsunckawa, & Nagata, 2006; Murayama et al., 2008).
Evaluation of the mechanisms of drug induced liver injury and release of solu-
ble products including AST, ALT, arginase, and ALP indicates that mitochondria
are critical targets for drug toxicity, either directly or indirectly through the for-
mation of reactive metabolites such as NAPQI. The consequence of these modi-
cations is generally a mitochondrial oxidant stress and peroxynitrite formation,
which leads to structural alterations of proteins and mitochondrial DNA. In ad-
dition, the release of intermembrane proteins, such as apoptosis-inducing factor
and endonuclease G, and their translocation to the nucleus, leads to nuclear DNA
fragmentation. Together, these events trigger necrotic cell death. Alternatively, the
release of cytochrome C and other proapoptotic factors from mitochondria can pro-
mote caspase activation and apoptotic cell death. Drug toxicity can also induce an
inammatory response with the formation of reactive oxygen species by Kupffer
cells and neutrophils. If not properly detoxied, these extracellularly generated ox-
idants can diffuse into hepatocytes and trigger mitochondrial dysfunction and oxi-
dant stress, which then induces necrotic cell death (Jaeschke et al., 2012). Moreover,
APAP treatment has been shown to elevate mRNA level of CYP2E1 (Ito, Abril,
Bethea, McCuskey, & McCuskey, 2006; Yamaura, Shimada, Nakayama, & Ueno,
2011) with concomitant generation of the highly reactive intermediate NAPQI and
apoptosis in HepG2 cells (Cederbaum, 1998). Free radicals resulted from NAPOI
and CYP2E1 production; induce peroxidation of polyunsaturated fatty acids in the
hepatocytes membranes.
Malondialdhyde is a good indicator of lipid peroxidation. In the current study,
remarkable increase in MDA level was observed in APAP-treated rats as com-
pared to controls. This nding is consistent with Gamal El-din et al. (2003) and
Sabina et al. (2011). Increased lipid peroxidation is generally believed to be an im-
portant underlying cause of the initiation of oxidative stress related various tissues
injuries, cell death, and the progression of acute and chronic diseases (Halliwell,
1997). An interesting nding of this study was the elevation of total antioxidant ca-
pacity by APAP administration. This may be explained as a body defense mecha-
nism against APAP-induced oxidative stress. HO-1 has been shown tobe protective
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
Hepatoprotective Effects of Ginger in Acetaminophen Rats 203
in several hepatic injuries. It has been reported that expression of HO-1 is rapidly
up regulated in the liver and macrophages following administration of high doses
of acetaminophen (Noriega, Ossola, Tomaro, & Batlle, 2000; Chiu, Brittinghum,
& Laskin, 2002; Lee et al., 2012). Thus, HO-1 induction by APAP may be, at least
in part, the reason for the increase of total antioxidant capacity. Hemoglobin was
considered as antioxidant through transitional iron compartmentalization mecha-
nism (Heffner & Repine, 1989). However, the emerged data showed nonsignicant
changes in hemoglobin levels in various groups. Ginger treatment extremely de-
creased lipid peroxidation but signicantly increased total antioxidant capacity as
compared to those of rats given APAP only. These ndings are consistent with those
obtained by (Onwuka, Erhabor, Eteng, & Umoh, 2011; Sharma & Singh, 2012).
APAP treatment signicantly reduced albumin and protein concentrations com-
pared to normal. These results are in consistence with Parameshappa et al., (2012).
Impairment of the hepatic synthetic function by APAP may be the cause of de-
creased protein level and the hypoalbuminemia. Also, the excessive reactive elec-
trophiles and free radicals originated from cytochrome P450 mediated oxidation of
APAP, attack the macromolecules causing oxidation to proteins (Sun, Suqivama,
Masuda, Och, & Takeuchi, 2011). Moreover, APAP enhanced protein catabolism
as indicated from increased level of blood urea nitrogen after APAP treatment
(Gopi, Reddy, Jyothi, & Kumar, 2010). Regarding the effects of APAP adminis-
tration on triacylglycerols; our results showed signicant increase in TAGs level.
This nding was in agreement with the results of Chen et al. (2009), Gopi et al.
(2010), and Oyagbemi and Odetola (2010). APAP induces disruption of free fatty
acid β-oxidation. It is known that mitochondrial proteins are the main targets
of APAP-elicited covalent binding (Qiu, Benet, & Burlingame, 1998). Therefore,
inhibition of fatty acid β-oxidation might be a consequence of APAP-elicited
structural damage to the mitochondria or a direct effect of inhibiting enzymes and
transporters involving in β-oxidation pathway by the reactive metabolites and ROS,
generated by CYP2E1-mediated reactions. Moreover, this oxidative stress stim-
ulates over production of epinephrine (Rosano & Whitley, 1999) which in turn
markedly suppresses insulin secretion with concomitant increase in glucagon, such
hormonal changes induce hormone sensitive lipase activity which greatly enhances
lipolysis in adipose tissue and liver increasing the concentration of free fatty acids in
circulation. Consequently, the conversion of free fatty acids to TAGs was associated
with concomitant increase in VLDL levels (Ghebremeskel et al., 2002).
Previous studies investigated the hepatoprotective effects of ginger against liver
injury induced by ethanol, carbon tetrachloride, and bromobenzene (Yemitan &
Izegbu, 2006; Mallikarjuna, Sahitya, Sathyavelu, & Rajendra, 2008; El-Sharaky
et al., 2009).
Polyphenolic compounds, avonoids and vitamin C present in ginger contribute
to ginger antioxidant activity (Oboh, Akinyemi, & Ademiluyi, 2012). Ginger, pre-
vents the production of free radicals, neutralizes, and scavenges free radicals pro-
duced in the body and chelats prooxidant transition metals as iron. Gingerols and
shogaols are the major bioactive avonoids present in ginger, they suppress the ac-
cumulation of reactive oxygen and/or nitrogen species in the cells (Dugasani et al.,
2010). Moreover, ginger enhances the activities of antioxidant enzymes; superoxide
dismutase, and glutathione peroxidase and increases GSH level (El-Sharaky et al.,
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
204 Abdel-Azeem et al.
2009). Gingerol treatment upregulated mRNA and protein expression of antiox-
idant enzymes such as γ-glutamyl-cysteine ligase the rate-limiting enzyme in the
glutathione biosynthesis (Lee, Park, Kim, & Jang, 2011).
APAP overdose was reported to increase inammatory cytokines such as
interleukin-6 (IL-6) and several proinammatory factors including tumor necrosis
factor α(TNF-α), and interferon-gamma (IFN-gamma) (Bourdi et al., 2007). The
6-gingerol treatment regulate the overexpression of TNF-αand IL6 in arsenic in-
toxicated mice (Chakraborty et al., 2012). Both gingerols and shagaols inhibit pro-
duction of inammatory mediators such as nitrite and prostaglandin E signicantly
and dose dependently (Dugasani et al., 2010). Ginger was found to inhibit the in-
duction of several genes involved in the inammatory response, and some of these
genes encode cytokines, chemokines, and COX-2 (Grzanna, Phan, Polotsky, Lind-
mark, & Frondoza, 2004). It also suppresses leukotriene biosynthesis by inhibiting
5-lipoxygenase (Nurtjahja-Tjendraputra, Ammit, Roufogalis, Tran, & Duke, 2003).
Constitutive androstane receptor (CAR), xenobiotic receptor, is a key regula-
tor of APAP metabolism and hepatotoxicity (Zhang, Huang, Chua, Wei, & Moore,
2002). Ginger may reduce the cellular uptake of APAP possibly due to steric mod-
ication of CAR. Besides, Ginger enhances APAP catabolism by inducing the
drug metabolizing enzymes (Nakamura, Murakami, Ohigashi, Osawa, & Uchide,
2004; El-Sharaky et al., 2009; Jackie, Haleagrahara, & Chakravarthi, 2011). Also 6-
gingerol pretreatment protects against amyloid-induced cytotoxicity and apoptotic
cell death (Lee et al., 2011).
Our emerged data demonstrated that pretreatment of ginger aqueous suspension
prior to APAP-treated rats signicantly increased total protein which is in agree-
ment with Al-Amin, Thomson, Al-Qattan, Peltonen-Shalaby, & Ali, (2006). Mean-
while, it markedly decreased plasma TAGs concentrations; that is consistent with
Fuhrman et al. (2000), Goyal and Kadnur (20 06), and Chakraborty et al. (2012). Al-
Amin et al. (2006) demonstrated that ginger treatment to diabetic rats signicantly
declined urine protein levels indicating decreased protein catabolism by ginger. On
the other hand, the decrease in TAGs may be attributed to the signicant improve-
ment of insulin sensitivity in ginger treated rats (Chakraborty et al., 2012), which
results in increased clearance of VLDL-Ch, consequently, TAGs level was dropped.
Moreover, insulin stimulates lipogenesis in adipose tissue and induces lipoprotein
lipase both virtually decreased TAGs. Sahebkar (2011) proved that ginger could
activate peroxisome receptor Y that induces adiponectin and downregulates pro-
inammatory cyt/okines, changing the balance between adiponectin and TNF-αin
favor of adiponectin promoting considerable antioxidant effects, antidysplipidemic
properties, and reducing hepatic TAGs.
The effect of ginger was comparable to that of standard antioxidant vitamin E.
The potential therapeutic benets of oral vitamin E supplementation have been
evaluated in several liver diseases including viral autoimmune hepatitis, alcoholic
liver diseases, nonalcoholic steatohepatitis, and cholestatic liver diseases (Medina
and Moreno-Otero, 2005). Besides, vitamin E had a remarkable protective effect
against toxins-induced hepatotoxicity (Awodele, Akintonwa, Osunkalu, & Coker,
2010; Uzunhisarcikli & Kalender, 2011; Asuku et al. 2012). Our data are in ac-
cordance with the above mentioned authors as vitamin E supplementation to our
rats prior to APAP showed noticeable hepatoprotection manifested by reduction
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
Hepatoprotective Effects of Ginger in Acetaminophen Rats 205
of hepatic enzyme activities and total bilirubin level. In parallel, it signicantly de-
creased MDA level. Treatment with vitamin E could efciently downregulate in-
ammatory pathways important in liver injury. Furthermore, vitamin E is a potent
agonist of the nuclear hormone receptor, pregnane x, which regulates the transcrip-
tion of cytochrome P450s and other enzymes involved in xenobiotic metabolism
(Kim and Lee, 2006; Park, Kim, & Lee, 2009). Thus, vitamin E could enhance the
detoxication of APAP and reduce hepatic injury in this fashion.
Vitamin E dual actions; enhancement of liver status and its antioxidant activity
shared the amelioration of total proteins after APAP treatment. This nding is con-
sistent with Kalender et al. (2010). It was notable that plasma TAGs concentration
was signicantly reduced by vitamin E treatment prior to APAP administration that
is in agreement with Augusti et al. (2005). The hypolipidemic effect of vitamin E
may be attributed to the increased insulin sensitivity by vitamin E via increasing
the release of hepatic insulin-sensitizing substance (Ming et al., 2006).
In conclusion, our data have shown that, ginger can improve the likelihood of
subsequent hepatic changes following administration of high dose of APAP. It can
ameliorate both histological and biochemical hepatic alterations. Ginger hepatic
protection, in major part, caused by decreasing oxidative stress and by increasing
total antioxidant capacity. The therapeutic efcacy of ginger was comparable to that
of vitamin E. From this point of view, combination regimens containing suitable
doses of ginger and acetaminophen could be advantageous, particularly in treating
patients who are susceptible to liver function disorders or when high doses of APAP
are needed. However, further detailed clinical studies are required to establish its
application.
Declaration of interest: The authors report no conicts of interest. The authors
alone are responsible for the content and writing of the article.
ABOUT THE AUTHORS
Amal S Abdel-Azeem, is a Researcher of Nutrition, Department of Food Science
& Nutrition, National Research Centre, Dokki, Cairo, Egypt. Amany M Hegazy,
is a Researcher of Nutrition, Department of Food Science & Nutrition, National
Research Centre, Dokki, Cairo, Egypt. Khadiga S Ibrahim, is a Professor of Bio-
chemistry, Department of Environmental & Occupational Medicine, National Re-
search Centre, El-Behoos St. (Tahrir St.Prev.), Dokki, Cairo 12311, Egypt. Abdel-
Razik H. Farrag, is a Professor of pathology, Department of Pathology, National
Research Centre, Dokki, Cairo, Egypt. Eman M. El-Sayed, is a Professor of Bio-
chemistry Nutrition, Department of Food Science & Nutrition, National Research
Centre, Dokki, Cairo, Egypt.
REFERENCES
Ajith TA, Nivitha V, Usha S. Zingiber ofcinale roscoe alone and in combination with alpha to-
copherol protect the kidney against cisplatin induced acute renal failure. Food Chem Toxicol.
2007;45(6):921–927.
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
206 Abdel-Azeem et al.
Al-Amin ZM, Thomson M, Al-Qattan KK, Peltonen-Shalaby R, Ali M. Antidiabetic and
hypolipidemic properties of ginger in streptozotocin-induced diabetic rats. Br J Nutr.
2006;96:660–666.
Ali BH, Blunden G, Tanira MD, Nemmar A. Some phytochemical pharmacological and toxi-
cological properties of Zingiber ofcinale Roscoe: a review of recent research. Food Chem
Toxicol. 2008;46:409–420.
Amar PJ, Schiff ER. Acetaminophen safety and hepatotoxicity-where do we go from here? Ex-
pert Opinion on Drug Safety. 2007;6(4):341–355.
Asuku O, Atawodi SE, Onyike E. antioxidant hepatoprotective and ameliorative effects of
methanolic extract of leaves of Grewia mollis juss on carbon tetrachloride treated albino rats.
J Med Food. 2012;15(1):83–88.
Augusti KT, Prabha SP, Smitha KB, Sudheesh M, George A, Joseph MC. Nutraceutical effects of
garlic oil its nonpolar fraction and a cus avonoid as compared to vitamin E in CCL4induced
liver damage in rats. Indian J Exp Biol. 2005;43(5):437–444.
Awodele O, Akintonwa A, Osunkalu VO, Coker HA. Modulatory activity of antioxidants against
the toxicity of Rifampicin in vivo. Rev Inst Med Trop Sao Paulo. 2010;52(1):43–46.
Beleld A, Goldberg MD. Revised assay for serum phenylphosphate activity using 4-amino-
antipyrine. Enzyme. 1971;12:561–573.
Bourdi M, Eiras DP, Holt MP, Webster MR, Reilly TP, Welch KD, Pohl LR. Role of IL-6 in an IL-
10 and IL-4 double Knockout mouse model uniquely susceptible to acetaminophen-induced
liver injury. Chem Res Toxciol. 2007;20(2):208–216.
Bradford MM. A rapid and sensitive method for the quantitation of micrograph of protein uti-
lizing the principle of protein dye binding. Anal Biochem. 1976;72:248–254.
Cederbaum AI. Ethanol-related cytotoxicity catalyzed by CYP2E1-dependent generation of
reative oxygen intermediates in transduced HePG2 cells. Biofactors. 1998;8(1-2):193–196
Chakraborty D, Mukherjee A, Sikdar S, Paul A, Ghosh S, Khuda-Bukhsh AR. 6-Gingerol iso-
lated from ginger attenuates sodium arsenite induced oxidative stress and plays a corrective
role in improving insulin signaling n mice. Toxciol Lett. 2012;210(11):34–43.
Chen C, Krausz KW, Shah YM, Idle JR, Gonzalez F. Serum metabolomics reveals irre-
versible inhibition of fatty acid β-oxidation through the suppression of APARαactivation
as a contributing mechanism of acetaminophen-induced hepatotoxicty. Chem Res Toxicol.
2009;22(4):699–707.
Chiu H, Brittinghum JA, Laskin DL. Differential induction of heme oxygenase-1 in macrophages
and hepatocytes during acetaminophen induced hepatotoxicity in the rat: effects of hemin and
biliverdin. Tox Appl Pharma. 2002;181(2):106–115
Doumas BT, Waston WA, Biggs HG. Albumin standards and the measurement of serum albumin
with bromocresol green. Clin Chem Acta. 1971;31:87–96.
Drury RA, Wallington EA. Carleton’s histopathological technique (pp. 234–239, 4th ed.). Oxford,
New York, Toronto: Oxford University Press, 1980.
Dugasani S, Pichika MR, Nadarajah VD, Balijephalli MK, Tandra S, Korlakunta JN. Compar-
ative antioxidant and anti-inammatory effects of 6-gingerol 8-gingerol 10-gingerol and 6-
shogaol. J Ethnopharmacol. 2010;127(2):515–520.
El-Sharaky AS, Newairy AA, Kamel MA, Eweda SM. Protective effects of ginger ex-
tract against broomobenzene induced hepatotoxicity in male rats. Food Chem Toxicol.
2009;47(7):1584–1590.
Fossati P, Prencipe L. Serum triglycerides determined colorimetrically with an enzyme that pro-
duce hydrogen peroxide. Clin Chem. 1982;28(10):2077–2080.
Fuhrman B, Rosenblat M, Hayek T, Coleman R, Aviram M. Ginger extract consumption reduced
plasma cholesterol inhibits LDL oxidation and attenuates development of atherosclerosis in
atherosclerotic apolipoprotein E decient mice. J Nutr. 2000;130:1124–1131.
Gamal El-Din AM, Mostafa AM, Al-Shabanah OA, Al-Bekairi AM, Nagi MM. Protective ef-
fect of Arabic gum against acetaminophen-induced heatotoxicity in mice. Pharmacological
Research. 2003;48(60):631–635.
Ghebremeskel K, Bitsanis D, Koukkon E, Lowy C, Poston L, Crawford MA. Liver triacylglyc-
erols and free fatty acids in streptozotocin induced diabetic rats have typical n-6 and n-3 pat-
tern. Comp Biochem Physiol. 2002;132:349–354.
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
Hepatoprotective Effects of Ginger in Acetaminophen Rats 207
Gonzalez FF, Tukey RH. Drug metabolism. In: L Brunton, J Lazo and K Parker (Eds.)
Goodman & gilman’s. The pharmacological basis of therapeutics (11th ed). The McGraw:
Hill Companies, Inc, China, 2006. http://0-www.accessmedicine.com.lilac.une.edu/content.
aspx?aID=943955. Accessed January 26, 2009.
Gonzalez FJ. Human cytochrome P4SO: problems and prospects. Trends Pharmacol Sci.
1992;13:346–435.
Gopi KS, Reddy AG, Jyothi K, Kumar BA. Acetaminophen-induced hepato and nephrotoxicity
and amelior-ation by silymarin and Terminalia chebula in rats. Toxicol Int. 2010;17(2):64–66.
Goyal RK, Kadnur SV. Benecial effects of Zingiber ofcinale on goldthioglucose induced obe-
sity. Fitoterapia. 2006;77:160–163.
Grzanna R, Lindmark L, Frondoza CG. Ginger an herbal medicinal product with broad anti-
inammatory actions. J Med Food. 2005;8:125–132.
Grzanna R, Phan P, Polotsky A, Lindmark L, Frondoza CG. Ginger extract inhibit beta-amyloid
peptide-induced cytokine and chemokine express ion in cultured THP-1 monocytes. J Altern
Complement Med. 2004;10:1009–1013.
Halliwell B. Antioxidants and human disease: a general introduction. Nutr Rev. 1997;55:544–549.
Hazai E, Monostory K, Bakos A, Zacher G, Vereczkey L. About paracetamol again. Orv Hetil.
2001;142(7):345–349.
Heffner JE, Repine JE. Pulmonary strategies of antioxidant defence. Am Rev Respir Dis.
1989;140(2):531–554.
Hinson JA, Reif AB, McCullough SS, James LP. Acetaminophen-induced hepatotoxicity role of
metabolic activation reactive oxygen/nitrogen species and mitochondrial permeability transi-
tion. Drug Metabol Rev. 2004;36(3-4):805–822.
Hwang J, Chang YH, Park JH, Kim SY, Chung H, Shim E, Hwang HJ. Dietary saturated and
monosaturated fats protect against acute acetaminophen hepatotoxicity by altering fatty acid
composition of liver microsomal membrane in rats. Lipids in Health Dis. 2011;10:184–192.
Ito Y, Abril ER, Bethea NW, McCuskey MK, McCuskey RS. Dietary steatotic liver attenuates
acetaminophen hepatotoxicaty in mice. Microcirculation. 2006;13:19–27.
Jackie T, Haleagrahara N, Chakravarthi S. Antixoidant effects of Etlingera elatior ower extract
against lead acetate induced-perturbations in free radical scavenging enzymes and lpid perox-
idation in rats. BMC Res Notes. 2011;4:67.
Jaeschke H. Reactive oxygen and mechanisms of inammatory liver injury. J Gast and Hepat.
2000;15(7):718–724.
Jaeschke H, Knight TR, Bajt ML. The role of oxidant stress and reactive nitrogen species in
acetaminophen hepatotoxicity. Toxicol Lett. 2003;144(3):279–288.
Jaeschke H, McGill MR, Ramachundran A. Oxidant stress mitochondria and cell death mecha-
nisms in drug induced liver injury: lessons learned from acetaminophen hepatotoxicity. Drug
Metab Rev. 2012;44(1):88–106.
Jolad SD, Lantz RC, Chen GJ, Bates RB, Timmermann BN. Commerically processed dry ginger
(Zingiber ofcinale): composition and effect on LPS-Stimulated PGE2 production. Phyto-
chemistry. 2005;66:1614–1635.
Kalender S, Uzun FG, Ourak D, Demir F, Kalender Y. Malathion-induced hepatotoxicity in rats:
the effect of vitamin C and E. Food Chem Toxicol. 2010;48(2):633–638.
Kim JY, Lee SM. Vitamins C and E protect hepatic cytochrome P450 dysfunction induced by
polymicrobial sepsis. Eur J Pharmacol. 2006;534:202–209.
Koracevic D, Koracevic G, Djordjevic V, Andrejevic S, Cosic V. Method for the measurement
of antioxidant activity in human uid. J Clin Pathol. 2001;54: 356–361.
Laine JE, Auriola S, Pasanen M, Juvonen RO. Acetaminophen bioactivation by human cy-
tochrome P4SO enzymes and animal microsomes. Xenobiotica. 2009;39(1):11–21.
Lee C, Park GH, Kim CY, Jang JH. 6-Gingerol attenuates B-amyloid-induced oxidative cell death
via fortifying cellular antoxidant defense system. Food Chem Toxicol. 2011;49(6):1261–1269.
Lee NH, Seo CS, Lee HY, Lee JK, Lee JA, Song KY, Shin HK, Lee MY, Seo YB, Kim H, Ha H.
Hepatoprotective and antioxidantive activities of cornus ofcinalis against acetoaminophen-
induced hepatotoxicity in mice. Evid Based Complement Alternat Med. 2012; Published on-
line 2011 August 17. Volume 2012 (2012), 8 pages. doi: 10.1155/2012/804924
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
208 Abdel-Azeem et al.
Mallikarjuna K, Sahitya CP, Sathyavelu RK, Rajendra W. Ethanol toxicity: rehabilitation of hep-
atic antioxidant defense system with dietary ginger. Fitoterapia. 2008;79:174–178.
Manju V, Nalini N. Effect of ginger on lipid peroxidation and antioxidant status in 1,2-
dimethylhydrazine induced experimental colon carcinogenesis. J Biochem Tech. 2010;
2(2):161–167.
March WH, Fingerhut B, Miller H. Automated and manual direct methods for the determination
of blood urea. Clin Chem. 1965;11:624–627.
Medina J, Moreno-Otero R. Pathophysiological basis for antioxidant therapy in chronic liver
disease. Drugs. 2005;65:2445–2461
Ming Z, Fan YJ, Yang X, Lautt WW, Synergistic protection by S-adenosylmethionine with
vitamin C and E on liver injury induced by thioacetamide in rats. Free Radic Biol Med.
2006;40(4):617–624.
Murayama H, Ikemoto M, Fukuda Y, Nagata A. Superiority of serum type-1 arginase and or-
nithine carbamyltransferase in the detection of toxicant-induced acute hepatic injury in rats.
Clin Elim Acta. 2008;391:31—35.
Murayama H, Ikemoto M, Fukuda Y, Tsunckawa S, Nagata A. Advantage of serum type-1
arginase and ornithne carbamoyltransferase in the evaluation of acute and chronic liver dam-
age induced by thioacetamide in rats. Clin Chim Acta. 2006;375: 63–68.
Nakamura Y, Yoshida C, Murakami A, Ohigashi H, Osawa T, Uchide K. Zerumbone a trop-
ical ginger sesquiterpene activates phase II drug metabolizing enzymes. FEBS Lett. 2004;
572:245–250.
Noriega GO, Ossola JO, Tomaro ML, Batlle AM. Effect of acetaminophen n heme metabolism
in rat liver. Int J Biochem and Cell Biol. 2000;32(9):983–991.
Nurtjahja-Tjendraputra E, Ammit AJ, Roufogalis BO, Tran LVH, Duke CC.Effects antiplatelet
and COX-1 enzyme inhibitors from pungent constituents of ginger. Thrombosis Res.
2003;111:259–265.
Oboh G, Akinyemi AJ, Ademiluyi AO. Antioxidant and inhibitory effect of red ginger (Zingiber
ofcinale Var Ruba) and white ginger (Zingiber ofcinale Roscoe) on Fe2+, induced lipid
peroxidation in rat brain in vitro. Exp Toxicol Pathol. 2012;64:31–36
Onwuka FC, Erhabor O, Eteng MU, Umoh IB. Protective effects of ginger toward cadmium-
induced testes and kidney lipid peroxidation and hematological impairment in albino rats. J
Med Food. 2011;14(7–8):817–821.
Oyagbemi AA, Odetola AA. Hepatoprotective effects of ethanolic extract of Cnidos-
colous aconitifolius on paracetamol-induced hepatic damage in rats. Pak J Biol Sci.
2010;13(4):164–169.
Parameshappa B, Ali Basha MS, Sen S, Chakraborty Kumar GV, Sugar GV, Sowmya L, Raju
KK, Sesh PK, Lakshmi AV. Acetaminophen-induced nephrotoxicity in rats: protective role of
Cardiospermum halicacabum. Pharm Biol. 2012;50(2):247–253
Park S, Kim AJ, Lee M. Synergic effects of α-tocopherol and β-carotene on tertbutylhydro-
peroxide-induced HepG2 cell injury. Toxicol Ind Health. 2009;25(4–5):311–320.
Qiu Y, Benet LZ, Burlingame AL, Identication of the hepatic protein targets of the reactive
metabolites of acetaminophen in vivo in mice using two-dimensional gel electrophoresis and
mass spectrometry. J Biol Chem. 1998;273:19740–17953.
Reitmen S, Frankel S. Colorimetric method for aspartate and alanine aminotransferases. Am J
Clin Path. 1957;28:56–72.
Reuge C. Standardization method for hemoglobin determination. Rev Fr Transfus. 1968;11:45–50.
Rosano TG, Whitley RJ. Catecholamines and serotonin In: E R Ashwood, Eds. Tietz textbook
of clinical chemistry (3rd ed.), pp. 1570–1600. Burtis CA, London: WB Saunders, 1999.
Sabina EP, Pragasam SJ, Kumar S, Rasool M. 6-Gingerol an active ingredient of ginger,
protects acetaminophen-induced hepatotoxicity in mice. Zhong Xi Yi Jie He Xue Bao.
2011;9(11):1264–1269.
Sahebkar A. Potential efcacy of ginger as a natural supplement for non alcoholic fatty liver
disease. World J Gastroenterol. 2011;17(2):271–272.
Satoh K. Serum lipid peroxide in cerebrovascular disorders determined by a new colormetric
method. Clinca Chimica Acta. 1978;90:37–48.
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
Hepatoprotective Effects of Ginger in Acetaminophen Rats 209
Sharma P, Singh R. Dichlorvos and Lindane Induced oxidative stress n rat brain: rotedctive ef-
fects of ginger. Pharmacognosy Res. 2012;4(1):27–32.
Sun J, Suqivama A, Masuda A, Och T, Takeuchi T. Expressions of protein oxidation markers
dityrosine and advanced oxidation protein products in acetaminophen-induced liver injury in
rats. J Vet Med Sci. 2011;73(9):1185–1190.
Tapsell LC, Hemphill I, Lobiac L, Parch CS, Sullivan DR, Fenech M, Roodenrys S, Keogh JB,
Clifton PM, Williams PG, Fazio VA, Inge KE. Health benets of herbs and spices: The past,
the present, the future. Med J Aust. 2006;185(4):54–524.
Uzunhisarcikli M, Kalender K. Protective effects of vitamin C and E against hepatotoxicity in-
duced by methyl parathion in rats. Ecotoxicol Environ Saf. 2011;74(7):2112–2118.
Walter M, Gerard H. Ultramicromethod for the determination of conjugated and total bilirubin
in serum or plasma. Microchem J. 1970;15:231–236.
Yamaura K, Shimada M, Nakayama N, Ueno K. Protective role of goldenseal (Hydrastis
Canadensis L.) on acetaminophen-induced hepatotoxicity through inhibition of CYP2E1 in
rats. Pharmacognosy Res. 2011;3(4):250–255.
Yassin NAZ, El-Rokh EM, El-Shenaw SM, Ehasn NA, Sayed WH, Hassanein HMD, Ibrahim
BMM. Study f the hepatoprotective effect tof ginger aqueous infusion in rats. J Chem Pharm
Res. 2010;2(4):476–488.
Yemitan OK, Izegbu MC. Protective effect of Zingiber ofcinale (Zingiberaccae) against
carbon tetrachloride and acetaminophen-induced hepatotoxicity in rats. Phytother Res.
2006;20:997–1002.
Zhang J, Huang W, Chua SS, Wei P, Moore DD. Modulation of acetaminophen-induced hepa-
totoxicity by xenobiotic receptor CAR. Science. 2002;298(5592):422–424.
J Dietary Suppl Downloaded from informahealthcare.com by Lulea University Of Technology on 08/20/13
For personal use only.
... The reducing power of a compound is an indicator of its potential antioxidant activity. Ginger exhibits excellent antioxidant activity and is a source of natural antioxidants that regulate lipid peroxidation and reduce the production of reactive oxygen species (Abdel-Azeem, Hegazy, Ibrahim, Farrag, & El-Sayed, 2013;Nayak et al., 2016;Zagórska et al., 2023). ...
... Here, we identified the volatiles of So, 55,53,49,48,57,and 55 compounds,respectively (Table 5). Nitrile compounds contributed to the special aroma of sesame oil (Abdel-Azeem et al., 2013), and various compounds were found, including 2-ethyl-6-methyl-pyrazine, 2,5-dimethyl pyrazine, 2,5-dimethyl-3-ethylpyrazine, 3,5-diethyl-2methyl pyrazine and methyl pyrazine in this study. Hexanal, nonanal, benzaldehyde, and benzeneacetaldehyde exhibited the highest odor activity value in sesame oil (Jia et al., 2019). ...
Article
Full-text available
Ginger-infused sesame oil enriches the nutrition and provides enhanced flavor for the foods. An original processing procedure and module for evaluation were established in this study, using different raw materials (Guangdong and Chu ginger) and treatments (ginger powder, extract, and both). The quality, functionality, and flavor of the infused oils were evaluated. Ginger-infused sesame oil contained 0.58–3.22 µg/g of 6-gingerol, 0.21–0.88 µg/g of 6-shogaol. The number range of volatile compounds from 48 to 55 identified by gas chromatography-mass spectrometry varies depending on different process procedures. Agglomerative hierarchical clustering analysis revealed the flavor profiles were clustered by different varieties, while gingerol and phytosterol was by different treatments. In conclusion, sesame oil was an appropriate carrier for gingerol and phytosterol, which are characterized by higher antioxidant capacities (p < 0.05). These results show the benefits of developing infused oil products with enhanced functional and sensory properties.
... (2009) and Abdel-Azeem et al.(2013). When butter was added, mice's ALT enzyme activity was significantly reduced[38,39,40]. ...
Research Proposal
Full-text available
Hypercholesterolemia is characterized by elevated blood cholesterol levels, which are treated with synthetic medications like statins. Plant extracts like ginger and cinnamon can lower blood triglycerides and total cholesterol while increasing HDL cholesterol. Studies show that cinnamon and ginger increase high-density lipoprotein cholesterol and decrease blood triglycerides and total cholesterol. The current study aims to assess the antihyperlipidemic impact of administering ginger and cinnamon together on hypercholesterolemic rats' salivary gland.
... However, after administration of Gin and/or Sor, there was an observed improvement in albumin levels, with the most significant increase revealed in the group treated with both Gin and Sor. These findings are consistent with the results of previous research that have been published [26][27][28]. ...
Article
Full-text available
Ginger (Gin) has numerous therapeutic properties. One of Gin’s most potent components is 6-gingerol, a naturally occurring phenol. This study aimed to investigate the therapeutic impact of gingerol and/or sorafenib on the ATG4/CASP3 and COIIV/COX-2/NF-B Expression as a potential therapy for DAB-induced HCC. Gin was administered to HCC mice induced by p-Dimethylaminoazobenzene (DAB) alone or combined with sorafenib (Sor). Superoxide dismutase (SOD), catalase (CAT), and oxidative stress malondialdehyde (MDA), as well as biochemical markers including AST, ALT, ALP, Albumin, and Bilirubin, were examined. The expression of oncogenes (COIIV, COX-2, NF-κB, and survivin) and tumor suppressor genes (ATG4 and CASP3) was evaluated using qPCR. According to the results, the levels of MDA have been markedly decreased, while SOD and CAT have been increased. Further, the expression levels of tumor suppressor genes were upregulated, whereas the expression levels of oncogene genes were downregulated. Furthermore, in a dose-dependent manner, gingerol has shown the potential to alleviate hepatic portal vein (PV) dilatation and could offer a reliable therapy for HCC. This suggests combining the two compounds may be more effective than alone and that Gin could be a promising therapeutic option for HCC. The binding of Gin and Sor to the active sites of the target genes prevents them from functioning normally, which in turn stops the pathways from carrying out their oncogenic functions. Additionally, COX-2 inhibition reduces the production of certain pro-inflammatory compounds, which further averts oncogenesis. Conclusively, this study indicated that Gin has cytoprotective properties and anti-cancer activity that may be related to controlling oxidative stress. This effect may be achieved by suppressing the COIIV/COX-2/NF-κB pathway and upregulating the ATG4 /CASP3 pathways. Graphical abstract
... So, the antioxidant characters of ginger could protect DNA and other important molecules from oxidation and damage and can improve liver function. Ginger exhibited DNA protection, blocked lipid peroxidation & decrease apoptosis and free radical scavenging [54,55] . ...
... Minimal toxicity to the liver hepatocytes were observed in rats treated with ginger. The hepato-protective effects of ginger have been attributed to its antioxidant activity which inhibits the membranous lipid peroxidation from free radicals that usually cause cell damage and necrosis [34,35]. Treatment with garlic and lemon caused prominent damage to the liver cells in the rats used in this study and an extended synergic effect seen in the group given the mixture. ...
Article
Full-text available
https://www.eaht.org/m/journal/view.php?number=936
Article
Background: Controlled and targeted drug delivery to treat nonalcoholic fatty liver disease (NAFLD) can benefit from additive attributes of natural formulation ingredients incorporated into the drug delivery vehicles. Methods: Lovastatin (LVN) loaded, bile acid (BA) and fatty acid (FA) integrated nanoemulsomes (NES) were formulated by thin layer hydration technique for synergistic and targeted delivery of LVN to treat NAFLD. Organic phase NES was comprised of stearic acid with garlic (GL) and ginger (GR) oils, separately. Ursodeoxycholic acid and linoleic acid were individually incorporated as targeting moieties. Results: Stability studies over 90 days showed average NES particle size, surface charge, polydispersity index, and entrapment efficiency values of 270 ± 27.4 nm, -23.8 ± 3.5 mV, 0.2 ± 0.04 and 81.36 ± 3.4%, respectively. Spherical NES were observed under a transmission electron microscope. In-vitro LVN release depicted non-fickian release mechanisms from GL and GR oils-based NES. Ex-vivo permeation of BA/FA integrated NES through isolated rat intestines showed greater flux than non-integrated ones. Conclusion: Liver histopathology of experimental rats together with in-vivo lipid profiles and liver function tests illustrated that these NES possess the clinical potential to be promising drug carriers for NAFLD.
Article
Etlingera Giseke, a genus of approximately 150 aromatic ginger species, is predominantly distributed in the Indomalayan realms. The pharmacological properties and ethnomedicinal uses of the genus are well proven as many of its species are used for the treatment of dermatosis, conjunctivitis, malignancy, myalgia, pharyngitis, hyperbilirubinemia, gastrointestinal disorders, and cardiotonic conditions. Ethnomedicinal studies revealed that about 32 species of Etlingera are traditionally used for different ailments, while their rich flavors and fragrances make them widely employed as spices and condiments worldwide. Considering the high aroma present in the rhizomes of Etlingera, this genus holds promise as a lucrative cash crop for the flavor and fragrance industries. To date, approximately 410 phytocompounds have been identified from the various plant parts where around 209 compounds are reported with various biological activities. Studies revealed that pharmacologically E. elatior is the most utilized species as its phytoconstituents show highest number of biological activities, including antibacterial, cytotoxic, antityrosinase, hepatoprotective, antiinflammatory, and antioxidant effects. Species E. elatior is ornamental and is dominated in Borneo, Java, Lesser Sunda Islands, Malaya, Sumatera, and Thailand. From the literature investigation, it has been reported that only E. elatior and E. hemisphaerica have an ornamental or market value (per bulb approx. 12-15$) while there is no documentation of market value for the remaining 148 species. Notably, 24 of these species exhibit significant pharmaceutical activities. The exploration of biogeography, ethnobotany, phytochemistry, and pharmacology holds the potential to uncover significant relevance. It may yield more comprehensive data that supports the appropriate utilization of a majority of species and enhances our understanding of the therapeutic efficacy within the genus. In conclusion, this genus holds promising pharmacological properties that need great attention, which could play a significant role in advancing the field of pharmaceutical development. Despite of its versatile applications, inadequate information and lack of attention keep most of Etlingera sp. in underutilized category. Recognizing immense potential of the genus, the present review mainly focuses on discussing the ethnobotany, phytochemistry, pharmacology and biogeography of the genus Etlingera as it expands our understanding of the genus, emphasizing potential avenues for future research.
Article
As plantas medicinais têm sido utilizadas como fonte alternativa no tratamento de doenças hepáticas a milhares de anos, atualmente a planta chamada Cardo Mariano (silimarina) e o Zingiber officinale (gengibre) vem apresentando evidências devido ao efeito hepatoprotetor. Objetivo: Essa pesquisa objetivou apresenta através de uma abordagem qualitativa descritiva buscar evidências científicas acerca da efetividade hepatoprotetora da planta Silimarina e do Zingiber officinale e como a mesma pode contribuir para a redução, tratamento e prevenção da Doença Hepática Gordurosa Não Alcoólica (DHGNA). Para isso foram utilizados dados secundários, os quais foram buscados em fontes bibliográficas no Scientific Lirary Online (Scielo), LILACS/BIREME, PubMed/Medline, Google Acadêmico. Resultados e discussão: Constatou-se que a utilização da planta medicinal Silimarina e Zingiber officinale mostraram avanços a respeito de seu efeito anti-inflamatório, antioxidante, antiviral, antifibrotico e hepatoprotetor. Os estudos comprovam que a Silimarina e Zingiber officinale podem ser boas alternativas e coadjuvantes para o tratamento da DHGNA. Conclusão: Conclui-se que a utilização de fitoterápicos tem sido eficaz nos tratamentos da Doença Hepática Gordurosa Não Alcoólica, além de ser utilizado na prevenção. Contudo, necessita mais estudos clínicos a fim de evidenciar a forma mais segura de sua eficácia, efeitos nos distúrbios hepáticos e principalmente em pacientes com DHGNA.
Article
Herbs and spices have a traditional history of use, with strong roles in cultural heritage, and in the appreciation of food and its links to health. Demonstrating the benefits of foods by scientific means remains a challenge, particularly when compared with standards applied for assessing pharmaceutical agents. Pharmaceuticals are small-molecular-weight compounds consumed in a purified and concentrated form. Food is eaten in combinations, in relatively large, unmeasured quantities under highly socialised conditions. The real challenge lies not in proving whether foods, such as herbs and spices, have health benefits, but in defining what these benefits are and developing the methods to expose them by scientific means. CULTURAL ASPECTS: The place of herbs and spices in the diet needs to be considered in reviewing health benefits. This includes definitions of the food category and the way in which benefits might be viewed, and therefore researched. Research may focus on identifying bioactive substances in herbs and spices, or on their properties as a whole food, and/or be set in the context of a dietary cuisine. THE ROLE OF HERBS AND SPICES IN HEALTH: The antioxidant properties of herbs and spices are of particular interest in view of the impact of oxidative modification of low-density lipoprotein cholesterol in the development of atherosclerosis. There is level III-3 evidence (National Health and Medical Research Council [NHMRC] levels of evidence) that consuming a half to one clove of garlic (or equivalent) daily may have a cholesterol-lowering effect of up to 9%. There is level III-1 evidence that 7.2 g of aged garlic extract has been associated with anticlotting (in-vivo studies), as well as modest reductions in blood pressure (an approximate 5.5% decrease in systolic blood pressure). A range of bioactive compounds in herbs and spices have been studied for anticarcinogenic properties in animals, but the challenge lies in integrating this knowledge to ascertain whether any effects can be observed in humans, and within defined cuisines. Research on the effects of herbs and spices on mental health should distinguish between cognitive decline associated with ageing and the acute effects of psychological and cognitive function. There is level I and II evidence for the effect of some herbal supplements on psychological and cognitive function. There is very limited scientific evidence for the effects of herbs and spices on type 2 diabetes mellitus, with the best evidence being available for the effect of ginseng on glycaemia, albeit based on four studies. More research is required, particularly examining the effects of chronic consumption patterns. With increasing interest in alternatives to non-steroidal anti-inflammatory agents in the management of chronic inflammation, research is emerging on the use of food extracts. There is level II evidence for the use of ginger in ameliorating arthritic knee pain; however, the improvement is modest and the efficacy of ginger treatment is ranked below that of ibuprofen. More definitive research is required. PUBLIC HEALTH AND DIETARY IMPLICATIONS: Recommendations for intakes of food in the Australian guide to healthy eating do not yet include suggested intakes of herbs and spices. Future consideration should be given to including more explicit recommendations about their place in a healthy diet. In addition to delivering antioxidant and other properties, herbs and spices can be used in recipes to partially or wholly replace less desirable ingredients such as salt, sugar and added saturated fat in, for example, marinades and dressings, stir-fry dishes, casseroles, soups, curries and Mediterranean-style cooking. Vegetable dishes and vegetarian options may be more appetising when prepared with herbs and spices. FUTURE DIRECTIONS: As several metabolic diseases and age-related degenerative disorders are closely associated with oxidative processes in the body, the use of herbs and spices as a source of antioxidants to combat oxidation warrants further attention. Immediate studies should focus on validating the antioxidant capacity of herbs and spices after harvest, as well as testing their effects on markers of oxidation. This will work in parallel with clinical trials that are aiming to establish antioxidants as mediators of disease prevention. From a dietary perspective, the functionality of herbs and spices will be exposed through consideration of their properties as foods. As with most foods, the real benefits of including them in the diet are likely to emerge with a better understanding of the attributes of health that are best supported by food, and in methodological developments addressing the evidence base for their effects. These developments are well underway through evidence-based frameworks for substantiating health claims related to foods. At present, recommendations are warranted to support the consumption of foods rich in bioactive components, such as herbs and spices. With time, we can expect to see a greater body of scientific evidence supporting the benefits of herbs and spices in the overall maintenance of health and protection from disease.