ArticlePDF Available

Thymoquinone mitigate ischemia-reperfusion-induced liver injury in rats: a pivotal role of nitric oxide signaling pathway

Authors:

Abstract and Figures

Oxidative and nitrosative stress-induced endothelial cell damage play an essential role in the pathogenesis of hepatic ischemia-reperfusion (IR) injury. IR is associated with reduced eNOS expression and exacerbated by superimposed stress. NOSTRIN induces intracellular endothelial nitric oxide synthase (eNOS) translocation and inducible nitric oxide synthase (iNOS) increases nitric oxide (NO) production. Our aim was to assess hepatic expression of iNOS, eNOS, and NOSTRIN in IR with or without N-acetylcysteine (NAC) or thymoquinone (TQ) pretreatment and to compare their hepatoprotective effects. Surgical induction of IR was performed by occlusion of hepatic pedicle for 30 min with mini-clamp and reperfused for 30 min. The effects of TQ (20 mg/kg/day) or NAC (300 mg/kg/day) administered orally for 10 days were evaluated by serum ALT and AST, oxidative stress parameters, NO production, and histopathological analysis. Also, localization and expression of iNOS, eNOS, and NOSTRIN were assessed by immunofluorescence. TQ or NAC pretreatment significantly decreased elevated serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and myeloperoxidase (MPO) activities, malondialdehyde (MDA) level, and NO production. In addition, they restored the depleted GSH content and alleviated histopathological changes. Furthermore, they up-regulated eNOS and down-regulated iNOS and NOSTRIN expressions. TQ exerts its hepatoprotective effect, at least in part, by nitric oxide signaling pathway through modulation of iNOS, eNOS, and NOSTRIN expressions as well as suppression of oxidative stress.
This content is subject to copyright. Terms and conditions apply.
ORIGINAL ARTICLE
Thymoquinone mitigate ischemia-reperfusion-induced liver
injury in rats: a pivotal role of nitric oxide signaling pathway
Mohamed Abd-Elbaset
1
&El-Shaimaa A. Arafa
1,2
&Gamal A. El Sherbiny
3
&
Mohamed S. Abdel-Bakky
4,5
&Abdel Nasser A.M. Elgendy
6
Received: 27 June 2016 /Accepted: 23 September 2016 /Published online: 7 October 2016
#Springer-Verlag Berlin Heidelberg 2016
Abstract Oxidative and nitrosative stress-induced endotheli-
al cell damage play an essential role in the pathogenesis of
hepatic ischemia-reperfusion (IR) injury. IR is associated with
reduced eNOS expression and exacerbated by superimposed
stress. NOSTRIN induces intracellular endothelial nitric oxide
synthase (eNOS) translocation and inducible nitric oxide syn-
thase (iNOS) increases nitric oxide (NO) production. Our aim
was to assess hepatic expression of iNOS, eNOS, and
NOSTRIN in IR with or without N-acetylcysteine (NAC) or
thymoquinone (TQ) pretreatment and to compare their hepa-
toprotective effects. Surgical induction of IR was performed
by occlusion of hepatic pedicle for 30 min with mini-clamp
and reperfused for 30 min. The effects of TQ (20 mg/kg/day)
or NAC (300 mg/kg/day) administered orally for 10 days were
evaluated by serum ALT and AST, oxidative stress parame-
ters, NO production, and histopathological analysis. Also, lo-
calization and expression of iNOS, eNOS, and NOSTRIN
were assessed by immunofluorescence. TQ or NAC pretreat-
ment significantly decreased elevated serum aspartate amino-
transferase (AST), alanine aminotransferase (ALT), and
myeloperoxidase (MPO) activities, malondialdehyde (MDA)
level, and NO production. In addition, they restored the de-
pleted GSH content and alleviated histopathological changes.
Furthermore, they up-regulated eNOS and down-regulated
iNOS and NOSTRIN expressions. TQ exerts its hepatoprotec-
tive effect, at least in part, by nitric oxide signaling pathway
through modulation of iNOS, eNOS, and NOSTRIN expres-
sions as well as suppression of oxidative stress.
Keywords Thymoquinone .Hepatic ischemiareperfusion .
iNOS .eNOS .NOSTRIN
Introduction
Hepatic ischemiareperfusion (IR) injury is one of the major
obstacles in different clinical conditions such as liver surgery,
especially in transplantation, hepatic resection, and trauma.
Several relevant factors and mediators such as nitric oxide
(NO) were involved in the ischemic injury of the liver. The
inducible nitric oxide synthase (iNOS) and the endothelial
nitric oxide synthase (eNOS) are two isoforms of nitric oxide
synthase that play an important but opposed role in liver IR
(Trocha et al. 2010). NO derived from iNOS and its oxidative
byproduct peroxynitrite cause increased hepatic injury and
systemic hypotension which reduce liver blood flow
(Jaeschke 2003). On the contrary, NO produced by eNOS
has a physiological protective role by maintaining liver blood
flow, inhibition of leukocyte adhesion, platelet aggregation,
and scavenging of free radicals (Shah and Kamath 2003).
Whereas, eNOS traffic inducer (NOSTRIN)-involved in shut-
tling of eNOS was originally described as an F-BAR domain-
*El-Shaimaa A. Arafa
elshimaa.arafa@pharm.bsu.edu.eg
1
Department of Pharmacology and Toxicology, Faculty of Pharmacy,
Beni-Suef University, Beni-Suef 62514, Egypt
2
Department of Pharmacology and Toxicology, College of Pharmacy
and Health Sciences, Ajman University of Science and Technology,
Ajman, United Arab Emirates
3
Department of Pharmacology and Toxicology, Faculty of Pharmacy,
Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
4
Department of Pharmacology and Toxicology, Faculty of Pharmacy,
Al-Azhar University, Cairo, Egypt
5
Department of Pharmacology, College of pharmacy, Al Jouf
University, Al Jouf, Kingdom of Saudi Arabia
6
Department of Pharmacology, Faculty of Veterinary medicine,
Beni-Suef University, Beni-Suef 62514, Egypt
Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976
DOI 10.1007/s00210-016-1306-7
containing protein which binds to eNOS and strongly attenu-
ated eNOS-dependent NO production (Zimmermann et al.
2002). Nostrin binds to eNOS causing decrease of eNOS en-
zyme activity. Moreover, caveolin and nostrin can augment
the binding of each others to distinct sites on eNOS
(Schilling et al. 2006). Nostrin may work as a scaffold protein
that recruits eNOS, dynamin-2, and Wiskott-Aldrich syn-
drome protein type N (N-WASP) and controls their internali-
zation via caveolar endocytosis (Zimmermann et al. 2002).
Other various mechanisms had been implicated in liver IR
injury such as abundant amounts of reactive oxygen species
(ROS) (Jaeschke 2003), activation of Kupffer cells, and in-
flammatory cytokines release (Montalvo-Jave et al. 2008).
However, the mechanisms that coordinate and integrate this
pathologic response are still unclear. It was evident that oxi-
dative stress plays a major role in tissue injury including IR
(Zimmermann BJ and Granger DN 1994). Thymoquinone
(TQ) is the main constituent of Nigella sativa, with a rich
background in history and religion, used to promote health
and for the treatment of different diseases (El-Ghany et al.
2009). Recent research revealed that TQ possesses a strong
antioxidantproperty(Gali-Muhtasibetal.2006), neuroprotec-
tive effects against transient forebrain ischemia (Al-Majed
et al. 2006), cardioprotective effects against oxidative damage
induced by doxorubicin (NAGI and MANSOUR 2000)and
gastroprotective effects against IR injury (El-Abhar et al.
2003). Moreover, it exerts antitumor, immunomodulatory,
and anti-inflammatory effects (Gali-Muhtasib et al. 2006).
N-acetylcysteine (NAC) is a thiol-containing compound. It
is extensively used in the management of paracetamol over-
dose induced fulminant liver failure (Prescott et al. 1979)and
as a hepatoprotective agent in liver transplantation (Thies et al.
1998). Furthermore, it has a therapeutic value for reducing
endothelial dysfunction, inflammation, and fibrosis
(Zafarullah et al. 2003). Finally, the use of antioxidants for
liver IR is still ongoing looking for any beneficial effects
and their underlying mechanisms. The aim of our study is to
investigate the possible hepatoprotective effect of TQ in IR-
induced liver injury through its endothelial NO-regulatory,
anti-inflammatory, and/or antioxidant activity.
Materials and methods
Chemicals
Thymoquinone (TQ), N-acetylcysteine (NAC), 4,6-diamidino-
2-phenylindole (DAPI), O-dianisidine, thiobarbituric acid, sul-
fanilamide, and 55- dithiobis-(2-nitrobenzoic acid)(DTNB)
were purchased from Sigma-Aldrich, USA. Mouse polyclonal
anti-inducible nitric oxide synthase (anti-iNOS) and mouse
polyclonal anti-endothelial nitric oxide synthase (eNOS) were
purchased from BD-Biosciences, USA. Rabbit polyclonal anti-
eNOS trafficking inducer (anti-NOSTRIN, 506 aa and 58 kDa
variant) was purchased from Proteintech, USA. Rabbit anti-
mouse Alexa Fluor 488 secondary antibody was purchased
from Invitrogen, USA. Goat anti-rabbit Cy3 secondary antibody
was purchased from Jackson ImmunoResearch Laboratories,
USA. Aspartate aminotransferase (AST) reagent kit and
Alanine aminotransferase (ALT) reagent kit were purchased
from Spinreact, Spain. The highest analytical grades were se-
lected for other chemicals.
Animals
Adult male Wister albino rats, weighing 300350 g, were
randomly selected. The protocol of this study has been ap-
proved by Research Ethics Committee of Faculty of
Pharmacy, Beni-Suef University, Egypt. Rats were housed in
metabolic cages under controlled environmental conditions
(25 °C and a 12 h light/dark cycle), allowed free access to
pulverized standard food pellet and tap water ad libitum and
animals were left for 2 weeks of acclimation to animal house
conditions.
Experimental protocol
Rats were randomly divided into four groups (10 rats each):
group I: sham-operated control group (received normal saline
and 1 % Tween 80 orally) and underwent a surgical procedure
similar to the other groups but the entire hepatic pedicule was
not occluded, group II: IR-induced injury group (received
normal saline and 1 % Tween 80 orally) and subjected to
30-min ischemia followed by 30-min reperfusion (Oguz
et al. 2013), group III: rats were pretreated with NAC freshly
dissolved in normal saline (300 mg/kg/day) (Hemalatha et al.
2013) orally for 10 days before subjection to IR, and group IV:
rats were pretreated with TQ suspended in normal saline and
1 % tween 80 (20 mg/kg/day) (El-Ghanyet al. 2009) orally for
10 days before subjection to IR.
Induction of hepatic ischemia-reperfusion
A complete midline laparotomy was performed under intra-
peritoneal injection of ketamine (50 mg/kg) and xylazine HCl
(10 mg/kg) anesthesia (Ara et al. 2005). Surgical induction of
hepatic ischemia was performed by occlusion of hepatic
pedicule (hepatic artery, portal vein, and bile duct), which
supplied the left and medial lobes, with a mini-clamp for
30 min. The vascular clamp was removed and the liver was
perfused for 30 min followed by collection of blood samples
from the retro-orbital plexus for the assay of serum ALT and
AST, then the rats were sacrificed and the liver was quickly
removed, rinsed in ice cold saline. Some portions fixed in
Davidson-Solution for immunofluorescence assessment; oth-
er portions fixed in 10 % formalin saline for histopathological
70 Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976
examination or were weighed and homogenized in normal
saline, 25 % w/v. Homogenates were then centrifuged for
15 min at 10,000×gand the supernatant was used for deter-
mination of other biochemical parameters.
Estimation of liver functions
Measurement of serum aspartate aminotransferase (AST) and
alanine aminotransferase (ALT) was determined by the meth-
od described by manufacturers instructions using activity col-
orimetric assay kits (Spin react, Spain). The absorbance was
measured spectrophotometrically at 340 nm. The activities
were expressed in U/L.
Estimation of malondialdehyde and reduced glutathione
contents
Malondialdehyde (MDA) was measured by the method of
Mihara and Uchiyama (1978). 1,1,3,3-tetramethoxypropane
used to construct the standard curve. MDA content was cal-
culated and expressed in nmol/g tissue. The difference in ab-
sorbance measured spectrophotometrically at 535 and 520 nm
was used to calculate the content of MDA in each sample.
Glutathione (GSH) content was measured according to the
method described by Beutler et al. (1963). The resulting yel-
low color absorbance was measured spectrophotometrically
within 5 min at 412 nm. GSH content was expressed in mg/
gtissue.
Estimation of myeloperoxidase activity and nitric oxide
production
Myeloperoxidase (MPO) activity was determined in tissue by
the method of Krawisz et al. (1984). The absorbance was
measured spectrophotometrically at 460 nm. MPO activity
was calculated according to the following equation: MPO ac-
tivity (U/g tissue) = ΔA/min × D. F / extinction coefficient,
where dilution factor = reciprocal of the relative sample vol-
ume in the reaction mixture × dilution factor of the homoge-
nate × 1000. Extinction coefficient = 1.13 × 104 Cm
1
M
1
.
Nitric oxide was measured in tissue according to the method
described by Miranda et al. (2001). Nitric oxide (NO) produc-
tion was calculated according to the standard curve using
NaNO
3
as a standard and expressed in μM/g tissue. The ab-
sorbance was measured spectrophotometrically at 540 nm.
Immunofluorescence assessment of iNOS, eNOS,
and NOSTRIN expressions
Paraffin embedded tissue sections (4 μm thin) were put in oven at
60 °C for 20 min, then deparaffinized in xylene 100 % two times,
15 min each to dissolve wax. A rehydration step was performed
by using graded ethanol 100 % for 5 min, 90, 70, 50, and 30 %,
5 min each. After rehydration, tissue was washed with distilled
water for 5 min and tissue section was incubated in Dako solution
(citrate buffer, pH 6) in microwave at 500 watt for 20 min. After
cooling of the solution, slides were washed three times by PBST
(0.05 % of tween 20 in phosphate buffer saline (PBS) pH 7.4),
3 min each. Fixation was done using methanol for 30 min. After
washing, slides were blocked by 10 % horse serum in 1 % bovine
serum albumin of PBS blocking solution to block the non-specific
binding of antibodies. Tissue sections were incubated with the
primary antibodies (mouse polyclonal iNOS or eNOS or rabbit
polyclonal NOSTRIN antibodies) in the concentration of 1:50 in
cool place overnight. Bound antibodies were detected by rabbit
anti-mouse Alexa fluor 488 (green) or goat anti-rabbit Cy3 (red)
secondary antibodies for 30 min, respectively. Nuclei were
stained with DAPI and washed by PBST for 30 min. Slides were
mounted with Fluoromount and evaluated by fluorescence mi-
croscopy (Leica DM 5000B).
Histopathological examination
The liver of the animals was removed and fixed in 10 % formalin,
dehydrated with graded ethanol (30, 50, 70, 90 and 100 %), and
cleared the in two changes of xylene. Samples were then steeped
with two times in molten paraffin wax. Paraffin sections (5 μm)
thick were stained with hematoxylin and eosin (H&E) according to
Jones et al. (2008). Stained tissues of sham-operated control and
treated rats were analyzed for alterations in the architecture,inflam-
matory infiltration, congestion of the central vein, pyknosis, and
cytoplasmic degeneration. The zero score was given to normal
tissues with no alteration, + indicates slight histopathological alter-
ations, ++ indicates moderate histopathological alterations, +++
referred to severe histopathological alterations while ++++ direct-
ed to very severe histopathological alterations. All experiments
were conducted in accordance with the guidelines approved by
the University of Beni-Suef Ethical Committee.
Statistical analysis
Statistical analysis was performed using SPSS statistical soft-
ware (version 22.0). Data were expressed as means ± standard
error (S.E.). Statistical evaluation of data was performed using
one-way analysis of variances (ANOVA), with Tukey post
hoc test to compare the means. A P < 0.05 was considered
statistically significant.
Results
Effect of TQ pretreatment on liver enzymes in IR operated
rats
The activities of liver toxicity marker enzymes, AST and ALT,
are represented in Table 1. IR group exhibited significant
Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976 71
elevation in serum activities of both enzymes to
914.16 ± 46.88 and 846.46 ± 91.92 %, respectively, compared
to the corresponding sham-operated control group. TQ pre-
treatment significantly decreased the elevated liver enzymes
to 52.31 ± 2.39 and 71.72 ± 1.03 %, respectively, similarly IR
operated rats pretreated with NAC also showed lowered AST
and ALT activities to 66.04 ± 2.42 and 67.10 ± 5.22 %, re-
spectively, compared to IR group.
Effect of TQ pretreatment on hepatic lipid peroxidation
and GSH contents in IR operated rats
Climbing to approximately 240.10 ± 24.51 % in IR operated
rats compared to sham-operated rats, Hepatic MDA content
significantly decreased to 79.70 ± 9.82 and 66.76 ± 3.78 % in
NAC and TQ-pretreated rats respectively compared to IR
group. In contrast, from a depletion of 20.24 ± 0.96 % in IR
group compared to sham-operated group, the reduced GSH
content significantly increased to 476.47 ± 23.53 and
388.23 ± 41.18 % in NAC and TQ-pretreated group respec-
tively compared to IR group (Table 1).
Effect of TQ pretreatment on hepatic MPO activity
and NO production in IR operated rats
IR group showed a significant increase in MPO activity and NO
production to 617.65 ± 70.59 and 254.79 ± 6.99 % respectively
compared to sham-operated group. NAC-pretreated rats sharply
declined to 43.81 ± 5.71 and 78.98 ± 4.03 % respectively com-
pared to IR group. Likewise, TQ-pretreated group dramatically
decreased these values to 25.71 ± 1.90 and 69.83 ± 4.28 %,
respectively (Table 1).
Effect of TQ pretreatment on protein expressions of iNOS,
eNOS, and NOSTRIN in IR operated rats
Negative iNOS and NOSTRIN protein expressions were seen in
sham-operated rats. On the other hand, livers of rats subjected to
IR revealed diffused up-regulation of iNOS and NOSTRIN
expression in the pericentral hepatocytes compared to sham-
operated rats. While TQ or NAC-pretreated groups exhibited
decreased expression of iNOS and NOSTRIN in the sinusoidal
areas closed to the central area compared to the IR group. Semi-
quantitative analysis of the immunofluorescence staining
expressed as fluorescence intensity demonstrated that IR oper-
ated group showed a significant increase in the hepatic iNOS
and NOSTRIN expression to approximately 153.10 ± 9.68 and
197.14 ± 6.50 %, respectively, compared to sham-operated rat
values. Conversely, the expression of iNOS and NOSTRIN in
the hepatic tissues of IR pretreated with TQ or NAC significant-
ly decreased to 69.87 ± 2.82 and 53.64 ± 3.84 % or 72.61 ± 3.13
and 55.07 ± 3.18 % respectively compared to IR group (Figs. 1
and 2, respectively).
Sinusoidal eNOS protein expression was seen in sham-
operated rats while liver sections from animals exposed to
IR revealed strong down-regulation of eNOS expression com-
pared to sham-operated rats. A dramatic rise in eNOS expres-
sion was seen in the sinusoidal areas as well as in periportal
and pericentral hepatocytes in TQ and NAC-pretreated IR-
exposed animals compared to the IR group. Fluorescence in-
tensity of eNOS revealed that IR processed group showed a
significant reduction in the hepatic eNOS protein expression
to 51.75 ± 2.29 % compared to sham-operated rats
(26.60 ± 1.18 %). Conversely, the expression of eNOS in
the hepatic tissues of IR pretreated with TQ or NAC group
significantly up-regulated to 174.70 ± 9.10 and
153.27 ± 7.48 %, respectively (Fig. 3).
Histopathological examination of liver sections
TheeffectsofTQorNACinIRoperatedratsonliver
histopathological features are illustrated in Table 2and
presented in Fig. 4. The sham-operated group revealed normal
hepatic architecture with central vein (CV) and radiating cords
of normal hepatocytes with central rounded vesicular nuclei
and prominent nucleoli. Hepatic cords are separated by blood
sinusoids (interstitial spaces) lined with endothelium and
Kupffer cells (KC). In contrast, liver sections of rats subjected
Tabl e 1 Effect of TQ pretreatment on liver enzymes and oxidative stress parameters in IR operated rats
Parameters Groups Sham IR NAC + IR TQ + IR
AST (U/L) 143.00 ± 3.05 1307.25 ± 67.04
*
863.33 ± 29.24
*@
682.33 ± 31.15
*@
ALT (U/L) 127.00 ± 10.15 1075.00 ± 116.74
*
721.33 ± 56.17
*@
771.00 ± 11.06
*@
MDA (nmol/g tissue) 17.73 ± 1.81 42.57 ± 3.60
*
33.93 ± 4.18
*@
28.42 ± 1.61
*@
GSH (mg/g tissue) 0.84 ± 0.04 0.17 ± 0.03
*
0.81 ± 0.04
@
0.66 ± 0.07
*@
MPO (U/g tissue) 0.17 ± 0.01 1.05 ± 0.12
*
0.46 ± 0.06
*@
0.27 ± 0.02
@
NO (μM/g tissue) 583.00 ± 38.31 1485.42 ± 40.74
*
1173.19 ± 59.91
*@
1037.21 ± 63.59
*@
All data were expressed as means ± standard error (S.E.) of six rats per group
*Significantly different from sham-operated group at P < 0.05
@
Significantly different from IR group at P < 0.05
72 Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976
to IR injury showed massively congested CV and dilated in-
terstitial spaces lined with activated KC, in addition many
hepatocytes showed pyknotic nuclei, cytoplasmic degenera-
tion, and inflammatory cell infiltration. On the other hand,
pretreatment with NAC or TQ resulted in a marked attenua-
tion of liver injury, as the majority of hepatic lobules revealed
normal liver picture while the former was associated with a
slight congestion of CV and interstitial spaces and the latter
was accompanied by just a moderate congestion of CV and a
slight dilation of blood sinusoids.
Discussion
Liver injury induced by IR remains an important clinical prob-
lem during liver transplantation, hemorrhagic shock, and
surgical procedures. The proposed mechanisms in the patho-
physiology of IR injury included massive production of ROS,
NO, and their reaction product of peroxynitrite (Montalvo-
Jave et al. 2008). Moreover, leukocytic infiltration induced
an inflammatory reaction proceeding oxidative stress upon
reperfusion (Sewerynek et al. 1995). It is apparent from the
present study that IR-induced liver injury is confirmed bio-
chemicallyby a considerable elevation in ALT, AST and MPO
activities as well as MDA level while reduced GSH content is
significantly depleted. Also, IR caused a marked histopatho-
logical alteration including pyknosis, degeneration of cyto-
plasm, congestion of CV, and PMNs infiltration in hepato-
cytes. Further and even more importantly, though, it signifi-
cantly increased liver iNOS expression along with NO over-
production. Meanwhile, it obviously increased NOSTRIN ex-
pression in the liver tissues while it was associated with a
Sham IR
NAC+IR TQ+IR
A
B
Fig. 1 Effect of TQ pretreatment on iNOS localization and expression in
hepatic tissues of IR operated rats. aImmunofluorescence staining of
liver sections obtained from control rats, showing negative expression
of iNOS while IR markedly increased expression of iNOS. TQ or NAC
pretreatment reduced the expression level of iNOS. Scale bar = 100 μm. b
Quantitative image analysis of immunofluorescence staining expressed as
fluorescence intensity of iNOS expression of sham-operated control and
IR with or without NAC or TQ pretreatment obtained from eight fields
from each rats section (minimally three rats in each group) using ImageJ
software (National Institutes of Health, Bethesda, MD). *Significantly
different from sham-operated group at P < 0.05.
@
Significantly different
from IR group at P<0.05
Sham IR
TQ+IR
NAC+IR
A
B
Fig. 2 Effect of TQ Pretreatment on NOSTRIN Localization and
Expression in Hepatic Tissues of IR Operated Rats. aControl rats,
revealing negligible expression of NOSTRIN while its expression in IR
subjected animals is concentrated in the nuclei of the hepatocytes as the
red fluorescence is co-localized with DAPI stain of the nuclei (arrows).
TQ or NAC pretreatment markedly reduced NOSTRIN expression in the
sinusoidal areas closed to the central area. Scale bar = 50 μm. b
Fluorescence intensity of NOSTRIN expression of sham-operated control
and IR with or without NAC or TQ pretreatment obtained from eight
fields from each rats section (minimally three rats in each group) using
ImageJ software (National Institutes of Health, Bethesda, MD).
*Significantly different from sham-operated group at P < 0.05.
@
Significantly different from IR group at P < 0.05
Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976 73
discernible decline in liver eNOS expression. These findings
are in harmony with our recent study (Abd-Elbaset et al.
2015). In the current study, pretreatment with TQ significantly
alleviated biochemical and histopathological status of the IR-
induced liver damage through a significant decline in ALT and
AST levels and restoration of normal hepatocyte architecture.
In addition, it significantly mitigated inflammatory infiltration
in the rat liver induced by IR as manifested by the reduced
hepatic MPO activity. It has been shown earlier that Nigella
sativa significantly lowers ALT, AST, and MPO activities, in
addition it decreases pathological changes in rats subjected to
IR-induced liver damage (Yildiz et al. 2008). TQ, meanwhile,
significantly decreased lipid peroxidation and enhanced the
antioxidant capacity by restoring reduced GSH content.
These results are comparable with a previous study
demonstrating that TQ pretreatment restored GSH depletion
and decreased MDA elevation in the rat hippocampus subject-
ed to transient forebrain ischemia (Al-Majed et al. 2006). An
interesting finding of our study is the suppression of iNOS
expression and reduction of NO overproduction, hence its
byproduct, peroxynitrite, which were deleterious and might
initiate hepatic inflammatory reaction. El-Mahmoudy et al.
(2002) stated that TQ decreased immunoreactivity of iNOS
in peritoneal macrophages and decreased nitrite production, a
stable product of NO parameter, induced by lipopolysaccha-
ride (LPS). We also found that TQ pretreatment increases the
expression of eNOS in the endothelial cells of hepatic blood
vessels compared to IR alone as revealed by immunofluores-
cence. This may explain the beneficial role of TQ during IR-
induced liver injury. TQ caused a marked increase in eNOS
activity in isolated rabbit aorta subjected to endothelial dys-
function induced by pyrogallol (El-Agamy and Nader 2012).
Moreover, the co-localization of NOSTRIN with DAPI indi-
cated nuclear translocation of NOSTRIN in hepatocytes of
rats subjected to IR alone while TQ decreased its nuclear
expression, indicating no more eNOS translocation from the
plasma membrane to vesicle-like subcellular structures and
leading to augmentation of eNOS-dependent NO production.
Mookerjee et al. (2007) demonstrated that increased
NOSTRIN may partly explain the decreased enzymatic activ-
ity of eNOS in diseased liver. Furthermore, increased gene and
protein expression of NOSTRIN has been observed in liver
samples of alcoholic hepatitis patients, which may contribute
to the reduced eNOS activity and increased intrahepatic resis-
tance in these chronically cirrhotic livers. In our study, we
reported an obvious increase of NO level in IR operated rats.
Additionally, it has been reported that NOSTRIN poses a sig-
nificant inhibition of NO release by direct inhibition of eNOS
activity/modulation of regulatory mechanism such as phos-
phorylation or protein association (Icking et al. 2005). The
increased NOSTRIN protein expression and NO production
along with the reduced eNOS expression in the current work
may be explained by the inhibitory effect of NOSTRIN me-
diating NO release which is mostly due to its influence on
eNOS cellular trafficking. These findings displayed that NO
could be good or bad according to NOS isoforms and the
stimuli triggering its production (Thippeswamy et al. 2006).
Administration of NAC prior to IR significantly decreased
serum AST and ALT activities, MDA level, and MPO activity.
Moreover, it strongly restored GSH content. In consistentwith
our results, Sener et al. (2003) stated that NAC alone or in
combination with melatonin was effective in bringing signif-
icant reduction in AST, ALT, MDA, and MPO as well as
significantly restoring GSH content in a model of hepatic
IR. In addition, histopathological examination of NAC pre-
treatment showed a marked improvement in the liver cell
structure. Similar result has been reported by Attri et al.
(2000) who reported that NAC faithfully maintained normal
Sham IR
NAC+IR TQ+IR
A
B
Fig. 3 Effect of TQ pretreatment with on eNOS localization and
expression in hepatic tissues of IR operated rats. aControl rats,
showing constitutive expression of eNOS and it is highly concentrated
in hepatocytes around the central vein. The expression is obviously
downregulated in the pericentral hepatocytes in IR subjected animals. In
TQ + IR and NAC + IR groups, the expression is concentrated in the
sinusoidal areas as well as in periportal and pericentral hepatocytes as the
green fluorescence is located in the blood vessels endothelial cells. Scale
bar = 100 μm. bFluorescence intensity of eNOS expression of sham-
operated control and IR with or without NAC or TQ pretreatment
obtained from eight fields from each rats section (minimally three rats
in each group) using ImageJ software (National Institutes of Health,
Bethesda, MD). *Significantly different from sham-operated group at
P<0.05.
@
Significantly different from IR group at P < 0.05
74 Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976
morphology in a model of isoniazid and rifampicin induced
hepatic injury. Furthermore, NAC decreased iNOS protein
expression along with reducing NO production. The same
observation was reported by Bergamini et al. (2001)who
demonstrated that NAC administration inhibited iNOS ex-
pression and the massive NO production induced by LPS.
Controversially, NAC restored eNOS protein expression, the
finding which was in harmony with a study demonstrating that
cultured vascular endothelial cell (ECV304) treated withNAC
increased eNOS expression along with NO production
resulting in improved cell viability and reduced apoptosis in-
duced by TNF-α(Xia et al. 2006). Thus, there is no significant
difference between pretreatment with TQ or NAC, which was
used as standard treatment in our study. In conclusion, our
findings highlight the ability of TQ to ameliorate the oxidative
stress, nitrosative stress, and inflammatory responses second-
ary to IR. Therefore, using TQ as a potential candidate as
supplement or adjunct therapy in patients subjected to liver
transplantation or surgery is strongly recommended.
References
Abd-Elbaset M, Arafa E-SA, El Sherbiny GA, Abdel-Bakky MS,
Elgendy ANA (2015) Quercetin modulates iNOS, eNOS and
NOSTRIN expressions and attenuates oxidative stress in warm he-
patic ischemia-reperfusion injury in rats Beni-Suef University.
Journal of Basic and Applied Sciences 4:246255
Al-Majed AA, Al-Omar FA, Nagi MN (2006) Neuroprotective effects of
thymoquinone against transient forebrain ischemia in the rat hippo-
campus. Eur J Pharmacol 543:4047
Ara C et al (2005) Protective effect of resveratrol against oxidative stress in
cholestasis. J Surg Res 127:112117. doi:10.1016/j.jss.2005.01.024
Attri S, Rana SV, Vaiphei K, Sodhi CP, Katyal R, Goel RC, Nain CK, Singh
K (2000) Isoniazid- and rifampicin-induced oxidative hepatic injury
protection by N-acetylcysteine. Hum Exp Toxicol 19(9):517522
Bergamini S, Rota C, Canali R, Staffieri M, Daneri F, Bini A, Giovannini
F, Tomasi A, Iannone A (2001) N-acetylcysteine inhibits in vivo
nitric oxide production by inducible nitric oxide synthase. Nitric
Oxide 5(4):34960
Beutler E, Duron O, Kelly B (1963) Improved method for the determi-
nation of blood glutathione. J Lab Clin Med 61:882888
El-Abhar H, Abdallah D, Saleh S (2003) Gastroprotective activity
of < i > Nigella sativa</i > oil and its constituent, thymoquinone,
against gastric mucosal injury induced by ischaemia/reperfusion in
rats. J Ethnopharmacol 84:251258
El-Agamy DS, Nader MA (2012) Attenuation of oxidative stress-induced
vascular endothelial dysfunction by thymoquinone. Exp Biol Med
237:10321038
Tabl e 2 Histopathological
examination of the effect of TQ
pretreatment in IR operated rats
Findings Groups Sham IR NAC + IR TQ + IR
Inflammatory infiltration 0 ++++ 0 0
Congestion of CV 0 ++++ + ++
Sinusoidal (interstitial) dilation 0 +++ + +
Pyknosis 0 ++++ 0 0
Cytoplasmic degeneration 0 +++ + +
IR was performed by (30-min ischemia + 30-min reperfusion). NAC (300 mg/kg, orally) or TQ (20 mg/kg, orally)
was given once daily for ten consecutive days followed by performing IR
0Non histopathological alterations
+Slight histopathological alterations
++Moderate histopathological alterations
+++Severe histopathological alterations
++++Very severe histopathological alterations
A
B-1 B-2
C D
Fig. 4 Histopathological examination of liver sections. ×400, H&E stain.
aPhotomicrograph of sham group shows normal hepatic architecture.b
Photomicrograph of I R operated rats (B-1) shows very severely congested
central vein, hepatocytes (H) are separated by dilated blood sinusoids (S)
with aggregated kupffer cells (white arrow) and extensive inflammatory
infiltration (encircled). An additional photomicrograph of IR group (B-2)
shows pyknotic nuclei and cytoplasmic degeneration (encircled). c
Photomicrograph of NAC-pretreated group shows normal hepatic
architecture with slightly congested central vein and slightly dilated
blood sinusoids. dPhotomicrograph of TQ-pretreated group shows
normal hepatic architecture with moderately congested central vein and
slightly dilated blood sinusoids
Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976 75
El-Ghany R, Sharaf N, Kassem L, Mahran L, Heikal O (2009)
Thymoquinone triggers anti-apoptotic signaling targeting death li-
gand and apoptotic regulators in a model of hepatic ischemia reper-
fusion injury. Drug discoveries & therapeutics 3:296306
El-Mahmoudy A, Matsuyama H, Borgan MA, Shimizu Y, El-Sayed MG,
Minamoto N, Takewaki T (2002) Thymoquinone suppresses
expression of inducible nitric oxide synthase in rat macrophages.
Int Immunopharmacol 2:16031611
Gali-Muhtasib H, Roessner A, Schneider-Stock R (2006) Thymoquinone: a
promising anti-cancer drug from natural sources. Int J Biochem Cell Biol
38:12491253
Hemalatha P, Reddy AG, Reddy YR, Shivakumar P (2013) Evaluation of
protective effect of N-acetyl cysteine on arsenic-induced hepatotoxicity.
JNatSciBiolMed4:393395. doi:10.4103/0976-9668.116986
Icking A, Matt S, Opitz N, Wiesenthal A, Müller-Esterl W, Schilling K
(2005) NOSTRIN functions as a homotrimeric adaptor protein fa-
cilitating internalization of eNOS. J Cell Sci 118:50595069
Jaeschke H (2003) Molecular mechanisms of hepatic ischemia-
reperfusion injury and preconditioning. Am J Physiol Gastrointest
Liver Physiol 284:G15G26. doi:10.1152/ajpgi.00342.2002
Jones LM, Bancroft JD, Gamble M (2008) Theory and practice of histo-
logical techniques. 6th ed. London: Churchill Livingstone, p 744
Krawisz JE, Sharon P, Stenson WF (1984) Quantitative assay for acute
intestinal inflammation based on myeloperoxidase activity.
Assessment of inflammation in rat and hamster models
Gastroenterology 87(6):1344-50
Mihara M, Uchiyama M (1978) Determination of malonaldehyde precur-
sor in tissues by thiobarbituric acid test. Anal Biochem 86(1):271-8
Miranda KM, Espey MG, Wink DA (2001) A rapid, simple spectropho-
tometric method for simultaneous detection of nitrate and nitrite.
Nitric Oxide 5(1):62-71
Montalvo-Jave EE, Escalante-Tattersfield T, Ortega-Salgado JA, Piña E,
Geller DA (2008) Factors in the pathophysiology of the liver
ischemia-reperfusion injury. J Surg Res 147:153159
Mookerjee RP, Wiesenthal A, Icking A, Hodges SJ, Davies NA, Schilling
K, Sen S, Williams R, Novelli M, ller-Esterl W, Jalan R (2007)
Increased gene and protein expression of the novel eNOS regulatory
protein NOSTRIN and a variant in alcoholic hepatitis.
Gastroenterology 132(7):2533-2541
NAGI MN, MANSOUR MA (2000) Protective effect of thymoquinone
against doxorubicininduced cardiotoxicity in rats: a possible mech-
anism of protection. Pharmacol Res 41:283289
Oguz A et al (2013) The effects of curcumin on the liver and remote
organs after hepatic ischemia reperfusion injury formed with
Pringle manoeuvre in rats. Eur Rev Med Pharmacol Sci 17:457466
Prescott L, Illingworth R, Critchley J, Stewart M, Adam R, Proudfoot A
(1979) Intravenous N-acetylcystine: the treatment of choice for para-
cetamol poisoning. BMJ 2:10971100
Schilling K, Opitz N, Wiesenthal A, Oess S, Tikkanen R, Muller-Esterl
W, Icking A (2006) Translocation of endothelial nitric-oxide syn-
thase involves a ternary complex with caveolin-1 and NOSTRIN.
MolBiolCell17:38703880. doi:10.1091/mbc.E05-08-0709
Sener G, Tosun O, Sehirli AO, Kaçmaz A, Arbak S, Ersoy Y, Ayanoğlu-
Dülger G (2003) Melatonin and N-acetylcysteine have beneficial
effects during hepatic ischemia and reperfusion. Life Sci 72(24):
2707-2718
Sewerynek E, Reiter RJ, Melchiorri D, Ortiz GG, Lewinski A (1995)
Oxidative damage in the liver induced by ischemia-reperfusion: protec-
tion by melatonin. Hepato-Gastroenterology 43:898905
Shah V, Kamath PS (2003) Nitric oxide in liver transplantation: pathobi-
ology and clinical implications. Liver Transpl 9:111
Thies J et al (1998) The efficacy of N-acetylcysteine as a hepatoprotective
agent in liver transplantation. Transpl Int 11:S390S392
Thippeswamy T, McKay JS, Quinn JP, Morris R (2006) Nitric oxide, a
biological double-faced janusis this good or bad? Histol
Histopathol 21(4):445458
Trocha M, Merwid-Lad A, Szuba A, Chlebda E, Piesniewska M,
Sozanski T, Szelag A (2010) Effect of simvastatin on nitric oxide
synthases (eNOS, iNOS) and arginine and its derivatives (ADMA,
SDMA) in ischemia/reperfusion injury in rat liver. Pharmacological
reports : PR 62:343351
Xia Z, Liu M, Wu Y, Sharma V, Luo T, Ouyang J, McNeill JH (2006) N-
acetylcysteine attenuates TNF-α-induced human vascular endothe-
lial cell apoptosis and restores eNOS expression. Eur J Pharmacol
550:134142
Yildiz F et al (2008) Nigella sativa relieves the deleterious effects of
ischemia reperfusion injury on liver. World journal of gastroenterol-
ogy: WJG 14:5204
Zafarullah M, Li WQ, Sylvester J, Ahmad M (2003) Molecular
mechanisms of N-acetylcysteine actions. Cell Mol Life Sci
60:620
Zimmerman BJ, Granger DN (1994) Mechanisms of reperfusion injury.
Am J Med Sci 307(4):284-292
Zimmermann K, Opitz N, Dedio J, Renne C, Muller-Esterl W,
Oess S (2002) NOSTRIN: a protein modulating nitric oxide
release and subcellular distribution of endothelial nitric ox-
ide synthase. Proc Natl Acad Sci U S A 99:1716717172.
doi:10.1073/pnas.252345399
76 Naunyn-Schmiedeberg's Arch Pharmacol (2017) 390:6976
... In addition, overexpression of endothelial nitric oxide synthase (eNOS) and down-regulation of iNOS occurred after thymoquinone administration. The results suggested that the hepatoprotective effects of thymoquinone were probably mediated via regulating the NO signaling pathway and improving oxidative stress [41]. The liver plays a key role in lipid homeostasis. ...
... Crocin (20 mg/kg for 30 d) has been found to decrease the bisphenol A-triggered liver toxicity in rats. The ameliorative effect of crocin against liver damage was mediated by the reduction of TG and liver enzymes, inhibition of oxidative stress, decrease of miR-122 expression, and up-regulation of JNK, extracellular signal-regulated kinase 1/2 (ERK1/2), and [41] Alcohol-caused injury Mouse 20 and 40 mg/kg Increase of LKB1, AMPK, and PPARs activity and up-regulation of SIRT1 [47] Streptozocin-induced diabetes Rat 20 mg/kg Improvement of hepatic injury through alleviating streptozocin-induced diabetes [50] Acetaminophen-induced damage Mouse 20 mg/kg Modulation of JNK and AMPK signaling and inhibition of CYP2E1 [54] High-fat high-cholesterol diet-induced damage Rat 10 and 20 mg/kg Amelioration of insulin resistance and blood glucose, reduction of total cholesterol and triglycerides, and elevation of HDL [56] Thioacetamide-induced damage Rat 20 mg/kg Down-regulation of Bcl-2, Bcl-XL, and TGF-β1 expression and up-regulation of TRAIL-linked apoptosis [61] IR: ischemia-reperfusion, NO: Nitric oxide, LKB1: Liver kinase B1, AMPK: AMP-activated protein kinase, PPARs: Peroxisome proliferator-activated receptors, SIRT1: Sirtuin 1, JNK: c-Jun N-terminal kinase, CYP2E1: Cytochrome 2E1, HDL: High-density lipoprotein, TGF: Transforming growth factor, TRAIL: Tumor necrosis factor-related apoptosis-inducing ligand. mitogen-activated protein kinase (MAPK) [74]. ...
Article
Full-text available
Medicinal plants are rich in nutrients and phytochemicals which prevent and treat a wide range of ailments. Accumulating experimental studies exhibit that some bioactive ingredients extracted from medicinal plants have suitable therapeutic effects on hepatic and renal injuries. This review focuses on the hepato- and reno-protective effects of thymoquinone, crocin, and carvacrol. The relevant literature was retrieved from PubMed, Scopus, Web of Science, and Google Scholar databases from the beginning of 2015 until the end of November 2021. According to the scientific evidence, the considered phytochemicals in this review have been applied with useful therapeutic effects on hepatic and renal damage. These therapeutic effects were mainly mediated through the amelioration of oxidative stress, suppression of inflammatory responses, and inhibition of apoptosis. Intracellular signaling pathways linked to nuclear factor kappa B (NF-κB), adenosine monophosphate-activated protein kinase, c-jun N-terminal kinase, and extracellular signal-regulated kinase 1/2 and Toll-like receptors are the most important pathways targeted by these phytochemicals. Up-regulation of transcription factor Nrf2 and down-regulation of transforming growth factor-beta 1 by these natural compounds also contribute to the alleviation of hepatic and renal injuries.
... The antioxidant property is the main effect of TQ. Previous studies attributed the capability of TQ to protect against several conditions such as hepatic cancer and ischemiainduced liver injury to oxidative stress suppression as TQ could decrease NO synthesis through direct suppression of nitric oxide synthases (iNOS and eNOS) besides TQ's ability to normalize the depleted GSH level and antioxidant enzymes [53]. Moreover, previous studies confirm the potency of TQ to protect against oxidative stress induced by several hepatotoxic agents such as CB 1954 [54], cisplatin [55], cyclophosphamide [56], Aflatoxins [57], and carbon tetrachloride [58] through decreasing lipid peroxidation and NO synthesis along with increased expression of antioxidant enzymes. ...
Article
Full-text available
Silver nanoparticles (AgNPs) are the most common nanomaterials in consumer products. Therefore, it has been crucial to control AgNPs toxicological effects to improve their safety and increase the outcome of their applications. This work investigated the possible protective effect of thymoquinone (TQ) against AgNPs-induced hepatic and renal cytotoxicity in rats. Serum markers of liver and kidney functions as well as liver and kidney oxidative stress status, pro-inflammatory cytokines, apoptosis markers, and histopathology were assessed. TQ reversed AgNPs-induced elevation in serum liver and kidney function markers, including aspartate transaminase, alanine transaminase, urea, and creatinine. Moreover, TQ co-administration with AgNPs alleviates hepatic and renal oxidative insults by decreasing MDA and NO levels with a significant increase in the activity of antioxidant enzymes (superoxide dismutase, catalase, and glutathione recycling enzymes peroxidase and reductase) compared to AgNPs-treated rats. Besides, TQ upregulated hepatic and renal Nrf2 gene expression in AgNPs-intoxicated rats. Furthermore, TQ co-administration decreased the hepatic and renal pro-inflammatory mediators represented by IL-1β, TNF-α, TGF-β, and NF-κB levels. Besides, TQ co-administration decreased apoptotic protein (Bax) levels and increased the anti-apoptotic protein (Bcl-2) levels. These findings were confirmed by the histopathological examination of hepatic and renal tissues. Our data affirmed the protective effect of TQ against AgNPs cytotoxicity and proposed a possible mechanism of TQ antioxidant, anti-inflammatory, and anti-apoptotic effects. Consequently, we could conclude that using TQ might control AgNPs toxicological effects, improve their safety, and increase the outcome of their applications.
... 138,139 Thymoquinone treatment also significantly decreases endothelial cell damage to alleviate hepatic IR injury. 140 Small molecules with similar skeleton exert protective property against IR injury via autophagy. The flavonoid 4,4′-dimethoxychalcone (DC), isolated from Angelica keiskei koidzumi, is identified as a natural autophagy inducer and alleviates prolonged myocardial ischemia. ...
Article
Ischemia/reperfusion (IR) injury contributes to disability and mortality worldwide. Due to the complicated mechanisms and lack of proper therapeutic targets, few interventions are available that specifically target the pathogenesis of IR injury. Regulated cell death (RCD) of endothelial and parenchymal cells is recognized as the promising intervening target. Recent advances in IR injury suggest that small molecules exhibit beneficial effects on various RCD against IR injury, including apoptosis, necroptosis, autophagy, ferroptosis, pyroptosis, and parthanatos. Here, we describe the mechanisms behind these novel promising therapeutic targets and explain the machinery powering the small molecules. These small molecules exert protection by targeting endothelial or parenchymal cells to alleviate IR injury. Therapies of the ideal combination of small molecules targeting multiple cell types have shown potent synergetic therapeutic effects, laying the foundation for novel strategies to attenuate IR injury.
... In this study, a significant increase in creatinine and BUN serum levels was observed in the GM-treated group compared with the control group. These results conform to many studies that have demonstrated that over-discharge of creatinine and urea into the serum could indicate reduced glomerular filtration rate (GFR) and impaired renal function, especially proximal tubule function (Abd-Elbaset et al. 2017;Liu et al. 2020;Talib and AbuKhader 2013). Moreover, elevated creatinine and BUN levels were considered the most important hallmarks of GM-induced renal toxicity (Al-Malki and Sayed 2014; Shehata et al. 2022). ...
Article
Full-text available
Gentamicin (GM) is an aminoglycoside antibiotic used to treat bacterial infections. However, its application is accompanied by renal impairments. Apigenin is a flavonoid found in many edible plants with potent therapeutic values. This study was designed to elucidate the therapeutic effects of apigenin on GM-induced nephrotoxicity. Animals were injected orally with three different doses of apigenin (5 mg kg⁻¹ day⁻¹, 10 mg kg⁻¹ day⁻¹, and 20 mg kg⁻¹ day⁻¹). Apigenin administration abolished the alterations in the kidney index and serum levels of kidney-specific functions markers, namely blood urea nitrogen and creatinine, and KIM-1, NGAL, and cystatin C following GM exposure. Additionally, apigenin increased levels of enzymatic (glutathione reductase, glutathione peroxidase, superoxide dismutase, and catalase) and non-enzymatic antioxidant proteins (reduced glutathione) and decreased levels of lipid peroxide, nitric oxide, and downregulated nitric oxide synthase-2 in the kidney tissue following GM administration. At the molecular scope, apigenin administration was found to upregulate the mRNA expression of Nfe2l2 and Hmox1 in the kidney tissue. Moreover, apigenin administration suppressed renal inflammation and apoptosis by decreasing levels of interleukin-1β, tumor necrosis factor-alpha, nuclear factor kappa-B, Bax, and caspase-3, while increasing B-cell lymphoma-2 compared with those in GM-administered group. The recorded data suggests that apigenin treatment could be used to alleviate renal impairments associated with GM administration.
Article
Several pharmacological effects were described for Nigella sativa (N. sativa) seed and it has been used traditionally to treat various diseases. In this review article, the updated and comprehensive anti-oxidant effects of N. sativa and its main constituent, thymoquinone (TQ), on various disorders are described. The relevant articles were retrieved through PubMed, Science Direct, and Scopus up to December 31, 2023. Various extracts and essential oils of N. sativa showed anti-oxidant effects on cardiovascular, endocrine, gastrointestinal and liver, neurologic, respiratory, and urogenital diseases by decreasing and increasing various oxidant and anti-oxidant marketers, respectively. The main constituent of the plant, TQ, also showed similar anti-oxidant effects as the plant itself. The anti-oxidant effects of different extracts and essential oils of N. sativa were demonstrated in various studies which were perhaps due to the main constituent of the plant, TQ. The findings of this review article suggest the possible therapeutic effect of N. sativa and TQ in oxidative stress disorders.
Article
Full-text available
Pain continues to be an enormous global health challenge, with millions of new untreated or inadequately treated patients reported annually. With respect to current clinical applications, opioids remain the mainstay for the treatment of pain, although they are often associated with serious side effects. To optimize their tolerability profiles, medicinal chemistry continues to study novel ligands and innovative approaches. Among them, natural products are known to be a rich source of lead compounds for drug discovery, and they hold potential for pain management. Traditional medicine has had a long history in clinical practice due to the fact that nature provides a rich source of active principles. For instance, opium had been used for pain management until the 19th century when its individual components, such as morphine, were purified and identified. In this review article, we conducted a literature survey aimed at identifying natural products interacting either directly with opioid receptors or indirectly through other mechanisms controlling opioid receptor signaling, whose structures could be interesting from a drug design perspective.
Article
In the present study, a self-nano-emulsifying drug delivery system (SNEDDS) was developed to evaluate the efficiency of thymoquinone (TQ) in hepatic ischemia/reperfusion. SNEDDS was pharmaceutically characterized to evaluate droplet size, morphology, zeta potential, thermodynamic stability, and dissolution/diffusion capacity. Animals were orally pre-treated during 10 days with TQ-loaded SNEDDS. Biochemical analyses, hematoxylin-eosin staining, indirect immunofluorescence, and reverse transcription polymerase chain reaction (RT-PCR) were carried out to assess cell injury, oxidative stress, inflammation, and apoptosis. The TQ formulation showed good in vitro characteristics, including stable nanoparticle structure and size with high drug release rate. In vivo determinations revealed that TQ-loaded SNEDDS pre-treatment of rats maintained cellular integrity by decreasing transaminase (ALT and AST) release and preserving the histological characteristics of their liver. The antioxidant ability of the formulation was proven by increased SOD activity, reduced MDA concentration, and iNOS protein expression. In addition, this formulation exerted an anti-inflammatory effect evidenced by reduced plasma CRP concentration, MPO activity, and gene expressions of TLR-4, TNF-α, NF-κB, and IL-6. Finally, the TQ-loaded SNEDDS formulation promoted cell survival by enhancing the Bcl-2/Bax ratio. In conclusion, our results indicate that TQ encapsulated in SNEDDS significantly protects rat liver from I/R injury.
Article
Full-text available
Monarda fistulosa is a plant often used in traditional medication with numerous benefits. This review paper aims to elaborate on the phytochemicals of Monarda fistulosa that contribute to its pharmacological activity. Numerous studies have found the bioactive compounds of Monarda fistulosa including carvacrol, thymol, thymoquinone, flavonoid, ????-pinene, caryophyllene oxide, limonene, and geraniol. Researchers have found several pharmacological activities in relation to these compounds including antimicrobial, antidiabetic, anticancer, anti-inflammatory, antioxidants, and immunomodulatory properties. Evidence shows that Monarda fistulosa has potential for applications in many areas. Nevertheless, further clinical studies still have to be conducted to assess the effects of this plant.
Article
Full-text available
Warm ischaemia is usually induced by the Pringle manoeuver (PM) during hepatectomy. Currently, there is no widely accepted standard protocol to minimise ischaemia-related injury, so reducing ischaemia-reperfusion damage is an active area of research. This systematic review and meta-analysis focused on inducible nitric oxide synthase (iNOS) as an early inflammatory response to hepatic ischaemia reperfusion injury (HIRI) in mouse- and rat-liver models. A systematic search of studies was performed within three databases. Studies meeting the inclusion criteria were subjected to qualitative and quantitative synthesis of results. We performed a meta-analysis of studies grouped by different HIRI models and ischaemia times. Additionally, we investigated a possible correlation of endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) regulation with iNOS expression. Of 124 included studies, 49 were eligible for the meta-analysis, revealing that iNOS was upregulated in almost all HIRIs. We were able to show an increase of iNOS regardless of ischemia or reperfusion time. Additionally, we found no direct associations of eNOS or NO with iNOS. A sex gap of primarily male experimental animals used was observed, leading to a higher risk of outcomes not being translatable to humans of all sexes.
Article
Full-text available
The pathogenesis of hepatic ischemia/reperfusion (I/R) is mediated through the generation of oxidative and nitrosative stress-induced cell injury. Hence, the present study was designed to evaluate the hepatoprotective effect of quercetin (QR) compared to N-acetylcysteine (NAC) against hepatic I/R injury in rats and to assess iNOS, eNOS and NOSTRIN protein expressions, as a possible mechanism of its hepatoprotective effect. Hepatic ischemia was surgically performed by occlusion of hepatic pedicle (hepatic artery, portal vein, bile duct) that supplies the left and medial lobes (approximately 70% of the total liver mass), for 30 minutes with a vascular clamp followed by releasing the clamp and the liver was reperfused for 30 minutes. QR-pretreatment increased eNOS protein expression with simultaneous decrease in iNOS and NOSTRIN protein expressions. It also decreased serum aspartate aminotransferase (AST), alanine aminotransferases (ALT) and hepatic myeloperoxidase (MPO) activities. In addition, it restored the depleted content of reduced glutathione (GSH) and decreased malondialdehyde (MDA) and nitric oxide (NO) levels. A notable finding is that QR alleviated I/R-induced histopathological changes. The present study illustrates the hepatoprotective effect of quercetin compared to N-acetylcysteine against ischemia/reperfusion-induced liver injury by inhibiting oxidative stress and by modulating iNOS, eNOS and NOSTRIN protein expressions.
Article
Full-text available
div> In this paper, Frechet distribution under Bayesian paradigm is studied. Posterior distributions are derived by using Gumbel Type-II and Levy prior. Quadrature numerical integration technique is utilized to solve posterior distribution. Bayes estimators and their risks have been obtained by using four loss functions. Prior predictive distributions are derived for elicitation of hyperparameters. The performance of Bayes estimators are compared by using Monte Carlo simulation study. </div
Article
Full-text available
The present study was aimed to study protective role of N-acetyl cysteine (NAC) was assessed against arsenic (As)-induced hepatotoxicity in rats. Twenty four male Wistar rats were divided into 4 groups of 6 animals each and treated as follows: Group 1: sham control, 2: arsenic control (sodium arsenite @ 10 mg/kg b. wt orally for 4 wks), 3: Pre-treatment with NAC (@ 300 mg/kg orally for 2 wks) followed by sodium arsenite along with NAC (as per above doses) and 4: Sodium arsenite + NAC (as per above doses for 4 wks). The concentration of thiobarbituric acid reacting substances (TBARS) and protein carbonyls was significantly (P<0.05) increased, while the concentration of reduced glutathione (GSH), and the activity of CYP450, Na+ - K+ ATPase and Mg2+ ATPase in liver were significantly (P<0.05) reduced in group 2 as compared to control. Groups 3 and 4 revealed improvement in the parameters in study. The study revealed that arsenic induces hepatotoxicity by inducing oxidative stress and supplementation of NAC is beneficial in countering the adverse effects.
Article
Full-text available
We aimed to investigate the effects of curcumin on ischemia/ reperfusion (IR) injury of the liver and distant organs resulting from liver blood flow arrest. Totally 40 rats, divided into four groups, each included 10 rats were used. Group I as only laparatomy, Group II laparatomy and curcumin application, Group III hepatic IR; and Group IV as hepatic IR and curcumin application group. Ischemia was generated by hepatoduedonal ligament clamping for 30 minutes and then reperfusion is started. Curcumin capsules were opened and appropriate dose had been created within weighing scales. After calculations, the powder was diluted with saline. Fifteen minutes before the ischemia, curcumin was applied via oral gavage. Blood samples were taken from the animals for biochemical analysis at 60th minutes of the experiment in the first and second groups; 30 minutes after beginning reperfusion in the third and forth groups. Simultaneously, liver, lung and kidney tissues were sampled for biochemical and histopathological examinations. Plasma malondialdehyde levels were found to be higher (p < 0.001), but total antioxidant activity values were not different in IR group compared with IR + curcumin group (p > 0.05). Biochemical and histopathological evaluation of tissue samples revealed that there were no differences in total antioxidant activity, total oxidant activity and histopathologic scores in IR + curcumin group compared with values of IR group (p > 0.05). Curcumin did not reduce the effects of hepatic ischemia reperfusion injury on the liver and distant organs including kidneys and lungs significantly.
Article
This study aimed to assess the effects of thymoquinone (TQ) on pyrogallol-induced endothelial dysfunction in isolated rabbit aorta. The protective effects of TQ were examined by incubating aortic rings in TQ concomitant with pyrogallol. The results revealed that pyrogallol produced significant enhancement of phenylephrine-induced contraction and impairment of acetylcholine-induced relaxation. Pyrogallol caused a significant increase in lipid peroxidation and reduction in the level of superoxide dismutase and reduced glutathione in the aortic homogenates. In addition, pyrogallol produced a significant decrease in nitrite/nitrate concentrations (NOx), constitutive nitric oxide synthase (cNOS) activity and an increase in inducible nitric oxide synthase (iNOS) activity in the aortic homogenates. These changes were counteracted by TQ co-incubation as TQ attenuated pyrogallol-induced impairment in vascular reactivity in a dose-dependent manner. Furthermore, TQ showed potent antioxidant activity as well as causing a significant increase in NOx and cNOS activity, and depression in iNOS activity. These results suggest that TQ can protect against pyrogallol-induced endothelial dysfunction which probably results from its potent antioxidant capacity that leads to an increase in NO production as well as its ability to enhance the generation and bioavailability of NO.
Article
Abstract One of the most common complications after liver transplantation is primary graft dysfunction which results from severe deterioration of the microcirculation. The data obtained from our experimental studies indicate that N-acetylcysteine (NAC) is able to reduce the severity of ischemia/reperfusion injury and improves postoperative graft function after liver transplantation in rats. The aim of this pilot study was to evaluate the efficacy of NAC as a hepatoprotective agent under clinical conditions. A group of 30 liver transplanted patients were treated with NAC, and 30 patients (control group) were treated with a 5 % solution of glucose only. In the NAC group we observed a distinct reduction in ischemia/reperfusion injury and improved liver function with less elevated peak transaminases, better macrocirculation, improved liver synthesis function and a lower incidence of primary nonfunction compared with the control group. We conclude that NAC is a very promising substance for reducing graft dysfunction in clinical liver transplantation.