ArticlePDF Available

Comparative Study of the Possible Protective Effects of Cinnamic Acid and Cinnamaldehyde on Cisplatin-Induced Nephrotoxicity in Rats

Authors:

Abstract and Figures

This study aimed to assess the protective effect of cinnamic acid (CA) and cinnamaldehyde (CD) against cisplatin-induced nephrotoxicity. A single dose of cisplatin (5 mg/kg), injected intraperitoneally to male rats, caused significant increases in serum urea, creatinine levels, and lipid peroxides measured as the malondialdehyde content of kidney, with significant decreases in serum albumin, reduced glutathione, and the activity of antioxidant enzymes (catalase, superoxide dismutase, and glutathione peroxidase) of kidney as compared with the control group. On the other hand, administration of CA (50 mg/kg, p.o.) or CD (40 mg/kg, p.o.) for 7 days before cisplatin ameliorated the cisplatin-induced nephrotoxicity as indicated by the restoration of kidney function and oxidative stress parameters. Furthermore, they reduced the histopathological changes induced by cisplatin. In conclusion, CA and CD showed protective effects against cisplatin-induced nephrotoxicity where CD was more effective than CA; affects that might be attributed to their antioxidant activities.
Content may be subject to copyright.
J BIOCHEM MOLECULAR TOXICOLOGY
Volume 27, Number 12, 2013
Comparative Study of the Possible Protective Effects of
Cinnamic Acid and Cinnamaldehyde on
Cisplatin-Induced Nephrotoxicity in Rats
El-Sayed M. El-Sayed,1Ola M. Abd El-Raouf,2Hala M. Fawzy,2
and Mohamed F. Manie2
1Pharmacolgy and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Nasr-City, Cairo, Egypt;
E-mail: elsayed200_1956@hotmail.com
2Pharmacolgy Department, National Organization for Drug Control and Research, Cairo, Egypt
Received 29 April 2013; revised 21 June 2013; accepted 20 July 2013
ABSTRACT: This study aimed to assess the protec-
tive effect of cinnamic acid (CA) and cinnamaldehyde
(CD) against cisplatin-induced nephrotoxicity. A single
dose of cisplatin (5 mg/kg), injected intraperitoneally
to male rats, caused significant increases in serum
urea, creatinine levels, and lipid peroxides measured
as the malondialdehyde content of kidney, with signif-
icant decreases in serum albumin, reduced glutathione,
and the activity of antioxidant enzymes (catalase, su-
peroxide dismutase, and glutathione peroxidase) of
kidney as compared with the control group. On the
other hand, administration of CA (50 mg/kg, p.o.) or
CD (40 mg/kg, p.o.) for 7 days before cisplatin ame-
liorated the cisplatin-induced nephrotoxicity as indi-
cated by the restoration of kidney function and ox-
idative stress parameters. Furthermore, they reduced
the histopathological changes induced by cisplatin. In
conclusion, CA and CD showed protective effects
against cisplatin-induced nephrotoxicity where CD
was more effective than CA; affects that might be at-
tributed to their antioxidant activities. C2013 Wiley Pe-
riodicals, Inc. J. Biochem. Mol. Toxicol. 27:508–514, 2013;
View this article online at wileyonlinelibrary.com. DOI
10.1002/jbt.21515
KEYWORDS: Cinnamic Acid; Cinnamaldehyde; Anti-
Oxidant; Cisplatin; Nephrotoxicity
INTRODUCTION
Cisplatin (cis-diamminedichloroplatinum (II)) is
one of the most effective chemotherapeutic agents
Correspondence to: El-Sayed M. El-Sayed.
C2013 Wiley Periodicals, Inc.
widely used in the treatment of a variety of malig-
nancies, including head and neck, ovarian, and tes-
ticular cancers [1, 2]. However, the full clinical utility
of cisplatin is limited by its nephrotoxicity [3]. Ap-
proximately 28% to 36% of patients receiving an ini-
tial dose (50–100 mg/m2) of cisplatin develop acute
renal failure. The vigorous hydration has not been ef-
fective in eliminating cisplatin toxicity. Also, the use
of diuretics may complicate the electrolyte disturbance
induced by cisplatin. The discontinuation of cisplatin
remains the only option in cases of progressive renal
failure [4].
In addition to direct tubular toxicity in the form of
apoptosis and necrosis [5], vascular factors [6], and in-
flammation [7] that have been implicated in the patho-
genesis of cisplatin-mediated nephrotoxicity, several
other studies have also demonstrated that cisplatin-
induced oxidative stress is involved in the development
of renal tubule injury [8,9]. The involvement of oxida-
tive stress was further supported by the fact that free
radical scavengers and antioxidants prevent cisplatin-
induced nephrotoxicity [10–13].
Many antioxidant agents were investigated for
their preventive abilities against cisplatin-induced
nephrotoxicity. Some researches advised the use of en-
riched diets with natural antioxidants like vitamin E,
ascorbic acid, and methionine [14, 15]. Other studies
reported that the use of sulfhydryl-containing drugs,
such as captopril, diethyldithiocarbamate, sodium
thiosulfate, N-acetylcysteine, and lipoic acid, could also
exert antioxidant activity [16, 17].
Cinnamon, scientifically named Cinnamomum spp,
is a plant with many uses as a herbal medicine, con-
taining mucilage, tannin, sugar, resin, and essential
oil, among which the essential oil is the most impor-
tant part, a substantial portion of which is made up
508
Volume 27, Number 12, 2013 CINNAMON PREVENTS NEPHROTOXICITY 509
of cinnamaldehyde (CD) which possesses antioxidant,
antibacterial and anti-inflammatory effects [18, 19]. In
addition, cinnamic acid (CA), a major active phenolic
ingredient in cinnamon displays many pharmacolog-
ical properties, such as antioxidant and antimicrobial
activity [20, 21].
Therefore, the aim of this study was to assess
the possible protective effects of CA and CD against
cisplatin-induced nephrotoxicity in rats.
MATERIALS AND METHODS
Chemicals
Cisplatin was obtained from EIMC United Phar-
maceuticals, Egypt, and was given intraperitoneally
(i.p.) in a single dose of 5 mg/kg [22]. CA was ob-
tained from Qualikem Fine Chemicals (New Delhi, In-
dia), and was given orally in a dose of 50 mg/kg daily
for 7 days before the cisplatin injection [23, 24]. CD
was obtained from LLUCH Essence (Barcelona, Spain),
and was given orally in a dose of 40 mg/kg daily for
7 days before the cisplatin injection. CA and CD were
dissolved in dimethylsulphoxide 50% (DMSO), which
was obtained from Sigma–Aldrich Corporation (Lyon,
France). All other chemicals were of the highest analyt-
ical grades available commercially.
Animals
Male adult Sprague–Dawley rats weighing 230–
260 g were obtained from the breeding colony main-
tained at the animal house of the National Organiza-
tion for Drug Control and Research, Egypt. Animals
were caged in seven groups, given food and water ad
libitum and maintained at 21C–24C and 40%–60%
relative humidity with 12-h light–dark cycles. Animals
were subjected to an adaptation period of 2 weeks
in the animal house before experiments. The exper-
iments were conducted in accordance with the ethi-
cal guidelines for investigations in laboratory animals
and were approved by the Ethical Committee of Fac-
ulty of Pharmacy, Cairo University, Egypt, and com-
ply with the Guide for the Care and Use of Laboratory
Animals [25].
Experimental Design
Fifty-six male adult Sprague–Dawley rats were al-
located into seven groups (eight rats each); two rats
from each group were used for histopathological ex-
amination as follows:
Group 1: Received saline and served as control.
Group 2: Received DMSO 50% (as a solvent).
Group 3: Received CA in a dose of 50 mg/kg, p.o.
Group 4: Received CD in a dose of 40 mg/kg, p.o.
Group 5: Received cisplatin in a single dose of 5 mg/kg,
i.p.
Group 6: Pretreated with CA in a dose of 50 mg/kg,
p.o., for 7 days, followed by a single dose of cisplatin
(5 mg/kg, i.p.).
Group 7: Pretreated with CD in a dose of 40 mg/kg,
p.o., for 7 days, followed by a single dose of cisplatin
(5 mg/kg, i.p.).
Serum and Tissue Preparation
On the seventh day after the cisplatin injection,
blood samples were taken under light ether anesthe-
sia in nonheparinized tubes. Serum was separated by
centrifugation for 20 min at 4000×gand stored at
20C.
The kidneys were rapidly isolated and washed
with ice-cold isotonic saline (0.9%). Then they were
stored at –80C till they were homogenized in 50 mM
phosphate buffer (pH 7.4) using an electronic homoge-
nizer (Ezister Daihan Scientific Company Ltd., Korea)
to prepare 10% w/v homogenate. The homogenate was
then made into aliquots and was used for the determi-
nation of kidney contents of malondialdehyde (MDA)
and reduced glutathione (GSH), and enzymatic activi-
ties of catalase (CAT), superoxide dismutase (SOD) and
glutathione peroxidase (GPx).
Biochemical Analysis
Serum urea nitrogen, creatinine, and albumin were
estimated colorimetrically according to methods of
Fawcettet and Scott [26], Bartles et al. [27], and Doumas
and Peters [28], respectively.
The kidney homogenate was used for the determi-
nation of thiobarbituricacid-reactive substances levels
measured as MDA according to the method of Satoh
[29], using 1,1,3,3-tetraethoxypropane as a standard.
Reduced GSH contents were assessed by the method
of Beutler et al. [30], using 5–5’ dithiobis (2-nitrobenzoic
acid) as a substrate. CAT activity was determined col-
orimetrically using hydrogen peroxide as a substrate
according to the method of Aebi [31]. SOD activity
was determined using the method of Nishikimi et al.
[32], which relies on the ability of the enzyme to in-
hibit the phenazinemethosulphate mediated-reduction
of nitrobluetetrazolium dye. GPx activity was mea-
sured by applying the method of Paglia and Valentine
[33] using hydrogen peroxide as a substrate.
J Biochem Molecular Toxicology DOI 10.1002/jbt
510 EL-SAYED ET AL. Volume 27, Number 12, 2013
TABL E 1. Effect of Pretreatment with CA or CD on Serum Urea, Creatinine, Albumin Levels, Final Body Weight, and Kidney–
Body Weight Ratio in Cisplatin-Treated Rats
Parameters
Groups Urea (mg/dl) Creatinine (mg/dl) Albumin (mg/dl) Final Body Weight (g) Kidney–Body Weight Ratio (1000×)
Control saline 25.37 ±2.11 0.48 ±0.04 4.29 ±0.19 235.30 ±3.99 5.60 ±0.11
DMSO 27.15 ±2.30 0.75 ±0.06 4.37 ±0.20 234.70 ±1.76 6.18 ±0.14
CA 25.25 ±2.22 0.74 ±0.06 4.28 ±0.09 243 ±6.23 6.22 ±0.12
CD 23.64 ±0.60 0.50 ±0.03 3.98 ±0.15 257.70 ±4.05 6.20 ±0.09
Cisplatin 127.10 ±0.79a4.98 ±0.34a1.37 ±0.06a202.2 ±1.83a11.3 ±0.17a
CA +cisplatin 69.27 ±3.83a,b0.74 ±0.05b3.68 ±0.15b249.30 ±9.45b9.35 ±0.19a,b
CD +cisplatin 46.91 ±1.06a,b,c0.77 ±0.03b4.07 ±0.15b256 ±6.01b7.13 ±0.24a,b,c
Data are expressed as means ±SEMofsixratspergroup.
aSignificantly different from the control saline group.
bSignificantly different from the cisplatin-treated group.
cSignificantly different from the CA +cisplatin-treated group using one-way ANOVA followed by the Tukey–Kramer test for multiple comparison test at p0.05.
Histopathological Examination
of the Kidney
Autopsy samples were taken from the kidney
of rats in different groups, and fixed in 10% neutral
buffered formalin for 24 h; and decalcification was car-
ried out on formic acid. Washing was done in tap water,
and then serial dilutions of alcohol (methyl, ethyl, and
absolute ethyl) were used for dehydration. Specimens
were cleared in xylene and embedded in paraffin at
56C in hot air oven for 24 h. Paraffin bees wax tis-
sue blocks were prepared for sectioning at 4 μm thick-
ness by sledge microtome. The obtained tissue sections
were collected on glass slides, deparaffinized, stained
by hematoxylin and eosin stain, and then examination
was done through the light electric microscope [34].
Statistical Analysis
All values were presented as means ±standard
error of the means (SEM). Statistical analysis was per-
formed using GraphPad Prism version 5 (GraphPad,
San Diego, CA). Comparison between different groups
was carried out using one-way analysis of variance
(ANOVA), followed by Tukey–Kramer’s multiple com-
parison tests. Difference was considered significant
when p0.05.
RESULTS
Table 1 shows that injection of cisplatin (i.p.) in
a single dose of 5 mg/kg caused significant increases
in serum urea (401%), creatinine (938%), and kidney–
body weight ratio (102%), and significant decreases in
body weight (14%) and serum albumin level (68%) after
7 days of treatment as compared with the control group.
Moreover, cisplatin (5 mg/kg) produced a signifi-
cant increase in MDA (124%) and significant decreases
in the GSH renal content (69%) and the enzymatic an-
tioxidant parameters in the kidney, that is, SOD, CAT,
and GPx (56%, 57%, and 77%, respectively, in compar-
ison with the control group) (Table 2).
In contrast, administration of CA for 7 days before
cisplatin significantly reduced the elevated levels of
urea and creatinine in serum by 46% and 85%, respec-
tively, as well as kidney–body weight ratio (17%), and
significantly increased the body weight (23%) and the
TABL E 2. Effect of Pretreatment With CA or CD on Kidney Contents of MDA and GSH as well as the Levels of Enzymatic
Antioxidants in Cisplatin-Treated Rats
Parameters
Groups MDA (nmol/g Tissue) GSH (mg/g Tissue) SOD (U/g Tissue) CAT (U/g Tissue) GPx (U/g Tissue)
Control saline 21.22 ±0.84 14.79 ±0.86 463.50 ±20.76 1.38 ±0.05 17.16 ±0.89
DMSO 23.74 ±0.16 13.63 ±0.64 395 ±16.86 1.39 ±0.08 13.05 ±0.73
CA 22.86 ±0.87 12.85 ±0.59 505.10 ±35.60 1.01 ±0.07 15.98 ±1.08
CD 19.68 ±0.69 14.04 ±2.22 385 ±13.88 0.88 ±0.03 11.78 ±1.80
Cisplatin 47.55 ±2.56a4.64 ±0.12a201.80 ±21.66a0.60 ±0.05a3.98 ±0.19a
CA +cisplatin 13.42 ±0.96a,b13.51 ±0.83b459.60 ±40.50b0.76 ±0.04a9.40 ±0.97a,b
CD +cisplatin 18.72 ±0.36b,c21.69 ±1.22a,b,c625.50 ±22.76a,b,c0.98 ±0.07a,b9.23 ±1.26a,b
Data are expressed as means ±SEMofsixratspergroup.
aSignificantly different from the control saline group.
bSignificantly different from the cisplatin-treated group.
cSignificantly different from the CA +cisplatin-treated group using one-way ANOVA followed by the Tukey–Kramer test for multiple comparison test at p0.05.
J Biochem Molecular Toxicology DOI 10.1002/jbt
Volume 27, Number 12, 2013 CINNAMON PREVENTS NEPHROTOXICITY 511
TABL E 3. Effect of Pretreatment with CA or CD on Histopathological Findings of Kidney Tissues of Cisplatin-Treated Rats.
Groups
Histopathological Control Cisplatin CD (40 mg/kg) +CA (50 mg/kg) +
Findings (Saline) (5 mg/kg) Cisplatin (5 mg/kg) Cisplatin (5 mg/kg)
Tubular cougulative necrosis 0 ++++ + ++
None Very severe Mild Moderate
Inflammatory cell infiltration 0 ++++ + ++
None Very severe Mild Moderate
Haemorrahage 0 ++++ + ++
None Very severe Mild Moderate
Renal casts 0 ++ + ++
None Moderate Mild Moderate
Tot al 0 14 ( +)4(+)8(+)
serum albumin level (169%) (in comparison with the
cisplatin-treated group). Furthermore, it decreased the
MDA content by 72% and increased GSH, SOD, CAT,
and GPx (191%, 128%, 27% and 136%) levels in kid-
ney tissue, respectively, in comparison with cisplatin-
treated group (Table 1 & 2). Similarly, treatment of an-
imals with CD (40 mg/kg) for 7 days before cisplatin
significantly reduced the elevated levels of urea and
creatinine in serum by 63% and 85%, respectively, as
well as kidney–body weight ratio (37%), and signifi-
cantly increased the body weight (27%) and the serum
albumin level (197%) (in comparison with the cisplatin-
treated group), while it decreased the MDA content by
61% and increased GSH, SOD, CAT, and GPx (367%,
FIGURE 1. Histology of kidney samples of the control (saline), cisplatin-treated group, CD +cisplatin-treated group, or CA +cisplatin-treated
group. (A) Control group: normal histological appearance of renal tubules (rt); (B) Cisplatin-treated group: inflammatory cells infiltration (m) in
between the cystic (s) and necrosed (n) tubules; (C) CD +Cisplatin-treated group: necrosis in some few individual tubules (n) with hyperplasia
and dysplasia in the lining epithelium (a) of other tubules; (D) CA +Cisplatin-treated group: degeneration and cystic (s) dilatation in most tubules
at the cortex. Hematoxylin–eosin staining, magnifications: ×40.
J Biochem Molecular Toxicology DOI 10.1002/jbt
512 EL-SAYED ET AL. Volume 27, Number 12, 2013
210%, 63%, and 132%, respectively) levels in kidney
tissue, in comparison with the cisplatin-treated group.
Histopathological findings of kidney tissues are
illustrated in Table 3 & Figure 1. The histopathological
examination of kidney sections of the control group
(saline) showed a normal histological structure
(Figure 1A). On the other hand, administration of
cisplatin to rats revealed degenerative changes, in-
flammatory cell infiltration between the cystic dilated
and necrosed tubules, and focal haemorrhage detected
in between the tubules at the corticomedullary junc-
tion (Figure 1B). Pretreatment of the rats with CD
(Figure 1C) or CA (Figure 1D) obviously mitigated the
histopathological changes induced by cisplatin.
DISCUSSION
Cisplatin is a major antineoplastic weapon used
for the treatment of solid tumors. Its chief dose-limiting
side effect is nephrotoxicity, which requires a reduction
of dose or discontinuation of the treatment [35].
The present study was designed to investi-
gate whether CA or CD administration before cis-
platin could afford protection against cisplatin-induced
nephrotoxicity.
Our results revealed that cisplatin produced sig-
nificant elevation in serum creatinine, urea levels, and
kidney–body weight ratio and a significant decrease
in the serum albumin level. The increased urea and
creatinine levels suggest the reduction of glomerular-
filtration rate [36]. Also, the increase in kidney–body
weight ratio could be attributed to the reduction of
body weight. Furthermore, cisplatin caused a signifi-
cant decline in the activity of the antioxidant enzymes
(CAT, SOD, and GPx), significant depletion of GSH,
and enhancement of MDA production in the renal tis-
sue. These findings are consistent with those of Ali et al.
[37], Fouad et al. [38] and Yadav et al. [39].
It was evident that cisplatin nephrotoxicity occurs
as a result of oxidative stress and increased genera-
tion of superoxide anion, hydrogen peroxide, and hy-
droxyl radicals due to the increased activity of NADPH
oxidase, xanthine oxidase, and adenosine deaminase
[13, 40]. These free radicals damage the lipid compo-
nents of the cell membrane via peroxidation and de-
naturing its proteins, which subsequently lead to en-
zymatic inactivation [41]. Moreover, cisplatin-induced
tubular damage could be explained by the fact that,
as fast as cisplatin is in the interior of the cells, the
hydrolysis product (chloride atoms replaced by water
molecules) reacts with GSH in the cytoplasm and DNA
in the nucleus [42]. The produced cisplatin-DNA in-
trastrand cross-links result in cytotoxicity (apoptosis/
necrosis) [43].
The present study demonstrated that pretreatment
with CA or CD ameliorated cisplatin-induced alter-
ations in serum creatinine, urea and albumin levels,
and body weight and kidney–body weight ratio. In ad-
dition, CA or CD significantly mitigated the lipid per-
oxidation in the rat kidney induced by cisplatin as man-
ifested by the decreased MDA level, accompanied by
the increased GSH content and enhanced activities of
CAT, SOD, and GPx. These results could be attributed
to the potential antioxidant effect of CA [44] and CD
[19], and are in agreement with those obtained by Pa-
tra et al. [45] who demonstrated that CA protects mice
from cyclophosphamide-induced hepatotoxicity and
myelotoxicity. Moreover, our results are consistent with
Molania et al. [19] who revealed the promising pro-
tective effect of CD against gamma radiation-induced
mucositis.
The histopathological findings demonstrated that
administration of cisplatin induced various degenera-
tive changes in kidney cells, which confirmed the bio-
chemical evidence of the oxidative stress. In contrast,
pretreatment with CA or CD obviously mitigated the
histopathological changes induced by cisplatin.
Our data showed that as a nephroprotective, CD
was more effective than CA. This could be explained
by the fact that a major portion of CD is metabo-
lized into CA and a portion of the absorbed CD conju-
gates with blood proteins before entering the liver, and
most of these protein conjugates escape hepatic first-
pass metabolism and slowly regenerate CD. Thus, this
whole process maintains CD concentrations in blood
for a long period [46].
In conclusion, CA or CD protected the kidney tis-
sue against cisplatin-induced nephrotoxicity in rats,
where CD was more effective than CA. The antioxidant
activities might be considered the main factors respon-
sible for such nephroprotective effects. Therefore, CA
or CD represents a potential candidate to prevent re-
nal injury, which is a major and dose-limiting problem
during the cisplatin therapy.
ACKNOWLEDGMENT
We thank Professor Dr. Adel B. Kholoussy, De-
partment of Pathology, Faculty of Veterinary Medicine,
Cairo University, for his kind help in performing
histopathological results.
REFERENCES
1. Badary O, Abdel-Maksoud S, Ahmed W, Owieda G.
Naringenin attenuates cisplatin nephrotoxicity in rats.
Life Sci 2005;76:2125–2135.
J Biochem Molecular Toxicology DOI 10.1002/jbt
Volume 27, Number 12, 2013 CINNAMON PREVENTS NEPHROTOXICITY 513
2. Rabik CA, Dolan ME. Molecular mechanisms and toxic-
ity associated with platinating agents. Cancer Treat Rev
2007;33:9–23.
3. Schrier RW. Cancer therapy and renal injury. J Clin Invest
2002;100:743–745.
4. Lebwohl D, Canetta R. Clinical development of platinum
complexes in cancer therapy: an historical perspective
and an update. Eur J Cancer 1998;34:1522–1534.
5. Arany I, Safirstein RL. Cisplatin nephrotoxicity. Semin
Nephrol 2003;23:460–464.
6. Luke DR, Vadiel K, Lopez-BeresteinG. Role of vascular
congestion in cisplatin-induced acute renal failure in the
rat. Nephrol Dial Transplant 1992;7:1–7.
7. Ramesh G, Reeves WB. TNFR2-mediated apoptosis and
necrosis in cisplatin-induced acute renal failure. Am J
Physiol Renal Physiol 2003;285:F610–F618.
8. Pabla N, Dong Z. Cisplatin nephrotoxicity: mechanisms
and renoprotective strategies. Kidney Int 2008;73:994–
1007.
9. Chirino Y, Pedraza-Chaverri J. Role of oxidative and ni-
trosative stress in cisplatin-induced nephrotoxicity. Exp
Toxicol Pathol 2009;61:223–242.
10. Tsuruya K, Tokumoto M, Ninomiya T, Hirakawa M,
Masutani K, Taniguchi M, Fukuda K, Kanai H, Hirakata
H, Iida M. Antioxidant ameliorates cisplatin-induced re-
nal tubular cell death through inhibition of death re-
ceptor mediated pathways. Am J Physiol Renal Physiol
2003;285:F208–F218.
11. Weijl NI, Elsendoorn TJ, Lentjes EG, Hopman GD,
Wipkin-Bakker A, Zwinderman AH, Cleton FJ, Osanto
S. Supplementation with antioxidant micronutrients
and chemotherapy-induced toxicity in cancer patients
treated with cisplatin-based chemotherapy: a ran-
domised double blind placebo-controlled study. Eur J
Cancer 2004;40:1713–1723.
12. Dickey DT, Wu YJ, Muldoon LL, Neuwelt EA. Protec-
tion against cisplatin-induced toxicities by N-acetyl cys-
teine and sodium thiosulfate as assessed at the molec-
ular cellular and in vivo levels. J Pharmacol Exp Ther
2005;314:1052–1058.
13. Gulec M, Iraz M, Yilmaz HR, Ozyurt H, Temel I. The ef-
fects of Ginkgo biloba extract on tissue adenosine deam-
inase, xanthine oxidase, myeloperoxidase, malondialde-
hyde and nitric oxide in cisplatin-induced nephrotoxicity.
Toxicol Ind Health 2006;22:125–130.
14. Basinger MA, Jones MM, Holscher MA. L-methionine
antagonism of cis-platinum nephrotoxicity. Toxicol Appl
Pharmacol 1990;103(1):1–15.
15. Appenroth D, Frob S, Kersten L, Splinter FK,
Winnefeld K. Protective effects of vitamin E and C on
cisplatin nephrotoxicity in developing rats. Arch Toxicol
1997;71(11):677–683.
16. Somani SM, Husain K, Whitworth C, Trammell GL,
Malafa M, Rybak LP. Dose-dependent protection by
lipoic acid against cisplatin-induced nephrotoxicity in
rats: Antioxidant defense system. Pharmacol Toxicol
2000;86(5):234–241.
17. El-Sayed EM, Abd-Ellah MF, Attia SM. Protective effect
of captopril against cisplatin-induced nephrotoxicity in
rats. Pak J Pharm Sci 2008;21(3): 255–261.
18. Barceloux DG. Cinnamon (Cinnamomum species). Dis
Mon 2009;55(6):327–335.
19. Molania T, Moghadamnia AA, Pouramir M, Aghel S,
Moslemi D, Ghassemi L, Motallebnejad M. The effect of
Cinnamaldehyde on mucositis and salivary antioxidant
capacity in gamma-irradiated rats (a preliminary study).
DARU J Pharm Sci 2012;20:89–93.
20. Akaro Y, Maruyama H, Mtasumoto K, Ohguchi K,
Nishizawa K, Sakamoto T, Araki Y, Mishima S, Nozawa
Y. Cell growth inhibitory effect of cinnamic acid deriva-
tives from propolis on human tumor cell lines. Biol Pharm
Bull 2003;26:1057–1059.
21. Chen YL, Huang ST, Sun FM, Chiang YL, Chiang CJ,
Tsai CM, Weng CJ. Transformation of cinnamic acid
from trans- to cis-form raises a notable bactericidal
and synergistic activity against multiple-drug resistant
Mycobacterium tuberculosis. Eur J Pharm Sci 2011;43:
188–194.
22. Naghizadeh B, Boroushaki MT, Mashhadian NV,
Mansouri SMT. Protective effects of crocin against
cisplatin-induced acute renal failure and oxidative stress
in rats. Iran Biomed J 2008;12 (2): 93–100.
23. Fern´
andez-Mart´
ınez E, Bobadilla RA, Morales-R´
ıos
MS, Muriel P, P´
erez- ´
Alvarez VM. Trans-3-phenyl-2-
propenoic acid (cinnamic acid) derivatives: structure-
activity relationship as hepatoprotective agents. Med
Chem 2007;3(5):475–479.
24. Kasetti RB, Abdul Nabi S, Swapna S, Apparao C. Cin-
namic acid as one of the antidiabetic active principle(s)
from the seeds of Syzygiumalternifolium. Food Chem
Toxicol 2012;50:1425–1431.
25. Institute of Laboratory Animal Resources. Guide for
the Care and Use of Laboratory Animals, 8th edition.
Washington, D.C.: National Academy Press; 1996.
26. Fawcett JK, Scott JE. A rapid and precise method for the
determination of urea. J Clin Path 1960;13:156–159.
27. Bartles H, Bohmer M, Heirli C. Colorimetric kinetic
method for creatinine determination in serum and urine.
Clin Chem Acta 1972;37:193.
28. Doumas BT, Peters T. Serum and urine albumin: a
progress report on their measurement and clinical sig-
nificance. Clin Chim Acta 1971;31:87–96.
29. Satoh K. Serum lipid peroxides in cerebrovascular dis-
orders determined by a new colorimetric method. Clin
Chim Acta 1978;90:37–43.
30. Beutler E, Duron O, Kelly MB. Improved method for
the determination of blood glutathione. J Lab Clin Med
1963;61:882–8.
31. Aebi H. Catalase in vitro. Methods Enzymol
1984;105:121–126.
32. Nishikimi M, Roa NA, Yogi K. Measurement of su-
peroxide dismutase. Biochem Biophys Res Commun
1972;46:849–854.
33. Paglia DE, Valentine WN. Studies on the quantitative and
qualitative characterization of erythrocyte glutathione
peroxidase. J Lab Clin Med 1967;70:158–169.
34. Banchroft JD, Stevens A, Turner DR. Theory and prac-
tice of histological techniques, 4th edition. New York:
Churchil Livingstone; 1996.
35. Kuhad A, Tirkey N, Pilkhwal S, Chopra K. Renopro-
tective effect of Spirulinafusiformis on cisplatin-induced
oxidative stress and renal dysfunction in rats. Ren Fail
2006;28(3):247–254.
36. Naziroglu M, Karaoglu A, Aksoy AO. Selenium and
higher dose vitamin E administration protects cisplatin-
induced oxidative damage to renal, liver, lens tissues in
rats. Toxicology 2004;195: 221–230.
37. Ali BH, Al Moundhri MS, Tag El-din MT, Nemmar
A, Tanira MO. The ameliorative effect of cysteine pro-
drug L-2-oxothiazolidine-4-carboxylic acid on cisplatin-
J Biochem Molecular Toxicology DOI 10.1002/jbt
514 EL-SAYED ET AL. Volume 27, Number 12, 2013
induced nephrotoxicity in rats. Fundam Clin Pharmacol
2007;l21:547–553.
38. Fouad AA, Al-Sultan AI, Refaie SM, Yacoubi MT. Coen-
zyme Q10 treatment ameliorates acute cisplatin nephro-
toxicity in mice. Toxicology 2010;274(1–3):49–56.
39. Yadav YC, Srivastav DN, Seth AK, Saini V, Yadav KS.
Nephropharmacological activity of ethanolic extract Le-
pidium Sativum L. seeds in albino rats using cisplatin
induced acute renal failure. Int J Pharm Sci Rev Res
2010;4(3):64–68.
40. ChirinoYI, Sanchez-Gonzalez DJ, Martinez CM, Cruz C,
Pedraza-Chaverri J. Protective effects of apocynin against
cisplatin-induced oxidative stress and nephrotoxicity.
Toxicology 2008;245:18–23.
41. Lalila A, Ola H, Hossam A, Mohamed M, Sayed A. Effect
of cremophor-EL on cisplatin-induced organ toxicity in
normal rat. J Egypt Natl Canc Inst 2001;(13):139–145.
42. Boulikas T, Vougiouka M. Cisplatin and platinum drugs
at the molecular level. Oncol Rep 2003;10(6):1663–1682.
43. Galea AM, Murray V. The interaction of cisplatin
and analogues with DNA in reconstituted chromatin.
Biochim Biophys Acta 2002;(2–3):142–152.
44. Dai A, Nie YX, Yu B, Li Q, Lu LY, Bai JG. Cinnamic acid
pretreatment enhances heat tolerance of cucumber leaves
through modulating antioxidant enzyme activity. Envi-
ron Exp Bot 2012;79:1–10.
45. Patra K, Bose S, Sarkar S, Rakshit J, Jana S, Mukher-
jee A, Roy A, Mandal DP, Bhattacharjee S. Ameliora-
tion of cyclophosphamide induced myelosuppression
and oxidative stress by cinnamic acid. Chem Biol Interact
2012;195:231–239.
46. Yuan JH, Dieter MP, Bucher JR, Jameson CW. Toxicoki-
netics of cinnamaldehyde in F344 rats. Food Chem Toxi-
col 1992;30(12): 997–1004.
J Biochem Molecular Toxicology DOI 10.1002/jbt
... The main chemical compound found in cinnamon is cinnamic acid (cin), which is one of the most common and basic phenolic acids found in nature (Hong et al. 2021;Ben Lagha et al. 2021). Cin possesses a lot of beneficial pharmacological actions such as antitumor, antimicrobial, antioxidant, and antiinflammatory activities (El-Sayed et al. 2013;Ruwizhi and Aderibigbe 2020). Consequently, hindering inflammation and oxidative stress was deliberated as a prospective beneficial goal in the treatment of fibrotic diseases. ...
... • Normal (saline) group: rats received saline orally daily. • Normal (DMSO) group: rats received 50% DMSO (solvent for cinnamic acid) orally daily (El-Sayed et al. 2013). • MTX group: rats received MTX, 14 mg/kg, orally once a week for 2 weeks; served as lung fibrotic group (Fikry et al. 2015). ...
... • Pir + MTX group: rats received pir (50 mg/kg/day) orally, whereas MTX (14 mg/kg) was orally given on the 5th and 12th days of the experiment; serve as a standard drug-treated group. • Cin group: rats were treated with cin (50 mg/kg/day) orally (El-Sayed et al. 2013). • Cin + MTX group: rats received cin in a dose of 50 mg/ kg/day orally, whereas MTX (14 mg/kg) was orally given on the 5th and 12th days of the experiment. ...
Article
Full-text available
Purpose Lung fibrosis is a heterogeneous lung condition characterized by excessive accumulation of scarred tissue, leading to lung architecture destruction and restricted ventilation. The current work was conducted to examine the probable shielding influence of cinnamic acid against lung fibrosis induced by methotrexate. Methods Rats were pre-treated with oral administration of cinnamic acid (50 mg/kg/day) for 14 days, whereas methotrexate (14 mg/kg) was orally given on the 5th and 12th days of the experiment. Pirfenidone (50 mg/kg/day) was used as a standard drug. At the end of the experiment, oxidative parameters (malondialdehyde, myeloperoxidase, nitric oxide, and total glutathione) and inflammatory mediators (tumor necrosis factor-α and interleukin-8), as well as transforming growth factor-β and collagen content, as fibrosis indicators, were measured in lung tissue. Results Our results revealed that cinnamic acid, as pirfenidone, effectively prevented the methotrexate-induced overt histopathological damage. This was associated with parallel improvements in oxidative, inflammatory, and fibrotic parameters measured. The outcomes of cinnamic acid administration were more or less the same as those of pirfenidone. In conclusion, pre-treatment with cinnamic acid protects against methotrexate-induced fibrosis, making it a promising prophylactic adjuvant therapy to methotrexate and protecting against its possible induction of lung fibrosis.
... Furthermore, there is evidence about the vasodilator activity of CA through the nitric oxide (NO)-cGMP-PKG pathway [32]. Recently, El-Sayed et al. showed the renoprotective effects of CA against renal injuries induced by cisplatin [33]. CA exhibited high antioxidant activity due to the presence of vinyl fragments in its structure. ...
... AST: Aspartate aminotransferase; ALT: Alanine aminotransferase (ALT); ALP: alkaline phosphatase (ALP). study of El-Sayed et al. representing the renoprotective effects of CA against renal injuries induced by cisplatin [33]. The decrease of serum urea and creatinine following CA treatment may be associated with improvement of GFR and blood renal filtration barrier function, as proposed for other antioxidants [59]. ...
... In addition, CA clearly recovered renal activities of CAT and GPX in CA þ GM treated animals. Our study is in accordance with previous studies showing that CA can ameliorate oxidative stress in different pathological conditions [23,26,33]. Amelioration of oxidative stress by CA can be associated with its ROS scavenging activity [61] and/or its ability to upregulation of mRNA expression of antioxidant enzymes, as indicated in this study ( Figure 6). ...
Article
Full-text available
This study was the first to evaluate the possible protective effects of cinnamic acid (CA) against Gentamicin (GM) induced liver and kidney dysfunctions in rats. Adult male Wistar rats were randomly assigned to 4 equal groups (n ¼ 8): Control group (saline, 0.5 ml/day), CA group (CA, 50 mg/kg/day), GM group (GM, 100 mg/kg/day), and GM þ CA group (100 & 50 mg/kg/day). Following 12 days of treatments, blood and 24 h urine samples were collected and kidneys were taken out for biochemical, histopathological, and molecular studies. Following CA treatment, renal function markers and transaminases activities including serum urea (59.92%) and creatinine (50.41%), protein excretion rate (43.67%), and serum activities of aspartate aminotransferase (AST) (54.34%) and alanine aminotransferase (ALT) (47.26%) significantly reduced in the treated group as compared with the GM group (P < 0.05). Also, CA could significantly ameliorate the levels of triglyceride (29.70%), cholesterol (13.02%), very low-density lipoprotein (29.69%) and high-density lipoprotein-cholesterol (7.28%). CA could also attenuate oxidative stress through a decrease of serum malondialdehyde (MDA) (50.86%) and nitric oxide (NO) (0.85%) and an increase of renal catalase (CAT) (196.14%) and glutathione peroxidase (GPX) activities (45.88%) as well as GPX mRNA expression (44.42-fold) as compared with the GM group (P < 0.05). Moreover, histopathological evaluations revealed attenuated tubular damages and reduced inflammatory cellular infiltration in CA treated animals. Overall, CA alleviates GM-induced nephrotoxicity and alterations in transaminases activities in rats through its antioxidant activities.
... Cinnamaldehyde is a phenylpropanoid phytoconstituents reported for reno-protective action via attenuation of oxidative and inflammatory stress. [39] In the cited study, it has been reported that cinnamaldehyde acts as a key regulator for interleukin's, TNF's, and p38 mitogen-activated protein kinase (MAPK), thereby improving signaling pathways involved in inflammations or even apoptosis. [40] It has further been documented to inhibit lipid peroxidation induced TLR-4 dimerization by cysteine residues modification into PI3K and phosphoinositide-dependent kinase-1 (PDK-1) and regulate upstream monocyte/macrophage-mediated immune responses via NF-κB signaling. ...
Article
Full-text available
Background: Tinospora cordifolia (Willd.) Miers (T. cordifolia) is a well-known Indian medicinal plant containing several nonpolar and polar constituents that play an important role to mitigate various ailments, such as diabetes, urinary disorders, and hepatoprotective. Due to the lack of evidence on phytopharmacological relevance to the unpredicted nonpolar matrix of T. cordifolia, the present study aimed to evaluate the metabolomic pattern of different fractions obtained from aqueous extract of T. cordifolia, which has been recommended in AYUSH for various ailments including kidney disorders. Materials and Methods: High-performance thin-layer chromatography and gas chromatography-mass spectrometry (GC-MS) analyses were performed on aqueous extracts and hexane, dichloromethane, and methanolic fraction of T. cordifolia aqueous extract to evaluate fingerprinting and metabolomic profile. Principal components and pharmacokinetic analysis were performed using XLSTAT and in-silico SwissADME tool to determine metabolite variability and pharmacokinetic relationship based on lipophilicity and drug-likeness. Further, network pharmacology analysis was performed to determine the exact biomolecular relationship of T. cordifolia in alleviating kidney disease. Results: The GC-MS metabolomics results showed several metabolites in different fractions with high variability of phytoconstituents in the methanolic fraction. In pharmacokinetics, each metabolite exhibited a direct correlation between drug lipophilicity and permeability. Network pharmacological suggested five fatty acids, which significantly interacted with the genes such as AGTR1, ATG, RELA, NOS3, NOS2, REN, INS, IL6, TNF, MAPK1, and CASP3, which could potentially regulate various pathophysiological conditions, such as hypertension, insulin resistance, oxidative and inflammatory stress, and electrolyte homeostasis, thereby strengthening the normal function of the kidney. Conclusion: The study showed that six metabolites of T. cordifolia play a multimechanistic role in alleviating kidney disease.
... antioxidant expression GSH levels, SOD, CAT, and GPx activities. [239] Ellagic acid 10 mg/kg/day, orally for 30 days. ...
Article
Full-text available
Abstract: Bee pollen is a hive product made up of flower pollen grains, nectar, and bee salivary secretions that beekeepers can collect without damaging the hive. Bee pollen, also called beecollected pollen, contains a wide range of nutritious elements, including proteins, carbs, lipids, and dietary fibers, as well as bioactive micronutrients including vitamins, minerals, phenolic, and volatile compounds. Because of this composition of high quality, this product has been gaining prominence as a functional food, and studies have been conducted to show and establish its therapeutic potential for medical and food applications. In this context, this work aimed to provide a meticulous summary of the most relevant data about bee pollen, its composition—especially the phenolic compounds—and its biological and/or therapeutic properties as well as the involved molecular pathways.
... Cisplatin is a platinum drug for tumor chemotherapy [3][4][5]. Cisplatin chemotherapy often induced nephrotoxicity and one-third of patients treated with Cisplatin develop AKI [6]. At present, there is no treatment to improve survival outcomes or protect the kidney from Cisplatin-induced AKI, despite advances in research. ...
Article
Full-text available
Acute kidney injury (AKI) is an important public health concern and characterized as tubular death involved in apoptosis and necrosis. Autophagy is rapidly induced in tubules and associates with renal tubular cells homeostasis to have a complex link with tubular death in AKI. Numb is a multifunctional protein and exerts protective role in tubular death in AKI induced by Cisplatin. However, the effect of Numb on tubular autophagy remains to be investigated. In the present study, the protein expression of LC3 and Beclin-1 related to autophagy was analyzed in Cisplatin-induced AKI mice with knocking down Numb. In model of tubular injury induced by Cisplatin in vitro, downregulation of Numb in NRK-52E cells also inhibited the activation of autophagy accompanied with the decreased protein level of p53. Overexpression of Numb in NRK-52E cells activated autophagy with increased LC3 and Beclin-1 expression accompanied with increased protein level of p53. Moreover, autophagy activation following Numb overexpression was suppressed by p53 inhibitor pifithrin-α. These data indicate that Numb promotes p53-mediated activation of tubular autophagy in AKI induced by Cisplatin and therefore may provide important targets for the treatment of AKI.
... Tohamy et al., 24 have conducted a study on the protective effect of CA on cis-platinum-induced liver injury in mice and have reported that cis-platinum elevates ALT and AST activities, and CREA level was decreased in the cis-platinum + CA group. El-Sayed et al., 25 have reported that there is an increase in CREA level in the cis-platinum group and a decrease in the cis-platinum + CA group compared to the cis-platinum group. These results are similar to ours. ...
Article
Full-text available
This study aimed to evaluate the protective effect of cinnamic acid (CA) on orthophenyl-phenol (OPP)-induced oxidative stress in rats. Thirty-two Sprague-Dawley male rats were divided into four groups as control, OPP, CA and OPP + CA groups. The animals in control, OPP and CA groups were received corn oil, OPP (700 mg kg-1 dissolved in corn oil) and CA (200 mg kg-1) by gavage for 21 days, respectively. The animals in OPP + CA group were received CA for 3 days and from day 4; OPP and CA were applied together daily until day 25. Blood and liver samples were collected at the end of experiment for measurement of aminotransferases, creatinine (CREA), catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), reduced glutathione (GSH) and malondialdehyde (MDA). The OPP-induced high serum activities of aminotransferases and level of CREA were reduced significantly by CA administration. The OPP induced significant increases of CAT activities and MDA levels in serum and liver tissue samples. Moreover, OPP significantly decreased GSH levels as well as GSH-Px and SOD activities. Pre-treatment with CA ameliorated the GSH levels along with GSH-Px and SOD activities compared to OPP-receiving rats. On the other hand, CAT activities and MDA levels significantly decreased following the pre-treatment with CA compared to OPP-receiving rats. It has been determined that OPP causes oxidative stress and lipid peroxidation in blood and liver tissues and creates changes in anti-oxidant defense enzymes. Pre-treatment with CA reduces lipid peroxidation and provides protective effect against oxidative stress.
... Interestingly, the two major constituents of Cinnamomum zeylanicum stem bark are cinnamaldehyde and eugenol. It has been well documented that they have various therapeutic properties like antioxidative and anti-inflammatory properties, as well as nephroprotective effects [25,[50][51][52]. ...
Article
Full-text available
Nephropathies and especially nephrotoxicity have become one of the serious causes of life-threatening conditions because of intensive exposure to xenobiotic whether by environmental pollution or by drug abuse. The present study was undertaken to assess the protective effects of Cinnamomum zeylanicum stem bark aqueous extract (AECZ) on gentamicin-induced nephrotoxicity. AECZ was prepared by maceration in water and tested orally at the doses of 200 and 400 mg/kg/day to prevent gentamicin-induced nephropathies in male Wistar rats. Gentamicin (100 mg/kg/day) was administered for 14 consecutive days by intraperitoneal route, concomitantly with AECZ or silymarin (50 mg/kg/day) used as reference drug. Animal body weight was monitored during the treatment. After the last treatment on the 14th day, animals were sacrificed. Blood was collected for the evaluation of hematological and renal function biomarkers. The homogenate of one kidney was used to assess oxidative stress markers and proinflammatory cytokines, while the other one was fixed in formaldehyde for histopathological studies. Gentamicin decreased body weight, serum total proteins, and calcium level but increased kidneys’ relative weight, serum creatinine, urea, and uric acid. Moreover, the levels of reduced glutathione, catalase, and superoxide dismutase activities were decreased, while an increase in malondialdehyde, proinflammatory cytokines (TNF-α, IL-1β, and IL-6), and nitrites was observed in the negative control group as compared to normal control. Histological analysis of the kidney revealed the presence of tubular necrosis, glomerular degeneration, and macrophage infiltration in the gentamicin-treated group. All these impairment parameters were prevented by AECZ and silymarin treatments. AECZ has a protective effect against gentamicin-induced nephrotoxicity. The antioxidant and anti-inflammatory potentials of this extract may highly contribute to its nephroprotective activity.
... Additionally, several compounds, which were reported in this study as AMSE metabolites, were also documented to exhibit strong antioxidant and anti-inflammatory properties. These include ferulic acid (Erseçkin et al., 2020), p-coumaric acid (Navaneethan and Rasool, 2014), vanillic acid (Sindhu et al., 2015), cinnamic acid (El-Sayed et al., 2013) acids), diarylheptanoids (Zhou et al., 2007;Rajaganapathy et al., 2013), oleic and linoleic acids (Marineli et al., 2012;Priyadarshini et al., 2012;Nafiu et al., 2019). Interestingly, 6-paradol, 6-shogoal (Bak et al., 2012;Peng et al., 2015), and phenolic acids (Juurlink et al., 2014) were reported to prevent oxidative stress-mediated disorders through the activation of Nrf2 transcription and its downstream genes. ...
Article
Ethnopharmacological relevance In Africa, Aframomum species have been traditionally used to treat illnesses such as inflammation, hypertension, diarrhea, stomachache and fever. Moreover, Aframomum melegueta seed extracts (AMSE) are used in traditional medicine to relieve stomachaches and inflammatory diseases. Aim Chronic administration of diclofenac (DIC) has been reported to cause acute kidney injury (AKI), which is a serious health condition. The nephroprotective effect of AMSE is yet to be elucidated. Accordingly, this study aims to investigate the phytoconstituents of standardized AMSE, evaluate its nephroprotective effects against DIC-induced AKI in rats, and elaborate its underlying molecular mechanisms. Materials and methods The quantitative estimation of major AMSE constituents and profiling of its secondary metabolites were conducted via RP-HPLC and LC-ESI/Triple TOF/MS, respectively. Next, DIC (50 mg/kg)-induced AKI was achieved in Sprague-Dawley rats and DIC-challenged rats were administered AMSE (100 and 200 mg/kg) orally. All treatments were administered for five consecutive days. Blood samples were collected and the sera were used for estimating creatinine, urea and, kidney injury molecule (KIM)-1 levels. Kidney specimens were histopathologically assessed and immunohistochemically examined for c-Myc expression. A portion of the kidney tissues was homogenized and examined for levels of oxidative stress markers (MDA and GSH). Heme oxygenase (HO)-1, TNF-α, IL-6, Bax, Bcl2 and caspase-3 renal levels were quantified by ELISA. Moreover, the protein expression levels of NF-ҡB was quantified using western blot analysis, whereas mRNA expression levels of AMPK, SIRT-1, nuclear factor erythroid-2-related factor (Nrf2) and STAT3 were detected using qRT-PCR) in the remaining kidney tissues. Results Standardized AMSE was shown to primarily contain 6-gingerol, 6-shogaol and 6-paradol among the 73 compounds that were detected via LC-ESI/Triple TOF/MS including phenolic acids, hydroxyphenylalkanes, diarylheptanoids and fatty acids. Relative to DIC-intoxicated rats, AMSE modulated serum creatinine, urea, KIM-1, renal MDA, TNF-α, IL-6, Bax, and caspase-3 levels. AMSE has also improved renal tissue architecture, enhanced GSH and HO-1 levels, and upregulated renal Nrf2, AMPK, and SIRT-1 mRNA expression levels. Furthermore, AMSE suppressed p-NF-ҡB65 protein and STAT3 mRNA expression, and further reduced c-myc immunohistochemical expression in renal tissues. Overall, our findings revealed that AMSE counteracted DIC-induced AKI via its antioxidant, anti-inflammatory, and antiapoptotic activities. Moreover, AMSE activated Nrf2/HO1 and AMPK/SIRT1, and inhibited NF-ҡB/STAT3 signaling pathways. Therefore, AMSE is a promising agent for inhibiting DIC-induced nephrotoxicity.
Preprint
Full-text available
Purpose Levels of reactive oxygen species (ROS) are the primary determinants of pulmonary fibrosis. It was discovered that antioxidants can ameliorate pulmonary fibrosis caused by prolonged paraquat (PQ) exposure. However, research into the precise mechanisms by which antioxidants influence the signaling pathways implicated in pulmonary fibrosis induced by paraquat is still insufficient. This research utilized a rat model of pulmonary fibrosis induced by PQ to examine the impacts of Silibinin (Sil) and cinnamic acid (CA) on pulmonary fibrosis, with a specific focus on pro-fibrotic signalling pathways and ROS-related autophagy. Methods Lung injury induced by paraquat was demonstrated to be associated with oxidative stress and inflammation of the lungs, downregulated (miR-193a) and upregulated PI3K/AKT/mTOR signaling lung tissues. Expression levels of miR-193a were determined with quantitative real-time PCR, protein level of protein kinase B (Akt), phosphoinositide 3-Kinase (PI3K) were determined by western blot analysis. Hydroxyproline levels (HYP) and Transforming growth factor-β1 (TGF-β1) were measured by ELISA, malondialdehyde(MDA), total antioxidant capacity (TAO), glutathione peroxidase (GSH) and catalase and were measured in lung tissue homogenates colorimetrically using spectrophotometer. Results Long-term exposure to paraquat resulted in decreased PI3K/AKT signalling, decreased cell autophagy, increased oxidative stress, increased and helped pulmonary fibrosis formation. Silibinin and cinnamic acid also decreased oxidative stress by increasing autophagy and miR-193a expression, which in turn decreased pulmonary fibrosis. These effects were associated by low TGF- β1. Conclusions Silibinin and cinnamic acid inhibited PQ-induced PI3K/AKT by stimulating miR-193-a expression, thus attenuating PQ- induced pulmonary fibrosis.
Article
The present study was designed to investigate to possible potential nephrocurative & nephroprotective activity of 200mg/kg ethanolic extract of Lepidium sativum L. seed was use to against cisplatin (5mg/kg, i.p.) induced nephrotoxicity. The experimental protocol designed as the animals were divided into four groups (n=6) like control, model control, curative (200mg/kg) and protective groups (200mg/kg) were received vehicle, cisplatin, cisplatin + extract, and extract + cisplatin respectively. After 6th days, blood collected from retro-orbital sinus of rats and determined urea and creatinine level in serum of each group after then rats were sacrificed for quantitative estimation of various enzymes and ATPase content in kidney tissue. A single dose of cisplatin induced loss in body weight, increase urine excretion, increased urea & creatinine level in serum; it was significantly recovered by 200mg/kg in curative and protective groups. The enzyme estimation in kidney tissue it found that increase malondialdehyde, superoxide dismutase, catalase and reduced glutathione level, it was significantly monitored by 200mg/kg in curative and protective groups. The level of brush border enzymes like Na+/K+ ATPase, Ca++ ATPase and Mg++ATPase were found significantly reduced after single dose cisplatin injection. It was overcome by treatment of same extract in curative and protective groups. Finally it is concluded that the present study data conformed nephrotoxicity induced by cisplatin due oxidative stress and ethanolic extract of Lepidium sativum L. seeds may have nephroprotective and curative activity.
Article
To investigate the physiological mechanism of heat stress mitigated by exogenous cinnamic acid (CA), seedlings of a cucumber cultivar (Cucumis sativus cv. Jinchun no. 4) were pretreated with CA for 2 d and then were exposed to normal (25/18 °C) and elevated (42/38 °C) temperatures for 3 d. 50 μM CA was chosen since it improved growth more than other CA concentrations under heat stress. At 2 d, supply of 50 μM CA elevated the activities of antioxidant enzymes superoxide dismutase (SOD, EC 1.15.1.1), catalase (CAT, EC 1.11.1.6), guaiacol peroxidase (GPX, EC 1.11.1.7), glutathione peroxidase (GSH-Px, EC 1.11.1.9), ascorbate peroxidase (APX, EC 1.11.1.11), monodehydroascorbate reductase (MDHAR, EC 1.6.5.4), dehydroascorbate reductase (DHAR, EC 1.8.5.1) and glutathione reductase (GR, EC 1.6.4.2) in cucumber leaves, and it also increased the contents of reduced glutathione (GSH) and ascorbate (AsA). When the CA-pretreated cucumber seedlings have been exposed to heat stress, their antioxidant activities were changed further and were higher than heat treatment alone. Compared to heat treatment, the combination of CA pretreatment and heat also increased relative water content (RWC) of leaves, enhanced levels of proline and soluble sugars, elevated contents of CA and vanillic acid (VA) in leaves, and decreased levels of malonaldehyde (MDA), superoxide radical (O2−) and hydrogen peroxide (H2O2), while it resulted in 8.33% of leaf edges being dried up. Meantime, heat had adverse effects on plant growth, RWC, CA and VA contents in leaves, and it as well increased the levels of MDA, O2− and H2O2 and made the dried leaf edges to be 79.17%. We conclude that pretreatment with 50 μM CA elevates CA and VA contents in leaves, enhances antioxidant activities under heat stress and decreases lipid peroxidation to some extent, accompanying with increasing proline and soluble sugar levels in heat-stressed leaves, thus the pretreatment enhances heat tolerance of cucumber seedlings.
Article
Cisplatin-induced toxicities are mainly caused by the formation of free radicals, leading to oxidative organ damage. Plasma concentrations of antioxidants decrease significantly during cisplatin chemotherapy for cancer. Forty-eight cancer patients treated with cisplatin-based chemotherapy were randomised in a double-blind manner to receive either supplementation with vitamin C, vitamin E and selenium dissolved in a beverage or to receive a placebo beverage. Primary outcome measures were the amount of nephrotoxicity and ototoxicity induced by cisplatin. No significant differences were found between the two study groups with respect to these primary outcome measures. However, patients who achieved the highest plasma concentrations of the three antioxidant micronutrients had significantly less loss of high-tone hearing. In addition, significant correlations were found between the reduced/oxidised vitamin C ratio and malondialdehyde (MDA), markers of oxidative stress, and cisplatin-induced ototoxicity and nephrotoxicity. The lack of protection against cisplatin-induced toxicities in patients in the intervention arm may be related to poor compliance and/or inadequate supplementation. Supplementation with a higher dose (intensity) and in combination with other antioxidants should be investigated further.
Article
A simple method for the determination of creatinine is described. At a picric acid concentration of 5.7 mmoles/l a high specificity of the reaction is obtained. The colour produced during the reaction can be estimated at 492 nm. No precipitation of serum proteins is observed.ZusammenfassungEs wird eine einfache Bestimmungsmethode für das Serumkreatinin beschrieben. Durch Verwendung einer 5.7 mM Pikrinsäurelösung wird eine im Verhältnis zu herkömmlichen Techniken grössere Richtigkeit erhalten. Die niedere Konzentration der Pikrinsäure erlaubt bei 492 nm zu messen. Eine Präzipitation von Eiweiss im Messansatz tritt nicht auf.
Article
Among various phenolic compounds, caffeic acid (3,4-dihydroxycinnamic acid) exhibited pharmacological antioxidant, anticancer and antimutagenic activities. The antioxidant properties of phenolic compounds depend on their chemical structure, however, the role of the ethylenic side chain in the radical scavenging activity remains controversial. Thus, the aim of this study consisted to test cinnamic acid and 15 cinnamic acid derivatives in the well known CCl4-induced acute liver damage model, which is dependent on oxidative stress mechanisms. Cinnamic acid and 15 cinnamic acid derivatives (50 mg/kg, p.o.) were administered to male Wistar rats intoxicated with CCl4 (4 g/kg, p.o.). The activities of gamma-glutamyl transpeptidase, alkaline phosphatase and alanine aminotransferase were measured in serum. The lipid peroxidation products were determined in liver. Compounds with a methoxy group at position 3 or 4, or a 3,4-methylenedioxy moiety were the most active ones. Also, we observed that the monosubstituted 3 or 4 hydroxy, or the bulky 3,4 dibenzyloxy substituted compounds showed lower activity. The poorest activity was displayed by disubstituted 3,4-dihydroxy, dimethoxy or diacetyl cinnamic acid derivatives, the ester derived from cinnamic acid with an 8 carbon chain and N-dimethyl substituted compound. Thus, the methoxy substituted group at positions 3 or 4 or the 3,4-methylenedioxy moiety in the caffeic acid derivatives; seem to be the main features required for the hepatoprotection in this model.