ArticlePDF Available

Rosmarinic acid ameliorates acute liver damage and fibrogenesis in carbon tetrachloride-intoxicated mice

Authors:
  • Medical Faculty University of Rijeka

Abstract and Figures

The aim of this study was to investigate the therapeutic potential of rosmarinic acid (RA), a natural phenolic, in the treatment of acute liver toxicity. RA at 10, 25 and 50 mg/kg was administered by gavage once daily for 2 consecutive days, 6 h after CCl(4) intoxication. CCl(4) intoxication caused hepatic necrosis and increased serum ALT activity. In the livers, oxidative/nitrosative stress was evidenced by increased 3-nitrotyrosine (3-NT) and thiobarbituric acid reactive substances (TBARS) formation and a significant decrease in Cu/Zn superoxide dismutase (SOD) activity. CCl(4) administration triggered inflammatory response in mice livers by activating nuclear factor-kappaB (NF-κB), which coincided with the induction of tumor necrosis factor-alpha (TNF-α) and cyclooxygenase-2 (COX-2). RA improved histological and serum markers of liver damage and significantly ameliorated oxidative/nitrosative stress and inflammatory response in liver tissue. Additionally, RA prevented transforming growth factor-beta1 (TGF-β1) and alpha-smooth muscle actin (α-SMA) expression, suggesting suppression of profibrotic response. Furthermore, RA significantly inhibited the CCl(4)-induced apoptosis, which was evident from decreased cleavage of caspase-3. The hepatoprotective activity of RA coincided with enhanced NF-E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression. The results of this study indicates that RA possesses antioxidant, anti-inflammatory, antiapoptotic and antifibrotic activity against acute liver toxicity.
Content may be subject to copyright.
Nephroprotective activities of rosmarinic acid against cisplatin-induced
kidney injury in mice
Robert Domitrovic
´
a,
, Iva Potoc
ˇnjak
b
,Z
ˇeljka Crnc
ˇevic
´-Orlic
´
c
, Marko Škoda
b
a
Department of Chemistry and Biochemistry, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
b
Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
c
Department of Endocrinology, Clinical Hospital Rijeka, 51000 Rijeka, Croatia
article info
Article history:
Received 24 September 2013
Accepted 2 February 2014
Available online 8 February 2014
Keywords:
Rosmarinic acid
Cisplatin
Nephrotoxicity
Oxidative stress
Inflammation
Apoptosis
abstract
Rosmarinic acid (RA) is a natural phenolic compound with a broad range of applications, from food pre-
servatives to cosmetics. Increasing amounts of evidence suggests its beneficial effects against various
pathological conditions. The aim of this study was to investigate the therapeutic activity of rosmarinic
acid (RA) against cisplatin (CP)-induced nephrotoxicity. RA was administered by oral gavage at doses
of 1, 2 and 5 mg/kg for two successive days, 48 h after intraperitoneal CP injection (13 mg/kg). Twenty
four hours later, mice were sacrificed. Treatment with RA significantly ameliorated histopathological
changes and the increase in serum creatinine and blood urea nitrogen (BUN) induced by CP. Oxidative
stress induced by CP, evidenced by increased renal 4-hydroxynonenal (4-HNE), cytochrome P450 2E1
(CYP2E1) and heme oxygenase (HO-1) expression, was significantly reduced by RA administration. More-
over, RA inhibited the expression of nuclear factor-kappaB (NF-
j
B) and tumor necrosis factor-
a
(TNF-
a
),
indicating the inhibition of inflammation. Additionally, RA exhibited antiapoptotic activity through the
reduction of p53, phosphorylated p53 and active caspase-3 expression in the kidneys. These findings
show that RA ameliorates CP-induced oxidative stress, inflammation and apoptosis in the kidneys. The
nephroprotective activity of RA could be, at least in part, attributed to reduced CYP2E1 expression.
Ó2014 Elsevier Ltd. All rights reserved.
1. Introduction
Cisplatin (cis-diamminedichloroplatinum(II), CP) is an antitu-
mor drug commonly used in the treatment of testicular, ovarian,
bladder, cervical, esophageal, head and neck and small cell lung
cancer (Giaccone, 2009). Unfortunately, numerous side effects are
related to CP therapy, including ototoxicity, gastrointestinal toxic-
ity, myelosuppression, neurotoxicity and kidney injury (Miller
et al., 2010; Hartmann and Lipp, 2003). Kidneys represent the main
route of CP excretion, with proximal tubule cells as a primary site
of CP accumulation (Yao et al., 2007). Thus, nephrotoxicity is one of
the most serious dose-limiting side effects in CP chemotherapy.
The major mechanisms of CP-induced nephrotoxicity include tubu-
lar necrosis, oxidative stress, inflammation and apoptosis (Miller
et al., 2010).
Natural phenolics, such as hesperidin, rutin, silymarin and gan-
istein, were shown to ameliorate CP-mediated nephrotoxicity
(Sahu et al., 2013; Kang et al., 2011; Ninsontia et al., 2011;Sung
et al., 2008). Moreover, quercetin prevented the nephrotoxic
activity of CP without affecting its anti-tumor activity (Sanchez-
Gonzalez et al., 2011). These findings indicate that natural phenolic
compounds could be utilized as a nephroprotective agents against
CP-induced kidney injury.
Rosmarinic acid (RA), an ester of caffeic acid and 3,4-dihydroxy-
phenyllactic acid, is a widely occurring natural product with a
broad range of applications, from food preservatives to cosmetics
(Petersen and Simmonds, 2003). It possesses numerous biological
activities, including antioxidative (Zhang et al., 2010), anti-inflam-
matory (Chu et al., 2012), antiapoptotic (Lee et al., 2008), antitu-
mor (Venkatachalam et al., 2013), antialergic (Costa et al., 2012),
antibacterial (Moreno et al., 2006) and antiviral (Swarup et al.,
2007). The antioxidant activity of RA may be attributed to its
phenolic structure. Phenolic compounds can easily donate elec-
trons or hydrogen atoms to neutralize free radicals, whereas
resulting phenoxyl radicals could be enzymatically recycled to par-
ent phenolic (Sakihama et al., 2002). The antioxidant capacity of
phenolics seems greatly dependent on the number and even more
the position of hydroxyl groups. Hydroxyl groups in the ortho posi-
tion of the aromatic ring, such as in RA, can greatly enhance the
http://dx.doi.org/10.1016/j.fct.2014.02.002
0278-6915/Ó2014 Elsevier Ltd. All rights reserved.
Corresponding author. Address: Department of Chemistry and Biochemistry,
School of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia.
Tel.: +385 51651135; fax: +385 51678895.
E-mail address: robert.domitrovic@medri.uniri.hr (R. Domitrovic
´).
Food and Chemical Toxicology 66 (2014) 321–328
Contents lists available at ScienceDirect
Food and Chemical Toxicology
journal homepage: www.elsevier.com/locate/foodchemtox
antioxidant capacity (Sroka and Cisowski, 2003). RA and luteolin
with four phenolic hydroxyl groups, including one catechol
structure, showed similar lipid peroxidation inhibition and free
radical scavenging activity in vitro (Özgen et al., 2011). Antioxidant
activity of RA makes this compound a good drug candidate for
treatment of oxidative stress-related pathological conditions. Pre-
viously, we showed that RA and luteolin ameliorated acute liver
damage in mice through the suppression of oxidative stress,
inflammation and fibrogenesis (Domitrovic
´et al., 2009, 2013). Re-
cently, Tavafi and Ahmadvand (2011) demonstrated that RA inhib-
its gentamicin-induced renal oxidative damage in rats. However,
the nephroprotective activity of RA against CP-induced kidney in-
jury has not been studied previously.
In the current study, we investigated the therapeutic activity of
RA against oxidative stress, inflammation and apoptosis induced
by administration of CP as a possible mechanisms of the nephro-
protective activity of RA.
2. Materials and methods
2.1. Chemicals
Rosmarinic acid (96%), cis-diamineplatinum(II) dichloride and dimethyl sulfox-
ide (DMSO) were purchased from Sigma–Aldrich (Steinheim, Germany). Diagnostic
kits for blood urea nitrogen (BUN) and creatinine were from Dijagnostika (Sisak,
Croatia). Radioimmunoprecipitation assay (RIPA) buffer (sc-24948) was purchased
from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Polyvinylidene difluoride
(PVDF) membrane and blocking reagent were obtained from Roche Diagnostics
GmbH (Mannheim, Germany). Mouse monoclonal antibodies to tumor necrosis
factor-alpha (TNF-
a
) (ab1793) and p53 (ab26), and rabbit polyclonal antibodies
to nuclear factor-kappa B (NF-
j
B) p65 (ab7970), heme oxygenase-1 (HO-1)
(ab13243), 4-hydroxynonenal (4-HNE) (ab46545) and cytochrome P450 2E1
(CYP2E1) (ab19140), HRP-conjugated anti-mouse IgG (ab97023) and HRP-conju-
gated anti-rabbit IgG (ab97051) for Western blotting were purchased from Abcam
(Cambridge, UK). Rabbit polyclonal antibodies to cleaved caspase-3 (Asp175,
#9661) and phospho-p53 (p-p53) (Ser15, #9284) were from Cell Signaling Technol-
ogy (Danvers, MA, USA). DAKO EnVision + System, Peroxidase/DAB kit with second-
ary antimouse/antirabbit antibodies (K500711) were from DAKO Corporation
(Carpinteria, CA, USA). Enhanced chemiluminescence (ECL) substrate was pur-
chased from Pierce Chemical Co. (Rockford, IL, USA). Anesthetic and analgesic
(Narketan 10 and Xylapan, respectively) were purchased from Vetoquinol (Bern,
Switzerland). All other chemicals were of the highest grade commercially available.
2.2. Animals
Male BALB/cN mice from our breeding colony, 14 week old, weighting 24-28 g,
were divided into 6 groups with 5 animals per group. Mice were fed a standard
rodent diet (pellet, type 4RF21 GLP, Mucedola, Italy), and water ad libitum. The
animals were maintained at 12 h light/dark cycle, at constant temperature
(20 ± 1 °C) and humidity (50 ± 5%). All experimental procedures were performed
in compliance with the Declaration of Helsinki and approved by the Ethical Com-
mittee of the Medical Faculty, University of Rijeka.
2.3. Experimental design
Group I (control group) received DMSO diluted with saline (5% DMSO, v/v) by
oral gavage. Group II was treated with RA (5 mg/kg) dissolved in the vehicle. Group
III received CP (13 mg/kg) dissolved in 5% DMSO (v/v) immediately before its
administration as a single intraperitoneal (ip) injection. Groups IV, V and VI were
treated with RA solution orally by gavage at doses of 1, 2 and 5 mg/kg, respectively,
for two consecutive days, two days after CP, whereas groups I, II and III received the
vehicle only. We minimized a possible effect of DMSO on cisplatin by reducing its
content. Additionally, controls received the same amount of DMSO as the CP- and
CP + RA-treated animals. RA or vehicle were given to mice after ip administration
of the combination of anesthetic and analgesic. We performed a preliminary inves-
tigation using RA in a dose range of 1–30 mg/kg with small groups of animals to
establish the suitable doses for usage in the main study. Several studies (Brahmi
et al., 2012; Sahu et al., 2011; Vijayan et al., 2007) demonstrated that pre-treatment
of animals with natural antioxidants was similarly or less renoprotective than post-
treatment. Since kidney damage develops gradually, we administered RA on day 2
and 3 following CP injection, targeting ongoing kidney injury. Twenty-four hours
after the last dose of RA or vehicle, mice were sacrificed. Previously, blood was
collected from retro-orbital sinus and serum was separated to determine serum
creatinine and blood urea nitrogen (BUN) concentration. The abdomen was open
and kidneys were removed. One kidney was frozen are later used for Western blot-
ting and other was immersed in buffered 4% paraformaldehyde solution for histo-
logical sections.
2.4. Serum markers of kidney damage
The level of serum markers of kidney function, BUN and creatinine, was mea-
sured by using a Bio-Tek EL808 Ultra Microplate Reader (BioTek Instruments,
Winooski, VT, USA) according to manufacturer’s instructions.
2.5. Histopathology
Paraformaldehyde-fixed tissues were processed as described previously (Dom-
itrovic
´et al., 2012). Histopathological changes in the kidneys were evaluated in
4
l
m thick deparaffinized sections stained by hematoxylin and eosin (HE). Tubular
damage was assessed by scoring tubular dilatation, necrosis, apoptosis and cast for-
mation in 10 different fields (Olympus BX51 microscope, Tokyo, Japan, 400origi-
nal magnification) in the corticomedullary junction of the kidneys (Leemans et al.,
2005). Histopathological changes were blindly scored by a pathologist on a 5-point
scale: 0 = no damage, 1 = 10% of the corticomedullary junction injured, 2 = 10–25%,
3 = 25–50%, 4 = 50–75%, 5 = more than 75%.
2.6. Immunohistochemistry
Immunohistochemical analysis on deparaffinized tissues sections was per-
formed using the primary antibodies against 4-HNE (1:1000), NF-
j
B p65
(1:1000), and cleaved caspase-3 (1:200), the secondary antimouse/antirabbit anti-
bodies and the DAKO EnVision kit, as described previously (Domitrovic
´et al.,
2011). Stained slides were analyzed by light microscopy (Olympus BX51, Tokyo,
Japan).
2.7. Western blot
Kidneys were lysed in RIPA buffer as described previously (Domitrovic
´et al.,
2011). Volume equivalents of 50
l
g of proteins were separated by 12% sodium
dodecyl sulfate polyacrylamide gel electrophoresis for 2.5 h at 4 °C under
reducing and denaturing conditions. Gels were blotted onto the PVDF membrane
for 45 min at room temperature, incubated in 1% blocking reagent for 1 h at
37 °C and incubated with the primary antibodies against CYP2E1 (1:5000),
HO-1 (1:2000), TNF-
a
(1:1000), p53 (1:1000) and p-p53 (1:1000) overnight at
4°C. The membranes were washed in Tris-buffered saline plus Tween 20 (TBST)
and incubated with the secondary antibodies for 1 h at 37 °C. Finally, the
membranes were exposed to ECL substrate, bands were detected and scanned
(Allianze 4.0, Cambridge, UK). The intensity of the bands was assayed by com-
puter image analysis software (NIH Image J software, available at http://rsb.info.
nih.gov/ij).
2.8. Statistical analysis
Data were analyzed using StatSoft STATISTICA version 12.0 software by Krus-
kal-Wallis test and post hoc comparisons were carried out with Dunn’s multiple
comparison test. Results of multiple comparisons tests were indicated by different
letters. Means with letters in common are not significantly different from each
other. Values in the text are means ± standard deviation (SD). Differences with
P< 0.05 were considered to be statistically significant.
3. Results
3.1. Kidney weight and serum markers of kidney damage
Both body weight and relative kidney weight of control and RA-
treated mice were similar (Table 1). CP-intoxication resulted in a
significant body weight reduction and increased relative kidney
weight. These changes were significantly ameliorated by RA. The
serum creatinine and BUN levels were significantly higher in CP-
treated animals when compared to control mice. Treatment with
RA significantly decreased the creatinine and BUN levels in a
dose-dependent manner.
3.2. Kidney histopathology
Normal tubular morphology was observed in cortical and
medullary regions of kidney in control mice (Fig. 1A) and mice
treated with RA (Fig. 1B). CP administration resulted in
322 R. Domitrovic
´et al./ Food and Chemical Toxicology 66 (2014) 321–328
histopathological changes in the corticomedullary junction of mice
kidneys, including tubular epithelial damage, tubular dilatation
and intratubular cast formation (Fig. 1C). Treatment with RA re-
sulted in a dose-dependent restoration of kidney morphology
(Fig. 1D and E), with only occasional degenerating tubules in
CP + RA5 treated mice (Fig. 1F).
3.3. Markers of oxidative stress
Oxidative stress, including lipid peroxidation, is a common fea-
ture of renal injury following cisplatin administration. The kidneys
of control mice and mice treated with RA were 4-HNE immunoneg-
ative (Fig. 2A and B), whereas CP administration induced strong
immunoreactivity toward this oxidative stress marker (Fig. 2C).
High 4-HNE immunopositivity was still present in mice receiving
the lowest dose of RA (Fig. 2D) but subsequently decreased by
higher doses (Fig. 2E, F and G). Similarly, administration of CP in-
creased CYP2E1 and HO-1 expression in mouse kidney lysates
(Fig. 2H) when compared to control mice and RA-treated animals.
CYP2E1 and HO-1 immunoreactivity decreased following RA
administration, with the expression level similar to control mice
by the highest RA dose.
Table 1
Body weight change, relative kidney weight and serum markers of kidney damage.
Body weight change (%) Relative kidney weight Creatinine (
l
mol/L) BUN (mmol/L)
Control +0.88 ± 0.7
a
8.17 ± 0.15
a
31.2 ± 0.9
a
16.3 ± 2.8
a
RA 5 mg/kg +0.16 ± 1.2
a
8.07 ± 0.38
a
33.2 ± 2.1
a
16.8 ± 1.7
a
CP 13 mg/kg 20.6 ± 3.1
b
9.35 ± 0.48
b
165.4 ± 14.7
b
89.6 ± 3.9
b
CP 13 mg/kg + RA 1 mg/kg 17.9 ± 1.8
b
9.26 ± 0.11
b
130.1 ± 9.7
c
69.2 ± 2.8
c
CP 13 mg/kg + RA 2 mg/kg 14.3 ± 2.2
c
8.56 ± 0.26
c
60.6 ± 10.5
d
29.2 ± 4.1
d
CP 13 mg/kg + RA 5 mg/kg 11.42 ± 1.7
c
8.04 ± 0.20
a
35.3 ± 4.6
a
14.8 ± 1.8
a
Mice were administered RA orally once daily for two consecutive, 48 h after intraperitoneal injection of cisplatin (CP). Animals were sacrificed 4 days after injection of CP.
Relative kidney weight is expressed as [(kidney weight/body weight) * 1000]. Each value represents the mean ± SD for 5 mice. Means within columns sharing the same letter
are not significantly different from each other (P<0.05).
Fig. 1. Representative histological abnormalities in the corticomedullary junction of mice kidneys. Mice treated with vehicle (A), RA 5 mg/kg (B), CP (C), CP + RA 1 mg/kg (D),
CP + RA 2 mg/kg (E) and CP + RA 5 mg/kg (F). Arrows show damaged tubular cells, stars show intratubular cast formation. Representative results from 5 similarly treated mice.
HE staining. Original magnification 400. Tubular injury score (G). Each value represents the mean ± SD for 5 mice. Means sharing the same letter are not significantly
different from each other (P< 0.05).
R. Domitrovic
´et al./ Food and Chemical Toxicology 66 (2014) 321–328 323
3.4. Inflammatory response
Immunohistochemical evaluation showed more intense expres-
sion of NF-
j
B p65 subunit in kidneys of mice exposed to CP
(Fig. 3C) when compared to controls (Fig. 3A) or RA-treated mice
(Fig. 3B). NF-
j
B p65 overexpression was noticed both in the
cytoplasm and the nuclei of tubular cells. Treatment with RA ame-
liorated NF-
j
B immunopositivity in a dose-dependent manner
(Fig. 3D, E, F and G). The lowest dose of RA reduced nuclear
NF-
j
B p65 immunopositivity, which diminished by higher doses
of RA, with concomitant reduction of the cytoplasmic staining
intensity. Western blot analysis revealed increased TNF-
a
expres-
sion in the kidneys of CP-treated mice when compared to control
and RA-treated mice (Fig. 3H).
3.5. Apoptosis
Apoptosis in the kidneys was determined by measuring the
expression of proapoptotic p53 protein and apoptotic executor
cleaved caspase-3. We subsequently determined p53 phosphoryla-
tion, which represents an important regulatory mechanism for
p53. As shown in Fig. 4A and B, the immunohistochemical analysis
revealed no detectable active caspase-3 staining in either cortical
or medullar structures of kidneys in control and RA-treated mice.
However, active caspase-3 staining was detected in the corticome-
dullary junction of CP-treated animals (Fig. 4C). The antiapoptotic
activity of RA was evident by dose-dependent reduction in cleaved
caspase-3 expression (Fig. 4D, E, F and G), which coincided with
decreased expression of p53 and p-p53 in the kidneys (Fig. 4H).
4. Discussion
CP is still a frequently used chemotherapeutic agent, despite of
its frequent adverse effects, including nephrotoxicity (Miller et al.,
2010). The toxic effects of CP occurs through increased oxidative
stress, tubulointerstitial inflammation and apoptosis (Sahu et al.,
2011). Therefore, combinatorial strategies which target multiple
mechanisms, including suppression of free radical production,
Fig. 2. 4-Hydroxynonenal (4-HNE) expression in the corticomedullary junction of mice kidneys. Mice treated with vehicle (A), RA 5 mg/kg (B), CP (C), CP + RA 1 mg/kg (D),
CP + RA 2 mg/kg (E) and CP + RA 5 mg/kg (F). Arrows show 4-HNE immunopositive cells. Representative results from 5 similarly treated mice. Immunohistochemistry
staining, original magnification 400. Measurement of the intensity of 4-HNE immunostaining (G). Western blot analysis of cytochrome P450 2E1 (CYP2E1) and heme
oxygenase-1 (HO-1) expression in mice kidney lysates (H). Each value represents the mean ± SD for 5 mice. Means sharing the same letter are not significantly different from
each other (P< 0.05).
324 R. Domitrovic
´et al./ Food and Chemical Toxicology 66 (2014) 321–328
anti-inflammatory and cytoprotective activities in the kidney, may
represent an optimal strategy for the prevention of CP-mediated
nephrotoxicity (Sahu et al., 2013). Based on antioxidant, anti-
inflammatory and antiapoptotic properties of RA, the present study
was undertaken to examine the protective effects of RA against CP-
induced kidney injury. The results of the current study showed that
RA may ameliorate tubular damage in CP-intoxicated animals,
which was achieved through the reduction of oxidative stress,
inflammation and apoptosis in the kidneys.
In the current study, CP administration resulted in a severe
nephropathy, accompanied by impaired histological features of
the kidneys. Body weight loss was used as a measure of CP-induced
toxicity (Leonetti et al., 2003). Increased creatinine and BUN in CP-
treated animals indicated reduction in the glomerular filtration
rate (Coca and Parikh, 2008). Similarly, increased relative kidney
weight was attributed to urine retention due to casts-induced
tubular obstruction (Schrier et al., 2004). However, post-treatment
with RA markedly ameliorated these pathological changes.
Oxidative stress plays a key role in the pathophysiology of
CP-mediated kidney injury (Chirino et al., 2008b; Matsushima
et al., 1998). CP induces the overproduction of free radicals,
including superoxide anion ðO
2
Þand hydroxyl radical ð
OHÞ(Shi-
no et al., 2003; Davis et al., 2001). Free radical scavengers prevent
acute renal failure through attenuation of tubular damage, en-
hanced regenerative response of tubular cells and preservation
of renal blood flow (Matsushima et al., 1998). As shown in
Fig. 2, renal oxidative stress induced by CP administration, dem-
onstrated as 4-HNE formation, was associated with kidney
parenchimal cells. This is in agreement with similar findings by
other authors, which demonstrated increased lipid peroxidation
in mice and rat kidneys and proximal tubular cells in vitro (Tanabe
et al., 2012; Pan et al., 2009; Chirino et al., 2008a). The increase in
4-HNE formation was subsequently decreased by RA, suggesting
the improvement in antioxidant defense system. Additionally,
RA ameliorated the increase in CYP2E1 expression. The catalytic
activity of CYP family of enzymes is associated with increased
production of free radicals and deterioration of antioxidant de-
fense (Kusirisin et al., 2009). Moreover, the interaction of CYP2E1
by CP is considered as the central pathway by which CP promotes
oxidative stress in kidneys (Liu and Baliga, 2003). In turn, CYP2E1
Fig. 3. Nuclear factor kappaB (NF-
j
B) cellular localization in the corticomedullary junction of mice kidneys. Mice treated with vehicle (A), RA 5 mg/kg (B), CP (C), CP + RA
1 mg/kg (D), CP + RA 2 mg/kg (E) and CP + RA 5 mg/kg (F). Arrows show NF
j
B immunopositive nuclei. Representative results from 5 similarly treated mice.
Immunohistochemistry staining, original magnification 400. Measurement of the intensity of NF-
j
B immunostaining (G). Western blot analysis of tumor necrosis
factor-alpha (TNF-
a
) expression in kidney lysates (H). Each value represents the mean ± SD for 5 mice. Each value represents the mean ± SD for 5 mice. Means sharing the
same letter are not significantly different from each other (P< 0.05).
R. Domitrovic
´et al./ Food and Chemical Toxicology 66 (2014) 321–328 325
inhibitors were shown to reduce free radical production and ame-
liorate tubular cell cytotoxicity (Liu et al., 2002). The increase in
expression of CYP2E1 in the kidneys of CP-treated animals ob-
served in this study is in agreement with similar findings by
Wu et al. (2011). Nevertheless, Liu and Baliga (2003) reported de-
creased levels of CYP2E1 in the kidneys of CP-treated mice, how-
ever, this could be attributed to a strain-specific differences in
renal expression of CYP isoforms (Muller et al., 2007). Notably,
the inhibition of renal CYP2E1 expression by RA agree with CYP
inhibiting properties of natural antioxidant compounds observed
in various cell types and tissues (Wu et al., 2011; Kimura et al.,
2010; Kusirisin et al., 2009; Wang et al., 2006). The increase in
CYP2E1 expression in the kidneys of CP-treated mice observed
in the current study was accompanied by the HO-1 overexpres-
sion, suggesting that the induction of CYP2E1 by CP resulted in
the mobilization of this cytoprotective enzyme. Nevertheless, RA
acted as an inhibitor of both CYP2E1 and HO-1 expression, sug-
gesting that the decrease in CYP2E1 expression plays an impor-
tant role in the nephroprotective activity of RA.
Inflammation plays an important role in the pathogenesis of CP
nephrotoxicity. It has been suggested that oxidative stress causes
local inflammation through up-regulation of pro-inflammatory
cytokines, including TNF-
a
(Hassan et al., 2012). Administration
of nephrotoxic doses of CP frequently results in the activation of
the NF-
j
B signaling pathway and increased expression of pro-
inflammatory mediators in renal parenchimal cells both in vivo
and in vitro (Jia et al., 2011; Pan et al., 2009; Ramesh and Reeves,
2004). Consequently, NF-
j
B inhibitors have shown protective
activity against CP-induced nephrotoxicity (Francescato et al.,
2007) and the suppression of pro-inflammatory mediators resulted
in reduced kidney tubular damage. Most recently, Amin et al.
(2012) showed that CP-mediated inflammation in rats was amelio-
rated by Ginkgo biloba extract through the inhibition of the NF-
j
B
pathway (Amin et al., 2012). Similarly, the inhibition of TNF-
a
by
salycilates resulted in decreased CP-mediated nephrotoxicity (Ra-
mesh and Reeves, 2004). In the current study, administration of
RA to CP-treated mice reduced NF-
j
B p65 nuclear translocation
in proximal tubular cells and expression of TNF-
a
, indicating the
Fig. 4. Cleaved caspase-3 expression in the corticomedullary junction of mice kidneys. Mice treated with vehicle (A), RA 5 mg/kg (B), CP (C), CP + RA 1 mg/kg (D),CP+RA
2 mg/kg (E) and CP + RA 5 mg/kg (F). Arrows show cleaved caspase-3 positive cells. Representative results from 5 similarly treated mice. Immunohistochemistry staining,
original magnification 400. Measurement of the intensity of cleaved caspase-3 immunostaining (G). Western blot analysis of p53 and p-p53 (Ser15) expression in kidney
lysates (H). Each value represents the mean ± SD for 5 mice. Each value represents the mean ± SD for 5 mice. Means sharing the same letter are not significantly different from
each other (P< 0.05).
326 R. Domitrovic
´et al./ Food and Chemical Toxicology 66 (2014) 321–328
anti-inflammatory effect of RA, which is in agreement with similar
findings in CCl
4
-induced liver injury (Domitrovic
´et al., 2013). We
hypothesize that inflammatory response is the result of oxidative
stress which occurs due to cisplatin metabolisation. The ability of
RA to abrogate the induction of the NF-
j
B pathway suggests its po-
tential use as an anti-inflammatory agent in CP-induced kidney
injury.
Reactive oxygen species were also suggested as an initiator of
apoptotic cell death triggered by CP (Kannan and Jain, 2000). Apop-
tosis of cancer cells is considered as the key mechanism of CP che-
motherapeutic activity (Bragado et al., 2007). Nevertheless, the
same mechanism seems to be involved in the development of kid-
ney injury. Thus, caspase-3, the executive cell death protease, has
been activated by CP treatment in renal proximal tubular cells
(Kaushal et al., 2001) and rat kidneys (Park et al., 2012). The tumor
suppressor p53, which plays an essential role in the induction of
apoptosis (Amaral et al., 2010; Shen and White, 2001), is rapidly
up-regulated following CP treatment and the inhibition of p53
can inhibit CP-induced apoptosis (Wei et al., 2007). p53 stability
and transcriptional activity are modulated by post-translational
modifications including phosphorylation (Sakaguchi et al., 1998).
Phosphorylation at N-terminal Ser15 resulted in reduced DNA re-
pair, enhanced DNA-binding activity and the induction of apopto-
sis (Offer et al., 2002). Previously, Pabla et al. (2008) and Jiang et al.
(2004) suggested a role for p-p53 (Ser15) in CP-induced renal cell
apoptosis in vitro and in vivo. Consistently with previous findings,
in the present study, the expression of p53, p-p53 (Ser15) and
cleaved caspase-3 increased in the kidneys of CP-treated mice. Or-
ally administered RA reduced the overexpression of p53, p-p53 and
cleaved caspase-3, suggesting the inhibition of tubular apoptosis.
In conclusion, the results of the current study suggest that the
suppression of oxidative stress, inflammatory response and apop-
totic cell death by RA could be the effective strategy for the treat-
ment of CP-induced kidney injury. However, controlled clinical
trials are necessary to confirm the therapeutic activity of RA in
the patients receiving CP chemotherapy.
Conflict of Interest
The authors declare that there are no conflicts of interest.
Transparency Document
The Transparency Document associated with this article can be
found in the online version.
Acknowledgements
This research was supported by grants from Ministry of Science,
Education and Sport, Republic of Croatia (project 062-0000000-
3554).
References
Amaral, J.D., Xavier, J.M., Steer, C.J., et al., 2010. The role of p53 in apoptosis. Discov.
Med. 9, 145–152.
Amin, A., Abraham, C., Hamza, A.A., et al., 2012. A standardized extract of Ginkgo
biloba neutralizes CP-mediated reproductive toxicity in rats. J. Biomed.
Biotechnol. 2012, 362049.
Bragado, P., Armesilla, A., Silva, A., et al., 2007. Apoptosis by cisplatin requires p53
mediated p38alpha MAPK activation through ROS generation. Apoptosis 12,
1733–1742.
Brahmi, D., Ayed, Y., Hfaiedh, M., et al., 2012. Protective effect of cactus cladode
extract against cisplatin induced oxidative stress, genotoxicity and apoptosis in
balb/c mice. combination with phytochemical composition. BMC Complement.
Altern. Med. 12, 111.
Chirino, Y.I., Trujillo, J., Sánchez-González, D.J., et al., 2008a. Selective iNOS
inhibition reduces renal damage induced by cisplatin. Toxicol. Lett. 176, 48–57.
Chirino, Y.I., Sánchez-González, D.J., Martínez-Martínez, C.M., et al., 2008b.
Protective effects of apocynin against cisplatin-induced oxidative stress and
nephrotoxicity. Toxicology 245, 18–23.
Chu, X., Ci, X., He, J., et al., 2012. Effects of a natural prolyl oligopeptidase inhibitor,
rosmarinic acid, on lipopolysaccharide-induced acute lung injury in mice.
Molecules 17, 3586–3598.
Coca, S.G., Parikh, C.R., 2008. Urinary biomarkers for acute kidney injury:
perspectives on translation. Clin. J. Am. Soc. Nephrol. 3, 481–490.
Costa, R.S., Carneiro, T.C., Cerqueira-Lima, A.T., et al., 2012. Ocimum gratissimum
Linn. and rosmarinic acid, attenuate eosinophilic airway inflammation in an
experimental model of respiratory allergy to Blomia tropicalis. Int.
Immunopharmacol. 13, 126–134.
Davis, C.A., Nick, H.S., Agarwal, A., 2001. Manganese superoxide dismutase
attenuates cisplatin-induced renal injury: importance of superoxide. J. Am.
Soc. Nephrol. 12, 2683–2690.
Domitrovic
´, R., Škoda, M., Vasiljev Marchesi, V., 2013. Rosmarinic acid ameliorates
acute liver damage and fibrogenesis in carbon tetrachloride-intoxicated mice.
Food Chem. Toxicol. 51, 370–378.
Domitrovic
´, R., Jakovac, H., Marchesi, V.V., et al., 2012. Preventive and therapeutic
effects of oleuropein against carbon tetrachloride-induced liver damage in
mice. Pharmacol. Res. 65, 451–464.
Domitrovic
´, R., Jakovac, H., Blagojevic
´, G., 2011. Hepatoprotective activity of
berberine is mediated by inhibition of TNF-
a
, COX-2 and iNOS expression in
CCl
4
-intoxicated mice. Toxicology 280, 33–43.
Domitrovic
´, R., Jakovac, H., Milin, C
ˇ., Radoševic
´-Stasic
´, B., 2009. Dose- and time-
dependent effects of luteolin on carbon tetrachloride-induced hepatotoxicity in
mice. Exp. Toxicol. Pathol. 61, 581–589.
Francescato, H.D.C., Costa, R.S., Scavone, C., et al., 2007. Parthenolide reduces
cisplatin-induced renal damage. Toxicology 230, 64–75.
Giaccone, G., 2009. Clinical perspectives on platinum resistance. Drugs 59, 9–17.
Hartmann, J.T., Lipp, H.P., 2003. Toxicity of platinum compounds. Expert Opin.
Pharmacother. 4, 889–901.
Hassan, I., Chibber, S., Khan, A.A., et al., 2012. Riboflavin ameliorates cisplatin
induced toxicities under photoillumination. PLoS One 7, e36273.
Jia, Z., Wang, N., Aoyagi, T., et al., 2011. Amelioration of cisplatin nephrotoxicity by
genetic or pharmacologic blockade of prostaglandin synthesis. Kidney Int. 79, 77–
88.
Jiang, M., Yi, X., Hsu, S., Wang, C.Y., et al., 2004. Role of p53 in cisplatin-induced
tubular cell apoptosis: dependence on p53 transcriptional activity. Am. J.
Physiol. Renal Physiol. 287, F1140–F1147.
Kang, K.P., Park, S.K., Kim, D.H., et al., 2011. Luteolin ameliorates cisplatin-induced
acute kidney injury in mice by regulation of p53-dependent renal tubular
apoptosis. Nephrol. Dial. Transplant. 26, 814–822.
Kannan, K., Jain, S.K., 2000. Oxidative stress and apoptosis. Pathophysiology 7, 153–
163.
Kaushal, G., Kaushal, V., Hong, X., et al., 2001. Role and regulation of activation of
caspases in cisplatin-induced injury to renal tubular epithelial cells. Kidney Int.
60, 1726–1736.
Kimura, Y., Ito, H., Ohnishi, R., et al., 2010. Inhibitory effects of polyphenols on human
cytochrome P450 3A4 and 2C9 activity. Food Chem. Toxicol. 48, 429–435.
Kusirisin, W., Jaikang, C., Chaiyasut, C., et al., 2009. Effect of polyphenolic compounds
from Solanum torvum on plasma lipid peroxidation, superoxide anion and
cytochrome P450 2E1 in human liver microsomes. Med. Chem. 5, 583–588.
Lee, H.J., Cho, H.S., Park, E., et al., 2008. Rosmarinic acid protects human
dopaminergic neuronal cells against hydrogen peroxide-induced apoptosis.
Toxicology 250, 109–115.
Leemans, J.C., Stokman, G., Claessen, N., et al., 2005. Renal-associated TLR2 mediates
ischemia/reperfusion injury in the kidney. J. Clin. Invest. 115, 2894–2903.
Leonetti, C., Biroccio, A., Gabellini, C., et al., 2003. Alpha-tocopherol protects against
cisplatin-induced toxicity without interfering with antitumor efficacy. Int. J.
Cancer 104, 243–250.
Liu, H., Baliga, M., Baliga, R., 2002. Effect of cytochrome P450 2E1 inhibitors on
cisplatin-induced cytotoxicity to renal proximal tubular epithelial cells.
Anticancer Res. 22, 863–868.
Liu, H., Baliga, R., 2003. Cytochrome P450 2E1 null mice provide novel protection
against cisplatin-induced nephrotoxicity and apoptosis. Kidney Int. 63, 1687–
1696.
Matsushima, H., Yonemura, K., Ohishi, K., et al., 1998. The role of oxygen free
radicals in cisplatin-induced acute renal failure in rats. J. Lab. Clin. Med. 131,
518–526.
Miller, R.P., Tadagavadi, R.K., Ramesh, et al., 2010. Mechanisms of cisplatin
nephrotoxicity. Toxins (Basel) 2, 2490–2518.
Moreno, S., Scheyer, T., Romano, C.S., et al., 2006. Antioxidant and antimicrobial
activities of rosemary extracts linked to their polyphenol composition. Free
Radic. Res. 40, 223–231.
Muller, D.N., Schmidt, C., Barbosa-Sicard, E., et al., 2007. Mouse Cyp4a isoforms:
enzymatic properties, gender- and strain-specific expression, and role in renal
20-hydroxyeicosatetraenoic acid formation. Biochem. J. 403, 109–118.
Ninsontia, C., Pongjit, K., Chaotham, C., et al., 2011. Silymarin selectively protects
human renal cells from cisplatin-induced cell death. Pharm. Biol. 49, 1082–
1090.
Offer, H., Erez, N., Zurer, I., et al., 2002. The onset of p53-dependent DNA repair or
apoptosis is determined by the level of accumulated damaged DNA.
Carcinogenesis 23, 1025–1032.
R. Domitrovic
´et al./ Food and Chemical Toxicology 66 (2014) 321–328 327
Özgen, U., Mavi, A., Terzi, Z., et al., 2011. Relationship between chemical structure
and antioxidant activity of luteolin and its glycosides isolated from Thymus
sipyleus subsp. sipyleus var. sipyleus. Rec. Nat. Prod. 5, 12–21.
Pabla, N., Huang, S., Mi, Q.S., et al., 2008. ATR-Chk2 signaling in p53 activation and
DNA damage response during cisplatin-induced apoptosis. J. Biol. Chem. 283,
6572–6583.
Pan, H., Shen, Z., Mukhopadhyay, P., et al., 2009. Anaphylatoxin C5a contributes to
the pathogenesis of cisplatin-induced nephrotoxicity. Am. J. Physiol. Renal
Physiol. 296, F496–F504.
Park, J.W., Cho, J.W., Joo, S.Y., et al., 2012. Paricalcitol prevents cisplatin-induced
renal injury by suppressing apoptosis and proliferation. Eur. J. Pharmacol. 683,
301–309.
Petersen, M., Simmonds, M.S., 2003. Rosmarinic acid. Phytochemistry 62, 121–125.
Ramesh, G., Reeves, W.B., 2004. Salicylate reduces cisplatin nephrotoxicity by
inhibition of tumor necrosis factor-
a
. Kidney Int. 65, 490–498.
Sakaguchi, K., Herrera, J.E., Saito, S., et al., 1998. DNA damage activates p53 through
a phosphorylation-acetylation cascade. Genes Dev. 12, 2831–2841.
Sakihama, Y., Cohen, M.F., Grace, S.C., et al., 2002. Plant phenolic antioxidant and
prooxidant activities: phenolics-induced oxidative damage mediated by metals
in plants. Toxicology 177, 67–80.
Sahu, B.D., Kuncha, M., Sindhura, G.J., et al., 2013. Hesperidin attenuates cisplatin-
induced acute renal injury by decreasing oxidative stress, inflammation and
DNA damage. Phytomedicine 20, 453–460.
Sahu, B.D., Reddy, K.K.R., Putcha, U.K., et al., 2011. Carnosic acid attenuates renal
injury in an experimental model of rat cisplatin-induced nephrotoxicity. Food
Chem. Toxicol. 49, 3090–3097.
Sanchez-Gonzalez, P.D., Lopez-Hernandez, F.J., Perez-Barriocanal, F., et al., 2011.
Quercetin reduces cisplatin nephrotoxicity in rats without compromising its
anti-tumour activity. Nephrol. Dial. Transplant. 26, 3484–3495.
Schrier, R.W., Wang, W., Poole, B., et al., 2004. Acute renal failure: definitions,
diagnosis, pathogenesis, and therapy. J. Clin. Invest. 114, 5–14.
Shen, Y., White, E., 2001. P53-dependent apoptosis pathways. Adv. Cancer Res. 82,
55–84.
Shino, Y., Itoh, Y., Kubota, T., et al., 2003. Role of poly(ADP-ribose)polymerase in
cisplatin-induced injury in LLC-PK1 cells. Free Radic. Biol. Med. 35, 966–997.
Sroka, Z., Cisowski, W., 2003. Hydrogen peroxide scavenging, antioxidant and anti-
radical activity of some phenolic acids. Food Chem. Toxicol. 41, 753–758.
Sung, M.J., Kim, D.H., Jung, Y.J., et al., 2008. Genistein protects the kidney from
cisplatin-induced injury. Kidney Int. 74, 1538–1547.
Swarup, V., Ghosh, J., Ghosh, S., et al., 2007. Antiviral and antiinflammatory effects
of rosmarinic acid in an experimental murine model of Japanese encephalitis.
Antimicrob. Agents Chemother. 51, 3367–3370.
Tanabe, K., Tamura, Y., Lanaspa, M.A., et al., 2012. Epicatechin limits renal injury by
mitochondrial protection in cisplatin nephropathy. Am. J. Physiol. Renal Physiol.
303, F1264–F1274.
Tavafi, M., Ahmadvand, H., 2011. Effect of rosmarinic acid on inhibition of
gentamicin induced nephrotoxicity in rats. Tissue Cell 43, 392–397.
Venkatachalam, K., Gunasekaran, S., Jesudoss, V.A., et al., 2013. The effect of
rosmarinic acid on 1,2-dimethylhydrazine induced colon carcinogenesis. Exp.
Toxicol. Pathol. 65, 409–418.
Vijayan, F.P., Rani, V.K., Vineesh, V.R., et al., 2007. Protective effect of Cyclea peltata
Lam on cisplatin-induced nephrotoxicity and oxidative damage. J. Basic Clin.
Physiol. Pharmacol. 18, 101–114.
Wang, Y.,Lee, K.W., Chan, F.L., et al.,2006. The red wine polyphenolresveratrol displays
bilevel inhibition on aromatase in breast cancercells. Toxicol. Sci. 92, 71–77.
Wei, Q., Dong, G., Yang, T., et al., 2007. Activation and involvement of p53 in
cisplatin-induced nephrotoxicity. Am. J. Physiol. Ren. Physiol. 293, F1282–
F1291.
Wu, C.T., Sheu, M.L., Tsai, K.S., et al., 2011. Salubrinal, an eIF2
a
dephosphorylation
inhibitor, enhances cisplatin-induced oxidative stress and nephrotoxicity in a
mouse model. Free Radic. Biol. Med. 51, 671–680.
Yao, X., Panichpisal, K., Kurtzman, N., et al., 2007. Cisplatin nephrotoxicity: a review.
Am. J. Med. Sci. 334, 115–124.
Zhang, Y., Li, X., Wang, Z., 2010. Antioxidant activities of leaf extract of Salvia
miltiorrhiza Bunge and related phenolic constituents. Food Chem. Toxicol. 48,
2656–2662.
328 R. Domitrovic
´et al./ Food and Chemical Toxicology 66 (2014) 321–328
... Carbon tetrachloride destroys the mitochondrial phospholipid bilayer in hepatocytes and induces caspase 3 dependent apoptosis (Domitrović et al., 2013;Tao et al., 2012). In vitro and in vivo studies have shown that hepatocyte apoptosis is determined immunohistochemically with caspase activity in CCl4 induced liver damage (Domitrović et al., 2013;Hassan et al., 2012;Tao et al., 2012). ...
... Carbon tetrachloride destroys the mitochondrial phospholipid bilayer in hepatocytes and induces caspase 3 dependent apoptosis (Domitrović et al., 2013;Tao et al., 2012). In vitro and in vivo studies have shown that hepatocyte apoptosis is determined immunohistochemically with caspase activity in CCl4 induced liver damage (Domitrović et al., 2013;Hassan et al., 2012;Tao et al., 2012). Researchers have reported increased caspase 3 activity in these studies. ...
... Researchers have reported increased caspase 3 activity in these studies. In the present study, in CCl4 administered groups, caspase 3 was evaluated and was consistent with the study results of the mentioned researchers (Domitrović et al., 2013;Hassan et al., 2012). It is suggested that CCl4 induced free radical formation, by decreasing endogenous antioxidant enzymes, induces hepatocyte apoptosis by caspase 3 in CCl4 toxicity (Jiang et al., 2012). ...
... Thus, ALT and AST are hepatic enzymes located in the cytoplasm of hepatocytes [42]. Hence, the elevated serum activities of ALT and AST indicating hepatic structural damage and impairment of the liver functions [43]. BUN is the first marker of renal injury, while SCr is the most reliable renal marker, and those parameters only increased when the bulk of renal functions is lost [44]. ...
Article
Full-text available
Background Hepatorenal syndrome is a life-threatening medical complication of liver cirrhosis. Hepatic cirrhosis is commonly accompanied by rapid failure of renal functions. Thioacetamide (TAA) is a potent hepatotoxin and a class 2-type carcinogen. Ubiquinone (Coq 10 ) and lactoferrin (LF) are potent antioxidants with antifibrotic and antiinflammatory effects. However, whether Coq 10 and LF reduce the hepatorenal injury induced by TAA remains unclear. Here, we investigated the potential protective effect of both/or Coq 10 and LF in ameliorating TAA-induced hepatorenal injury and the role of WNT4 gene expression in detecting TAA-induced renal injury in rats. Seventy healthy and mature male Sprague Dawley rats, weighting (200 g ± 20 g) and aging (4–6) weeks were randomly divided into seven groups ( n = 10): control, Coq 10 , LF, TAA, TAA + Coq 10 , TAA + LF, and TAA + Coq 10 + LF. The hepatorenal injury was induced through intraperitoneal (i.p.) injection of TAA (150 mg/kg/twice/weekly) for nine weeks. Coq 10 (10 mg/kg/day) and LF (200 mg/kg/day) were orally administered for nine weeks. Results TAA induced marked hepatorenal damage, evident by the significant increase in the alanine aminotransferase (ALT), aspartate transaminase (AST), serum creatinine (SCr) activities, and the blood urea nitrogen (BUN) level. Besides, the significant increases in concentrations of malondialdehyde (MDA) and nitric oxide (NOx) together with significant decreases in the activities of catalase (CAT) and superoxide dismutase (SOD). The histopathological analysis of the TAA group showed obvious fibrosis, steatosis, and inflammation of the hepatic parenchyma as well as severe glomerular and tubular damage of the renal parenchyma. In addition, TAA induced marked ultrastructural alterations and up-regulation in the expression of the WNT4 gene in the kidney. Meanwhile, the biochemical, histopathological, and ultrastructural alterations were significantly decreased with significant down-regulation in the expression of WNT4 in the groups exposed to TAA and treated with Coq 10 and LF. Conclusion Our data suggested that Coq 10 and LF could have protective effects on TAA hepatorenal damage, through improving the hepatic and renal functions, reduction of oxidative stress, structural and ultrastructural alterations, besides down-regulation in the expression of WNT4.
... RosA, a phenolic compound, has shown promising potential in the treatment of liverrelated diseases [16]. It has been observed to exhibit an anti-fibrotic effect against carbon tetrachloride-induced liver fibrosis [31], an anti-inflammatory effect in autoimmune hepatitis [32], and the ability to stimulate hepatocyte proliferation through activation of the mTOR/S6K pathway [33]. RosA has been found to possess the ability to inhibit apoptosis in a mouse model of nonalcoholic steatohepatitis through the SIRT1/NF-κB pathway [34], and RosA was observed to attenuate chromium-induced hepatic oxidative damage [35]. ...
Article
Full-text available
The objective of this study is to examine the potential protective effect of rosmarinic acid (RosA) encapsulated within nanoliposomes (RosA-LIP) on hepatic damage induced by iron overload. The characteristics, stability, and release of RosA-LIP in vitro were identified. The mice were randomly assigned to five groups: Control, Model, Model+DFO (DFO), Model+RosA (RosA), and Model+RosA-LIP (RosA-LIP). The iron overload model was induced by administering iron dextran (i.p.). The DFO, RosA, and RosA-LIP groups received iron dextran and were subsequently treated with DFO, RosA, and RosA-LIP for 14 days. We developed a novel formulation of RosA-LIP that exhibited stability and controlled release properties. Firstly, RosA-LIP improved liver function and ameliorated pathological changes in a mouse model of iron overload. Secondly, RosA-LIP demonstrated the ability to enhance the activities of T-SOD, GSH-Px, and CAT, while reducing the levels of MDA and 4-HNE, thereby effectively mitigating oxidative stress damage induced by iron overload. Thirdly, RosA-LIP reduced hepatic iron levels by downregulating FTL, FTH, and TfR1 levels. Additionally, RosA-LIP exerted a suppressive effect on hepcidin expression through the BMP6-SMAD1/5/8 signaling pathway. Furthermore, RosA-LIP upregulated FPN1 expression in both the liver and duodenum, thereby alleviating iron accumulation in these organs in mice with iron overload. Notably, RosA exhibited a comparable iron chelation effect, and RosA-LIP demonstrated superior efficacy in mitigating liver damage induced by excessive iron overload. RosA-LIP exhibited favorable sustained release properties, targeted delivery, and efficient protection against iron overload-induced liver damage. Graphical Abstract A schematic representation of the proposed protective mechanism of rosmarinic acid liposome during iron overload. Once RosA-LIP is transported into cells, RosA is released. On the one hand, RosA attenuates the BMP6-SMAD1/5/8-SMAD4 signaling pathway activation, leading to inhibiting hepcidin transcription. Then, the declined hepcidin contacted the inhibitory effect of FPN1 in hepatocytes and duodenum, increasing iron mobilization. On the other hand, RosA inhibits TfR1 and ferritin expression, which decreases excessive iron and oxidative damage.
... Among these components, rosmarinic acid was found to exhibit both antioxidant [22] and anti-inflammatory effects [75]. In detail, rosmarinic acid elicits neuroprotection in ischemic stroke via Nrf2/HO-1 pathway [76] and ameliorates acute liver damage and fibrogenesis accompanied by enhanced Nrf2/HO-1 expression [77]. Rosmarinic acid is also known to inhibit LPS-induced NO production and iNOS in RAW264.7 macrophage via suppressing NF-κB signaling [78]. ...
Article
Full-text available
Ehretia tinifolia (E. tinifolia) L., an evergreen tree with substantial biological activity, including antioxidant and anti-inflammatory effects, has been used in many herbal and traditional medicines. To elucidate its antioxidant and anti-inflammatory activity and the underlying mechanisms, we applied a methanol extract of E. tinifolia (ETME) to lipopolysaccharide (LPS)-stimulated mouse immortalized Kupffer cells. ETME suppressed the LPS-induced increase in nitric oxide, a mediator for oxidative stress and inflammation, and restored LPS-mediated depletion of total glutathione level by stabilizing antioxidative nuclear factor erythroid 2-related factor 2 (Nrf2) and the subsequent increase in heme oxygenase-1 levels. Furthermore, ETME inhibited the LPS-induced production of pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin (IL)-1β, and IL-6. The inhibitory effects of ETME on pro-inflammatory responses were regulated by ETME-mediated dephosphorylation of mitogen-activated protein kinases (MAPKs: p38, p44/p42, and stress-associated protein kinase/c-Jun N-terminal kinase) and inhibition of nuclear localization of nuclear factor kappa B (NF-κB). These results suggest that ETME is a possible candidate for protecting Kupffer cells from LPS-mediated oxidative stress and excessive inflammatory responses by activating antioxidant Nrf2/HO-1 and inhibiting pro-inflammatory NF-κB and MAPKs, respectively.
... Rats were orally administered distilled water (control group, HCl group) or RA (50 mg/kg/day; Carbosynth Ltd., Berkshire, UK, #FR02310; HCl + RA group) under isoflurane inhalation anesthesia once a day immediately after surgery and for the following three days (a total of four times). The dose of RA was set by referring to previous studies in rats [18] and mice [19]. Three days after catheter implantation, intravesical pressure was measured in conscious rats. ...
Article
Full-text available
Shiso (Perilla frutescens var crispa f. purprea) is a traditional medicinal herb that exerts anti-inflammatory effects and alleviates lower urinary tract symptoms. In this study, we examined the effects of rosmarinic acid, a major polyphenol in shiso, on urinary function and the bladder in a rat hydrochloric acid-induced cystitis model. Sprague-Dawley rats were administered intravesically with hydrochloric acid or saline solution (control) to induce cystitis. Afterwards, the rats were administered orally with distilled water or rosmarinic acid for three days and then the intravesical pressure was measured, a stretch stimulation test was performed using the harvested bladder, and histological and biochemical analyses were performed. In addition, we investigated the effects of rosmarinic acid on the expression of inflammation-related molecules in normal human bladder epithelial cells. Rosmarinic acid ameliorated hydrochloric acid-induced shortening of micturition interval by 49%. In hydrochloric acid-treated bladders, significantly more prostaglandin E2 was released after stretching; however, rosmarinic acid suppressed its release to control levels. Rosmarinic acid also reduced hydrochloric acid-induced epithelial thickening and the levels of inflammatory molecules in the bladder. Furthermore, rosmarinic acid suppressed interleukin 1β-induced increases in Cox2 and Il6 expression in bladder epithelial cells. These findings indicate that rosmarinic acid can ameliorate hydrochloric acid-induced cystitis in rats and that these effects are due, at least in part, to its anti-inflammatory effects on the bladder and inhibition of stretch-induced prostaglandin E2 release.
... It is recognized for its antioxidant qualities which allow it to neutralize free radicals, restricting oxidative effects in human cells and the proliferation of cancer cell lines [5]. Moreover, many other therapeutic effects have been reported, such as hepatoprotective [6,7], lung protective [8,9], and cardioprotective [10,11] responses, highly connected with antioxidant and anti-inflammatory activities [12]. In addition, Dahchour et al. de-scribed that this phenolic compound presents benefits for anxiety and depression [3]. ...
Article
Full-text available
Sweet basil (Ocimum basilicum) leaves are rich in bioactive compounds that present therapeutic benefits for human health. Ultrasonic-assisted extraction (UAE) is frequently used to obtain phenolic compounds from plants/herbal sources. However, few works have developed multi-variable studies to find the optimal conditions to extract the maximum amount of compounds, especially when applied to UAE via a sonotrode. The purpose of this work was to perform a multi-variable study by employing a Box-Behnken design to collect the highest active compound content from Ocimum basilicum leaves. The efficacy of the design was endorsed by ANOVA. The studied parameters for UAE via a sonotrode were the ethanol/water ratio, amplitude, and time. The analyzed responses were the rosmarinic acid, the sum of phenolic acids, and the sum of phenolic compounds content. The optimal conditions were found to be 50% ethanol/water, 50% amplitude, and 5 min. Twenty bioactive compounds were identified by HPLC-ESI-TOF-MS when the extract was collected by applying the optimal conditions. Ocimum basilicum may be appreciated as a valuable source of important bioactive substances for pharmaceutical use.
... Each assay was performed in triplicate. As reported in the literature studies, Dimethyl sulfoxide (DMSO, Millipore Sigma, Germany) was used as a solvent to dissolve both TQ [23] and RA [24] for treatment. Specifically, samples were dissolved in 100% DMSO diluted using Dulbecco′s Modified Eagle's Medium (DMEM, Gibco, UK) from solutions to IC50 doses, and the final concentration of DMSO in each group was adjusted to be 0.5%. ...
Article
Full-text available
Purpose Thymoquinone (TQ) and rosmarinic acid (RA) are two biologically active compounds found in plants and that possess remarkable anti-oxidant and anti-inflammatory properties. The present study aimed to investigate the potential protective effects of RA and TQ, which have known anti-inflammatory and anti-oxidant effects, on retinal damage by establishing a wound healing model for retinal pigment epithelial cells (ARPE-19). Method To this end, IC50 doses of RA and TQ in ARPE-19 cells were calculated by MTT assay. Both agents were administered at IC50, IC50/2 and IC50/4 doses for wound healing assay, and wound closure percentages were analyzed. Since the best wound healing was found at IC50/4 dose (low dose) for both agents, other biochemical and molecular analyses were planned to be performed using these doses. Following low dose RA and TQ treatments, the cells were lysed and TGF-β1 and MMP-9 levels were analyzed by ELISA technique from the cell lysates obtained. In addition, the mRNA expression levels of TLR3, IFN-γ and VEGF were calculated by RT-PCR technique. Results Low dose of RA and TQ dramatically increased wound healing. RA may have achieved this by increasing levels of MMP-9 and TLR-3. In contrast, the mRNA expression level of VEGF remained unchanged. TQ accelerated wound healing by increasing both the protein levels of TGF-β1 and MMP-9. Furthermore, low dose of TQ decreased both TLR3 and IFN-γ mRNA expression levels. Conclusion Low doses of RA and TQ were clearly demonstrated to have protective properties against possible damage to retinal pigment epithelial cells.
Article
Rosmarinic acid is an ester of caffeic acid and 3,4-dihydroxyphenyllactic acid. It is commonly found in species of the Boraginaceae and the subfamily Nepetoideae of the Lamiaceae. However, it is also found in species of other higher plant families and in some fern and hornwort species. Rosmarinic acid has a number of interesting biological activities, e.g. antiviral, antibacterial, antiinflammatory and antioxidant. The presence of rosmarinic acid in medicinal plants, herbs and spices has beneficial and health promoting effects. In plants, rosmarinic acid is supposed to act as a preformed constitutively accumulated defence compound. The biosynthesis of rosmarinic acid starts with the amino acids l-phenylalanine and l-tyrosine. All eight enzymes involved in the biosynthesis are known and characterised and cDNAs of several of the involved genes have been isolated. Plant cell cultures, e.g. from Coleus blumei or Salvia officinalis, accumulate rosmarinic acid in amounts much higher than in the plant itself (up to 36% of the cell dry weight). For this reason a biotechnological production of rosmarinic acid with plant cell cultures has been proposed.
Article
Oxidative stress and apoptosis
Article
Nephrotoxicity is an important complication in cancer patients undergoing cisplatin therapy. Oxidative stress, inflammation and apoptosis/necrosis are the major patho-mechanisms of cisplatin induced nephrotoxicity. In the present study, hesperidin, a naturally-occurring bioflavonoid has been demonstrated to have protective effect on cisplatin-induced renal injury in rats. Cisplatin intoxication resulted in structural and functional renal impairment which was revealed by massive histopathological changes and elevated blood urea nitrogen and serum creatinine levels, respectively. Renal injury was associated with oxidative stress/lipid peroxidation as evident by increased reactive oxygen species (ROS) and malondialdehyde (MDA) formation with decreased levels of antioxidants such as reduced glutathione, vitamin C, catalase, superoxide dismutase, glutathione reductase, glutathione peroxidase and glutathione-S-transferase. Cisplatin administration also triggered inflammatory response in rat kidneys by inducing pro-inflammatory cytokine, TNF-α, with the increased expression of myeloperoxidase (MPO). Furthermore, cisplatin increased the activity of caspase-3 and DNA damage with decreased tissue nitric oxide levels. Hesperidin treatment significantly attenuated the cisplatin-induced oxidative stress/lipid peroxidation, inflammation (infiltration of leukocytes and pro-inflammatory cytokine), apoptosis/necrosis (caspase-3 activity with DNA damage) as well as increased expression of nitric oxide in the kidney and improved renal function. Thus, our results suggest that hesperidin co-administration may serve as a novel and promising preventive strategy against cisplatin-induced nephrotoxicity.