Journal Pre-proof Nicorandil combats doxorubicin–induced nephrotoxicity via amendment of TLR4/P38 MAPK/NFκ-B signaling pathway Ali Khames Abd El-twab, Marwa M. Khalaf, Amany M. Gad, Ola M. Abd El-raouf, Mohamed Ahmed Kandeil PII:
S0009-2797(19)30720-3
DOI:
https://doi.org/10.1016/j.cbi.2019.108777
Reference:
CBI 108777
To appear in:
Chemico-Biological Interactions
Received Date: 1 May 2019 Revised Date:
16 July 2019
Accepted Date: 31 July 2019
Please cite this article as: A. Khames Abd El-twab, M.M. Khalaf, A.M. Gad, O.M. Abd El-raouf, M.A. Kandeil, Nicorandil combats doxorubicin–induced nephrotoxicity via amendment of TLR4/P38 MAPK/NFκ-B signaling pathway, Chemico-Biological Interactions (2019), doi: https://doi.org/10.1016/ j.cbi.2019.108777. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier B.V.
Editors-in-Chief
1
Chemico-biological interactions
2
Dear Editor:
3
Kindly find our attached revised manuscript, entitled: Nicorandil combats
4
doxorubicin-induced
TLR4/P38
5
MAPK/NFκ-B signaling pathway which we submit for consideration for
6
publication following revision in Chemico-biological interactions
7
We believe that this study would be of interest to your readers. The current
8
study aims to highlight on the ameliorative effect of nicorandil (anti-
9
anginal and cardioprotective drug) on doxorubicin-induced nephrotoxicity
10
experimentally in rats. This manuscript focuses on describing the
11
underlying mechanisms that mediate its activities targeting inflammatory &
12
oxidative stress and apoptotic markers mentioning previous trials and what
13
is added by our study, and focusing on the pathway that is included greatly
14
by doxorubicin; TLR4/MAPK P38/NFκ-B expression pathway that is
15
significantly induced by doxorubicin and is expected to be suppressed by
16
nicorandil. We emphasized our results and conclusions by western blot
17
technique.
this manuscript is complied with the scope of "Chemico-
18
biological interactions" journal because it discusses how nicorandil
19
prevented doxorubicin toxicity and this is the scopes of "Chemico-
20
biological interactions" journal.
21
nephrotoxicity
1
via
amendment
of
Thank you for your consideration.
22
Best regards
23
Correspondence: All correspondence should be addressed to:
24
Dr. Marwa Mahmoud Khalaf,
25
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-
26
Suef University.
27
Postal code: 62514, Salah Salem Street, Beni-Suef, Egypt
28
E-mail:
[email protected]
29
[email protected]
30
Tel.: 00201002784548
31
• Nicorandil ameliorated doxorubicin-induced nephrotoxicity.
32
• Nicorandil restored the oxidant/antioxidant balance.
33
• Nicorandil suppressed inflammatory signaling pathway TLR4/MAPK
34
P38/NF-κb/TNF-α.
35
Nicorandil regulated BAX/Bcl-2 apoptotic pathway
36
Title: Nicorandil
37
combats
doxorubicin–induced
nephrotoxicity
via
38
amendment of TLR4/P38 MAPK/NFκ-B signaling pathway
39
Ali Khames Abd El-twab1, Marwa M. Khalaf2, Amany M. Gad3, Ola M.
40
Abd El-raouf4, Mohamed Ahmed Kandeil5.
41
2
1
Department of Pharmacology and Toxicology, Faculty of Pharmacy,
42
Deraya university, Minia, Egypt.
43
1,3,4
44
Department of Pharmacology, National Organization for Drug Control
and Research (NODCAR), Cairo, Egypt.
45
2,5
46
Department of Pharmacology and Toxicology, Faculty of Pharmacy,
Beni-Suef University, Beni-Suef, Egypt.
47
Running Title: Nicorandil combats doxorubicin-induced nephrotoxicity
48
via amendment of TLR4/P38 MAPK/NFκ-B signaling pathway
49
* Correspondence: All correspondence should be addressed to:
50
Dr. Marwa Mahmoud Khalaf,
51
Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-
52
Suef University.
53
Postal code: 62514, Salah Salem Street, Beni-Suef, Egypt
54
E-mail:
[email protected]
55
[email protected]
56
Tel.: 00201002784548
57
3
Abstract
58
Nicorandil ameliorated doxorubicin-induced nephrotoxicity; this study
59
aimed to show and explain the mechanism of this protection. A precise
60
method was elucidated to study the effect of nicorandil on doxorubicin-
61
induced nephrotoxicity in rats depending on the critical inflammation
62
pathway TLR4/ MAPK P38 / NFκ-B. Adult male rats were subdivided
63
into four groups. The 1st group was normal control, the 2nd group received
64
nicorandil (3 mg/kg; p.o., for 4 weeks), the 3rd group received doxorubicin
65
(2.6 mg/kg, i.p., twice per week for 4 weeks), and the fourth group was
66
combination of doxorubicin and nicorandil for 4 weeks.
67
Nephrotoxicity was assessed by biochemical tests through measuring
68
Kidney function biomarkers such as [serum levels of urea, creatinine,
69
albumin and total protein] besides renal kidney injury molecule-1 (KIM-1)
70
and cystatin C], oxidative stress parameters
such as [renal tissue
71
malondialdehyde (MDA), reduced glutathione (GSH), SOD, catalase and
72
nrf-2], mediators of inflammation such as [Toll like receptor 4 (TLR-4),
73
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB),
74
p38 MAPK, Interleukin 1 beta (IL-1 β), and Tumor necrosis factor alpha
75
(TNF-α)] and markers of apoptosis [BAX and Bcl-2 in renal tissue].
76
Finally, our data were supported by histopathology examination.
77
4
Nicorandil pretreatment resulted in a significant decrease in nephrotoxicity
78
biomarkers, oxidative stress markers, inflammatory mediators and
79
prevented apoptosis through decreasing BAX and increasing Bcl-2 in renal
80
tissues. Nicorandil prevented all the histological alterations caused by
81
doxorubicin. Nicorandil is a promising antidote against doxorubicin-
82
induced nephrotoxicity by neutralizing all toxicity mechanisms caused by
83
doxorubicin through normalizing inflammatory cascade of TLR4/ MAPK
84
P38 / NFκ-B.
85
Key words: Nicorandil, Doxorubicin, Nephrotoxicity, Inflammation,
86
Apoptosis.
87
5
1. Introduction
88
Doxorubicin (DOX), an anthracycline derivative, is commonly
89
used to treat a variety of malignant neoplasms including, solid tumors such
90
as breast, pulmonary, uterine, ovary and cervix cancer as well as leukemia
91
and haematopoietic tumors (Akindele et al., 2018). Its continuous usage
92
alone in clinical settings is hindered owing to its life-threatening outcomes
93
on organs such as heart, kidney, liver and testis (Ren et al., 2016, Nagai et
94
al., 2018, Zidan et al., 2018).
95
Doxorubicin
nephrotoxicity
is
a
critical
factor
during
96
chemotherapy (Lee and Harris, 2011). Previous studies focused on
97
doxorubicin nephrotoxicity but there is no antidote that can prevent this
98
toxicity completely. Doxorubicin nephrotoxicity can be attributed to the
99
mitochondrial electron transfer reactions besides accumulated calcium ions
100
(Danmaigoro et al., 2018). In addition, inflammatory reaction and
101
apoptosis are causative factors (Shaker et al., 2018). Previous studies
102
evaluated the doxorubicin nephroathy mainly through measuring oxidative
103
stress parameters (Nagai et al., 2018), while others studied the effect of
104
hyperuricemia (Khames et al., 2017).
105
Several efforts have recently been made to ameliorate the
106
adverse effects of DOX. An approach to ameliorate doxorubicin related
107
toxicity is to use drug which engender a pharmacological intervention and
108
6
promising therapeutic strategy for the treatment or prevention of renal
109
disorders related to ROS overproduction.
110
Nicorandil, a mitochondrial ATP-dependent potassium (KATP)
111
channel opener, was used for the treatment of angina through increasing
112
coronary circulation (Simons and Laham, 2016, Kallistratos et al., 2017)
113
and was considered to be cardioprotective agent on ischaemic myocardium
114
(Abdel‐‐Raheem et al., 2013, Zhang et al., 2018). beside inhibiting
115
apoptosis in cardiac tissues (Nagata et al., 2003).
116
Nicorandil is a powerful anti-oxidant drug that has been previously
117
reported to exert great protection against oxidative stress in several
118
conditions (Qi et al., 2015, Ravindran et al., 2017) such as prevention of
119
oxidative stress in streptozotocin-induced diabetes (Mano et al., 2000).
120
Nicorandil was also used for the prevention of contrast‐induced
121
nephropathy (Ma et al., 2018). In addition, nicorandil has anti-
122
inflammatoy effects as shown by its anti-neuroinflammation effects in
123
astrocytes (Dong et al., 2016). Through anti-apoptotic effects, nicorandil
124
was pereviously reported to improve cardiac function and integrity after
125
coronary micro embolisation (He et al., 2018).
126
Our study assessed the involvement of toll like receptor 4
127
(TLR4) as a novel and significant inflammatory pathway. Toll like receptor
128
4 is a part of the innate immunity (Nair, 2015) and mediate inflammatory
129
7
response (Molteni et al., 2016) by inducing nuclear factor kappa B (NFκ-
130
B).
131
Currently, there is no information available about the protective effects of
132
the KATP channel openers on doxorubicin injury in the kidney. In the
133
present study, we hypothesized that nicorandil might prevent the renal
134
injury caused by doxorubicin and we investigated the effects and the
135
possible mechanisms of action of the KATP channel opener, nicorandil, on
136
the DOX injury in the rat kidney through TLR4 and NFκ-B signalling
137
pathway.
138
2. Material and Methods:
139
2.1. Ethics statement:
140
Animals were adapted for two weeks in the animal house before the start of
141
the study. Experimental methods were conducted according to the ethical
142
guidelines for investigations in experimental rats and were advisable by the
143
Research Ethical Committee of Faculty of Pharmacy, Beni-Suef University
144
(Beni-Suef, Egypt) to agree with the instructions of the Care and Use of
145
experimental rats (ILAR, 1996). Unnecessary aggressive deal with animals
146
was avoided. Animals were quietly handled; vigorous handling, pressure,
147
and tough maneuver was avoided.
148
2 materials:
149
8
- Doxorubicin: 2.62 mg/kg i.p twice weekly for 4 weeks (Elsherbiny
150
and El-Sherbiny, 2014a).
151
- Nicorandil: 3 mg/kg/day orally for 4 weeks (Ahmed and El-maraghy,
152
2013a).
153
All drugs and chemicals will be obtained from Sigma Chemical
154
Company (St Louis, USA).
155
2.3. Animals
156
Male adult Sprague-Dawley rats (200g ± 20) were purchased from the
157
animal house of national organization for drug control and research, Dokki,
158
Egypt. Animals were adapted at the experimental area of the National
159
Organization for Drug Control and Research (NODCAR, Dokki, Egypt).
160
Animals had no limitations to food and drink. They were kept at 20–25˚C
161
with 12-h light–dark cycle
162
2.4. Experimental design and sampling
163
Rats were randomly divided into four groups (n=10-12 per each),
164
The 1st group served as normal control group received saline (vehicle for
165
nicroandil) orally for 4 weeks and administered saline (0.5 ml/Rat, i.p.)
166
twice a week as vehicle for Dox.
167
9
The 2nd group received nicorandil only (3 mg/kg/day, orally for 4 weeks)
168
(Ahmed and El-Maraghy, 2013b).
169
The 3rd group were injected with a cumulative dose of doxorubicin 21
170
mg/kg, i.p. twice per week, for 4 weeks) (Elsherbiny and El-Sherbiny,
171
2014b).
172
The 4th group received doxorubicin and nicorandil for 4 weeks.
173
Nicorandil was given one hour before doxorubicin injection. At the end
174
of the experiment blood samples were collected from retro-orbital sinus
175
plexus and sera were separated by centrifugation at 1000g for 10 min and
176
stored at −80°C for serological measurements of the kidney function tests.
177
Afterwards, animals were sacrificed by decapitation under anesthesia, both
178
kidneys of each animal were dissected out, and portions of them were fixed
179
in 10% formalin solution for histopathological examination while the
180
remainder portions were homogenized in 50 mM phosphate buffer (pH
181
7.4), and stored at -80°C till estimations of biochemical and western blot
182
examinations.
183
2.5. Biochemical analysis
184
2.5.1. Assessment of renal functions
185
Colorimetric assay kits for the measurement of the level of blood
186
urea nitrogen (BUN), serum creatinine, serum albumin and total protein
187
10
using (biomed diagnostics, Cairo, Egypt). Rat Cystatin-C (Cys-C) as well
188
as Rat Kidney injury molecule (KIM-1) ELISA Kits were obtained from
189
Mybiosource (San Diego, CA, USA) and were used for measurement of
190
serum cystatin and KIM-1, respectively. All procedures were performed
191
according to the manufacturers’ instructions.
192
2.5.2. Measurement of renal oxidative stress estimation
193
The renal content of thiobarbituric acid reactive substances (TBARS)
and
glutathione
(GSH),
activity of
195
myeloperoxidase, catalase (CAT), and superoxide dismutase (SOD) were
196
measured using the Mybiosource ELISA kits (San Diego, CA, USA)
197
according to the manufacturer’s instructions.
198
2.5.3. Measurement of renal Toll like receptor 4 (TLR-4), Nuclear
199
factor kappa-light-chain-enhancer of activated B cells (NF-κB),
200
Nuclear factor erythroid 2–related factor 2 (Nrf-2) and P38 mitogen-
201
activated protein kinases (p38-MAPK)
202
Protein levels of TLR-4, NF-κB, Nrf-2, and p38-MAPK were assessed
203
using Western blot technique using TGX Stain-Free™ Fast Cast™
204
Acrylamide Kit (SDS-PAGE) which was provided by Bio-Rad
205
Laboratories, TNC, USA Catalog. NO. 161-0181.
206
11
as
well
as
the
194
The Western Blot analysis technique was applied (using V3 Western
207
WorkflowTM Complete System, Bio-Rad R _ Hercules, CA). Briefly,
208
proteins were extracted from tissue homogenates using ice-cold radio-
209
immuno precipitation assay buffer supplemented with phosphatase and
210
protease
mM
211
phenylmethylsulphonyl fluoride, 2 mg/mL aprotinin, and 0.5 mg/mL
212
leupeptin) then centrifugated at 12,000 rpm for 20 min. Bradford assay
213
method was used to estimate the protein concentration of each sample. The
214
same amounts of protein (20–30 ??g of total protein) were isolated by gel
215
electrophoresis (10% acrylamide gel) depending on a Bio-Rad Mini-
216
Protein II system. The protein was moved to polyvinylidene difluoride
217
membranes (Pierce, Rockford, IL) with a Bio- Rad Trans-Blot system.
218
Then, PBS was used for washing membranes then blocked for 1 h at
219
ambient temperature with 5% (w/v) skimmed milk powder in PBS.
220
Following blocking, the blots were developed using antibodies for TLR4,
221
NF-κB, Nrf-2, p38-MAPK and beta actin supplied by Thermoscientific
222
(Rockford, IL) and incubated overnight at pH 7.6 at 4◦C with gentle
223
shaking. After washing, peroxidase-labeled secondary antibodies were
224
added and the membranes were incubated at 37◦C for 1 h. Band intensity
225
examined and analyzed by the imaging system ChemiDocTM with Image
226
LabTM software version 5.1 (Bio-Rad Laboratories, Hercules, CA). The
227
inhibitors
(50
mmol/L
12
sodium
vanadate,
0.5
results are expressed as arbitrary units after comparison to the normal -
228
actin protein expression
229
2.5.4. Measurement of (TNF-α) and Interleukin 1 beta (IL-1 β)
230
Rat TNF-α and IL-1 β were estimated according to the ELISA kits
231
manufacturer guide obtained from RayBiotech Inc. (Parkway, LaneSuite
232
Norcross, GA).
233
2.5.5. Measurement of protein expression of Bax/Bcl2
234
B-cell lymphoma 2 (Bcl-2) protein, Bcl-2-Associated-X-protein (Bax) and
235
Bax/Bcl-2 ratio were assessed using western blot technique, Protein levels
236
of Bax/Bcl2 in renal tissues of different treatment groups (1–4) were
237
assessed using Western blot technique. Where proteins were extracted
238
using TRIzol reagent, and protein concentrations were estimated using
239
Bradford method. The primary antibodies used were raised in rabbit anti-
240
Bcl-2 antibody (13-8800 Thermo Fisher Scientific), anti-Bax antibody
241
(MA5-14003 Thermo Fisher Scientific).
242
2.5.6. Renal histopathological examination
243
Kidney specimens were taken from many rats in each group and were
244
stabilized in 10% formal saline for one day. Washing was carried out by
245
water and dehydrated by diluted methyl and ethyl alcohol that is serially
246
diluted and were used for dehydration. Samples were kept in paraffin wax
247
13
at 56 degree in oven for one day. Renal tissues blocks 4 microns thickness
248
were made by slidge microtome. Then the sections were added to glass
249
slides, paraffin is removed , hematoxylin & eosin stain are used for the
250
examination by light electric microscope (Suvarna et al., 2018).
251
2.6. Data and Statistical analysis
252
Results were expressed as mean ± SE. Statistical analysis was performed
253
using the SPSS version 16 (Chicago, IL, USA), while the graphs were
254
drawn using a prism computer program (GraphPad software Inc. V5, San
255
Diego, CA, USA). Statistical analysis was carried out using one-way
256
analysis of variance (ANOVA) followed by Tukey-Kramer Multiple
257
Comparison Test as a post hoc test. Probability values of less than 0.05
258
were considered statistically significant.
259
3. Results
260
3.1.
Effect of nicorandil on serum levels of urea nitrogen,
261
creatinine, total protein and albumin and on renal tissue
262
contents of KIM-1 and Cystatin C in doxorubicin-induced
263
nephrotoxicity in rats:
264
Normal control values for the serum levels of urea, creatinine, total protein
265
and albumin were 33.17 ± 1.9 mg/dl, 0.18 ± 0.013 mg/dl, 5.59 ± 0.15
266
mg/dl and 3.5 ± 0.25 mg/dl, respectively. Normal control values for the
267
14
renal contents of KIM-1 and cystatin C were 2.202 ± 0.09 pg/mg.tissue and
268
0.6617 ± 0.05 pg/mg.tissue. Doxorubicin significantly increased serum
269
levels of urea nitrogen and creatinine associated with a significant decline
270
in total protein and albumin levels as compared to the normal control
271
group. Additionally, doxorubicin significantly increased renal contents of
272
KIM-1 and cystatin C.
273
However, co-treatment with nicorandil significantly abolished the increase
274
of urea nitrogen, creatinine, KIM-1 and cystatin C levels and counteracted
275
the reduction of total protein and albumin, respectively, as compared to
276
doxorubicin group (Table 1).
277
3.2.
Effect of nicorandil on the renal oxidative stress biomarkers in
278
doxorubicin-induced nephrotoxicity in rats:
279
Normal control values for the renal contents of MDA and GSH and the
280
renal activities of SOD, CAT, Nrf-2 and MPO were 5.350 ± 0.41 nmol/g.
281
tissue, 71.15 ± 7.24 mg/g tissue, 5.89 ± 0.26 mg/g tissue, 120.0 ± 2.6 mg/g
282
tissue and 39.25 ± 5.86 mg/g tissue respectively.
283
Doxorubicin treatment resulted in a marked increase in the renal MDA
284
content and MPO activity associated with a marked decrease in renal GSH
285
content as well as a significant decrease in Nrf-2, SOD and CAT activities,
286
respectively, as compared to the normal control group
287
15
Treatment with nicorandil significantly decreased the renal MDA content
288
and MPO activity and nearly restored renal GSH content and Nrf-2, SOD
289
and CAT activities to normal control values as compared to doxorubicin
290
group (Fig. 1A-F).
291
3.3.
Effect of nicorandil on the renal inflammatory mediators in
292
doxorubicin-induced nephrotoxicity in rats:
293
Normal control values for TLR4, P38 MAPK, NF-κB, IL.1 β and TNF-α
294
were 1.00 ± 0.01, 1.005 ± 0.001, 1.007 ± 0.003, and 24.88 ± 2.121 pg/g
295
tissue, respectively.
296
Doxorubicin treatment resulted in a significant increase in renal content of
297
TLR4, P38 MAPK, NF-κB, IL.1 β and TNF-α as compared to the normal
298
control group.
299
Co-treatment with nicorandil significantly normalized renal content of
300
TLR4, P38 MAPK, NF-κB, IL.1 β and TNF-α as compared to doxorubicin
301
group (Fig. 2A-D).
302
3.4.
Effect of nicorandil on renal antiapoptotic markers in doxorubicin-induced nephrotoxicity in rats:
303 304
Normal control values for the pro-apoptotic protein BCL2 Associated X
305
protein (Bax), and anti-apoptotic protein B-cell lymphoma 2 (Bcl-2) were
306
1.012 ± 0.01 and 1.018 ± 0.016.
307 16
Doxorubicin increased apoptotic markers significantly as compared to the
308
normal control group.
309
As shown in Fig. (3A-C) after nicorandil administration the level of Bax
310
was significantly decreased while Bcl-2 level was significantly increased in
311
the renal tissue resulting in inhibition of apoptosis as compared with
312
doxorubicin group.
313
3.5. Effect of nicorandil on histopathological examination of renal tissues in doxorubicin-induced nephrotoxicity in rats.
314 315
As shown in Figure 4, kidney sections obtained from the normal control
316
and the control nicorandil groups showed normal histological appearance
317
of kidney architecture.
318
Meanwhile, doxorubicin-treated group showed focal inflammatory cells
319
infiltrations with few fibroblastic cells proliferation were detected in
320
between the tubules and glomeruli at the cortex. The endothelial cells
321
lining the tufts of the glomeruli showed vacuolization. The cortical stromal
322
blood vessels showed congestion as well as perivascular oedema. The
323
corticomedullary portion showed focal fibrosis and focal haemorrhage
324
inbetween the tubules. There was swelling in the lining tubular epithelium
325
with obliteration in the tubular lumen
326
17
while other tubules at the
corticomedullary portion had vacuolar degeneration in the lining
327
epithelium (Fig.4),(table 2)
328
However, nicorandil co-treated group showed mild focal inflammatory
329
cells infiltration, the tubules at the corticomedullary portion showed
330
vacuolar degeneration
331
4. Discussion
332
Cancer chemotherapy usually demolishes the normal physiological
333
homoeostasis and affects multiple organs during the course of treatment.
334
Despite its extensive clinical utilization in the fight against a variety of
335
human malignancies (Akindele et al., 2018), treatment with the
336
conventional doxorubicin (DOX) is limited because of its multiorgan
337
toxicities including renal damage and nephrotoxicity (Wang et al., 2000),
338
cardiac, pulmonary, testicular and hematological toxicities (Singal et al.,
339
2000a).
several
340
mechanisms including free radical production as well as inflammatory,
341
apoptotic and hyperuricemic effects (Khames et al., 2017b).
342
In the present study, i.p. doxorubicin produced severe increase in serum
343
levels of BUN, creatinine, KIM and cystatin C, while it significantly
344
decreased serum levels of albumin and total protein. These results are in
345
agreement with (Öz and İlhan, 2006, Kramer et al., 2009;, Jaćević et al.,
346
Doxorubicin
deteriorates
18
renal
function
through
2018, Qiao et al., 2018). These deteriorations in renal function are
347
suggested to be attributed mainly to the increased production of free
348
radicals and ROS in renal tissues that resulted in reaction of these free
349
radicals with proteins of the nephron causing structural and functional
350
changes of renal tubules and glomeruli.
351
In the present study, Kim-1 was significantly elevated in the Dox-treated
352
group. This result was consistent with the earlier reports of (Lateef et al.,
353
2014, Wang et al., 2015).
354
Kidney injury molecule-1 is a type 1 membrane protein that is expressed at
355
negligible levels in normal rat kidneys. However, it is reported to be
356
massively induced in tubules after ischemic or toxic injury in rats
357
(Ichimura et al., 1998). Tubular kidney injury molecule-1 (Kim-1) is
358
induced in acute renal injury and is known to be reversible. In addition,
359
Kim-1 is also induced in chronic renal damage (Huo et al., 2010). The
360
mechanism of Kim-1 induction by doxorubicin nephropathy was not
361
specifically studied. However, its expression was found to occur in a wide
362
array of renal conditions in association with early tubular damage (Kramer
363
et al., 2009). It is plausible in this study that proteinuria was the marker for
364
renal injury.
365
The present results also showed that the level of cystatin C was
366
significantly elevated in the Dox-treated group. In agreement, Wang et al.
367
19
(2015) reported that, animals treated with doxorubicin (6 mg-kg) for 15
368
days showed a significant increase in cystatin C level.
369
Cystatin C in the blood is usually filtered through the glomeruli, being
370
absorbed in the proximal tubule of the kidney. Dysfunction of the renal
371
tubule leads to raised level of cystatin C in the body. many studies proved
372
that serum cystatin C is preferred than creatinine as an indicator of renal
373
failure involving defective glomerular functions (Dharnidharka et al.,
374
2002).
375
Oxidative stress was incriminated to be the main mechanism responsible
376
for DOX-induced nephrtoxicity (Khames et al., 2017a). Mansouri et al.
377
(2017) reported that, the mechanisms of doxorubicin nephrotoxicity
378
included oxidative stress status characterized by high amount of the
379
produced free oxidative radicals and a defect in the endogenous antioxidant
380
defense mechanisms causing an imbalance in the normal oxygen metabolic
381
pathway. First, a semiquinone metabolite is formed after adding an electron
382
to the quinone form of doxorubicin and finally, the quinone structure is
383
rapidly reformed by reduction of the molecular oxygen to ROS (Liu et al.,
384
2009). ROS produces hydrogen peroxide (H2O2) and hydroxyl radicals
385
(OH· ) and these products can attack DNA, oxidize it and finally induce
386
apoptosis in both normal and tumoral tissue cells (Al-Dalaen and Al-
387
Qtaitat, 2014).
388
20
Results of the current investigation revealed that, DOX administration led
389
to a significant reduction in GSH content, Nrf-2 level as well as the
390
activities of SOD and catalase enzymes. However, there was a significant
391
increase in MDA content and MPO activity. These findings are in the
392
agreement with (Sun et al. 2016, Oyagbemi et al., 2017, Qiao et al.,
393
2018).
394
A possible explanation for these results may be the consumption of GSH as
395
result of DOX-induced lipid peroxidation directly through semiquinone
396
structure or indirectly through production of ROS (Ashour et al., 2011).
397
The oxidant metabolites of doxorubicin, the semiquinone form, in presence
398
of oxygen can produce hydrogen peroxide and other oxidative free
399
radicals. In addition, another mechanism for doxorubicin-induced oxidative
400
stress is dependent on the presence of iron, where doxorubicin-iron
401
complex oxidizes oxygen to give hydrogen peroxide and other ROS
402
resulting in accumulation of MDA and MPO with the consumption of
403
antioxidant enzymes (Deavall et al., 2012). Nuclear factor erythroid-2
404
related factor 2 (Nrf2) is a master transcription factor in controlling the
405
basal and inducible expression of a battery of antioxidant genes and other
406
cytoprotective phase II detoxifying enzymes (Li et al., 2009). Kabel et al.
407
(2018) and Zhao et al. (2018) reported that, administration of DOX caused
408
21
a significant decrease in Nrf-2 level resulting in hepatotoxicity and
409
cardiotoxicity.
410
Another important finding in the present study was that, DOX increased
411
the inflammatory mediators namely; TLR4, P38 MAPK, NF-κB and TNF-
412
α in renal tissues. These results are in harmony with (Zhu et al., 2015, Min
413
et al., 2016, Yao et al., 2017). This inflammatory response can be
414
explained on the basis of oxidative stress, because free radicals produced
415
by the semiquinone form of doxorubicin after consumption of natural anti-
416
oxidant enzymes induces renal tissue injury leading to inflammatory
417
response.
418
This finding provides a new answer to what causes systemic inflammation
419
in the cancer patients receiving doxorubicin treatment. Actually , evidences
420
obtained through clinical studies or in animal experiments have proved that
421
doxorubicin caused serious systemic inflammation in vivo, including
422
hepatitis, nephritis, phlebitis, and mucositis throughout the digestive tract,
423
with an increase in the inflammatory cytokines levels in the circulation
424
(Wang et al., 2016). LPS, The heat shock proteins, ROS and chemicals
425
can bind TLR4 starting an inflammatory cascade by activating mitogen-
426
protein kinases (MAPK) that activate NFκB transcriptin factor. Finally
427
stimulation of NFκB induces protein synthesis and increases the levels of
428
the inflammatory mediators IL-1β and TNF-α (Baeza-Raja and Munoz-
429
22
Cánoves, 2004, Vyas et al., 2014).
NF-κB activation through TLR4
430
stimulation by the anticancer drug induces the expression of many
431
cytokines such as interleukin and TNF-α inducing cellular apoptosis and
432
death (Kumar et al., 2004).
433
Results of the present investigation showed that, one of the important
434
causes of doxorubicin-induced nephrotoxicity is apoptosis and activating
435
proapoptic markers as evidenced by the significant increase observed in
436
BAX expression and the significant decrease observed in Bcl-2 expression.
437
These results are in agreement with (Hoshi et al., 2017, Sun et al., 2018).
438
Apoptosis induced by DOX is considered to be a logic response for
439
inflammatory and oxidative mediators which triggers and activate pro-
440
apoptic agents. MAPK can motivate NF-κB and activate downstream
441
genes to further modulate the inflammatory responses which would
442
regulate the pathological state. Therefore, ROS production, increased
443
inflammatory cytokines and MAPK signaling, facilitate the activation of
444
NF-κB transcription factors and apoptotic genes as well (Imam et al.,
445
2018). (Park et al., 2012) reported that, the activation of p38 MAPK
446
induces apoptosis. In addition, the pro-apoptotic proteins such as BAX and
447
the anti-apoptotic proteins such as Bcl-2, and the tumor suppressor protein
448
p53 are also involved in doxorubicin-induced apoptosis. These findings
449
were in full agreement with the results of the present study that, NF-κB and
450
23
the activation of MAPK pathways are involved in the development of
451
doxorubicin-induced nephropathy. It is also clear in this study that
452
apoptotic changes in renal tissue were due to ROS mediated NF-κB
453
activation. Moreover, the generation of ROS promotes lipid peroxidation
454
which is reported to induce apoptotic changes in the cells as reported
455
previously by (Park et al., 2014) that the apoptotic changes in the cells are
456
due to loss of mitochondrial membrane integrity and p53 activation.
457
The present study proved that, nicorandil pretreatment protected against
458
doxorubicin-induced nephrotoxicity as evidenced by the decreased plasma
459
levels of urea, creatinine, KIM and cystatin. This nephroprotective
460
potential of nicorandil could be explained by its vasdilatory effect
461
enhancing the renal perfusion and excretion. Similar results suggested that
462
nicorandil ameliorates nephrotoxicity in rat (Shimizu et al., 2011, Ozturk
463
et al., 2017).
464
Results of the present investigation showed that, nicorandil significantly
465
decreased the elevated MDA content and myeloperoxidase activity in the
466
kidney, while restored the depleted GSH content and the activities of the
467
anti-oxidant enzymes Nrf-2, SOD and catalase. These results are thinkable
468
to be due to the free radical scavenger property of nicoranil and its ability
469
to neutralize ROS produced by doxorubicin. This agreed with the results of
470
(Gupta and Sharma, 2014, Ozturk et al., 2017, Zhu et al., 2018).
471
24
In this study, the anti-inflammatory effect of nicorandil was proved
472
through its ability to decrease TLR 4, p38 MAPK, NF-κB , IL-1 beta and
473
TNF–α as compared to the doxorubicin group. This is in the harmoney
474
with the results of (Heywood and Thomas, 2002, Kawamura et al., 2005,
475
Zhao et al., 2014) and could be explained by the nitric oxide donating
476
effect of nicorandil (Naito et al., 1994, Wei et al., 2003) and its effect on
477
potassium channels (Hongo et al., 2005).
478
Additionally, it has been observed in the present study that nicorandil
479
significantly ameliorated the apoptotic effect of doxorubicin through
480
decreasing Bax content and increasing Bcl-2 content in renal tissues. The
481
strong antiapoptotic effect of nicorandil is thought to be through the anti-
482
oxidative and free radical capturing ability of nicorandil besides its ability
483
to inhibit p38 MAPK pathway resulting in inhibiting all signals of cell
484
death (Yu et al., 2013).Additionally, the released NO causes vasodilatory
485
effect and inhibits inflammation due to bradykinin that is decreased in
486
various models of renal tissue apoptosis (El-Kashef, 2018). This is similar
487
to the results of (Yu et al., 2015, He et al., 2018). (Nagata et al., 2003)
488
reported that, nicorandil prevents the inflammatory cascade caused by
489
doxorubicin and thereby prevents the activation of pro-apoptotic proteins
490
and oxidative stress induced apoptosis.
491
25
Histopathological results confirmed our findings through improvement of
492
structural changes of kidney induced by doxorubicin. Where nicorandil
493
prevented the congestion of the renal blood vessels,
the perivascular
494
edema, focal fibrosis and haemorrhage in between the tubules in addition
495
to maintaining the renal tubules integrity keeping the filtration,
496
reabsorptive power and renal function.
497
From the previous results and conclusions, it could be deduced that the
498
mechanisms behind doxorubicin nephrotoxicity include oxidative stress,
499
inflammation and apoptosis. Nicorandil is a promising drug that could be
500
used concomitantly with cancer chemotherapy in order to prevent the
501
expected toxicity through its anti-oxidant, anti-inflammatory and anti-
502
apoptotic mechanisms. Furthermore, the ability of nicorandil to prevent
503
doxorubicin-induced cardiotoxicity
504
gives it a great importance and
priority in treating doxorubicin adverse effects than any other agent.
505
In conclusion
506
The present study concluded that DOX induced severe nephrotoxicity by
507
promoting oxidative, inflammatory and apoptotic mechanisms. Nicroandil
508
attenuated DOX-induced apoptosis and inflammation through suppression
509
of oxidative stress mediated activation of TLR4/MAPKp38/NF-κB
510
signaling pathways. Therefore, it is advisable to be used concomitantly
511
with DOX to reduce its nephrotoxicity.
512
26
Acknowledgments:
513
The authors would like to thank Prof. Dr. A. Bakear (Pathology
514
Department, Faculty of Veterinary Medicine, Cairo University, Cairo,
515
Egypt) for assistance in the histopathological examinations.
516
Disclosure of Conflict of interest
517
The authors have read the journal's policy on disclosure of potential
518
conflicts of interest and they all declare no personal or financial conflict of
519
interest.
520
Authorship Statement
521
All authors have read the journal’s authorship statement and agree to it.
522
5. References
523
AHMED, L. A. & EL-MARAGHY, S. A. 2013a. Nicorandil ameliorates mitochondrial dysfunction in doxorubicin-induced heart failure in rats: possible mechanism of cardioprotection. Biochemical pharmacology, 86, 1301-1310. AHMED, L. A. & EL-MARAGHY, S. A. 2013b .Nicorandil ameliorates mitochondrial dysfunction in doxorubicin-induced heart failure in rats: possible mechanism of cardioprotection. Biochem Pharmacol, 86, 1301-10. AKINDELE, A. J., OLUDADEPO, G. O., AMAGON, K. I., SINGH, D. & OSIAGWU, D. D. 2018. Protective effect of carvedilol alone and coadministered with diltiazem and prednisolone on doxorubicin and 5-fluorouracil-induced hepatotoxicity and nephrotoxicity in rats. Pharmacol Res Perspect, 6. AL-DALAEN, S. & AL-QTAITAT, A. 2014. Review article: Oxidative stress versus antioxidants. American journal of bioscience and bioengineering, 2, 60-71. ASHOUR, O. M., ELBERRY, A. A., ALAHDAL, A. M., AL MOHAMADI, A. M., NAGY, A. A., ABDEL NAIM, A. B., ABDEL SATTAR, E. A. & MOHAMADIN, A. M. 2011. Protective effect of bilberry (Vaccinium myrtillus) against doxorubicin-induced oxidative cardiotoxicity in rats. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research, 17, 110-115. BAEZA-RAJA, B. & MUNOZ-CáNOVES, P. 2004. p38 MAPK-induced nuclear factor-κB activity is required for skeletal muscle differentiation: role of interleukin-6. Molecular biology of the cell, 15, 2013-2026. 27
524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543
DEAVALL, D. G., MARTIN, E. A., HORNER, J. M. & ROBERTS, R. 2012. Drug-induced oxidative stress and toxicity. Journal of toxicology, 2012. DHARNIDHARKA, V. R., KWON, C. & STEVENS, G. 2002. Serum cystatin C is superior to serum creatinine as a marker of kidney function: a meta-analysis. American Journal of Kidney Diseases, 40, 221-226. DONG, Y.-F., CHEN, Z-.Z., ZHAO, Z., YANG, D.-D., YAN, H., JI, J. & SUN, X.-L. 2016. Potential role of microRNA-7 in the anti-neuroinflammation effects of nicorandil in astrocytes induced by oxygen-glucose deprivation. Journal of neuroinflammation, 13, 60. EL-KASHEF, D. H. 2018 .Nicorandil ameliorates pulmonary inflammation and fibrosis in a rat model of silicosis. Int Immunopharmacol, 64, 289-297. ELSHERBINY, N. M. & EL-SHERBINY, M. 2014a. Thymoquinone attenuates Doxorubicin-induced nephrotoxicity in rats: Role of Nrf2 and NOX .4Chemico-biological interactions, 223, 102108. ELSHERBINY, N. M. & EL-SHERBINY, M. 2014b. Thymoquinone attenuates Doxorubicin-induced nephrotoxicity in rats: Role of Nrf2 and NOX4. Chem Biol Interact, 223, 102-8. GUPTA, S. & SHARMA, B. 2014. Protective effects of phosphodiesterase-1 (PDE1) and ATP sensitive potassium (KATP) channel modulators against 3-nitropropionic acid induced behavioral and biochemical toxicities in experimental Huntington ׳s disease. European journal of pharmacology, 732, 111-122. HE, W., SU, Q., LIANG, J., SUN, Y., WANG, X. & LI, L. 2018. The protective effect of nicorandil on cardiomyocyte apoptosis after coronary microembolization by activating Nrf2/HO-1 signaling pathway in rats. Biochemical and biophysical research communications, 496, 1296-1301. HEYWOOD, G. & THOMAS, P. 2002. Nicorandil inhibits degranulation and TNF-α release from RBL-2H3 cells. Inflammation Research, 51, 176-181. HONGO, M., MAWATARI, E., SAKAI, A., RUAN, Z., KOIZUMI, T., TERASAWA, F., YAZAKI, Y., KINOSHITA ,O., IKEDA, U. & SHIBAMOTO, T. 2005. Effects of nicorandil on monocrotaline-induced pulmonary arterial hypertension in rats. Journal of cardiovascular pharmacology, 46, 452-458. HOSHI, R., WATANABE, Y., ISHIZUKA, Y., HIRANO, T., NAGASAKI-MAEOKA, E., YOSHIZAWA, S., UEKUSA, S., KAWASHIMA, H., OHASHI, K. & SUGITO, K. 2017. Depletion of TFAP2E attenuates adriamycin-mediated apoptosis in human neuroblastoma cells. Oncology reports, 37, 2459-2464. HUO, W., ZHANG, K., NIE, Z., LI, Q. & JIN, F. 2010. Kidney injury molecule-1 (KIM-1): a novel kidney-specific injury molecule playing potential double-edged functions in kidney injury. Transplantation Reviews, 24, 143-146. ICHIMURA, T., BONVENTRE, J. V., BAILLY, V., WEI, H., HESSION, C. A., CATE, R. L. & SANICOLA, M . .1998Kidney injury molecule-1 (KIM-1), a putative epithelial cell adhesion molecule containing a novel immunoglobulin domain, is up-regulated in renal cells after injury. Journal of Biological Chemistry, 273, 4135-4142. IMAM, F., AL-HARBI, N. O., AL-HARBI, M. M., ANSARI, M. A., AL-ASMARI, A. F., ANSARI, M. N., ALANAZI, W. A., BAHASHWAN, S., ALMUTAIRI, M. M. & ALSHAMMARI, M. 2018. Apremilast prevent doxorubicin-induced apoptosis and inflammation in heart through inhibition of oxidative stress mediated activation of NF-κB signaling pathways. Pharmacological Reports, 70, 993-1000. JAĆEVIĆ, V., DRAGOJEVIĆ-SIMIĆ, V., TATOMIROVIĆ, Ž., DOBRIĆ, S., BOKONJIĆ, D., KOVAČEVIĆ, A., NEPOVIMOVA, E., VALIŠ, M. & KUČA, K. 2018. The Efficacy of Amifostine against Multiple-Dose Doxorubicin-Induced Toxicity in Rats. International journal of molecular sciences, 19, 2370. KABEL, A. M., ALZAHRANI, A. A., BAWAZIR, N. M., KHAWTANI, R. O. & ARAB, H. H. 2018. Targeting the proinflammatory cytokines, oxidative stress, apoptosis and TGF-β1/STAT-3 28
544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594
signaling by irbesartan to ameliorate doxorubicin-induced hepatotoxicity. Journal of Infection and Chemotherapy. KAWAMURA, T., KADOSAKI, M., NARA, N., WEI, J., ENDO, S. & INADA, K. 2005. Nicorandil attenuates NF-κB activation, adhesion molecule expression, and cytokine production in patients with coronary artery bypass surgery. Shock, 24, 103-108. KHAMES, A., GAD, A. M. & EL-RAOUF, O. M. A. 2017a. Ameliorative effects of sildenafil and/or febuxostat on doxorubicin-induced nephrotoxicity in rats. European journal of pharmacology, 805, 118-124. KHAMES, A., KHALAF, M. M., GAD, A. M. & ABD EL-RAOUF, O. M. 2017b. Ameliorative effects of sildenafil and/or febuxostat on doxorubicin-induced nephrotoxicity in rats. Eur J Pharmacol, 805, 118-124. KRAMER, A. B., VAN TIMMEREN, M. M., SCHUURS, T. A., VAIDYA, V. S., BONVENTRE, J. V., VAN GOOR, H. & NAVIS, G. 2009. Reduction of proteinuria in adriamycin-induced nephropathy is associated with reduction of renal kidney injury molecule (Kim-1) over time. American Journal of Physiology-Renal Physiology, 296, F1136-F1145. KUMAR, A., TAKADA, Y., BORIEK, A. M. & AGGARWAL, B. B. 2004. Nuclear factor-κB: its role in health and disease. Journal of Molecular Medicine, 82, 434-448. LATEEF, A., HASSAN, S. K., RASHID ,S., ALI, N., ZEESHAN, M. & SULTANA, S. 2014. D-limonene suppresses doxorubicin-induced oxidative stress and inflammation via repression of COX-2, iNOS, and NFiB in kidneys of Wistar rats. LI, J., ICHIKAWA, T., JANICKI, J. S. & CUI, T. 2009. Targeting the Nrf2 pathway against cardiovascular disease. Expert opinion on therapeutic targets, 13, 785-794. LIU, J., QU, W. & KADIISKA, M. B. 2009. Role of oxidative stress in cadmium toxicity and carcinogenesis. Toxicology and applied pharmacology, 238, 209-214. MANO, T., SHINOHARA, R., NAGASAKA, A., NAKAGAWA, H., UCHIMURA, K., HAYASHI, R., NAKANO, I., TSUGAWA, T., WATANABE, F. & KOBAYASHI, T. 2000. Scavenging effect of nicorandil on free radicals and lipid peroxide in streptozotocin-induced diabetic rats. Metabolism, 49, 427-431. MANSOURI, E., JANGARAN, A. & ASHTARI, A. 2017. Protective effect of pravastatin on doxorubicin-induced hepatotoxicity. Bratislavske lekarske listy, 118, 273-277. MIN, H. S., CHA, J. J., KIM, K., KIM, J. E., GHEE, J. Y., KIM, H., LEE, J .E., HAN, J.-Y., JEONG, L. S. & CHA, D. R. 2016. Renoprotective effects of a highly selective A3 adenosine receptor antagonist in a mouse model of adriamycin-induced nephropathy. Journal of Korean medical science, 31, 1403-1412. NAGATA, K., OBATA, K., ODASHIMA, M., YAMADA, A., SOMURA, F., NISHIZAWA, T., ICHIHARA, S., IZAWA, H., IWASE, M. & HAYAKAWA, A. 2003. Nicorandil inhibits oxidative stressinduced apoptosis in cardiac myocytes through activation of mitochondrial ATP-sensitive potassium channels and a nitrate-like effect. Journal of molecular and cellular cardiology, 35, 1505-1512. NAITO, A., ANIYA, Y. & SAKANASHI, M. 1994. Antioxidative action of the nitrovasodilator nicorandil: inhibition of oxidative activation of liver microsomal glutathione Stransferase and lipid peroxidation. The Japanese Journal of Pharmacology, 65, 209-213. OYAGBEMI, A. A., OMOBOWALE, T. O., OLOPADE, J. O. & FAROMBI, E. O. 2017. Kolaviron and Garcinia kola attenuate doxorubicin-induced cardiotoxicity in Wistar rats. Journal of Complementary and Integrative Medicine, 15. ÖZ, E. & İLHAN, M. N. 2006. Effects of melatonin in reducing the toxic effects of doxorubicin. Molecular and cellular biochemistry, 286, 11-15. OZTURK, H., FIRAT, T., TEKCE, B. K., YILMAZ, F. & OZTURK, H. 2017 .Effects of nicorandil on renal function and histopathology in rats with partial unilateral ureteral obstruction. The Kaohsiung journal of medical sciences, 33, 236-245.
29
595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644
PARK, E.-J., KWON, H.-K., CHOI, Y.-M., SHIN, H.-J. & CHOI, S. 2012. Doxorubicin induces cytotoxicity through upregulation of perk–dependent ATF3. PloS one, 7, e44990. PARK, S., YOON, J., BAE, S., PARK, M., KANG, C., KE, Q., LEE, D. & KANG, P. M. 2014. Therapeutic use of H2O2-responsive anti-oxidant polymer nanoparticles for doxorubicin-induced cardiomyopathy. Biomaterials, 35, 5944-5953. QIAO, J., LIU, Y., JIANG, Z., YANG, Y., LIU, W. & HAN, B. 2018. Preparation and renoprotective effects of carboxymethyl chitosan oligosaccharide on adriamycin nephropathy. Carbohydrate polymers, 201, 347.356SHIMIZU, S., SAITO, M., KINOSHITA, Y., OHMASA, F., DIMITRIADIS, F., SHOMORI, K., HAYASHI, A. & SATOH, K. 2011. Nicorandil ameliorates ischaemia-reperfusion injury in the rat kidney. British journal of pharmacology, 163, 272-282. SUN, Y., WANG, C., MENG, Q., LIU, Z., HUO, X., SUN, P., SUN, H., MA, X., PENG, J. & LIU, K. 2018. Targeting P-glycoprotein and SORCIN: Dihydromyricetin strengthens anti-proliferative efficiency of adriamycin via MAPK/ERK and Ca2+-mediated apoptosis pathways in MCF7/ADR and K562/ADR. Journal of cellular physiology, 233, 3066-3079. SUN, Z., YAN, B., YU, W. Y., YAO, X., MA, X., SHENG, G. & MA, Q. 2016. Vitexin attenuates acute doxorubicin cardiotoxicity in rats via the suppression of oxidative stress, inflammation and apoptosis and the activation of FOXO3a. Experimental and therapeutic medicine, 12, 1879-1884. SUVARNA, K. S., LAYTON, C. & BANCROFT, J. D. 2018. Bancroft's Theory and Practice of Histological Techniques E-Book, Elsevier Health Sciences. VYAS, D., LAPUT, G. & VYAS ,A. K. 2014. Chemotherapy-enhanced inflammation may lead to the failure of therapy and metastasis. OncoTargets and therapy, 7, 1015. WANG, L., CHEN, Q., QI, H., WANG, C., WANG, C., ZHANG, J. & DONG, L. 2016. Doxorubicininduced systemic inflammation is driven by upregulation of toll-like receptor TLR4 and endotoxin leakage. Cancer research. WANG, N., ZHANG, D. X., ZHANG, S. C., DU, M., LI, J. L. & SUN, Y. X. 2015. Biomarkers for kidney injury induced by gentamicin, doxorubicin and adenine in rats. WANG ,Y., WANG, Y. P., TAY, Y.-C. & HARRIS, D. C. 2000. Progressive adriamycin nephropathy in mice: sequence of histologic and immunohistochemical events. Kidney international, 58, 1797-1804. WEI, X., HEYWOOD, G., DI GIROLAMO, N. & THOMAS, P. 2003. Nicorandil inhibits the release of TNFα from a lymphocyte cell line and peripheral blood lymphocytes. International immunopharmacology, 3, 1581-1588. YAO, H., CAI, Z. & SHENG, Z. 2017. NAC attenuates adriamycin-induced nephrotic syndrome in rats through regulating TLR4 signaling pathway. Eur Rev Med Pharmacol Sci, 21, 19381943. YU, D., FAN, C., ZHANG, W., WEN, Z., HU, L., YANG, L., FENG, Y., YIN, K.-J. & MO, X. 2015. Neuroprotective effect of nicorandil through inhibition of apoptosis by the PI3K/Akt1 pathway in a mouse model of deep hypothermic low flow. Journal of the neurological sciences, 357, 119-125. YU, Y., XIAO, Y., WANG, H., LI, J., ZUO, X., WANG, H. & XIE, W. 2013. Protective effect of nicorandil on hypoxiainduced apoptosis in HPAECs through inhibition of p38 MAPK phosphorylation. Mol Med Rep, 7, 816-20. ZHAO, A. P., DONG, Y. F., LIU, W., GU, J. & SUN, X. L. 2014. Nicorandil Inhibits Inflammasome Activation and Toll-Like Receptor-4 Signal Transduction to Protect against Oxygen– Glucose Deprivation-Induced Inflammation in BV-2 Cells. CNS neuroscience & therapeutics, 20, 147-153. ZHAO, L., TAO, X., QI, Y., XU, L., YIN, L. & PENG, J. 2018. Protective effect of dioscin against doxorubicin-induced cardiotoxicity via adjusting microRNA-140-5p-mediated myocardial oxidative stress. Redox biology, 16, 189-198. 30
645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695
ZHU, F., ZHONG, X., ZHOU, Y., HOU, Z., HU, H., LIANG, L., CHEN, J., CHEN, Q., JI, X. & SHANG, D. 2018. Protective effects of nicorandil against cerebral injury in a swine cardiac arrest model. Experimental and Therapeutic Medicine, 16, 37-44. ZHU, Y., GUAN, F., NA, A., BAI, S. & YANG, S. 2015. Total glucosides of paeony inhibits adriamycin-induced nephropathy in rats through blocking TLR4/NF-κB/TGF-β1 signaling. Chinese Journal of Biochemical Pharmaceutics, 43.46-
696 697 698 699 700 701 702
31
Table1: Table1: Effect of Nicorandil on serum levels of urea, creatinine, total protein, albumin, albumin, Kidney injury moleculemolecule-1 and cystatin in doxorubicindoxorubicin-induced nephrotoxicity in rats.
Group
Control saline Nicorandil Doxorubicin Nicorandil + Doxorubicin
Serum urea
Serum
Serum total
Serum albumin
Kidney injury
Serum cystatin
(mg/dl)
creatinine
protein
g/dl
moleculemolecule-1
Unit
(mg/dl)
g/dl
pg/ml
pg/ml
33.17 ± 1.9
0.18 ± 0.013
5.59 ± 0.15
3.48 ± 0.25
2.2 ± 0.09
0.66 ± 0.05
30.33 ± 1.35b
0.45 ± 0.01b
5.51 ± 0.14b
3.5 ± 0.34b
2.13 ± 0.1b
0.63 ± 0.06 b
83.83 ± 2.79a
1.64 ± 0.12 a
3.77 ± 0.06a
2.4 ± 0.22a
12.17 ± 0.35a
2.862 ± 0.11a
58.83 ± 2.13ab
0.70 ± 0.07ab
4.98 ± 0.03ab
3.3 ± 0.23ab
5.65 ± 0.37ab
1.23 ± 0.17ab
1
Table 2: Effect of Nicorandil on histopathological alterations in doxorubicin-induced nephrotoxicity in rats.
Group
Control saline
Nicorandil
Doxorubicin
Doxorubicin + nicorandil
Histopathlgical alteration Tubular degeneration
_
_
++
_
Focal inflammatory cell infiltration
_
_
++
+
Focal fibrosis
_
_
++
_
Vaculisation of glomerular endothelium
_
_
+++
_
Congestion in blood vessels
_
_
++
_
Perivascular edema
_
_
++
Focal fibrosis
_
_
++
_
Focal haemorrhage
_
_
++
_
Degeneration in the tubules
_
_
++
_
+++ severe ++ moderate + mild 0 none 2
Statistical analysis was carried out by one way ANOVA followed by Tukey- Multiple Comparison Test. Each value represents the standard deviation of 8 rats (S.E.). .a Significantly different from normal control group value at p < 0.05 .b Significantly different from doxorubicin group value at p < 0.05
3
8
a
40
4
a
a 60 40
ab 20
0
0
b
1.5
ab
b
ab
100
100
b
Catalase ng/ g tissue
a
b
0
150
150
MPO ng/ g tissue
ab
2
20
0
6
MDA nmol/g tissue
ab
60
80
a 50
0
Nrf2 pg/ g tissue
GSH mg/g tissue
80
50
b
b
SOD Pg/ g tissue
100
1.0
ab 0.5
a 0.0
Groups
Figure 1 A-F : Effect of Nicorandil on renal content of GSH, MDA, SOD, MPO, Nrf2 and catalase in doxorubicin-induced nephrotoxicity in rats. Each value represents the mean of 8-10 rats ± standard error of the mean (SE.). Statistical analysis was carried out by one way ANOVA followed by Tukey Multiple Comparison Test. a b
Significantly different from normal control group value at p < 0.05. Significantly different from doxorubicin group value at p < 0.05.
nrf2 b actin
b
2 0
ab 4
b
2
a
100
100
ab
0
b
2
150
a
50
ab
4
0
0
150
IL.1B pg/ g tissue
P38 ng/ g tissue
ab
4
a
6
6
TNF Pg/ g tissue
NFĸb Pg/ g tissue
a 6
8
a
8
TLR4 ng/ g tissue
8
b
ab b
50
NFĸb P38
0
TLR4 b actin Groups
Figure 2A-D : Effect of Nicorandil on renal content of NFKb, P38 MAPK, TLR4, IL-1β and TNF-α in doxorubicininduced nephrotoxicity in rats. Each value represents the mean of 8-10 rats ± standard error of the mean (SE.). Statistical analysis was carried out by one way ANOVA followed by Tukey Multiple Comparison Test. a b
Significantly different from normal control group value at p < 0.05. Significantly different from doxorubicin group value at p < 0.05.
Bax pg/ g tissue
6
ab 4 2
b
0
BcL-2 pg/ g tissue
a
a
b
30
1.0
ab 0.5
a
Bax /Bcl-2 ratio
1.5
8
20
10
ab b
0
0.0
Bax BcL-2
Groups
b actin
b actin
Figure (3A-C) : Effect of Nicorandil on renal content of Bax and Bcl in doxorubicin-induced nephrotoxicity in rats. Each value represents the mean of 8-10 rats ± standard error of the mean (SE.). Statistical analysis was carried out by one way ANOVA followed by Tukey Multiple Comparison Test. a b
Significantly different from normal control group value at p < 0.05. Significantly different from doxorubicin group value at p < 0.05.
g
Figure 4: Effect of Nicorandil on kidney sections in doxorubicin-induced nephrotoxicity in rats stained with hematoxylin/eosin (H/E) and examined under the light microscope. Doxorubicin treated rats showed degenerative changes; focal inflammatory cells infiltration (DOX 1), The endothelial cells lining the tufts of the glomeruli showed vacuolization (DOX 2), The cortical stromal blood vessels showed congestion (DOX 3) , as well as perivascular oedema (DOX 4), The corticomedullary portion showed focal fibrosis (DOX 5), and focal haemorrhages in between the tubules (DOX 6), There were swelling in the lining tubular epithelium with obliteration in the tubular lumen (DOX 7), while other
tubules at the corticomedullary portion had vacuolar degeneration in the lining epithelium (DOX 8), There was no histopathological alteration as recorded in (nicorandil only) , Focal inflammatory cells infiltration was detected in between the degenerated tubules at the cortex (nicorandil + DOX).
• Nicorandil ameliorated doxorubicin-induced nephrotoxicity.
1
• Nicorandil restored the oxidant/antioxidant balance.
2
• Nicorandil suppressed inflammatory signaling pathway TLR4/MAPK
3
P38/NF-κb/TNF-α.
4
• Nicorandil regulated BAX/Bcl-2 apoptotic pathway
1
5
Declaration of interests ☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. ☐The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: