Protective Role of CoQ10 or L-Carnitine On The Integrity of The Myocardium in Doxorubicin Induced Toxicity

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Tissue and Cell 49 (2017) 410–426

Contents lists available at ScienceDirect

Tissue and Cell


journal homepage: www.elsevier.com/locate/tice

Protective role of CoQ10 or L-carnitine on the integrity of the


myocardium in doxorubicin induced toxicity
Hesham N. Mustafa a,∗ , Gehan A. Hegazy b,c , Sally A. El Awdan d , Marawan AbdelBaset d
a
Anatomy Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
b
Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
c
Medical Biochemistry Department, National Research Centre, Cairo, Egypt
d
Pharmacology Department, National Research Center, Cairo, Egypt

a r t i c l e i n f o a b s t r a c t

Article history: Doxorubicin (DOX) is a chemotherapeutic agent used for treatment of different cancers and its clinical
Received 16 January 2017 usage is hindered by the oxidative injury-related cardiotoxicity. This work aims to declare if the harmful
Received in revised form 15 March 2017 effects of DOX on heart can be alleviated with the use of Coenzyme Q10 (CoQ10) or L-carnitine. The study
Accepted 31 March 2017
was performed on seventy two female Wistar albino rats divided into six groups, 12 animals each: Control
Available online 2 April 2017
group; DOX group (10 mg/kg); CoQ10 group (200 mg/kg); L-carnitine group (100 mg/kg); DOX + CoQ10
group; DOX + L-carnitine group. CoQ10 and L-carnitine treatment orally started 5 days before a single dose
Keywords:
of 10 mg/kg DOX that injected intraperitoneally (IP) then the treatment continued for 10 days. At the end
Dox
CoQ10 and L-carnitine of the study, serum biochemical parameters of cardiac damage, oxidative stress indices, and histopatho-
Cardiotoxicity logical changes were investigated. CoQ10 or L-carnitine showed a noticeable effects in improving cardiac
eNOS functions evidenced reducing serum enzymes as serum interleukin-1 beta (IL-1 ␤), tumor necrosis factor
Vimentin alpha (TNF-␣), leptin, lactate dehydrogenase (LDH), Cardiotrophin-1, Troponin-I and Troponin-T. Also,
alleviate oxidative stress, decrease of cardiac Malondialdehyde (MDA), Nitric oxide (NO) and restoring
cardiac reduced glutathione levels to normal levels. Both corrected the cardiac alterations histologically
and ultrastructurally. With a visible improvements in ␣-SMA, vimentin and eNOS immunohistochemical
markers. CoQ10 or L-carnitine supplementation improves the functional and structural integrity of the
myocardium.
© 2017 Elsevier Ltd. All rights reserved.

1. Introduction (Potemski et al., 2006). Cardiomyocytes suffer from relative defi-


ciency of antioxidant enzymes so they are more vulnerable for the
Chemotherapy is not a risk-free experience due to the unwanted deleterious effects produced by free radical result from DOX admin-
effects of these medications on the healthy cells. DOX (Adriamycin) istration (Dong et al., 2014). DOX-induced cardiotoxicity may result
is one of the chemotherapeutic medications that is widely used in evident cardiac damage before clinical signs become apparent,
and occurs within anthracycline list of antibiotics. It is produced by hence the importance of detecting biochemical markers possess
Streptomyces peucetius var caesius algae (Arcamone et al., 2000). It high sensitivity and specificity that indicates early damage to be
is used in the management of lymphomas, leukemia and different used for screening and early diagnoses of DOX-induced cardiotox-
solid tumors like carcinoma of ovaries, breast, lung and thyroid icity (Atas et al., 2015).
gland (Gianni et al., 2007). L-carnitine is a naturally occurring compound biosynthesized
DOX has a dose dependent cardiotoxicity (Elbaky et al., 2010). from the amino acids methionine and lysine in the kidneys and
DOX toxicity is responsible for 2–3% of all cases of heart trans- liver. It is found at a high concentration in skeletal and cardiac
plantation patients (Mitry and Edwards, 2016). DOX-induced muscles (Aliev et al., 2009). L-carnitine exerts an antioxidant action
cardiotoxicity is mainly mediated via oxidative stress and apoptosis that protect against lipid peroxidation of membrane phospholipid
(Guan et al., 2009). L-carnitine eliminates extracellular toxic acetyl-
coenzyme A that is responsible for mitochondrial ROS (reactive
oxygen species) (Agarwal and Said, 2004). L-carnitine revealed a
∗ Corresponding author at: King Abdulaziz University, Faculty of Medicine, varied range of biological actions comprising anti-inflammatory
Anatomy Department, Building No. 8 − P.O. Box: 80205, Jeddah, 21589, Saudi Arabia. and anti-apoptotic properties (İzgüt-Uysal et al., 2003).
E-mail address: hesham977@hotmail.com (H.N. Mustafa).

http://dx.doi.org/10.1016/j.tice.2017.03.007
0040-8166/© 2017 Elsevier Ltd. All rights reserved.
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 411

CoQ10 is a compound which is synthesized endogenously that At the end of treatment, the animals were kept for an overnight
is a potent lipophilic antioxidant capable of recycling and regen- fasting and the blood samples were collected from retroorbital
erating other antioxidants such as ascorbate and tocopherol. Also, plexus and allowed to clot for 30 min at room temperature. After
CoQ10 causes scavenging of free radicals, inhibition of lipid perox- blood collection, all animals were rapidly sacrificed and the hearts
idation (Zhang et al., 2013). CoQ10 is a cofactor that plays a crucial were dissected and immediately homogenized in 50 mM ice-cold
role in the mitochondria respiratory chain and ATP production phosphate buffer (pH 7.4) to give 10% homogenate (w/v). The
(Bhagavan and Chopra, 2006). homogenate was centrifuged at 3200 rpm for 20 min in cooling cen-
␣-Smooth muscle actin (␣-SMA) is a marker for myofibroblast- trifuge. The supernatant was used for the determination of different
like cells and hepatic stellate cells (HSC) (Mustafa, 2016). Vimentin parameters.
is an intermediate filament (IF) protein that is expressed frequently
in the cells of mesodermal origin as endothelial cells, that forms 2.5. Biochemical measurements
an irregular network in endomysium and perimysium sheaths of
the myocardium (Heling et al., 2000). Vimentin expression occurs 2.5.1. Measurement of malondialdehyde (MDA) and reduced
during myocardial stress as heart failure (Sharov et al., 2005). glutathione (GSH)
Endothelial nitric oxide synthase (eNOS) is shown in the Measurement of malondialdehyde (MDA) and reduced glu-
endothelium, within the heart, in cardiac conduction tissue and tathione (GSH) levels using colorimetric assay kits (Catalogues No.
in cardiac myocytes (Jones et al., 2004). In cardiac muscle, eNOS MD 25 29, GR 25 11 respectively) in accordance with the manufac-
arranges NO physiological action, as organizing endothelial func- turer’s instructions (Bio Diagnostic, Cairo, Egypt).
tion, platelet aggregation, vascular tone and cardiac contractility
(Pott et al., 2006). The expression of eNOS in the myocardium is 2.5.2. Measurement of Nitric oxide (NO)
modulated in dilated cardiomyopathy with evidence of heart fail- Nitric oxide metabolites (NO) were determined according to
ure (Crespo et al., 2008). Thus, the aim of this work is to declare the the method described by Miranda et al. (Miranda et al., 2001)
possible ameliorative potentials of CoQ10 or L-carnitine on DOX and expressed as ␮M/g wet tissue using colorimetric assay kits
induced cardiotoxicity. (Catalogue No. NO 25 33) in accordance with the manufacturer’s
instructions (Bio Diagnostic, Cairo, Egypt). Nitric oxide has a short
2. Material and methods biological half-life and is rapidly converted into its stable metabo-
lites, nitrite and nitrate. Determination of nitrite and nitrate (NOx)
2.1. Ethical approval in body fluid and tissues is widely used as a marker of NO produc-
tion Miranda et al. (Miranda et al., 2001). Nitric oxide measured
The study was conducted after approval by the Medical Research as nitrite was determined using Griess reagent, according to the
Ethics Committee of the National Research Centre, Cairo, Egypt and method of Moshage et al. (Moshage et al., 1995), where nitrite, sta-
followed the recommendations of the National Institutes of Health ble end product of nitric oxide radical, is mostly used as an indicator
Guide for Care and Use of laboratory Animals (NIH Publications No. for the production of nitric oxide.
8023, revised 1978).
2.5.3. Assessment of inflammatory cytokines
2.2. Animals Serum interleukin-1beta (IL-1 ␤), tumor necrosis factor (TNF-
␣) and Leptin levels were measured using an enzyme-linked
The study was performed on female Wistar albino rats (n = 72), immunosorbent assay (ELISA) kit (Catalogues No. RAB0277 Sigma,
8–10 weeks of age and weighed ranging 150–200 g that were bred RAB0479 Sigma and RAB0335 Sigma respectively) according to the
and obtained from Animal House Colony, National Research Centre, manufacturer’s instructions (Sigma-Aldrich, St. Louis, MO, United
Cairo, Egypt. All animals were housed in cages in a temperature con- States). All samples were tested in duplicate and averaged.
trolled (24 ± 1 ◦ C) with a 12 h light/dark cycle and 60 ± 5% humidity
and were provided with standard laboratory diet and water ad libi-
2.5.4. Assessment of cardiac markers
tum. DOX was provided by Sigma, which was dissolved in sterile
Lactate dehydrogenase (Catalog No. MA5-17242) and cardiac
saline. CoQ10 and L-carnitine were obtained from Mepaco, Egypt.
specific creatinine kinase levels were measured using commercial
kits (Catalog No. LF-MA0233) purchased from Invitrogen (Thermo
2.3. Experiment
Fisher Scientific, Inc., Waltham, MA, USA) according to the manu-
facturer’s protocol. All measurements were performed in duplicate.
Rats were divided into six groups, including 12 animals: Control
group; DOX group (10 mg/kg) (Mustafa et al., 2015); CoQ10 group
2.5.5. Quantitative estimation of serum troponin I
(200 mg/kg)(Mustafa et al., 2015); L-carnitine group (100 mg/kg)
Quantitative estimation of serum troponin I levels were car-
(Mescka et al., 2016); DOX + CoQ10 group; DOX + L-carnitine group.
ried out by ELISA technique-using kit (Catalog No. LS-F127394)
CoQ10 and L-carnitine treatment orally started 5 days before a sin-
purchased from Lifespan BioSciences international Inc., USA
gle dose of 10 mg/kg DOX that was injected intraperitoneally (IP)
then the treatment was continued for 10 days. At the beginning and
at the end of the study the animals body weights were measured. 2.5.6. Measurement of serum Troponin-T (cTnT) levels
Troponin-T (cTnT) levels were measured cTnT with a third-
2.4. Echocardiographic study generation cardio-specific assay (Catalog No. 04660307190)
(ElecsysR Troponin T STATimmunoassay manufactured by Roche
ECG was recorded at the beginning of the experiment to ensure Diagnostics, France)
the normal ECG pattern of the rats. At the last day of the experiment,
rats were anesthetized by dimethyl ether and ECG was recorded for 2.5.7. Measurement of Cardiotrophin-1
1 min. Heart rate, P duration, QRS Interval, QTc, and ST Height were Cardiotrophin-1 is measured based on the sandwich ELISA
monitored using ECG Powerlab module which consists of Power- principle following the manufacturer’s instructions using Rat
lab/8sp and Animal Bio-Amplifier, Australia, in addition to Lab Chart CTF1/Cardiotrophin-1 ELISA Kit (Catalog No. LS-F127394), pur-
7 software with ECG analyzer (Hajrasouliha et al., 2004). chased from LifeSpan BioSciences international Inc., USA.
412 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

Table 1
Effect of CoQ10 and L-carnitine on body weight, heart weight, heart/body weight% and mortality No.

Groups Control N = 12 CoQ10 N = 12 L-carnitine DOX N = 9 DOX + CoQ10 DOX + L-


N = 12 N = 11 carnitine
N = 11

Body weight 152.3 ± 2.31 153.01 ± 3.41 151.33 ± 5.61 135 ± 6.51 146.07 ± 3.80 144.4 ± 5.03
(g) NS NS 1
P ≤ 0.001 1
P ≤ 0.05 1
P ≤ 0.01
2
P ≤ 0.001 2
P ≤ 0.001
Heart weight 0.810 ± 0.019 0.79 ± 0.021 0.83 ± 0.017 0.482 ± 0.040 0.603 ± 0.011 0.594 ± 0.026
(g) NS NS P1 ≤ 0.001 1
P ≤ 0.001 1
P ≤ 0.001
2
P ≤ 0.001 2
P ≤ 0.001
Heart 0.532 ± 0.002 0.516 ± 0.006 0.548 ± 0.008 0.357 ± 0.007 0.413 ± 0.003 0.411 ± 0.001
weight/Body 1
P ≤ 0.001 1
P ≤ 0.001 1
P ≤ 0.001 1
P ≤ 0.001 1
P ≤ 0.001
weight% 2
P ≤ 0.001 2
P ≤ 0.001
Mortality No. 0 0 0 3 1 1

Values are means ± SD (Control n = 12& DOX = 9 & treated = 11). ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX.

Table 2
Comparison of electrocardiographic changes in different studied groups.

Groups Heart Rate P-R (s) QRS Interval (s) QTc duration P amplitude T amplitude S-T Height
(bpm) (s) (mV) (mV) (mV)

Control 318.386 ± 47.582 0.164 ± 0.017 0.015 ± 0.002 0.096 ± 0.005 0.061 ± 0.018 0.185 ± 0.043 0.035 ± 0.033
(n = 12)
DOX 281.930 ± 22.591 0.214 ± 0.019 0.016 ± 0.004 0.134 ± 0.077 0.057 ± 0.057 0.145 ± 0.105 0.090 ± 0.066
1 1 1 1 1 1 1
Significance P = 0.013 P = 0.0001 P = 0.624 P = 0.024 P = 0.743 P = 0.141 P = 0.006
(n = 9)
CoQ10 286.167 ± 10.913 0.187 ± 0.024 0.017 ± 0.002 0.101 ± 0.005 0.066 ± 0.007 0.174 ± 0.021 0.059 ± 0.030
1 1 1 1 1 1 1
Significance P = 0.061; P = 0.059; P = 0.275; P = 0.811; P = 0.796; P = 0.750; P = 0.405;
2 2 2 2 2 2 2
(n = 12) P = 0.815 P = 0.016 P = 0.443 P = 0.082 P = 0.625 P = 0.409 P = 0.116
L-carnitine 307.483 ± 44.887 0.182 ± 0.031 0.015 ± 0.001 0.103 ± 0.014 0.066 ± 0.009 0.209 ± 0.058 0.023 ± 0.019
1 1 1 1 1 1 1
Significance P = 0.520; P = 0.111; P = 0.698; P = 0.773; P = 0.774; P = 0.482; P = 0.335;
2 2 2 2 2 2 2
(n = 12) P = 0.161 P = 0.003 P = 0.447 P = 0.148 P = 0.604 P = 0.071 P = 0.002
DOX + CoQ10 340.989 ± 8.550 0.179 ± 0.007 0.017 ± 0.002 0.096 ± 0.011 0.047 ± 0.031 0.127 ± 0.063 0.065 ± 0.008
1 1 1 1 1 1 1
Significance P = 0.130; P = 0.123; P = 0.140; P = 0.975; P = 0.260; P = 0.022; P = 0.216;
2 2 2 2 2 2 2
(n = 11) P = 0.0001 P = 0.0001 P = 0.276 P = 0.009 P = 0.422 P = 0.486 P = 0.154
DOX + L- 311.536 ± 36.491 0.188 ± 0.022 0.014 ± 0.003 0.105 ± 0.036 0.068 ± 0.29 0.170 ± 0.075 0.037 ± 0.035
carnitine
1 1 1 1 1 1 1
Significance P = 0.560; P = 0.015; P = 0.336; P = 0.603; P = 0.588; P = 0.050; P = 0.755;
2 2 2 2 2 2 2
(n = 11) P = 0.030 P = 0.002 P = 0.119 P = 0.069 P = 0.386 P = 0.036 P = 0.004

Values are means ± SD. ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX.
Bpm: beat per minute. S: seconds. mV: millivolts.

2.6. Histological studies Carpinteria, California, USA] with a dilution of 1:1000; cellular
site was cytoplasmic) to evaluate the fibrosis. They were similarly
2.6.1. Light microscopic study incubated with vimentin (a Mouse monoclonal antibody [Dako,
Tissues were fixed in 10% neutral buffered formalin and 5 ␮m in Carpinteria, California, USA] with a dilution of 1:400; cellular site
thickness sections were prepared. For each specimen, at least three was cytoplasmic) as a cytoskeleton marker for cardiac fibroblasts
to five slides were stained with H&E (hematoxylin and eosin) for and endothelial cells and pericytes. They were incubated with
general examination, Masson’s trichrome stain to demonstrate col- eNOS (a rabbit polyclonal antibody [Santa Cruz Biotechnology, CA,
lagen fibers. Slides were observed with Olympus BX53 microscope USA] with a dilution of 1:50; cellular site was cytoplasmic) is
equipped with DP73 camera (Olympus, Tokyo, Japan) (Mustafa, involved in the modulation of cardiac myocyte function. Sections
2015). The scoring system for the severity of changes was quan- were incubated at room temperature with HRP (horseradish per-
titated from none (0) to severe (4) based on the degree of necrosis, oxidase) conjugate as a secondary antibody (Invitrogen, Zymed,
cytoplasmic vacuolations, myocardial disorganization, degenera- Burlington, ON, Canada). Sections were then incubated with DAB
tion edema and inflammatory cell infiltrate (Alpsoy et al., 2013; (3,3 -diaminobenzidine tetrachloride; Vector Laboratories, Orton
Dudka et al., 2012; Gala, 2013; Mandziuk et al., 2015). Southgate, Peterborough, United Kingdom) substrate chromogen
solution (1 drop of DAB chromogen/1 mL of substrate buffer)
2.6.2. Immunohistochemical study for 5 min to detect immunoreactivity. All sections were counter-
Streptavidin–biotin peroxidase technique was applied to stained with Mayer’s hematoxylin and negative control sections
paraffin-embedded tissue. 5 ␮ sections were de-waxed and pre- were prepared by omitting the primary antibody. While positive
treated with 3% H2 O2 (hydrogen peroxide) to block endogenous control standard slides were used to prove the success of the tech-
peroxidase activity. Microwave-assisted antigen retrieval was per- nique. All slides were examined and the presence of labeled cells
formed for 10 min in 0.01 M sodium citrate buffer (pH 6.0) at was documented. Absence of staining was recognized as a negative
95 ◦ C, and then, the slides were cooled at room temperature for result (−), while the presence of brown staining was recognized as
20 min. Blocking non-specific binding by incubating in 3% BSA/PBS positive result (+) (Mustafa, 2016).
(Bovine Serum Albumin/Phosphate buffered saline) for 10 min.
Then, slides were incubated overnight at 4 ◦ C with the primary
antibody against ␣-SMA (a mouse monoclonal antibody [Dako,
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 413

Fig. 1. Effect of CoQ10 and L-carnitine on body weight, heart weight and heart/body weight%.
Values are means ± SD (Control n = 12& DOX = 9 & treated = 11). ANOVA followed by Bonferroni’s post hoc test.
1P: compared to control. 2P: compared to DOX.

Table 3
Comparison of measured oxidative stress parameters in heart tissue homogenate in different studied groups.

Groups Malondialdehyde Reduced Nitric oxide


(nM/g) glutathione (␮M/g)
(␮M/g)

Control 11.79 ± 2.89 1.06 ± 0.19 10.00 ± 0.65


(n = 12)
DOX 65.60 ± 9.46 0.82 ± 0.17 27.71 ± 8.96
1 1 1
Significance P = 0.0001 P = 0.005 P = 0.0001
(n = 9)
CoQ10 17.18 ± 1.66 1.26 ± 0.11 10.49 ± 0.52
1 1 1
Significance P = 0.202; P = 0.022; P = 0.840;
2 2 2
(n = 12) P = 0.0001 P = 0.0001 P = 0.0001
L-carnitine 19.10 ± 2.24 1.31 ± 0.06 11.34 ± 0.59
1 1 1
Significance P = 0.101; P = 0.004; P = 0.579;
2 2 2
(n = 12) P = 0.0001 P = 0.0001 P = 0.0001
DOX + CoQ10 40.77 ± 7.44 1.15 ± 0.10 18.77 ± 1.38
1 1 3
Significance P = 0.0001; P = 0.266; P = 0.001;
2 2 2
(n = 11) P = 0.0001 P = 0.0001 P = 0.001
DOX + L- 41.03 ± 10.73 1.08 ± 0.08 16.29 ± 1.51
carnitine
1 1 1
Significance P = 0.0001; P = 0.840; P = 0.015;
2 2 2
(n = 11) P = 0.0001 P = 0.003; P = 0.0001

Values are means ± SD. ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX. nM/g: nanomolar/gram. ␮M/g: micromolar/gram.

2.6.3. Morphometric study Maryland, USA) and ImageJ (NIH, 1.51; Melville, NY, USA), which
Ten non-overlapping fields for each animal were selected ran- was calibrated for distance, color and area before its use (Mustafa
domly and analyzed to determine cardiomyocytes’ diameter of and Hussein, 2015).
H&E stained sections. Cardiomyocytes with centrally located visible
nuclei intact cell membrane were selected and the measurements
were done along their short axis (de Salvi Guimaraes et al., 2017; 2.6.4. Ultrastructure study
Nascimento et al., 2016; Pradegan et al., 2016). The area percent- One mm3 samples were immersed in 2.5% glutaraldehyde in
age of collagen fibers in Masson’s trichrome, ␣-SMA, vimentin and 0.1 M phosphate buffer at 4 ◦ C for 3 hs and post-fixed in 1% OsO4
eNOS-stained sections. Quantitative measurements were analyzed (osmium tetraoxide). Then, tissues were embedded in Epon 812 and
with the use of Image-Pro Plus v6 (Media Cybernetics Inc., Bethesda, semithin sections were prepared, stained with toluidine blue and
observed with a microscope. Ultrathin sections of 50–60 nm thick
414 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

Fig. 2. A: Comparison of electrocardiographic changes in different studied groups [Heart Rate (bpm)]. B: Comparison of electrocardiographic changes in different stud-
ied groups [P-R duration (seconds)]. C: Comparison of electrocardiographic changes in different studied groups [QRS Interval (seconds)]. D: Comparison of electrocardiographic
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 415

Table 4
Comparison of measured inflammatory parameters in different studied groups.

Groups Interleukin-1␤ Tumor necrosis Leptin (pg/ml) Lactate


(pg/ml) factor-␣ dehydrogenase
(pg/ml) (U/ml)

Control 40.35 ± 7.47 40.25 ± 4.48 27.12 ± 3.45 100.80 ± 12.24


(n = 12)
DOX 286.01 ± 24.55 198.00 ± 12.43 120.14 ± 10.51 403.40 ± 37.83
1 1 1 1
Significance P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
(n = 9)
CoQ10 45.43 ± 8.82 53.86 ± 22.80 27.54 ± 6.86 96.20 ± 18.10
1 1 1 1
Significance P = 0.597; P = 0.077; P = 0.932; P = 0.740;
2 2 2 2
(n = 12) P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
L-carnitine 42.28 ± 5.27 37.19 ± 3.93 27.36 ± 7.19 105.00 ± 17.92
1 1 1 1
Significance P = 0.840; P = 0.682; P = 0.961; P = 0.761;
2 2 2 2
(n = 12) P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
DOX + CoQ10 84.44 ± 8.82 72.24 ± 6.52 48.66 ± 6.35 131.60 ± 18.37
1 1 1 1
Significance P = 0.0001; P = 0.0001; P = 0.0001; P = 0.034;
2 2 2 2
(n = 11) P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
DOX + L- 143.51 ± 21.67 113.61 ± 7.90 84.72 ± 9.80 182.80 ± 15.51
carnitine
1 1 1 1
Significance P = 0.0001; P = 0.0001; P = 0.0001; P = 0.0001;
2 2 2 2
(n = 11) P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001

Values are means ± SD. ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX.
pg/ml: Picograms per Millilitre. U/ml: Units per Millilitre.

Table 5
Comparison of measured heart parameters in different studied groups.

Groups Cardiotrophin- Cardiac Troponin-I Troponin-T


1 specific- (ng/ml) (ng/ml)
(pg/ml) creatine kinase
(ng/ml)

Control 68.20 ± 8.47 100.80 ± 12.24 0.72 ± 0.06 0.39 ± 0.08


(n = 12)
DOX 237.36 ± 18.01 403.40 ± 37.83 5.80 ± 0.74 1.81 ± 0.55
1 1 1 1
Significance P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
(n = 9)
CoQ10 62.45 ± 6.19 96.20 ± 18.10 0.70 ± 0.09 0.43 ± 0.07
1 1 1 1
Significance P = 0. 0.375; P = 0.740; P = 0.952; P = 0.850;
2 2 2 2
(n = 12) P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
L-carnitine 64.08 ± 6.22 105.00 ± 17.92 0.68 ± 0.15 0.44 ± 0.13
1 1 1 1
Significance P = 0. 0.523; P = 0.761; P = 0.886; P = 0.792;
2 2 2 2
(n = 12) P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
DOX + COQ10 109.78 ± 9.10 131.60 ± 18.37 1.44 ± 0.38 0.98 ± 0.33
1 1 1 1
Significance P = 0.0001; P = 0.034; P = 0.011; P = 0.004;
2 2 2 2
(n = 11) P = 0.0001 P = 0.0001 P = 0.0001 P = 0.0001
DOX + L- 167.08 ± 7.05 182.80 ± 15.51 2.37 ± 0.55 1.39 ± 08
carnitine
1 1 1 1
Significance P = 0.0001; P = 0.0001; P = 0.0001; P = 0.0001;
2 2 2 2
(n = 11) P = 0.001 P = 0.0001 P = 0.0001 P = 0.23

Values are means ± SD. ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX. pg/ml: picograms per Millilitre. ng/ml: Nanograms per Millilitre.

were cut by ultramicrotome (NOVA, LKB 2188, Bromma, Sweden); tistical Package for the Social Sciences (SPSS), version 23. The values
and stained with uranyl acetate and lead citrate. Then tissues were considered significant when P < 0.05 (Mustafa, 2015).
were examined with Philips 201 transmission electron micro-
scope (Philips Industries, Eindhoven, Netherlands) at 60–80 kv at
the Transmission Electron Microscope Unit (Mustafa and Hussein,
2015).
3. Results

2.7. Statistical analysis 3.1. General assessment

Statistical Analysis. Quantitative data were expressed as the The results revealed a significant decrease in the heart/body
mean and standard deviations. Data were analyzed using a one- weight ratio in DOX group. The administration of CoQ10 or
way analysis of variance (ANOVA) followed by Bonferroni’s post L-carnitine significant increase in the heart/body weight ratio
hoc test. All statistical analyses were implemented using the Sta- (Table 1, Fig. 1).

changes in different studied groups [T amplitude (mV)]. E: Comparison of electrocardiographic changes in different studied groups [S-T Height (mV)]. (F&G): ECG of control
group. ECG of DOX group. Rats were anesthetized and ECG was recorded for 1 min. PVC: premature ventricular complex. AV block: Atrio-ventricular block.
416 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

3.2. ECG findings in the area percentage of collagen (P < 0.001) when compared with
the control (Tables 6, 7).
Heart rate was significantly lower in DOX group than control, DOX + CoQ10 group: H&E showed nearly normal microscopic
DOX + CoQ10 and DOX + L-carnitine groups. P-R duration was sig- architecture of cardiomyocytes with minimal changes in nuclei
nificantly higher in DOX + L-carnitine group versus control and was were observed (Fig. 6C). Masson’s trichrome stained sections
significantly higher in DOX group than control, CoQ10, L-carnitine, showed mild reaction (Fig. 7C). These results were confirmed
DOX + CoQ10 and DOX + L-carnitine groups. QTc was significantly by morphometric and statistical study (Tables 6, 7). DOX + L-
higher in DOX group than control and DOX + CoQ10. T amplitude carnitine group: H&E showed apparently normal microscopic
was significantly lower in DOX group than DOX + L-carnitine. S-T histo-architecture of cardiomyocytes with mild changes in nuclei
height was significantly lower in DOX group than control, L- were observed (Fig. 6D). Masson’s trichrome stained sections
carnitine and DOX + L-carnitine (Table 2, Fig. 2A–I). showed mild reaction (Fig. 7D). These results were confirmed by
morphometric and statistical study (Tables 6, 7). Cardiomyocytes
3.3. Heart tissue homogenate levels of oxidative stress markers diameter of groups treated with CoQ10 and L-carnitine showed a
significant improvement as compared with DOX group (Table 6,
Heart tissue homogenate levels of MDA was significantly higher Fig. 11). Area percentage of collagen (Masson’s trichrome stain) of
in DOX group than control, CoQ10, L-carnitine, DOX + CoQ10 groups treated with CoQ10 and L-carnitine showed a significant
and DOX + L-carnitine groups and in DOX + CoQ10 and DOX + L- improvement as compared with the DOX group (Tables 6, 7 Fig. 11).
carnitine groups versus control. Heart tissue homogenate levels
of NO was significantly higher in DOX group than control, CoQ10, 3.6.1. Immunohistochemical results for ˛-SMA
L-carnitine, DOX + CoQ10 and DOX + L-carnitine groups and in Immunohistochemical Results for ␣-SMA of the control group
DOX + CoQ10 and DOX + L-carnitine groups versus control. Heart revealed minimal immune expression (Fig. 8A). DOX group
tissue homogenate levels of reduced glutathione was significantly showed extensive immune expression (Fig. 8B). DOX + CoQ10 group
lower in DOX group than control, CoQ10, L-carnitine, DOX + CoQ10 revealed mild immune expression (Fig. 8C). DOX + L-carnitine
and DOX + L-carnitine groups but was significantly higher in CoQ10 group showed moderate immune expression (Fig. 8D). Mean area%
and L-carnitine groups versus control (Table 3, Fig. 3A–C). of ␣–SMA immunopositive cells of DOX group showed a signifi-
cant increase in the mean area% of ␣–SMA immunoreactivity when
3.4. Serum levels of inflammatory cytokines compared with the control. Also, groups treated with CoQ10 and
L-carnitine showed a significant improvement as compared with
Serum levels of IL-1 beta, TNF-␣ and leptin were significantly the DOX group (Table 7, Fig. 11).
higher in DOX group than control, CoQ10, L-carnitine, DOX + CoQ10
and DOX + L-carnitine groups and in DOX + CoQ10 and DOX and
3.6.2. Immunohistochemical results for vimentin
L-carnitine groups versus control. Serum level of LDH was sig-
Immunohistochemical Results for vimentin of the control
nificantly higher in DOX group than control, CoQ10, L-carnitine,
group revealed faint immune expression in the myocardium
DOX + CoQ10 and DOX + L-carnitine groups and in DOX + CoQ10 and
(Fig. 9A). DOX group showed wide positive immunoreactivity in
DOX + L-carnitine groups versus control (Table 4, Fig. 4A–D).
the myofibroblasts (Fig. 9B). DOX + CoQ10 group revealed mini-
mal immune expression (Fig. 9C). DOX + L-carnitine group showed
3.5. Serum levels of cardiac parameters
slight immune expression (Fig. 9D). Mean area% of vimentin of DOX
group showed a significant increase in the mean area% of vimentin
Serum levels of Cardiotrophin-1, Cardiac specific-creatine
immunoreactivity when compared with the control. In addition,
kinase, and Troponin-I were significantly lower in control, CoQ10,
groups treated with CoQ10 and L-carnitine showed a significant
L-carnitine, DOX +CoQ10 and DOX + L-carnitine than DOX group
improvement as compared with the DOX group (Table 7, Fig. 11).
while regarding Troponin-T no significant difference between DOX
group and DOX + L-carnitine group. Serum levels of Cardiotrophin-
1, Cardiac specific-creatine kinase, Troponin-I and Troponin-T in 3.6.3. Immunohistochemical Results for vimentin
DOX + CoQ10 and DOX + L-carnitine groups versus control showed Immunohistochemical Results for eNOS of the control group
a significant increase (Table 5, Fig. 5A–D). revealed faint or no immune expression (Fig. 10A). DOX group
showed strong immune expression (Fig. 10B, C). DOX + CoQ10
3.6. Histological studies group revealed minimal immune expression (Fig. 10D). DOX + L-
carnitine group showed moderate immune expression (Fig. 10E).
Control group. H&E stained sections of control heart tissues Mean area% of eNOS of DOX group showed a significant increase
showed normal cardiac myocytes with their centrally placed nuclei in the mean area% of eNOS immunoreactivity when compared
(Fig. 6A). Sections stained with Masson’s trichrome stain showed with the control. Also, groups treated with CoQ10 and L-carnitine
scanty green stained connective tissue surrounding the muscle showed a significant improvement as compared with the DOX
fibers (Fig. 7A). Groups treated with only L-carnitine and CoQ10 group (Table 7, Fig. 11).
revealed no significant differences between these groups and con-
trol as regard H&E and Masson’s trichrome stains. 3.6.4. Immunohistochemical Results for eNOS
DOX group. H&E showed necrosis and swollen of the car- Ultrastructural results. Control group showed normal architec-
diomyocytes with an increase in the diameter. Pyknotic nuclei, ture of the cardiomyocytes (Figs. 12 and 13A). DOX group revealed
mononuclear cellular infiltration and dilated blood vessels were degeneration and fragmentation of myofibrils and loss of light
observed (Fig. 6B). Masson’s trichrome stained sections showed bands with broadening and interruption of Z lines. The mitochon-
intense increase in collagen fibers of the surrounding endomysium dria appeared electron dense with a moth eaten appearance among
(Fig. 7B). These results were confirmed by morphometric and sta- the muscle fibers (Figs. 12 and 13B). DOX + CoQ10 group revealed
tistical study. Cardiomyocytes diameter of DOX group showed a well-organized myofibrils and the mitochondria looked normal
significant decrease in the mean cardiomyocyte diameter (P < 0.01) with tightly packed cristae (Figs. 12 and 13C). DOX + L-carnitine
when compared with the control. Area percentage of collagen (Mas- showed an improvement of the myofibrils organization (Figs. 12
son’s trichrome stain) of DOX group showed a significant increase and 13D).
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 417

Fig. 3. A: Comparison of measured oxidative stress parameters in heart tissue homogenate in different studied groups for Malondialdehyde (nM/g). B: Comparison of
measured oxidative stress parameters in heart tissue homogenate in different studied groups for reduced glutathione (␮M/g). C: Comparison of measured oxidative stress
parameters in heart tissue homogenate in different studied groups for Nitric oxide (␮M/g).
Values are means ± SD. ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX. nM/g: nanomolar/gram.
418 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

Fig. 4. A: Comparison of measured inflammatory parameters in different studied groups [Interleukin-1␤ (pg/ml)]. B: Comparison of measured inflammatory parameters in
different studied groups [Tumor necrosis factor-␣ (pg/ml)]. C: Comparison of measured inflammatory parameters in different studied groups [Leptin (pg/ml)]. D: Comparison
of measured inflammatory parameters in different studied groups [Lactate dehydrogenase (U/ml)].
Values are means ± SD. ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX.
pg/ml: Picograms per Millilitre.

Fig. 5. A: Comparison of measured heart parameters in different studied groups [Cardiotrophin-1 (pg/ml)]. B: Comparison of measured heart parameters in different studied
groups [Cardiac specific-creatine kinase (ng/ml)]. C: Comparison of measured heart parameters in different studied groups [Troponin-I (ng/ml)]. D: Comparison of measured
heart parameters in different studied groups [Troponin-T (ng/ml)].
Values are means ± SD. ANOVA followed by Bonferroni’s post hoc test.
1
P: compared to control. 2 P: compared to DOX. pg/ml: picograms per Millilitre.
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 419

Fig. 6. (A). Photomicrograph of control showed cardiac myocytes with centrally placed nuclei (arrow). (B). DOX treated group showed cardiac myocytes showing massive
necrosis with focal marked fragmentation and nuclear changes in the form of pyknosis (p), karyolysis (k) and chromatin margination (c). (C). CoQ10 and DOX showed nearly
normal architecture of the cardiac myocytes with focal necrosis. (D): L-carnitine and DOX showed apparently regular architecture of the cardiac myocytes with focal necrosis
(H&E, Scale bar 20 ␮m).

Fig. 7. (A). Photomicrograph of control showed scanty green colored collagen fibers (arrow) between the cardiomyocytes. (B). DOX treated group showed an intense of
greenish colored collagen fibers (arrow) between swollen cardiomyocytes. (C). CoQ10 and DOX showed mild reaction. (D): L-carnitine and DOX showed mild reaction
(Masson’s trichrome, Scale bar 20 ␮m). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.).

4. Discussion with a large number of mitochondria, which include cardiac and


liver cells. New approaches are therefore needed to decrease the
The cardiotoxicity of doxorubicin (DOX) limits its use in cancer oxidative side effects of doxorubicin (Chao et al., 2011). DOX pos-
chemotherapy; the cells that are most affected by DOX are those sesses cardiotoxic properties that affect both the conductivity and
420 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

Fig. 8. (A). Photomicrograph of control showed faint immunoreactivity in the myocardium (arrow). (B). DOX treated group showed wide positive immunoreactivity in the
myofibroblasts, which are attached together by their processes (arrow). (C). CoQ10 and DOX showed minimal immunoreactivity. (D). L-carnitine and DOX showed slight
immunoreactivity (arrow) (arrow) (␣-SMA. Scale bar 20 ␮m).

Fig. 9. (A) Photomicrograph of control showed minimal immune reaction in the blood capillaries wall (curved arrows) and interstitial cells (arrow). With an immune negative
cardiac muscle fibers (arrowhead). (B). DOX treated group showed strong immune reaction in endomysium and perimysium connective tissues (star), in the blood capillaries
wall (curved arrows), and interstitial cells (arrow). (C). CoQ10 and DOX showed mild immune reaction in the endomysium and perimysium (star), in the blood capillaries wall
(curved arrows) and interstitial cells (arrow). With an immune negative reaction in cardiac muscle fibers (arrowhead). (D): L-carnitine and DOX showed moderate immune
reaction (Vimentin. Scale bar 20 ␮m).
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 421

Fig. 10. (A). Photomicrograph of control showed faint or no immune reaction cardiac muscle fibers. (B, C). DOX treated group showed strong immune reaction (arrowhead)
in cardiac muscle fibers and endothelial cells of blood capillaries. (C). CoQ10 and DOX showed minimal immune reaction (arrowhead). (E): L-carnitine and DOX showed
moderate immune reaction (arrowhead) (eNOS. Scale bar 5 ␮m).

Table 6
Effect of CoQ10and L-carnitine on the heart tissues treated with DOX.

Groups Control DOX DOX + CoQ10 DOX + L-Carnitine

Necrosis 0* +4 +1 +1
Degeneration and vacuolations 0 +4 +1 +1
Edema 0 +3 +1 +1
Inflammatory cell infiltrate 0** +3 +1 +1

A single animal may be represented more than once in the listing of individual histological changes. *Massive necrosis/changes limited to single cardiomyocytes. **Massive
inflammatory infiltration/disseminate mononuclear cells between cadiomyocytes.

Table 7
Cardiomyocyte diameter, area percentage of collagen, vimentin, ␣-SMA and eNOS Immunohistochemistry of the different groups.

Groups Control N = 12 CoQ10 N = 12 L-Carnitine DOX N = 9 DOX + CoQ10 DOX + L-


N = 12 N = 11 Carnitine
N = 11

Cardiomyocyte 14.24 ± 2.71 15.35 ± 3.07 13.84 ± 2.3 10.01 ± 0.97 16.92 ± 1.082 16.25 ± 1.071
diameter (␮m) 1
P ≤ 0.001 1
P ≤ 0.01 1
P ≤ 0.05
2
P ≤ 0.001 2
P ≤ 0.001
Area 4.57 ± 1.52 3.82 ± 1.27 5.32 ± 1.77 23.17 ± 3.61 9.01 ± 0.70 11.72 ± 1.41
percentage of 1
P ≤ 0.001 1
P ≤ 0.001 1
P ≤ 0.001
collagen (␮m2 ) 2
P ≤ 0.001 2
P ≤ 0.001
Area 0.16 ± 0.05 0.91 ± 0.3 0.66 ± 0.22 12.67 ± 1.97 5.12 ± 1.36 6.32 ± 1.14
percentage of 1
P ≤ 0.001 1
P ≤ 0.001 1
P ≤ 0.001
␣-SMA 2
P ≤ 0.001 2
P ≤ 0.001
Area 0.37 ± 0.12 1.12 ± 0.37 0.87 ± 0.29 31.21 ± 4.45 10.02 ± 2.13 9.97 ± 1.08
percentage of 1
P ≤ 0.001 1
P ≤ 0.001 1
P ≤ 0.001
vimentin 2
P ≤ 0.001 2
P ≤ 0.001
Area 1.23 ± 0.36 1.48 ± 0.49 0.98 ± 0.33 6.45 ± 2.15 1.97 ± 0.32 1.64 ± 0.54
percentage of 1
P ≤ 0.001 1
P = NS 1
P = NS
eNOS 2
P ≤ 0.001 2
P ≤ 0.001

Values are means ± SD (Control n = 12& DOX = 9 & treated = 11). ANOVA followed by Bonferroni’s post hoc test.
1P: compared to control. 2P: compared to DOX.

rhythmicity of cardiac muscle, as shown by its effect on heart rate include reflected arrhythmias, conduction abnormalities, and the
in addition to the associated elongation of the corrected QT interval attenuation of left ventricular function (Mantawy et al., 2014).
(QTc), ST elevation, and shortening of the T amplitude (Mantawy This study illustrated that DOX induces oxidative damage and
et al., 2014). nitrosative stress in the cardiac muscle. These results align with
The results of this study showed significant abnormalities that those from other studies (Goyal et al., 2016; Jagetia and Venkatesh,
affected ECG in the DOX group in agreement with previous studies 2015). These results could be explained by the ability of DOX to gen-
(Goyal et al., 2016; Jagetia and Venkatesh, 2015). These changes erate ROS, which results in lipid peroxidation of both the cellular
and mitochondrial membrane, ending in the injury of myocardio-
422 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

Fig. 11. Cardiomyocyte diameter and Area% of collagen,␣-SMA, vimentin and eNOS. The mean is given in columns, and error bars represent the standard deviation (SD).

cytes (Sahu et al., 2016). Moreover, DOX creates free radicals that mitochondrial membrane’s permeability; and promoting the elim-
cause destruction in DNA and proteins and interfere with the struc- ination of free radicals (Chao et al., 2011).
ture of the cytoskeleton (Ikeda et al., 2010). Oxidative stress could CoQ10 plays an important role in energy metabolism and is
injure mitochondrial cell membranes, increasing the membrane’s part of the electron transport chain that is responsible for ATP
permeability and making it vulnerable to rupture (Viswanatha synthesis. Moreover, it is one of the most efficient endogenous
Swamy et al., 2011). antioxidants and protects cellular DNA, lipids, and protein from
L-carnitine produces its antioxidant effects through different oxidative damage (Garrido-Maraver et al., 2014). CoQ10 protects
mechanisms, including the scavenging of free radical activity either myocardial integrity through many mechanisms, including preser-
directly or by inhibition of its production, maintaining the effi- vation of myocardial ATP levels and powerful antioxidant effects.
ciency of the mitochondrial electron transport chain, stimulating CoQ10 may exert its effects directly by acting as a scavenger of free
the activation of antioxidant enzymes, and synthesis of antioxidant radicals or through the regeneration of tocopherol and ascorbic acid
molecules like reduced glutathione (Surai, 2015). from their oxidized state (Chen et al., 2017).
L-carnitine protects myocardial integrity by controlling the The results of this study confirm that DOX toxicity has specific
intra-mitochondrial percentage of acyl-CoA/CoA, resulting in elim- inflammatory effects, as evidenced by the significant increase in
ination of toxic compounds; maintaining the integrity of the
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 423

Fig. 12. (A). Electron micrograph of control showed a cardiomyocyte with an elongated nucleus (N) with an evenly dispersed chromatin pattern and regular nuclear membrane
(↑). Numerous mitochondria (M) appear with apparent cristae between the longitudinally arranged myofibrils. That exhibit a normal cross-striated pattern Z lines (Z). (B).
DOX treated group showed disorganized, fragmented, degenerated myofibrils with loss of cross striations (↑). Distorted mitochondria (M) with dense matrix, unapparent
cristea, with different shapes and sizes irregularly arranged between the myofibrils and wide intercellular spaces (star) in the sarcoplasm of the cardiac myocytes. (C). CoQ10
and DOX showed regularly arranged myofilaments between successive Z lines (Z) in the sarcomeres. Mitochondria (M) arranged in rows between the myofibrils. The nucleus
(N) of a cardiac muscle fiber with slightly irregular nuclear membrane (↑). (D): L-carnitine and DOX showed mitochondria (M) appear distorted, with different shapes and
sizes around the nucleus (N) and between the myofibrils. Nuclear membrane indentations is observed (↑). Note the wide intercellular space (*) between adjacent muscle
fibers. (Scale bar 2 ␮m).

inflammatory cytokines. These results are in agreement with other cardiotoxicity have been measured, including cardiac troponin
studies (Elsherbiny et al., 2016; Sun et al., 2016). I (cTnL), T (cTnT), and cardiotrophin-1. All of these parameters
These results might be explained by the fact that ROS produced showed significant elevation in the group treated with DOX (Atas
by DOX can initiate inflammatory responses, mainly via NF-␬B, et al., 2015). These results are concordant with those from other
which results in the release of cytokines such as tumor necrosis studies (Atas et al., 2015; Bertinchant et al., 2003; Reagan et al.,
factor-alpha [TNF-␣] and interleukin 1 beta [IL-1 ␤] (Sun et al., 2013) showing an increase in the levels of cTnI and cTnT, confirm-
2016). ing that these are sensitive and specific markers for cardiac injury
Leptin is considered one of acute response markers in oxida- that may be elevated in the blood of patients treated with DOX
tive stress; it is involved in the prediction of coronary heart disease before cardiac damage is evident. Therefore, these markers can
due to the known relation between C-reactive protein and leptin be used for the prediction of future left ventricular dysfunction.
(Ahmed et al., 2005). In addition, this study showed that DOX led They are used in early detection of necrosis, before CK-MB levels
to significant myocardial damage, as evidenced by increased serum significantly increase in the heart (Atas et al., 2015).
levels of both CK-MB and LDH. These results, in accordance with In the current study, the DOX group showed visible conges-
those of other studies (El-Agamy et al., 2016; Sun et al., 2016), can tion in between cardiomyocytes, a finding that coincided with
be explained by the increase in oxidative stress leading to lipid per- other findings that noted the presence of marked blood cells
oxidation and disruption of the cell membranes of myocardiocytes, in the peri-capillary space (Hadi et al., 2012). The vacuoles are
along with the release of biochemical markers in the serum and ascribed to the expansion of cytoplasmic membranous compo-
plasma. CK-MB is one of the most important biochemical diagnostic nents due to redistribution of the intra-cellular electrolytes and
markers for myocardial damage (El-Agamy et al., 2016). water (Balli et al., 2004). The diameter of cardiomyocytes was
Treatment with CoQ10 and L-carnitine resulted in a sig- increased, in agreement with other reports in which myocytic
nificant decrease of these enzymes that is attributable to a diameter increased, with the presence of hyperchromatic nuclei,
decrease in oxidative stress and stabilization of cardiomyocyte disorganization of myofibrils, and loss of cross-striation of car-
cell membranes. Furthermore, specific cardiac markers for acute diac myocytes (Rashikh et al., 2011). With CoQ10 or L-carnitine,
424 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

Fig. 13. (A). Electron micrograph of control showed a cardiomyocyte contains strands of myofibrils formed of light bands (I), Z lines (Z), dark bands (A), H zone (H), sarcomere
(S) and mitochondrial rows (M). (B). DOX treated group showed destruction, fragmentation and lysis of myofibrils (arrows) with absence of light bands and broadening of Z
lines (Z). Moth-eaten appearance of degenerated mitochondria (ME) with variable sizes were seen among the myofibrils. Note lipofuscin pigment (star). (C). CoQ10 and DOX
showed myofibrils with preserved cross-banding pattern, intercalated disc (IC) and euchromatic nucleus (N). The mitochondria (m) looks normal with tightly packed cristae
and relative increase in number. (D): L-carnitine and DOX showed well-organized myofibrils with few interrupted Z lines (arrow. Preserved healthy mitochondria (M) and
Dilated SER (SER). (Scale bar 500 nm).

the histopathological findings were improved such that they were Myofibroblasts also generate angiotensin II and fibrogenic growth
consistent with other studies that found decreases in myofibril dis- factors, which play a crucial role in fibrosis and collagen type I
organization, exudation, and inflammatory cell infiltration in the synthesis (Weber et al., 2013). With CoQ10 or L-carnitine, there
myocardium (Kwong et al., 2002). Analysis of the ultrastructure is a decrease in the transformation of fibroblasts to myofibroblasts,
morphology images showed peripheral chromatin condensation, which are a source of collagen, thus restraining cardiac fibrosis.
deformity and fragmentation of the nuclei, and apoptosis (Zhang In the current work, an increase in vimentin area percentage
et al., 2012), supporting the hypothesis that apoptosis is one of expressed in the arterial walls was observed in the DOX group;
the mechanisms of DOX-cardiotoxicity, DOX-induced lipid per- similar results were revealed in dilated cardiomyopathy, where
oxidation, reactive oxygen species (ROS) production, disturbed vimentin immunoreaction was increased in the interstitial tis-
mitochondrial metabolism, and direct cardiotoxicity (Oktem et al., sue cells (Di et al., 2000). The increased vimentin was linked
2012). to an increase of collagen and fibrosis (Schaper et al., 1991).
The results were in agreement with those of previous Scientists revealed an adverse connection between myocardial
researchers who observed that myocardial stress increases the vimentin overexpression and the sliding rate of actin myosin. It
mean number of ␣-SMA positive myofibroblasts. This was was proposed that disarrangement of cytoskeleton proteins occurs
attributed to myofibroblasts, which are considered the key cells with participation of vimentin in the modulations of coupling of
responsible for extracellular matrix and collagen deposition in myocytes to the extracellular matrix, myocyte functions, and intra-
myocardial fibrosis (Naugle et al., 2006). Other researchers have cellular signaling during cardiac failure and hypertrophy (Rastogi
observed a rise in fibronectin and in collagen types I and III, ascrib- et al., 2008). Moreover, investigators noticed the proliferation of
ing this to collagen synthesis related to ␤-adrenergic receptor T-tubules linked to vimentin overexpression in cardiomyopathy.
activation in fibroblasts (Yin et al., 2009). The contractile fibers of This can lead to recovery of an inappropriate cardiac function by
myofibroblasts contain ␣-SMA and are linked to exaggerated extra- substituting for the contractile elements (Di et al., 2000). Fibrosis
cellular matrix accumulation in pathological disorders (Ma et al., is responsible for an increased stiffness and decrease of ventricular
2014). In cardiac disease, cardiomyocytes are wasted due to necro- compliance (Heling et al., 2000).
sis, and myofibroblasts are stimulated to launch restorative fibrosis.
H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426 425

The interaction between DOX and NOS is a complex. DOX prostacyclin-mediated pathway in neonatal rat cardiomyocytes. Int. J. Cardiol.
converts eNOS from a nitric oxide donor to a superoxide gen- 146, 145–152.
Chatterjee, K., Zhang, J., Honbo, N., Karliner, J.S., 2010. Doxorubicin
erator (Octavia et al., 2012). DOX-induced hydrogen peroxide cardiomyopathy. Cardiology 115, 155–162.
creation (H2 O2 ) is responsible for apoptotic cell death and DOX- Chen, P.-Y., Hou, C.-W., Shibu, M.A., Day, C.H., Pai, P., Liu, Z.-R., Lin, T.-Y.,
toxicity (Octavia et al., 2012). In turn, H2 O2 promotes endothelial Viswanadha, V.P., Kuo, C.-H., Huang, C.-Y., 2017. Protective effect of Co-enzyme
Q10 On doxorubicin-induced cardiomyopathy of rat hearts. Environ. Toxicol.
nitric oxide synthase (eNOS) transcription in endothelial cells and 32, 679–689.
cardiomyocytes (Kalyanaraman et al., 2002). Up-regulated eNOS Crespo, M.J., Zalacain, J., Dunbar, D.C., Cruz, N., Arocho, L., 2008. Cardiac oxidative
expression can play a key role in DOX-cardiac dysfunction by stress is elevated at the onset of dilated cardiomyopathy in
streptozotocin-diabetic rats. J. Cardiovasc. Pharmacol. Ther. 13, 64–71.
affecting ROS-mediated apoptosis of endothelial cells (Neilan et al.,
Di, S., Marotta, M., Salvatore, G., Cudemo, G., Cuda, G., De Vivo, F., Di, B., Ciaramella,
2007). Genetic disruption of eNOS transcription protects against F., Caputo, G., de Divitiis, O., 2000. Changes in myocardial cytoskeletal
DOX-induced cardiotoxicity and mortality, while overexpression intermediate filaments and myocyte contractile dysfunction in dilated
cardiomyopathy: an in vivo study in humans. Heart 84, 659–667.
exaggerates the toxic effects of DOX (Šimůnek et al., 2009). Studies
Dong, Q., Chen, L., Lu, Q., Sharma, S., Li, L., Morimoto, S., Wang, G., 2014. Quercetin
about endothelial dysfunction have demonstrated a considerable attenuates doxorubicin cardiotoxicity by modulating Bmi-1 expression. Br. J.
attenuation of endothelial vasodilation after DOX administration, Pharmacol. 171, 4440–4454.
suggesting dysfunctional eNOS activity (Olukman et al., 2009). Dudka, J., Gieroba, R., Korga, A., Burdan, F., Matysiak, W., Jodlowska-Jedrych, B.,
Mandziuk, S., Korobowicz, E., Murias, M., 2012. Different effects of resveratrol
The current results provide proof that CoQ10 and L-carnitine on dose-related Doxorubicin-induced heart and liver toxicity. Evid. Based
attenuates DOX-induced generation of free radicals. Also, prevent Complement Alternat. Med. 2012, 606183.
eNOS uncoupling by reducing superoxide formation, increasing El-Agamy, D.S., Abo-Haded, H.M., Elkablawy, M.A., 2016. Cardioprotective effects
of sitagliptin against doxorubicin-induced cardiotoxicity in rats. Exp. Biol. Med.
NO bioavailability, and inhibiting upregulation of the activity and (Maywood) 241, 1577–1587.
expression of the vascular NAD (P) H oxidase (Chatterjee et al., Elbaky, N.A., Ali, A.A., Ahmed, R.A., 2010. Cardioprotective effect of simvastatin on
2010). doxorubicininduced oxidative cardiotoxicity in rats. J. Basic Appl. Sci. 6, 29–38.
Elsherbiny, N.M., Salama, M.F., Said, E., El-Sherbiny, M., Al-Gayyar, M.M., 2016.
Crocin protects against doxorubicin-induced myocardial toxicity in rats
5. Conclusion through down-regulation of inflammatory and apoptic pathways. Chem. Biol.
Interact. 247, 39–48.
Gala, A.A.A., 2013. Protective effect of Zingiber officinale (ginger) on doxorubicin
Supplementation with CoQ10 or L-carnitine defends the induced oxidative cardiotoxicity in rats. Life Sci. J. 10, 2924–2934.
myocardium through their antioxidant activity, as was proven by Garrido-Maraver, J., Cordero, M.D., Oropesa-Ávila, M., Fernández Vega, A., De La
Mata, M., Delgado Pavón, A., De Miguel, M., Pérez Calero, C., Villanueva Paz, M.,
the improvement of different biochemical markers and oxidative Cotán, D., 2014. Coenzyme q10 therapy. Mol. Syndromol. 5, 187–197.
status and the restoration of the myocardium’s structural integrity Gianni, L., Salvatorelli, E., Minotti, G., 2007. Anthracycline cardiotoxicity in breast
and function. cancer patients: synergism with trastuzumab and taxanes. Cardiovasc. Toxicol.
7, 67–71.
Goyal, S.N., Mahajan, U.B., Chandrayan, G., Kumawat, V.S., Kamble, S., Patil, P.,
Disclosure of interest Agrawal, Y.O., Patil, C.R., Ojha, S., 2016. Protective effect of oleanolic acid on
oxidative injury and cellular abnormalities in doxorubicin induced cardiac
toxicity in rats. Am. J. Transl. Res. 8, 60–69.
The authors declare that they have no conflicts of interest. Guan, Y., Zheng, X., Yang, Z., Li, S., 2009. [Protective effects of L-carnitine upon
testicular ischemia-reperfusion damage in rats]. Zhonghua yi xue za zhi 89,
1858–1861.
References Hadi, N., Yousif, N.G., Al-Amran, F.G., Huntei, N.K., Mohammad, B.I., Ali, S.J., 2012.
Vitamin E and telmisartan attenuates doxorubicin induced cardiac injury in rat
İzgüt-Uysal, V.N., Ağaç, A., Derin, N., 2003. Effect of L-Carnitine on through down regulation of inflammatory response. BMC Cardiovasc. Disord.
carrageenan-induced inflammation in aged rats. Gerontology 49, 287–292. 12, 1.
Šimůnek, T., Štěrba, M., Popelová, O., Adamcová, M., Hrdina, R., Geršl, V., 2009. Hajrasouliha, A.R., Tavakoli, S., Jabehdar-Maralani, P., Shafaroodi, H., Borhani, A.A.,
Anthracycline-induced cardiotoxicity: overview of studies examining the roles Houshmand, G., Sadeghipour, H., Dehghani, M., Dehpour, A.R., 2004. Resistance
of oxidative stress and free cellular iron. Pharmacol. Rep. 61, 154–171. of cholestatic rats against epinephrine-induced arrhythmia: the role of nitric
Agarwal, A., Said, T.M., 2004. Carnitines and male infertility. Reprod Biomed Online oxide and endogenous opioids. Eur. J. Pharmacol. 499, 307–313.
8, 376–384. Heling, A., Zimmermann, R., Kostin, S., Maeno, Y., Hein, S., Devaux, B., Bauer, E.,
Ahmed, H.H., Mannaa, F., Elmegeed, G.A., Doss, S.H., 2005. Cardioprotective activity Klovekorn, W.P., Schlepper, M., Schaper, W., Schaper, J., 2000. Increased
of melatonin and its novel synthesized derivatives on doxorubicin-induced expression of cytoskeletal, linkage, and extracellular proteins in failing human
cardiotoxicity. Bioorg. Med. Chem. 13, 1847–1857. myocardium. Circ. Res. 86, 846–853.
Aliev, G., Liu, J., Shenk, J.C., Fischbach, K., Pacheco, G.J., Chen, S.G., Obrenovich, M.E., Ikeda, Y., Aihara, K., Akaike, M., Sato, T., Ishikawa, K., Ise, T., Yagi, S., Iwase, T., Ueda,
Ward, W.F., Richardson, A.G., Smith, M.A., 2009. Neuronal mitochondrial Y., Yoshida, S., Azuma, H., Walsh, K., Tamaki, T., Kato, S., Matsumoto, T., 2010.
amelioration by feeding acetyl-L-carnitine and lipoic acid to aged rats. J. Cell. Androgen receptor counteracts Doxorubicin-induced cardiotoxicity in male
Mol. Med. 13, 320–333. mice. Mol. Endocrinol. 24, 1338–1348.
Alpsoy, S., Aktas, C., Uygur, R., Topcu, B., Kanter, M., Erboga, M., Karakaya, O., Jagetia, G., Venkatesh, P., 2015. An indigenous plant bael (Aegle marmelos (L.)
Gedikbasi, A., 2013. Antioxidant and anti-apoptotic effects of onion (Allium correa) extract protects against the doxorubicin-Induced cardiotoxicity in
cepa) extract on doxorubicin-induced cardiotoxicity in rats. J. Appl. Toxicol. 33, mice. Biochem. Physiol. 4, 2.
202–208. Jones, S.P., Greer, J.J., Kakkar, A.K., Ware, P.D., Turnage, R.H., Hicks, M., van Haperen,
Arcamone, F., Cassinelli, G., Fantini, G., Grein, A., Orezzi, P., Pol, C., Spalla, C., 2000. R., de Crom, R., Kawashima, S., Yokoyama, M., 2004. Endothelial nitric oxide
Adriamycin, 14-hydroxydaunomycin, a new antitumor antibiotic from S. synthase overexpression attenuates myocardial reperfusion injury. Am. J.
peucetius var. caesius. reprinted from biotechnology and bioengineering, vol. Physiol.-Heart Circulatory Physiol. 286, H276–H282.
XI, issue 6, pages 1101–1110 (1969). Biotechnol. Bioeng. 67, 704–713. Kalyanaraman, B., Joseph, J., Kalivendi, S., Wang, S., Konorev, E., Kotamraju, S.,
Atas, E., Kismet, E., Kesik, V., Karaoglu, B., Aydemir, G., Korkmazer, N., Demirkaya, 2002. Doxorubicin-induced apoptosis: implications in cardiotoxicity. Mol. Cell.
E., Karslioglu, Y., Yurttutan, N., Unay, B., Koseoglu, V., Gokcay, E., 2015. Cardiac Biochem. 234–235, 119–124.
troponin-I, brain natriuretic peptide and endothelin-1 levels in a rat model of Kwong, L.K., Kamzalov, S., Rebrin, I., Bayne, A.C., Jana, C.K., Morris, P., Forster, M.J.,
doxorubicin-induced cardiac injury. J. Cancer Res. Ther. 11, 882–886. Sohal, R.S., 2002. Effects of coenzyme Q(10) administration on its tissue
Balli, E., Mete, U.O., Tuli, A., Tap, O., Kaya, M., 2004. Effect of melatonin on the concentrations, mitochondrial oxidant generation, and oxidative stress in the
cardiotoxicity of doxorubicin. Histol. Histopathol. 19, 1101–1108. rat. Free Radic. Biol. Med. 33, 627–638.
Bertinchant, J.P., Polge, A., Juan, J.M., Oliva-Lauraire, M.C., Giuliani, I., Ma, Y., de Castro Brás, L.E., Toba, H., Iyer, R.P., Hall, M.E., Winniford, M.D., Lange,
Marty-Double, C., Burdy, J.Y., Fabbro-Peray, P., Laprade, M., Bali, J.P., Granier, C., R.A., Tyagi, S.C., Lindsey, M.L., 2014. Myofibroblasts and the extracellular
de la Coussaye, J.E., Dauzat, M., 2003. Evaluation of cardiac troponin I and T matrix network in post-myocardial infarction cardiac remodeling. Pflugers
levels as markers of myocardial damage in doxorubicin-induced Arch. 466, 1113–1127.
cardiomyopathy rats, and their relationship with echocardiographic and Mandziuk, S., Gieroba, R., Korga, A., Matysiak, W., Jodlowska-Jedrych, B., Burdan, F.,
histological findings. Clin. Chim. Acta 329, 39–51. Poleszak, E., Kowalczyk, M., Grzycka-Kowalczyk, L., Korobowicz, E., Jozefczyk,
Bhagavan, H.N., Chopra, R.K., 2006. Coenzyme Q10: absorption, tissue uptake, A., Dudka, J., 2015. The differential effects of green tea on dose-dependent
metabolism and pharmacokinetics. Free Radic. Res. 40, 445–453. doxorubicin toxicity. Food Nutr. Res. 59, 29754.
Chao, H.-H., Liu, J.-C., Hong, H.-J., Lin J.-w. Chen, C.-H., Cheng, T.-H., 2011.
L-carnitine reduces doxorubicin-induced apoptosis through a
426 H.N. Mustafa et al. / Tissue and Cell 49 (2017) 410–426

Mantawy, E.M., El-Bakly, W.M., Esmat, A., Badr, A.M., El-Demerdash, E., 2014. Pradegan, N., Vida, V.L., Geva, T., Stellin, G., White, M.T., Sanders, S.P., Padera, R.F.,
Chrysin alleviates acute doxorubicin cardiotoxicity in rats via suppression of 2016. Myocardial histopathology in late-repaired and unrepaired adults with
oxidative stress, inflammation and apoptosis. Eur. J. Pharmacol. 728, 107–118. tetralogy of Fallot. Cardiovasc. Pathol. 25, 225–231.
Mescka, C.P., Rosa, A.P., Schirmbeck, G., da Rosa, T.H., Catarino, F., de Souza, L.O., Rashikh, A., Abul Kalam, N., Akhtar, M., Mahmood, D., Pillai, K.K., Ahmad, S.J., 2011.
Guerreiro, G., Sitta, A., Vargas, C.R., Dutra-Filho, C.S., 2016. L-carnitine prevents Protective effects of aliskiren in doxorubicin-induced acute cardiomyopathy in
oxidative stress in the brains of rats subjected to a chemically induced chronic rats. Hum. Exp. Toxicol. 30, 102–109.
model of MSUD. Mol. Neurobiol. 53, 6007–6017. Rastogi, S., Mishra, S., Zaca, V., Mika, Y., Rousso, B., Sabbah, H.N., 2008. Effects of
Miranda, K.M., Espey, M.G., Yamada, K., Krishna, M., Ludwick, N., Kim, S., chronic therapy with cardiac contractility modulation electrical signals on
Jourd’heuil, D., Grisham, M.B., Feelisch, M., Fukuto, J.M., 2001. Unique oxidative cytoskeletal proteins and matrix metalloproteinases in dogs with heart failure.
mechanisms for the reactive nitrogen oxide species, nitroxyl anion. J. Biol. Cardiology 110, 230–237.
Chem. 276, 1720–1727. Reagan, W.J., York, M., Berridge, B., Schultze, E., Walker, D., Pettit, S., 2013.
Mitry, M.A., Edwards, J.G., 2016. Doxorubicin induced heart failure: phenotype and Comparison of cardiac troponin I and T, including the evaluation of an
molecular mechanisms. Int. J. Cardiol. Heart Vasc. 10, 17–24. ultrasensitive assay, as indicators of doxorubicin-induced cardiotoxicity.
Moshage, H., Kok, B., Huizenga, J.R., Jansen, P.L., 1995. Nitrite and nitrate Toxicol. Pathol. 41, 1146–1158.
determinations in plasma: a critical evaluation. Clin. Chem. 41, 892–896. Sahu, B.D., Kumar, J.M., Kuncha, M., Borkar, R.M., Srinivas, R., Sistla, R., 2016.
Mustafa, H.N., Hussein, A.M., 2016. Does allicin combined with vitamin B-complex Baicalein alleviates doxorubicin-induced cardiotoxicity via suppression of
have superior potentials than alpha-tocopherol alone in ameliorating lead myocardial oxidative stress and apoptosis in mice. Life Sci. 144, 8–18.
acetate-induced Purkinje cell alterations in rats? An immunohistochemical Schaper, J., Froede, R., Hein, S., Buck, A., Hashizume, H., Speiser, B., Friedl, A., Bleese,
and ultrastructural study. Folia Morphol (Warsz) 75 (1), 76–86. N., 1991. Impairment of the myocardial ultrastructure and changes of the
Mustafa, H.N., El Awdan, S.A., Hegazy, G.A., Jaleel, G.A.A., 2015. Prophylactic role of cytoskeleton in dilated cardiomyopathy. Circulation 83, 504–514.
coenzyme Q10 and Cynara scolymus L on doxorubicin-induced toxicity in rats: Sharov, V.G., Kostin, S., Todor, A., Schaper, J., Sabbah, H.N., 2005. Expression of
biochemical and immunohistochemical study. Indian J. Pharmacol. 47, cytoskeletal, linkage and extracellular proteins in failing dog myocardium.
649–656. Heart Fail. Rev. 10, 297–303.
Mustafa, H.N., 2015. Potential alleviation of chlorella vulgaris and zingiber Sun, Z., Yan, B., Yu, W.Y., Yao, X., Ma, X., Sheng, G., Ma, Q., 2016. Vitexin attenuates
officinale on lead-Induced testicular toxicity: an ultrastructural study. Folia acute doxorubicin cardiotoxicity in rats via the suppression of oxidative stress,
Biol. (Praha) 63, 269–278. inflammation and apoptosis and the activation of FOXO3a. Exp. Ther. Med. 12,
Mustafa, H.N., 2016. The role of curcumin in streptozotocin-induced hepatic 1879–1884.
damage and the trans-differentiation of hepatic stellate cells. Tissue Cell 48, Surai, P.F., 2015. Antioxidant action of carnitine: molecular mechanisms and
81–88. practical applications. EC Veterinary Sci. 2, 66–84.
Nascimento, A.R., Machado, M.V., Gomes, F., Vieira, A.B., Viswanatha Swamy, A.H., Wangikar, U., Koti, B.C., Thippeswamy, A.H., Ronad, P.M.,
Gonçalves-de-Albuquerque, C.F., Lessa, M.A., Bousquet, P., Tibiriçá, E., 2016. Manjula, D.V., 2011. Cardioprotective effect of ascorbic acid on
Central sympathetic modulation reverses microvascular alterations in a rat doxorubicin-induced myocardial toxicity in rats. Indian J. Pharmacol. 43,
model of high-Fat diet-Induced metabolic syndrome. Microcirculation 23, 507–511.
320–329. Weber, K.T., Sun, Y., Bhattacharya, S.K., Ahokas, R.A., Gerling, I.C., 2013.
Naugle, J.E., Olson, E.R., Zhang, X., Mase, S.E., Pilati, C.F., Maron, M.B., Folkesson, Myofibroblast-mediated mechanisms of pathological remodelling of the heart.
H.G., Horne, W.I., Doane, K.J., Meszaros, J.G., 2006. Type VI collagen induces Nat. Rev. Cardiol. 10, 15–26.
cardiac myofibroblast differentiation: implications for postinfarction Yin, W., Zhang, P., Huang, J.H., Zhang, Q.Y., Fan, R., Li, J., Zhou, J.J., Hu, Y.Z., Guo, H.T.,
remodeling. Am. J. Physiol.-Heart Circulatory Physiol. 290, H323–H330. Zhang, S.M., Wang, Y.M., Kaye, A.D., Gu, C.H., Liu, J.C., Cheng, L., Cui, Q., Yi, D.H.,
Neilan, T.G., Blake, S.L., Ichinose, F., Raher, M.J., Buys, E.S., Jassal, D.S., Furutani, E., Pei, J.M., 2009. Stimulation of kappa-opioid receptor reduces
Perez-Sanz, T.M., Graveline, A., Janssens, S.P., 2007. Disruption of nitric oxide isoprenaline-induced cardiac hypertrophy and fibrosis. Eur. J. Pharmacol. 607,
synthase 3 protects against the cardiac injury, dysfunction, and mortality 135–142.
induced by doxorubicin. Circulation 116, 506–514. Zhang, S., Liu, X., Bawa-Khalfe, T., Lu, L.S., Lyu, Y.L., Liu, L.F., Yeh, E.T., 2012.
Octavia, Y., Tocchetti, C.G., Gabrielson, K.L., Janssens, S., Crijns, H.J., Moens, A.L., Identification of the molecular basis of doxorubicin-induced cardiotoxicity.
2012. Doxorubicin-induced cardiomyopathy: from molecular mechanisms to Nat. Med. 18, 1639–1642.
therapeutic strategies. J. Mol. Cell. Cardiol. 52, 1213–1225. Zhang, Y.P., Song, C.Y., Yuan, Y., Eber, A., Rodriguez, Y., Levitt, R.C., Takacs, P., Yang,
Oktem, G., Uysal, A., Oral, O., Sezer, E.D., Olukman, M., Erol, A., Akgur, S.A., Bilir, A., Z., Goldberg, R., Candiotti, K.A., 2013. Diabetic neuropathic pain development
2012. Resveratrol attenuates doxorubicin-induced cellular damage by in type 2 diabetic mouse model and the prophylactic and therapeutic effects of
modulating nitric oxide and apoptosis. Exp. Toxicol. Pathol. 64, 471–479. coenzyme Q10. Neurobiol. Dis. 58, 169–178.
Olukman, M., Can, C., Erol, A., Öktem, G., Oral, O., 2009. Reversal of de Salvi Guimaraes, F., de Moraes, W.M., Bozi, L.H., Souza, P.R., Antonio, E.L.,
doxorubicin-induced vascular dysfunction by resveratrol in rat thoracic aorta: Bocalini, D.S., Tucci, P.J., Ribeiro, D.A., Brum, P.C., Medeiros, A., 2017.
is there a possible role of nitric oxide synthase inhibition? Anatol. J. Cardiol. 9, Dexamethasone-induced cardiac deterioration is associated with both calcium
260–266. handling abnormalities and calcineurin signaling pathway activation. Mol. Cell.
Potemski, P., Polakowski P. a. Wiktorowska-Owczarek, A.K., Owczarek, J., Biochem. 424, 87–98.
Pluzanska, A., Orszulak-Michalak, D., 2006. Amifostine improves hemodynamic
parameters in doxorubicin-pretreated rabbits. Pharmacol. Rep. 58, 966.
Pott, C., Steinritz, D., Bolck, B., Mehlhorn, U., Brixius, K., Schwinger, R.H., Bloch, W.,
2006. eNOS translocation but not eNOS phosphorylation is dependent on
intracellular Ca2+ in human atrial myocardium. Am. J. Physiol. Cell Physiol.
290, C1437–1445.

You might also like