ArticlePDF Available

Resveratrol solid lipid nanoparticles to trigger credible inhibition of doxorubicin cardiotoxicity

Taylor & Francis
International Journal of Nanomedicine
Authors:

Abstract and Figures

Background Doxorubicin (DOX), a broad-spectrum chemotherapy drug, is clinically employed to treat cancers especially for breast cancer and lung cancer. But its clinical applications are limited by the dose-dependent cardiac toxicity. Resveratrol (Res), a polyphenolic antitoxin, has been proved to be capable of improving the cardiomyocyte calcium cycling by up-regulating SIRT-1-mediated deacetylation to inhibit DOX-induced cardiotoxicity. Purpose The objective of this study was to develop a solid lipid nanoparticle (SLN) loaded with Res to trigger inhibition of DOX-induced cardiotoxicity. Methods Res-SLN was prepared by emulsification-diffusion method followed by sonication and optimized using central composite design/response surface method. The Res-SLN was further evaluated by dynamic light scattering, transmission electron microscopy for morphology and high performance liquid chromatography for drug loading and release profile. And the Res distribution in vivo was determined on rats while the effect of inhibit DOX-induced cardiotoxicity was investigated on mice. Results Res-SLN with homogeneous particle size of 271.13 nm was successfully formulated and optimized. The prepared Res-SLN showed stable under storage and sustained release profile, improving the poor solubility of Res. Heart rate, ejection fractions and fractional shortening of Res-SLN treating mice were found higher than those on mice with cardiac toxicity induced by single high-dose intraperitoneal injection of DOX. And the degree of myocardial ultrastructural lesions on mice was also observed. Conclusion Res-SLN has a certain therapeutic effect for protecting the myocardium and reducing DOX-induced cardiotoxicity in mice.
This content is subject to copyright. Terms and conditions apply.
ORIGINAL RESEARCH
Resveratrol solid lipid nanoparticles to trigger
credible inhibition of doxorubicin cardiotoxicity
This article was published in the following Dove Press journal:
International Journal of Nanomedicine
Lili Zhang
1
Kexin Zhu
1
Hairong Zeng
2
Jiaxin Zhang
1
Yiqiong Pu
3
Zhicheng Wang
4
To n g Z h a n g
3
Bing Wang
1,5
1
School of Pharmacy, Shanghai University
of Traditional Chinese Medicine,
Shanghai, Peoples Republic of China;
2
Department of Pharmacy, Putuo
Hospital, Shanghai University of
Traditional Chinese Medicine, Shanghai,
Peoples Republic of China;
3
Experiment
Center for Teaching and Learning,
Shanghai University of Traditional
Chinese Medicine, Shanghai, Peoples
Republic of China;
4
Department of
Laboratory Medicine, Huashan Hospital,
Shanghai Medical College, Fudan
University, Shanghai, Peoples Republic of
China;
5
Center for Pharmaceutics
Research, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences,
Shanghai, Peoples Republic of China
Background: Doxorubicin (DOX), a broad-spectrum chemotherapy drug, is clinically
employed to treat cancers especially for breast cancer and lung cancer. But its clinical
applications are limited by the dose-dependent cardiac toxicity. Resveratrol (Res),
a polyphenolic antitoxin, has been proved to be capable of improving the cardiomyocyte
calcium cycling by up-regulating SIRT-1-mediated deacetylation to inhibit DOX-induced
cardiotoxicity.
Purpose: The objective of this study was to develop a solid lipid nanoparticle (SLN) loaded
with Res to trigger inhibition of DOX-induced cardiotoxicity.
Methods: Res-SLN was prepared by emulsication-diffusion method followed by sonica-
tion and optimized using central composite design/response surface method. The Res-SLN
was further evaluated by dynamic light scattering, transmission electron microscopy for
morphology and high performance liquid chromatography for drug loading and release
prole. And the Res distribution in vivo was determined on rats while the effect of inhibit
DOX-induced cardiotoxicity was investigated on mice.
Results: Res-SLN with homogeneous particle size of 271.13 nm was successfully formu-
lated and optimized. The prepared Res-SLN showed stable under storage and sustained
release prole, improving the poor solubility of Res. Heart rate, ejection fractions and
fractional shortening of Res-SLN treating mice were found higher than those on mice with
cardiac toxicity induced by single high-dose intraperitoneal injection of DOX. And the
degree of myocardial ultrastructural lesions on mice was also observed.
Conclusion: Res-SLN has a certain therapeutic effect for protecting the myocardium and
reducing DOX-induced cardiotoxicity in mice.
Keywords: resveratrol, solid lipid nanoparticles, doxorubicin, heart failure
Introduction
Doxorubicin (DOX) is an anthraquinonoid broad-spectrum chemotherapy drug against
lymphoma, breast cancer and other malignant tumors. Actually, DOX is restricted on
clinical applications due to its severe cardiac toxicity.
1
The certain pathogenesis of
DOX-induced cardiotoxicity is still unclear, but intracellular calcium disorders, p53-
mediated cardiomyocyte apoptosis, oxidative stress and mitochondrial function dis-
orders are concerned.
25
Resveratrol (Res) is a polyphenolic antitoxin widely found in
natural plants, possessing the properties of anti-inammatory, anti-tumor, and protect-
ing cardiovascular pharmacological activity.
6,7
It is certain that Res can improve the
cardiomyocyte calcium cycling by up-regulating silent information regulator-1 (SIRT-
1)-mediated deacetylation, thereby inhibiting the production of reactive oxygen species
to protect the myocardium and improve DOX-induced cardiotoxicity.
8,9
Correspondence: Tong Zhang
Experiment Center for Teaching and
Learning, Shanghai University of
Traditional Chinese Medicine, 1200 Cailun
Road, Pudong New District, Shanghai
201203, Peoples Republic of China
Email zhangtdmj@hotmail.com
Bing Wang
Center for Pharmaceutics Research,
Shanghai Institute of Materia Medica,
Chinese Academy of Sciences, 501 Haike
Road, Pudong New District, Shanghai
201203, Peoples Republic of China
Email bwang@simm.ac.cn
International Journal of Nanomedicine Dovepress
open access to scientic and medical research
Open Access Full Text Article
submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2019:14 60616071 6061
DovePress © 2019 Zhang et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.
php and incorporate the Creative Commons Attribution Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the
work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For
permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
http://doi.org/10.2147/IJN.S211130
Although Res possesses the effect of attenuating DOX-
induced cardiotoxicity, Res is poorly soluble in water,
which makes it hard to achieve a satisfactory effect after
orally taken. To overcome this obstacle, solid lipid nano-
particle (SLN) is employed. SLN, drew up in the early
1990s, is a drug delivery system made up of skeleton
materials and drugs and can improve drugs solubility
and bioavailability.
10
The drugs are wrapped or embedded
in the lipid nucleus, consisting of physiological compat-
ibility, biodegradable natural or synthetic solid lipid with
high-melting point like lecithin or glycerol, and made into
particles with the size of about 501000 nm.
11
The reported Res-SLNs were prepared by the probe
ultrasonication method,
12
emulsication and solidication
method,
13,14
solvent injection method
15
or homogenization
method.
16
They were dividedly utilized for transdermal
drug delivery, treating breast cancer, anti-inammatory,
enhancing hepatoprotection and brain delivery, but nothing
about Res-SLN inhibiting DOX-induced cardiotoxicity
was discussed. In this study, we interrogated SLN as
a drug delivery carrier of Res for the potential treatment
of Dox-induced cardiotoxicity. The Res-SLN was prepared
by emulsication and sonication method, followed with
optimation of the formulations and pharmacodynamic eva-
luation. The prepared Res-SLN was able to improve the
bioavailability of Res for better protecting the myocardium
and inhibiting the DOX-induced cardiac toxicity
(Figure 1).
Materials and methods
Materials
Res was purchased from ZELANG (Nanjing, China) and
Res reference substance (M28F-Q3FG, purity 98.513%)
was procured from National Institutes for Food and Drug
Control of China. PC-98T egg yolk lecithin (AL15018, purity
98%) was obtained from A.V.T. Pharmaceutical Co., Ltd
(Shanghai, China). Glycerol monostearate and glycerol tris-
tearate were purchased from Sinopharm Chemical Reagent
Co., Ltd (Shanghai, China), while poloxamer 188 and sodium
dichromate were from Yuanye Bio. (Shanghai, China).
Fifty male Kunming mice (2025 g), 10 male and female
SpragueDawley(SD)rats(180220 g) were purchased from
Shanghai Slac Laboratory Animal Company and raised in
Laboratory Animal Center, Shanghai University of
Traditional Chinese Medicine. Experiments were performed
in compliance with the requirements of the animal ethics
committee of Shanghai University of Traditional Chinese
Medicine (Ethical Accreditation No. SZY201507002).
Preparation of Res solid lipid
nanoparticles
Res-loaded SLNs were prepared using emulsication-
diffusion method followed by sonication.
17
Briey,
a certain amount of egg yolk lecithin and Res were dis-
solved completely in 30 mL absolute alcohol and then
mixed with 120 mg molten glycerol monostearate (GMS)
i) Emulsification diffusion
Sustained release Res
Lipids
Res-SLNs protect myocardium from doxorubicin
Doxorubicin
Doxorubicin
20 mg/kg Res
1 g/kg Res-SLN
60 mg/kg digoxin
Res-SLNs treated
Res-SLN
Improved solubility
Better efficiency
ii) Sonication
270 nm
O
OOH
OH
OH
OH N+OO
OO
O
O
RR
R
OO-
P
HO
Glycerol monostearate (GMS)
Egg yolk lecithin
0
Control
Model
Res
Res-SLN
Digoxin
Control
Model
Res
Res-SLN
Digoxin
Control
Model
Res
Res-SLN
Digoxin
Ejection fractions (%)
Fractional shortening (%)
Heart rate (bpm)
20
40
60
80
00
200
400
600
20
40
60
80
100
Resveratrol (Res)
20 mg/kg
Figure 1 Schematic illustration of the preparation of Res-SLN and its effects against doxorubicin cardiotoxicity.
Zhang et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
6062
to form an oil phase. The organic solvent was removed by
vacuum rotary evaporation (Yarong, Shanghai, China) to
obtain a layer of lipid lm. The water phase containing
poloxamer 188 was added to the lipid lm in 40
mins through a needle under sustained ultrasound. The
Res-SLN was collected after intermittently sonicated by
a probe sonicator (Xinzhi, Ningbo, China) at 400 W for 12
mins. The amount of egg yolk lecithin, Res and the
volume of the water phase were optimized using central
composite design/response surface method (Table 1).
Morphological characterization of
Res-SLN
The Res-SLN was diluted by pure water. Particle size dis-
tribution and ζ-potential of Res-SLN were determined by
Nano ZS 90 Dynamic Light Scattering (Malvern
Instruments, Malvern, UK). The mean diameter of Res-
SLN stored hermetically at different temperatures on the
5th and 10th day was also determined to evaluate its stabi-
lity. After that, the Res-SLN was imaged under transmission
electron microscopy (TEM) to investigate its morphology
by the approach of a reported procedure.
18
Res-SLN was
diluted at 0.5 mg/mL and placed on a carbon-coated copper
grid, followed by being stained with phosphotungstic acid
solution (1%, w/v) for 1 min. The prepared samples were
imaged under TEM (FEI, Columbus, OH, USA).
Determination of drug loading and
entrapment efciency
To calculate the entrapment efciency and drug loading
of Res-SLN, the unencapsulated Res was isolated from
Res-SLN through 10 KD ultraltration centrifuge tube
(Millipore, Billerica, MA, USA) by centrifugation at
15,000 rpm for 30 mins (Anting, Shanghai, China) and
detected by HPLC. The HPLC system (Agilent, Santa
Clara, CA, USA) was under the following condition:
Topsil C
18
column (4.6×150 mm, 5 μm, Welch,
Shanghai, China); injection temperature, 25°C; mobile
phase, acetonitrilewater (25:75, v/v, 1.0 mL/min); detec-
tion wavelength, 305 nm; sample size, 10 μL.
Stability of Res-SLN
The effect of storage temperature on the stability of Res-SLN
was studied. Optimized Res-SLNs were prepared repeatedly
and stored at 4°C or 25°C. Samples were taken to determine
the amount of Res by HPLC on the 1st, 5th and 10th day.
In vitro release of Res-SLN
In vitro release study of Res from Res-SLN was performed by
the dialysis method; 30 mL Res-SLN injected into a dialysis
bag (800014,000 Da, molecular weight cut-off) was sunk
into 400 mL simulated gastric uid, stirred at the speed of
50 rpm and maintained at 37°C. At regular intervals, 2 mL
Res containing simulated gastric uid was taken out and
replaced by 2 mL fresh-simulated gastric uid. The aliquots
were ltrated with 0.45 μm Millipore ltration before injected
into the HPLC system.
In vivo pharmacokinetic evaluation
Pharmacodynamic study was assessed in SD rats. Ten rats
were randomly divided into two groups and received
intragastric administration (10 mL/kg weight) of Res-
SLN or Res-lipid physical mixture at 10.2 mg/mL doses.
At regular intervals, 0.5 mL blood was collected through
orbit at 0, 5, 10, 20, 30, 60, 90, 120, 180, 360, 720 and
1440 mins after administration into centrifuge tubes con-
taining heparin sodium. The blood samples were subse-
quently centrifuged at 3000 rpm for 10 min to obtain
plasma.
For determination of Res content in plasma, 50 μL genis-
tein as internal standard (IS) was added into 100 μL plasma
before mixed with 850 μL. The mixture was vortexed for 5
mins and centrifuged at 12,000 rpm for 10 mins. The super-
natant was collected and quantied by ultra-HPLC (UPLC)
in tandem with mass spectrometry (MS).
The Ultimate 3000 UPLC system (Thermo, Waltham,
MA, USA) was applied and chromatographic separation
was achieved on a Syncronis C
18
column (2.1×50 mm,
1.7 μm; Thermo). The LC gradient elution was pro-
grammed as 70% water:30% methanol (0 min), 5%
water:95% methanol (3 mins), and 70% water:30%
methanol (8 mins). The ow rate was 0.3 mL/min.
Injection volume was 10 μL. The MS condition for
Res and IS quantication of TSQ triple quadrupole
mass spectrometry (Thermo) with electrospray ioniza-
tion is shown in Table 2.
Table 1 Facts and levels for central composite design-response
surface method
Factors Levels and code values
-α1 0 +1 +α
Lecithin (mg) 24 67.78 132 196.22 240
Res (mg) 20 40.27 70 99.73 120
Water volume (mL) 100 140.54 200 259.46 300
Abbreviation: Res, resveratrol.
Dovepress Zhang et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 6063
Pharmacodynamic study of Res-SLN
Animal experiments
Male KM mice (2025 g) were kept in an experimental animal
barrier environment for 1 week adaptive feeding. Fifty mice
were randomly divided into 5 groups: control group (normal
saline), model group (normal saline), Res group (20 mg/kg
free Res solution), Res-SLN group (1 g/kg Res-SLN lyophi-
lized powder suspension, equivalent to 20 mg/kg free Res),
and positive control group (60 mg/kg Digoxin solution). After
3 days of administration, except the control group, all mice
were treated with single intraperitoneal injection of 20 mg/kg
DOX solution.
19
The control group was injected with the same
volume of normal saline. Thereafter, mice in each group
received the corresponding drug for another 5 days according
to the above-mentioned dose.
Measurement of heart functions
Echocardiography was applied to the measurement of heart
functions. Every mouse was treated with isourane for
anesthesia and placed in a supine position with abdominal
hair removed on a Philips Sonos 5500 color Doppler ultra-
sound system (Philips, Amsterdam, Netherlands). Heart rate
(HR), left ventricular ejection fractions (EF) and left ventri-
cular fractional shortening (FS) were recorded and analyzed
to gure the degree of heart failure out.
Hematoxylin and Eosin staining
After the treatment, the mice were sacriced and the hearts
were removed. The myocardial tissue was circumcised at 1/4
of the apex of the left ventricle and xed with 10% formal-
dehyde. After dehydration, it was embedded in conventional-
dehydrated parafn, cut into 5-μm slices, and stained with
Hematoxylin and Eosin. After that, it was made into
a pathological section and microscopically observed under
an optical microscope. Arrangement of cardiomyocytes,
interstitial cells, cell necrosis and inammatory cell inltra-
tion were observed under 200-fold microscopy.
Statistical analysis method
Statistical analysis was performed by SPSS22.0 statistical
software (SPSS Inc., Chicago, IL, USA). The data were
expressed as mean ± standard deviation (x ± s). If the data
is in a normal distribution and the variance is homogeneous,
the LSD method is used for comparison between the groups.
P<0.05 indicates that the difference is statistically signicant.
Results
Optimization and characterization of
Res-SLN
Central composite design-response surface method
As shown in Tab le 3,theinuence of three factors (A: lecithin
dosage; B: Res drug dosage, and C: water volume) and ve
levels on the response variables (Y
1
: drug loading and Y
2
:
particle size) were studied. The quadratic polynomial regres-
sion equation was tted by Design Export 7.0 software (Stat-
Ease Inc., Minneapolis, MN, USA). Regression analysis was
performed for determining the optimal region for response
studies (Figure 2A). The model equation is:
Y1¼þ18:30 4:13 Aþ8:61 B0:052 C
1:46 ABþ0:19 ACþ0:17 BC
þ1:10 A20:65 B21:83 C2
;
ðR2¼0:9767;P<0:001Þ
Y2¼þ293:91 45:85 Aþ95:58 B2:61 C
30:41 ABþ9:13 ACþ4:59 BC
þ0:2:A213:96 B22:66 C2
;
ðR2¼0:9599;P<0:001Þ:
The R
2
values are closer to 1 (>0.95), showing that the model
ts well. And, P-values are<0.001, which indicates that the
equation model is signicant and has better correlation.
Particle size, zeta potential and drug loading
efciency
The optimal prescription of Res-SLN was as follows: the
concentration of lecithin was 47.17%, the concentration of
Res was 23.98%, and the volume ratio of water phase and oil
phase was 6.174. The average diameter of Res-SLN was
271.13 nm with a ζ-potential of 25.8±0.33 mV (Figure 2B
and C). The TEM observations showed that the Res-SLN
Table 2 The MS condition for Res and IS quantication
MS condition Parameter settings
Res IS
Spray voltage 3500 3500
Vaporizer temperature 90 90
Sheath gas pressure 25 25
Aux. gas pressure 5 5
Capillary temperature 350 350
Tube lens 61 68
Collision pressure 2.0 2.0
Collision energy 28 35
Protonated molecular ions [M+H]
+
(m/z) 227.0 269.1
Main fragment ion peak (m/z) 143.0 133.1
Abbreviations: MS, mass spectrometry; Res, resveratrol; IS, internal standard.
Zhang et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
6064
was spherical solid particles with uniform size and essen-
tially no adhesion between nanoparticles (Figure 2B).
Stability
To quantitatively detect Res in non-biological samples,
HPLC was introduced. Res was in good linearity over the
concentration range of 1.0854.00 μg/mL and the linear
equation was Y ¼77:013X 1:1707 R ¼0:9999ðÞ.
The long-term storage experiment showed that the
particle size of Res-SLN would grow up from 271.1 to
389.8 nm as time increases at 25°C, and the drug loading
would be reduced slightly (Figure 2D). While both particle
size and drug loading were stable at 4°C (Figure 2D).
Obviously, the prepared Res-SLN is more stable at 4°C
which indicated that it is more suitable for storing at 4°C
rather than room temperature.
In vitro release of Res-SLN
The in vitro release of Res-SLN was investigated by
dialysis in simulated gastric juice. The release of opti-
mized Res-SLN in simulated gastric juice was relatively
stable. As shown in Figure 2E, over 80% of Res was
released into the medium after 30 hrs. The Res-SLN
release prole revealed a sustained property.
In vivo pharmacokinetics evaluation
UPLC-MS/MS
UPLC in tandem with MS was introduced to detect Res in rat
plasma. Res in rat plasma was in good linearity over the
concentration range of 1.368684 ng/mL with a linear equa-
tion of y=0.02083x-0.1167 (r=0.9996). The UPLC-MS/MS
method developed for Res was specic(Figure 3A)andin
good precision, accuracy, recovery and matrix effect which
listed in Tab le 4. Meanwhile, Res was stable in rat plasma at
room temperature, 20°C and under freezing and thawing
conditions. (Table 5)
Pharmacokinetics of Res-SLN in rats
The pharmacokinetics study was performed in rats follow-
ing intragastric administration of Res-SLN and Res-lipid
physical mixture, respectively. Concentrationtime proles
of Res are shown in Figure 3B, and the pharmacokinetics
parameters are presented in Table 6. Compared with the
Res-lipid physical mixture, C
max
and AUC of Res-SLN
were signicantly increased and T
max
was signicantly
shortened (P<0.05), suggesting that entrapping of Res in
the Res-SLN may promote the oral absorption of Res and
SLN as a carrier may promote oral bioavailability of
insoluble drugs.
Table 3 The factors, levels and results of central composite design-response surface method
Number A: lecithin (mg) B: resveratrol (mg) C: water volume (mL) Y
1
: drug loading (%) Y
2
: particle size (nm)
1 132.00 70.00 200.00 18.74 309.37
2 67.78 40.27 259.46 8.76 169.93
3 196.22 99.73 259.46 21.20 334.03
4 132.00 70.00 200.00 18.88 309.37
5 196.22 40.27 140.54 6.72 165.77
6 132.00 120.00 200.00 28.97 372.30
7 196.22 99.73 140.54 21.65 307.70
8 24.00 70.00 200.00 29.67 372.93
9 132.00 20.00 200.00 3.65 121.13
10 132.00 70.00 200.00 18.67 301.23
11 132.00 70.00 200.00 18.83 301.23
12 67.78 40.27 140.54 10.66 198.47
13 132.00 70.00 200.00 18.74 274.97
14 67.78 99.73 259.46 30.65 451.87
15 67.78 99.73 140.54 32.13 479.63
16 240.00 70.00 200.00 12.90 200.57
17 132.00 70.00 300.00 14.33 279.83
18 132.00 70.00 100.00 11.65 277.50
19 132.00 70.00 200.00 16.01 269.97
20 196.22 40.27 259.46 4.88 156.17
Dovepress Zhang et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 6065
Pharmacodynamic study of Res-SLN
Behavior, weight and survival rate
Fifty mice were divided equally into ve groups.
(Figure 4A) All of them were in good spirits and behaved
normally in the rst 3 days of the experiments. After
injected with Dox (or saline) to induce heart failure, the
mice in the model group started losing weight and the
action of them was slower (Figure 4B). At the 7th day,
the survival rate was lower. As for the groups injected with
different drugs, the mice were better than the model group
to some extent. The mice in Res-SLN group lost least
weight (P<0.01) and the survival rate was signicantly
higher than other drug groups. According to the data
showed in Figure 4C, Res owns the benet of inhibiting
Dox-induced heart failure and entrapping Res in the SLN
was helpful for improving its efciency.
A
BD
CE
Drug loading capacity (%)
X1 = A: lecithin (mg)
25.5
18
10.5
3
99.73
84.87
70.00
55.13
40.27
400
375
290
206
120
99.73
84.87
70.00
55.13
40.27 67.78
99.89
132.00
164.11
196.22
140.64
170.27
200.00
229.73
259.46
200
245
290
335
380
67.78
99.89
132.00
164.11
194.22
40.27
140.54
170.27
200.00
229.73
259.46
120
185
250
315
380
55.13
70.00
84.87
99.73
170.27
200.00
229.73
259.46
11
15.75
20.5
25.25
30
140.54 67.78
99.89
132.00
164.11
196.22
140.54
170.27
200.00
229.73
259.46
3
9.6
16
22.5
29
40.27
55.13
70.00
84.87
99.73
B: Res (mg) B: Res (mg)
C: Volume (mL)
C: Volume (mL)
C: Volume (mL)
B: Res (mg)
C: Volume (mL)
A: lecithin (mg)
B: Res (mg)
16
14
12
10
8
6
4
2100 nm
0
0.1
-200
0612 18 24
Time (h)
Time (days)
0510
25oC
4oC
0
10
20
30
100
80
60
40
20
0
40
30 36
0
200000
400000
600000
800000
1000000
1200000
-100 0 100 200
Zeta potential (mV)
Total counts
Resveratrol release (%) Drug loading (%)
110 100 1000 10000
Size (d.nm)
Intensity (%)
A: lecithin (mg) A: lecithin (mg)
A: lecithin (mg)
67.78
99.89
132.00
164.11
196.22
33
X2 = B: Res (mg)
Actual factor
C: Volume (mL) = 200.00
Drug loading capacity (%)
Particle size (nm)
Particle size (nm)
Particle size (nm)
Drug loading capacity (%)
Drug loading capacity (%)
X1 = A: lecithin (mg)
A: lecithin (mg) = 132.00
X2 = B: Res (mg)
Actual factor
B: Res (mg) = 70.00
32.13
3.65
Drug loading capacity (%)
32.13
3.65
X1 = B: Res (mg)
X2 = C: valume (mL)
Actual factor
Drug loading capacity (%)
32.13
3.65
Particle size (nm)
X1 = A: lecithin (mg)
X2 = B: Res (mg)
Actual factor
C: Volume (mL) = 200.00
X1 = A: lecithin (mg)
X2 = C: Volume (mL)
Actual factor Actual factor
B: Res (mg) = 70.00
479.633
121.133
Particle size (nm)
479.633
121.133
A: lecithin (mg) = 132.00
X2 = C: Volume (mL)
X1 = B: Res (mg)
Particle size (nm)
479.633
121.133
Figure 2 The preparation and characterization of Res-SLN. (A) The response surface and contour plots of the tting equations of the particle size and drug loading. (B)
Particle size and TEM photography of the optimized Res-SLN. Bar = 100 nm. (C) Zeta-potential of the optimized Res-SLN. (D) Stability on drug loading of optimized Res-
SLN at 4°C or 25°C during 10 days. (E) Drug release from the optimized Res-SLN in simulated gastric juice (n=3).
Abbreviations: Res, resveratrol; SLN, solid lipid nanoparticles; TEM, transmission electron microscopy.
Zhang et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
6066
Figure 3 Pharmacokinetics of Res-SLN in rats. (A) The mass pattern of Res reference solution (black peak), rat plasma containing Res (red peaks), and blank rat plasma
(green peaks) in 8-min run. (B) Pharmacokinetic curves of Res-SLN and Res-lipid physical mixture in rats (n=5).
Abbreviations: Res, resveratrol; SLN, solid lipid nanoparticles.
Table 4 The data of methodological verication of UPLC-MS/MS for Res
Number Concentration
(ng/mL)
Precision (%) Accuracy (%) Recovery (%, mean ± SD) Matrix effect (%, mean ± SD)
Intra-
day
Inter-
day
16.84 1.4 11.3 98.1101.4 87.1±9.3 94.7±3.5
268.4 1.9 9.9 96.4102.0 93.5±2.5 93.2±3.6
3684 1.2 7.8 98.9101.9 85.7±3.2 89.0±0.9
Abbreviations: UPLC, ultra-high performance liquid chromatography; MS, mass spectrometry; Res, resveratrol.
Table 5 The stability of Res in rat plasma under different
conditions (n=5)
Groups Concentration
(ng/mL)
Recovery
(%)
RSD
(%)
Room temperature 6.84 94.9±9.9 11.6
68.4 97.1±6.8 7.9
684 92.2±9.1 11.1
20 °C 6.84 89.6±4.3 10.3
68.4 89.7±6.0 6.7
684 89.0±9.3 13.2
Freeze thawing 6.84 84.3±4.9 14.2
68.4 82.6±11.6 14.1
684 73.2±4.4 6.0
Abbreviations: Res, resveratrol; RSD, relative standard deviation.
Table 6 Pharmacokinetic parameters of Res-SLN and Res-lipid
physical mixture in rats (n=5)
Parameter Res-SLN Res-lipid physical
mixture
t
1/2
(1/h) 0.033±0.012 0.023±0.008
AUC (h × ng/mL) 264.288±26.334* 218.380±14.700
Vd (mL) 0.475±0.109 0.559±0.053
CL (mL/h) 0.014±0.003 0.013±0.003
MRT (h) 8.931±0.480* 11.015±0.389
C
max
(ng/mL) 150.544±32.120* 14.145±2.639
T
max
(h) 0.083±0.000* 3.333±2.160
Notes: *P<0.05, Res-SLN group vs Res-lipid physical mixture group.
Abbreviations: Res, resveratrol; SLN, solid lipid nanoparticles; Vd, apparent
volume of distribution; CL, clearance; MRT, mean residence time.
Dovepress Zhang et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 6067
Cardiac function detection
The cardiac function was examined by Doppler ultrasonic
diagnostic apparatus and HR, EF and FS were recorded in
Figure 4DF.
Heart function and HR decreased (P<0.05) in mice in
the model group which was injected with Dox to induce
heart failure but not with therapeutic drugs. The HRs of
Res-SLN and Digoxin group were increased compared to
the model group (P<0.01) in which the HR of Res-SLN
was better. On the contrary, free Res cannot increase the
HR of Dox-induced heart failure mice (P>0.05).
The EF and FS in echocardiography are introduced as
indicators in the clinical evaluation of cardiac function.
20
EF
and FS were decreased in the model group (P<0.01), sug-
gested that the Dox had induced the heart failure in mice.
BothEFandFSinRes-SLNwereincreasedcomparedtothe
model group (P<0.01). Res-SLN can improve the EF and FS
of left ventricle, while Digoxin only improves the EF.
0123456789
25
600
400
200
80
60
40
20
80
100
60
40
20
0
80
90
100
AB
D
C
EF
70
60
50
0246810
Time (days)
0
0
Control
Model
Res
Res-SLN
Digoxin
Control
Model
Res
Res-SLN
Digoxin
Control
Model
Res
Res-SLN
Digoxin
Control
Model
Res
Res-SLN
Digoxin
Body weight (g)
Heart rate (bpm)
Fractional shortening (%)
Ejection fractions (%) Percent survival (%)
30
35
40
45
50
Figure 4 Res-SLN inhibits doxorubicin-induced cardiotoxicity. (A) Schematic representation of the reported treatment for doxorubicin-induced cardiotoxicity. (B) The body
weight of mice in different groups (n=10). (C) The survival rate of mice in different groups (n=10). (DF) The heart rate, ejection fractions and fractional shortening of mice
in different groups. (*P<0.05, **P<0.01, ***P<0.01).
Abbreviations: Res, resveratrol; SLN, solid lipid nanoparticles.
Zhang et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
6068
It can be concluded that Res-SLN can improve the
decrease of cardiac indexes (HR, EF, FS) induced by
DOX in mice.
Pathological observation
Microstructure of myocardial histopathology showed that the
myocardial bers of the mice in the control group were well
arranged and no abnormality appeared in the myocardial
cells (Figure 5A). In the model group, the myocardial bers
were distorted and the cells showed obvious vacuoles degen-
eration of different sizes (Figure 5B). After the mice were
treated with drugs (free Res, Res-SLN and Digoxin), the
myocardial bers were arranged regularly and a few vacuoles
degenerated in the myocardial cells (Figure 5CE).
Discussion
In recent years, studies on the antagonism of DOX cardiotoxi-
city by botanicals have become a hot topic,
21
such as some
important active ingredients, Astragalus polysaccharide,
22
oxymatrine,
23
baicalin,
24
rhamnosin,
25
Res,
26
etc. In this
study, we developed a Res-SLN for better absorption and
efcacy after orally taken of Res, directing against its poor
solubility. The presented results verify an important role as
Res-SLN to be a protector against Dox-induced cardiotoxicity.
A role for SLN is to be a submicron drug delivery
system for delivering Res, leading to release gradually and
distribute better in the body. Our Res-SLN made up with
egg yolk lecithin and GMS released approximately 80% of
Res within 24 hrs, which is similar to those with glyceryl
Figure 5 Res-SLN protects myocardium from doxorubicin-induced cardiotoxicity. Myocardial tissue biopsy (200X) of mice in control group (A), model group (B), Res group
(C), Res-SLN group (D), and Digoxin group (E) (the vacuoles were indicated by black arrows and circles).
Abbreviations: Res, resveratrol; SLN, solid lipid nanoparticles.
Dovepress Zhang et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 6069
behenate.
15,16
The ndings of pharmacokinetics demon-
strated that Res-SLN resulted in better distribution in rat
than Res-lipid physical mixture. Other strategy was
reported that Res-loaded liposome was also able to
improve the efcacy of Res on stimulating the prolifera-
tion but preventing its cytotoxicity.
27
This role for Res-SLN is not limited to it, Res-SLN
triggers more credible improvement on cardiac function of
cardio-damaged mice by DOX than free Res. An estab-
lished DOX-induced cardiotoxicity model was employed
to evaluate the Res-SLN; 20 mg/kg DOX was intraperito-
neally injected into mice, which caused severe damage to
cardiac myocytes. As reported, after treated by 20 mg/kg
Dox, the left ventricular myocardium of a rat appeared
massive fragmentation and lysis of the myobrils, which is
similar to the model mice in our study.
28
The most promis-
ing therapeutics meaning of our ndings is that Res-SLN
can mitigate the collapse of the heart caused by DOX,
evidenced by restoring the HR, ES, FE of heart, which is
also proved by the myocardial tissue biopsy of mice.
Conclusion
In our study, SLN encapsulating Res was successfully pre-
pared and optimized by emulsication-diffusion method
followed by sonication. The prepared Res-SLN has
a credible therapeutic effect on DOX-induced cardiotoxicity
in mice by protecting the cardiomyocytes and improving the
HR, EF, and FS values in mice. However, Res-SLNs
research on anti-myocardial toxicity still requires deeper
and multi-faceted mutual verication. Based on the promis-
ing evidence that we have already found, we will be able to
keep working on elucidating the correlation between multi-
ple mechanisms that Res-SLN is involved.
Abbreviation list
Dox, doxorubicin; Res, resveratrol; SLN, solid lipid nano-
particles; GMS, glycerol monostearate; TEM, transmission
electron microscopy; IS, internal standard; UPLC, ultra-
high performance liquid chromatography; HR, heart rate;
EF, ejection fractions; FS, fractional shortening.
Acknowledgments
The work was supported by grants from Shanghai
Committee of Science and Technology (17401902300 and
18401931400), the Program of Shanghai Academic/
Technology Research Leader (18XD1403700), Shanghai
Three-year Action Plan for the Development of Traditional
Chinese Medicine [ZY(2018-2020)-CCCX-2001-04] and the
National Scientic and Technological Major Special Project
of China (2018ZX09201008-002).
Disclosure
The authors report no conicts of interest in this work.
References
1. Barry E, Alvarez JA, Scully RE, et al. Anthracycline-induced cardiotoxi-
city: course, pathophysiology, prevention and management. Expert Opin
Pharmacother.2007;8:10391058. doi:10.1517/14656566.8.8.1039
2. Takemura G, Fujiwara H. Doxorubicin-induced cardiomyopathy from
the cardiotoxic mechanisms to management. Prog Cardiovasc Dis.
2007;49:330352. doi:10.1016/j.pcad.2006.10.002
3. Lin ST, Chou HC, Chen YW, Chan HL. Redox-proteomic analysis of
doxorubicin-induced altered thiol activity in cardiomyocytes. Mol
Biosyst.2013;9:447456. doi:10.1039/c2mb25367d
4. Tallaj JA, Franco V, Rayburn BK, et al. Response of doxorubicin-induced
cardiomyopathy to the current management strategy of heart failure.JHeart
Lung Transplant.2005;24:21962201. doi:10.1016/j.healun.2004.12.108
5. Zhang S, Liu X, Bawa-Khalfe T, et al. Identication of the molecular
basis of doxorubicin-induced cardiotoxicity. Nat Med.
2012;18:16391642. doi:10.1038/nm.2919
6. Oktem G, Uysal A, Oral O, et al. Resveratrol attenuates
doxorubicin-induced cellular damage by modulating nitric oxide
and apoptosis. Exp Toxicol Pathol.2012;64:471479. doi:10.1016/j.
etp.2010.11.001
7. Salehi B, Mishra AP, Nigam M, et al. Resveratrol: a double-edged
sword in health benets. Biomedicines.2018;6:E91. doi:10.3390/
biomedicines6030091
8. Zhang C, Feng Y, Qu S, et al. Resveratrol attenuates doxorubicin-induced
cardiomyocyte apoptosis in mice through SIRT1-mediated deacetylation
of p53. Cardiovasc Res.2011;80:538545. doi:10.1093/cvr/cvr022
9. Gu J, Hu W, Song ZP, et al. Resveratrol-induced autophagy promotes
survival and attenuates doxorubicin-induced cardiotoxicity. Int
Immunopharmacol.2016;32:17. doi:10.1016/j.intimp.2016.01.002
10. Khan AR, Yang X, Fu M, Zhai G. Recent progress of drug nanofor-
mulations targeting to brain. J Control Release.2018;291:3764.
doi:10.1016/j.jconrel.2018.10.004
11. Mehnert W, Mader K. Solid lipid nanoparticles: production, charac-
terization and applications. Adv Drug Deliv Rev.2001;47:165196.
12. Shrotriya SN, Ranpise NS, Vidhate BV. Skin targeting of resveratrol
utilizing solid lipid nanoparticle-engrossed gel for chemically
induced irritant contace dermatitis. Drug Deliv Transl Res.
2017;7:3752. doi:10.1007/s13346-016-0350-7
13. Wang W, Zhang L, Chen T, et al. Anticancer effects of
resveratrol-loaded solid lipid nanoparticles on human breast cancer
cells. Molecules.2017;22:E1814. doi:10.3390/molecules22111814
14. Barbosa JP, Neves AR, Silva AM, et al. Nanostructured lipid carriers
loaded with resveratrol modulate human dendritic cells.
Int J Nanomed.2016;11:35013516. doi:10.2147/IJN.S108694
15. Singh A, Ahmad I, Ahmad S, et al. A novel monolithic controlled delivery
system of resveratrol for enhanced hepatoprotection: nanoformulation
development, pharmacokinetics and pharmacodynamics. Drug Dev Ind
Pharm.2016;42:15241536. doi:10.3109/03639045.2016.1151032
16. Jose S, Anju SS, Cinu TA, et al. In vivo pharmacokinetics and biodis-
tribution of resveratrol-loaded solid lipid nanoparticles for brain delivery.
Int J Pharm.2014;474:613. doi:10.1016/j.ijpharm.2014.08.003
17. Oliveira RR, Carriao MS, Pacheco MT, et al. Triggered release of
paclitaxel from magnetic solid lipid nanoparticles by magnetic
hyperthermia. Mater Sci Eng C Mater Biol Appl.2018;92:547553.
doi:10.1016/j.msec.2018.07.011
Zhang et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
6070
18. Wu X, Hu Z, Nizzero S, et al. Bone-targeting nanoparticle to co-deliver
decitabine and arsenic trioxide for effective therapy of myelodysplastic
syndrome with low systemic toxicity. J Control Release.2017;268:92
101. doi:10.1016/j.jconrel.2017.10.012
19. Gu J, Fan YQ, Zhang HL, et al. Resveratrol suppresses
doxorubicin-induced cardiotoxicity by disrupting E2F1 mediated autop-
hagy inhibition and apoptosis promotion. Biochem Pharmacol.
2018;150:202213. doi:10.1016/j.bcp.2018.02.025
20. Wu Z, Chen Q, Ke D, et al. Emodin protects against diabetic
cardiomyopathy by regulating the AKT/GSK-3beta signaling path-
way in the rat model. Molecules.2014;19:1478214793.
doi:10.3390/molecules190914782
21. Shabalala S, Muller CJF, Louw J, Johnson R. Polyphenols, autophagy
and doxorubicin-induced cardiotoxicity. Life Sci.2017;180:160170.
doi:10.1016/j.lfs.2017.05.003
22. Cao Y, Shen T, Huang X, et al. Astragalus polysaccharide restores
autophagic ux and improves cardiomyocyte function in
doxorubicin-induced cardiotoxicity. Oncotarget.2017;8:48374848.
doi:10.18632/oncotarget.13596
23. Zhang YY, Yi M, Huang YP. Oxymatrine ameliorates doxorubicin-
induced cardiotoxicity in rats. Cell Physiol Biochem.2017;43:626635.
doi:10.1159/000480471
24. Sahu BD, Kumar JM, Kuncha M, et al. Baicalein alleviates
doxorubicin-induced cardiotoxicity via suppression of myocardial
oxidative stress and apoptosis in mice. Life Sci.2016;144:818.
doi:10.1016/j.lfs.2015.11.018
25. Sun J, Sun G, Meng X, et al. Isorhamnetin protects against
doxorubicin-induced cardiotoxicity in vivo and in vitro. PLoS One.
2013;8:e64526. doi:10.1371/journal.pone.0064526
26. Shoukry HS, Ammar HI, Rashed LA, et al. Prophylactic supplemen-
tation of resveratrol is more effective than its therapeutic use against
doxorubicin induced cardiotoxicity. PLoS One.2017;12:e0181535.
doi:10.1371/journal.pone.0181535
27. Caddeo C, Teskac K, Sinico C, Kristl J. Effect of resveratrol
incorporated in liposomes on proliferation and UV-B protection
of cells. Int J Pharm.2008;363:183191. doi:10.1016/j.ijpharm.
2008.07.024
28. Al-Harthi SE, Alarabi OM, Ramadan WS, et al. Amelioration of
doxorubicin-induced cardiotoxicity by resveratrol. Mol Med Rep.
2014;10:14551460. doi:10.3892/mmr.2014.2384
International Journal of Nanomedicine Dovepress
Publish your work in this journal
The International Journal of Nanomedicine is an international, peer-
reviewed journal focusing on the application of nanotechnology in
diagnostics, therapeutics, and drug delivery systems throughout the
biomedical eld. This journal is indexed on PubMed Central,
MedLine, CAS, SciSearch
®
, Current Contents
®
/Clinical Medicine,
Journal Citation Reports/Science Edition, EMBase, Scopus and the
Elsevier Bibliographic databases. The manuscript management system
is completely online and includes a very quick and fair peer-review
system, which is all easy to use. Visit http://www.dovepress.com/
testimonials.php to read real quotes from published authors.
Submit your manusc ript here: https:/ /www.dovepress.com/in ternational-jou rnal-of-nanome dicine-journal
Dovepress Zhang et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 6071
... Furthermore, they measured the severity of cardiac ultrastructural abnormalities in mice. Protecting the myocardium and lowering DOX-induced cardiotoxicity are two of Res-SLN's therapeutic effects in mice [65]. ...
... They can encapsulate drug molecules in synthetic or natural solid lipids, so colloidal structures can form and store lipophilic molecules [26]. It was observed in another study that preparing RES-SLNs significantly increases blood plasma concentration and area under the curve, reducing the maximum plasma time needed to reach [27]. Also, comparing free RES with RES-SLNs showed increased absorption of drugs by regulating down overexpression of drug resistance protein in breast cancer [28]. ...
Article
Purpose Although resveratrol (RES) is an efficacious molecule, its therapeutic activity is impeded by significant limitations, such as rapid oral absorption, poor oral bioavailability, and low water solubility. Therefore, the preparation of RES in different pharmaceutical carriers represents an important tool to enhance its therapeutic applications. This study aims to potentiate the anti-cancer activity of RES by formulating it into a novel nanocarrier called Smart Lipid. Methods RES-loaded Smart Lipids were prepared by high-shear hot homogenization method utilizing a 21 × 32 factorial design with three factors at different levels: the total lipid concentration, the concentration of surfactant, and the type of surfactant. The responses were evaluated based on entrapment efficiency percentages and particle size. Results Our novel optimized RES-loaded Smart Lipid formula showed small particle size (288.63 ± 5.55 nm), good zeta potential (−16.44 ± 0.99 mV), and an entrapment efficiency of 86.346 ± 3.61% with spherical, clearly distinct, and no signs of fusion by transmission electron microscopy. Further characterization was done using differential scanning calorimetry, which showed no interaction between the drug and other components as the optimum lyophilized formula showed a peak at 54.75°C, which represents the lipid mixture, with an undetectable characteristic peak of the drug, which indicates entrapment of the drug, and the structure of the compounds was confirmed by Fourier transform-infrared spectroscopy, in which the majority of the drug’s characteristic peaks disappeared when loaded into Smart Lipid, which may indicate Smart Lipid’s ability to reduce the stretching and bending between bonds in RES. In addition, the optimized formula showed a sustained release pattern compared to RES suspension. Finally, the cytotoxic activity of the optimized RES-loaded Smart Lipid on different cell lines (human breast adenocarcinoma (MCF7), human hepatocellular carcinoma (HepG2), and human colon cancer cells (HT29)) was assessed through MTT assay (7-fold reduction in the IC50, from 3.7 ± 0.5 μM for free RES to 0.5 ± 0.033 μM for Smart Lipid loaded formula against MCF7, 3-fold reduction in the IC50 against HepG2 cells, from 10.01 ± 0.35 to 3.16 ± 0.21 μMm, and a more than 10-fold reduction in the IC50 from more than 100 to 10 ± 0.57 μM against HT-29 cells) and its effect on cell cycle progression and apoptosis induction were assessed using flow cytometry and annexin V kit, respectively. Our results showed that RES-loaded Smart Lipid significantly reduced cell viability, induced cell cycle arrest at G0/G1 phase, and apoptosis compared to free formula and free RES suspension. Conclusion Loading RES into this novel kind of nanocarrier enhanced RES absorption, cellular accumulation, and improved its anticancer properties.
Article
Full-text available
Overcoming the limited bioavailability and extensive metabolism of effective in vitro drugs remains a challenge that limits the translation of promising drugs into clinical trials. Resveratrol, despite its well-reported therapeutic benefits, is not metabolically stable and thus has not been utilized as an effective clinical drug. This is because it needs to be consumed in large amounts to overcome the burdens of bioavailability and conversion into less effective metabolites. Herein, we summarize the more relevant approaches to modify resveratrol, aiming to increase its biological and therapeutic efficacy. We discuss combination therapies, derivatization, and the use of resveratrol nanoparticles. Interestingly, the combination of resveratrol with established chemotherapeutic drugs has shown promising therapeutic effects on colon cancer (with oxaliplatin), liver cancer (with cisplatin, 5-FU), and gastric cancer (with doxorubicin). On the other hand, derivatizing resveratrol, including hydroxylation, amination, amidation, imidation, methoxylation, prenylation, halogenation, glycosylation, and oligomerization, differentially modifies its bioavailability and could be used for preferential therapeutic outcomes. Moreover, the encapsulation of resveratrol allows its trapping within different forms of shells for targeted therapy. Depending on the nanoparticle used, it can enhance its solubility and absorption, increasing its bioavailability and efficacy. These include polymers, metals, solid lipids, and other nanoparticles that have shown promising preclinical results, adding more “hype” to the research on resveratrol. This review provides a platform to compare the different approaches to allow directed research into better treatment options with resveratrol.
Article
Resveratrol (RSV) is a polyphenolic phytoalexin, and belongs to the stilbene family. RSV has several therapeutic activities such as cardioprotective, anticancer, and antioxidant. Apart from its therapeutic benefits, its pharmacological uses are limited due to low solubility, poor bioavailability, and short biological halflife. A researcher continuously focuses on overcoming the limitations of RSV through nanotechnology platforms to get the optimum health benefits. In this context, nanocarriers are pioneering to overcome these drawbacks. Nanocarriers possess high drug loading capacity, thermal stability, low production cost, longer shelflife, etc. Fortunately, scientists were proficient in delivering resveratrol-based nanocarriers in the present scenario. Nanocarriers can deliver drugs to the target sites without compromising the bioavailability. Thus, this review highlights how the latest nanocarrier systems overcome the shortcomings of RSV, which will be good for improving therapeutic efficacy and bioavailability. Moreover, recent updates on resveratrol-based novel formulations and their clinical trials have been addressed to manage several health-related problems.
Article
Antioxidant nutraceuticals can be found in several dietary sources and have been utilized for various medical benefits including health promotion, disease prevention, and support for treatment of acute and/or chronic diseases. Nonetheless, there are some limitations in delivering antioxidants via oral administration such as low solubility and permeability, pH and enzyme degradation, and instability of the compounds along the gastrointestinal tract leading to low bioavailability. In order to tackle these challenges, the utilization of lipid nanoparticles has numerous advantages to the escalating delivery system of antioxidants in nutraceuticals across the gastrointestinal tract barrier. Nowadays, several types of lipid nanoparticles can be used in antioxidant nutraceutical delivery systems through the oral route, namely solid lipid nanoparticles and nanostructured lipid carriers. This review article aims to provide notable information on the importance and applications of lipid nanoparticles in antioxidant delivery systems from nutraceuticals by an oral route. The mechanism in enhancing antioxidant compound transport across the gastrointestinal tract can occur by elevating loading capacity, improving chemical and physical stability, and increasing its bioavailability. To date, lipid nanoparticle vehicles have been developed to improve the delivery of antioxidant compounds to enhance bioavailability via oral routes. Lipid nanoparticles have remarkable benefits in delivering antioxidant nutraceuticals via oral administration. Hence, scale-up and commercialization of antioxidant nutraceutical-loaded lipid nanoparticles have been a potential technology in recent years. Subsequently, several vegetable and natural oils with antioxidant activity can also be utilized for nanoparticle formulation lipid components to increase nutraceuticals’ antioxidant properties and bioavailability.
Article
Full-text available
Resveratrol (3,5,4′-trihydroxy-trans-stilbene) belongs to polyphenols’ stilbenoids group, possessing two phenol rings linked to each other by an ethylene bridge. This natural polyphenol has been detected in more than 70 plant species, especially in grapes’ skin and seeds, and was found in discrete amounts in red wines and various human foods. It is a phytoalexin that acts against pathogens, including bacteria and fungi. As a natural food ingredient, numerous studies have demonstrated that resveratrol possesses a very high antioxidant potential. Resveratrol also exhibit antitumor activity, and is considered a potential candidate for prevention and treatment of several types of cancer. Indeed, resveratrol anticancer properties have been confirmed by many in vitro and in vivo studies, which shows that resveratrol is able to inhibit all carcinogenesis stages (e.g., initiation, promotion and progression). Even more, other bioactive effects, namely as anti-inflammatory, anticarcinogenic, cardioprotective, vasorelaxant, phytoestrogenic and neuroprotective have also been reported. Nonetheless, resveratrol application is still being a major challenge for pharmaceutical industry, due to its poor solubility and bioavailability, as well as adverse effects. In this sense, this review summarized current data on resveratrol pharmacological effects.
Article
Full-text available
Most of the potential therapeutic agents capable to modulate the pathophysiology or treat the neurological disorders and brain tumors are useless in the current modern and advanced era of neuroscience due to the impeding action of biological barriers. Among various therapeutic strategies applied for translocation of drug delivery across the blood-brain barrier (BBB), nanoformulations set an excellent platform for brain targeting by overcoming the biological and chemical barriers and protecting drug from efflux to promote the optimum therapeutic drug concentration in brain parenchyma tissues. Nanocarriers are the most widely studied delivery vehicles for BBB translocation with the efficiency of selectively targeting or exploiting inherent biological molecules, receptors, carriers or mechanisms of the brain. Nearly all of the available drug delivery nanocarriers explored in recent years for brain therapeutics and theranostics are based on lipid or polymeric materials. Polymeric nanoparticles (NPs) and lipid based nanocarriers including liposomes, solid lipid NPs (SLNs) and micelles, etc. are under the direct focus of neuroscientists due to the promising attributes and vast applications in neurological disorders. Surface modification of nanovehicles with proper targeting moiety or coating with surfactants promotes the interaction with endothelial cells and passage of nanocarriers to the brain. This review comprehensively depicts challenges to the brain targeted drug delivery, mechanisms of drug transportation across the BBB, and potential contributions of endogenous cells as NPs delivery cells and novel targeting ligands decorated nanoformulations in imaging, treating and controlling neurological disorders.
Article
Full-text available
In this study, resveratrol-loaded solid lipid nanoparticles (Res-SLNs) were successfully designed to treat MDA-MB-231 cells. The Res-SLNs were prepared using emulsification and low-temperature solidification method. The Res-SLNs were spherical, with small size, negative charge, and narrow size distribution. Compared with free resveratrol, the Res-SLNs displayed a superior ability in inhibiting the proliferation of MDA-MB-231 cells. In addition, Res-SLNs exhibited much stronger inhibitory effects on the invasion and migration of MDA-MB-231 cells. Western blot analysis revealed that Res-SLNs could promote the ratio of Bax/Bcl-2 but decreased the expression of cyclinD1 and c-Myc. These results indicate that the Res-SLN may have great potential for breast cancer treatment.
Article
Full-text available
Background/aims: Doxorubicin-induced cardiac toxicity has been a major concern of oncologists and is considered the main restriction on its clinical application. Oxymatrine has shown potent anti-cancer, anti-fibrosis, and anti-oxidative effects. Recently, it has been reported that oxymatrine is protective against some cardiovascular diseases. In this study, we aimed to investigate the effects of oxymatrine on doxorubicin-induced cardiotoxicity in rat hearts and H9c2 cells. Methods: Creatine Kinase - MB (CK-MB) and Lactate Dehydrogenase (LDH) levels were determined using commercial kits. Biochemical indices reflecting oxidative stress, such as catalase (CAT), malonyldialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were also analyzed with commercial kits. Mitochondrial reactive oxygen species (ROS) 2',7'-dichlorofluorescin diacetate (DCFH-DA) was measured by fluorescence microscopy. Histological analyses were conducted to observe morphological changes, and apoptosis was measured using a commercial kit. Western blots were used to detect the level of expression of cleaved caspase-3. Results: Doxorubicin treatment significantly increased oxidative stress levels, as indicated by catalase, malonyldialdehyde, superoxide dismutase, glutathione peroxidase and reactive oxygen species. Doxorubicin also increased pathological damage in myocardial tissue, myocardial ROS levels, and malonyldialdehyde levels, and induced apoptosis in myocardial tissues and H9c2 cells. All of these doxorubicin-induced effects were attenuated by oxymatrine. Conclusion: These in vitro and in vivo findings indicate that oxymatrine may be a promising cardioprotective agent against doxorubicin-induced cardiotoxicity, at least in part mediated through oxymatrine's inhibition of cardiac apoptosis and oxidative stress.
Article
Full-text available
Resveratrol (RSV), a polyphenolic compound and naturally occurring phytoalexin, has been reported to exert cardio-protective effects in several animal studies. However, the outcome of initial clinical trials with RSV was less effective compared to pre-clinical studies. Therefore, RSV treatment protocols need to be optimized. In this study we evaluated prophylactic versus therapeutic effect of resveratrol (RSV) in mitigating doxorubicin (Dox)-induced cardiac toxicity in rats. To investigate prophylactic effects, RSV was supplemented for 2 weeks along with Dox administration. After 2 weeks, Dox treatment was stopped and RSV was continued for another 4 weeks. To study therapeutic effects, RSV treatment was initiated after 2 weeks of Dox administration and continued for 4 weeks. Both prophylactic and therapeutic use of RSV mitigated Dox induced deterioration of cardiac function as assessed by echocardiography. Also RSV treatment (prophylactic and therapeutic) prevented Dox induced myocardial damage as measured by cardiac enzymes (LDH and CK-MB) in serum. Which was associated with decrease in Dox induced myocardial apoptosis and fibrosis. Interestingly our study also reveals that prophylactic use of RSV was more effective than its therapeutic use in mitigating Dox induced apoptosis and fibrosis in the myocardium. Therefore, prophylactic use of resveratrol may be projected as a possible future adjuvant therapy to minimize cardiotoxic side effects of doxorubicin in cancer patients. © 2017 Shoukry et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Article
We developed a magnetic solid lipid nanoparticles formulation of paclitaxel (PTX-loaded MSLNs) via emulsification-diffusion method. The physicochemical characterization of PTX-loaded MSLNs was performed by AFM, DLS, determination of entrapment efficiency (EE) and drug loading (DL), DSC, VSM, and physical stability. The in vitro effect of temperature and pulsed magnetic hyperthermia on drug release were studied. PTX-loaded MSLNs had a particle diameter around 250 nm with a narrow size distribution, spherical morphology, EE of 67.3 ± 1.2% and a DL of 17.1 ± 0.4 μg/mg. A decrease of the melting point of the lipid was observed following the preparation of the MSLNs. A threefold increase in the in vitro drug release rate was seen when temperature was raised from 25 to 43 °C. The lipid coating of MPs confer a temperature-dependent drug release and magnetic hyperthermia was used to trigger controlled PTX release from MSLNs.
Article
The clinical use of doxorubicin (DOX) is limited by cardiotoxicity, involving the dysregulation of autophagy and apoptosis in the myocardium, which were partly reversed by resveratrol (RSV) supplement. However, a definitive mechanisms accounting for DOX's cardiotoxicity and the protective role of RSV remain poorly defined. The aim of the present study was to clarify the specific role of E2F transcription factor 1(E2F1) in regulating autophagy and apoptosis in DOX-induced cardiotoxicity as well as the protective effects of RSV. Autophagy and apoptosis were successfully induced by the serum deprivation strategy in H9c2 cardiomyocytes. DOX inhibited autophagy through activating E2F1/mammalian target of rapamycin complex 1 (mTORC1) pathway and further induced apoptosis by activating E2F1/AMP-activated protein kinase α2 (AMPKα2) pathway in starved H9C2 cells. And RSV supplement showed increased autophagy and decreased apoptosis, accompanied with inhibitory effect on E2F1/mTORC1 as well as E2F1/AMPKα2 pathway. Moreover, the favorable effect of RSV on autophagy and apoptosis was dependent on E2F1. The same result was observed in the mouse model of DOX-induced cardiotoxicity in both non-myocardial ischemia and myocardial ischemia condition. Injection with DOX and RSV in combination, resulted in a reduced apoptotic ratio and activated autophagy process compared with the DOX treatment alone. In conclusions, RSV, which has been suggested to attenuate DOX-induced cytotoxicity, significantly blocks induction of E2F1/mTORC1 and E2F1/AMPKα2 pathway by DOX, leading to acceleratory autophagy and inhibitory apoptosis. And E2F1 plays a key role for the protective effect of RSV.
Article
Myelodysplastic syndromes (MDS) are a diverse group of bone marrow disorders and clonal hematopoietic stem cell disorders characterized by abnormal blood cells, or reduced peripheral blood cell count. Recent clinical studies on combination therapy of decitabine (DAC) and arsenic trioxide (ATO) have demonstrated synergy on MDS treatment, but the treatment can cause significant side effects to patients. In addition, both drugs have to be administered on a daily basis due to their short half-lives. In addressing key issues of reducing toxic side effects and improving pharmacokinetic profiles of the therapeutic agents, we have developed a new formulation by co-packaging DAC and ATO into alendronate-conjugated bone-targeting nanoparticles (BTNPs). Our pharmacokinetic studies revealed that intravenously administered BTNPs increased circulation time up to 3days. Biodistribution analysis showed that the BTNP facilitated DAC and ATO accumulation in the bone, which is 6.7 and 7.9 times more than untargeted NP. Finally, MDS mouse model treated with BTNPs showed better restoration of complete blood count to normal level, and significantly longer median survival as compared to free drugs or untargeted NPs treatment. Our results support bone-targeted co-delivery of DAC and ATO for effective treatment of MDS.
Article
Irritant contact dermatitis (ICD) is a chronic and relapsing skin disease with severe eczematous lesions. Despite its growing prevalence, therapeutic treatments remain limited. Long-term topical corticosteroid treatment can induce skin atrophy, hypopigmentation, and increase in transepidermal water loss. An innovative dermal treatment is essential to reduce the side effects of corticosteroids. Topical resveratrol (RES), although effective for ICD, is a challenging molecule due to low solubility and poor bioavailability. The objective of this work was to build RES-loaded solid lipid nanoparticles (RES-SLNs) with skin targeting. For this purpose, RES-SLNs were prepared using the probe ultrasonication method utilizing Precirol ATO 5 and Tween 20. The RES-SLNs were evaluated for particle size, entrapment efficiency (EE), and transmission electron microscopy (TEM) studies. Further, RES-SLNs were incorporated into Carbopol gel and investigated for ex vivo skin permeation, deposition study on human cadaver skin, and finally skin irritation study on New Zealand White rabbits. It was further assessed for possible beneficial effects on ICD using BALB/c mice. RES-SLN showed mean size below 100 nm and 68–89% EE. TEM studies confirmed spherical particles in the nanometer range. An ex vivo study of RES-SLN-loaded gel exhibited controlled drug release up to 24 h; similarly, in vitro drug deposition studies showed potential of skin targeting with no skin irritation. RES-SLN gel confirmed competent suppression of ear swelling and reduction in skin water content in the BALB/c mouse model of ICD when compared to marketed gel. Thus, the formulated RES-SLN gel would be a safe and effective alternative to conventional vehicles for treatment of ICD.