ArticlePDF Available

Chitosan Nanoparticles of Gamma-Oryzanol: Formulation, Optimization, and In vivo Evaluation of Anti-hyperlipidemic Activity

Authors:

Abstract

The elevated blood levels of cholesterol and low-density lipoproteins result in hyperlipidemia. The available expensive prophylactic treatments are kindred with severe side effects. Therefore, we fabricated the polymeric nanoparticles of gamma-oryzanol to achieving the improved efficacy of drug. The nanoparticles were prepared by ionic gelation method and optimized using 2³ full factorial design taking drug/polymer ratio (X1), polymer/cross linking agent ratio (X2), and stirring speed (X3) as independent variables. The average particle size, percentage entrapment efficiency, and in vitro drug release at 2, 12, and 24 h were selected as response parameters. The factorial batches were statistically analyzed and optimized. The optimized nanoparticles were characterized with respect to particle size (141 nm) and zeta potential (+ 6.45 mV). Results obtained with the prepared and characterized formulation showed 83% mucoadhesion towards the intestinal mucosa. The in vitro findings were complemented well by in vivo anti-hyperlipidemic activity of developed formulation carried out in Swiss albino mouse model. The in vivo studies showed improved atherogenic index, malondialdehyde, and superoxide dismutase levels in poloxamer-407-induced hyperlipidemic animals when treated with oryzanol and gamma-oryzanol nanoformulation. Based on our findings, we believe that chitosan-mediated delivery of gamma-oryzanol nanoparticles might prove better in terms of anti-hyperlipidemic therapeutics.
Research Article
Chitosan Nanoparticles of Gamma-Oryzanol: Formulation, Optimization,
and In vivo Evaluation of Anti-hyperlipidemic Activity
Tejal Rawal,
1
Neha Mishra,
1
Abhishek Jha,
1
Apurva Bhatt,
1
Rajeev K. Tyagi,
2,3
Shital Panchal,
1
and Shital Butani
1,4
Received 25 December 2017; accepted 19 March 2018
Abstract. The elevated blood levels of cholesterol and low-density lipoproteins result in
hyperlipidemia. The available expensive prophylactic treatments are kindred with severe side
effects. Therefore, we fabricated the polymeric nanoparticles of gamma-oryzanol to achieving
the improved efcacy of drug. The nanoparticles were prepared by ionic gelation method and
optimized using 2
3
full factorial design taking drug/polymer ratio (X
1
), polymer/cross linking
agent ratio (X
2
), and stirring speed (X
3
) as independent variables. The average particle size,
percentage entrapment efciency, and in vitro drug release at 2, 12, and 24 h were selected as
response parameters. The factorial batches were statistically analyzed and optimized. The
optimized nanoparticles were characterized with respect to particle size (141 nm) and zeta
potential (+ 6.45 mV). Results obtained with the prepared and characterized formulation
showed 83% mucoadhesion towards the intestinal mucosa. The in vitro ndings were
complemented well by in vivo anti-hyperlipidemic activity of developed formulation carried
out in Swiss albino mouse model. The in vivo studies showed improved atherogenic index,
malondialdehyde, and superoxide dismutase levels in poloxamer-407-induced hyperlipidemic
animals when treated with oryzanol and gamma-oryzanol nanoformulation. Based on our
ndings, we believe that chitosan-mediated delivery of gamma-oryzanol nanoparticles might
prove better in terms of anti-hyperlipidemic therapeutics.
KEY WORDS: gamma-oryzanol; chitosan; nanoparticles; poloxamer-407; atorvastatin.
INTRODUCTION
The changes in the lifestyle lead to the prevalence of
hyperlipidemia. The hyperlipidemia is the presence of
abnormal levels of lipids in blood and marked by an elevated
cholesterol low-density lipoprotein (LDL) and low or unal-
tered high-density lipoproteins (HDL) levels in the blood [1].
Hyperlipidemia is treated by various classes of marketed
drugs, but the expensive treatment and side effects associated
with the long-term treatment are the major pitfalls [2].
Therefore, safe and cost-effective formulations are inevitably
required, and molecules which are generally recognized as
safe (GRAS) with no side effects might be good alternatives
to allopathic treatments [3]. The rice bran oil is considered as
a healthy vegetable oil [4], and gamma-oryzanol is a mixture
of ferulic acid esters of phytosterol and tri-terpene alcohols.
This is a group of compounds present in the rice bran oil
comprising anti-oxidant, anti-hyperlipidemic, immuno-modu-
latory, and anti-cancer activities [5,6]. The gamma-oryzanol
has low bioavailability majorly due to poor aqueous solubility
[7]. Moreover, poor physicochemical nature is one of the
reasons for the failure of many drugs to enter the clinics [8].
There has been lot of work being done for the
development of novel drug delivery systems (NDDS) which
are seen better in terms of targeting, improved bioavailabil-
ity, and reduced adverse effects over the conventional
therapy [9]. Owing to the poor water solubility of gamma-
oryzanol, Ghaderi et al. [10] prepared ethyl cellulose-based
gamma-oryzanol nanoparticles in order to improvise the
stability and functional properties of liquid foodstuffs
containing gamma-oryzanol. Seetapan et al. [11] fabricated
gamma-oryzanol solid lipid nanoparticles and assessed their
rheological behavior under different temperature conditions.
Many researchers are working on the development of
different formulations of gamma-oryzanol. Viriyaroj A.
et al. [12] developed gamma-oryzanol liposomes composed
of phosphatidylcholine and cholesterol and investigated
their physicochemical properties and antioxidant activity
for the cosmetic applications. The niosomal formulation of
1
Department of Pharmaceutical Technology, Institute of Pharmacy,
Nirma University, Ahmedabad, Gujarat 382481, India.
2
Institute of Science, Nirma University, Ahmedabad, Guajrat 382481,
India.
3
Department of Periodontics, College of Dental Medicine, Georgia
Regents University, Augusta, Georgia 30912, USA.
4
To whom correspondence should be addressed. (email:
shital.butani@nirmauni.ac.in)
AAPS PharmSciTech ( #2018)
DOI: 10.1208/s12249-018-1001-8
1530-9932/18/0000-0001/0 #2018 American Association of Pharmaceutical Scientists
gamma-oryzanol by Manosroi et al. [13] was prepared using
cholesterol and Tween 61 to enhance the transdermal
absorption. Sapino et al. [14] incorporated gamma-
oryzanol in β-cyclodextrin nanosponges in order to enhance
its stability and effectiveness. The nanoparticulate systems
are advantageous with respect to their stability over other
systems [15]. The systems such as liposomes and niosomes
get easily cleared by the reticulo-endothelial systems which
reduce their bioavailability. Therefore, nanoparticulate
systems are reportedly seen to evade the reticulo-
endothelial systems for bettering the bioavailability of
gamma-oryzanol [16].
Chitosan is a natural polymer widely used due to its
salient features of biodegradability, biocompatibility, low
toxicity, anti-bacterial, and anti-microbial properties [17,
18]. The mucoadhesive properties of chitosan help in the
fabrication of nanoparticles due to its cationic behavior. The
negatively charged intestinal mucosa will manifest a greater
uptake for the oppositely charged chitosan nanoparticles.
The hydrophobic polymers may lead to fast elimination of
gamma-oryzanol by reticulo-endothelial system (RES), and
therefore the use of hydrophilic polymeric carriers might
serve as a boon in the effective delivery of the drug.
Therefore, the chitosan is chosen as suitable polymer
candidate for the fabrication of nanoparticles of gamma-
oryzanol in order to protect the molecule from the RES
system to increase its bioavailability to achieving good anti-
hyperlipidemic activity.
Present work encompasses the formulation of chitosan
nanoparticles of gamma-oryzanol to improve its bioavail-
ability and efcacy. The nanoparticles were freeze-dried in
powder form and evaluated for their anti-hyperlipidemic
activity in an animal model. In the end, potency of drug-
loaded chitosan nanoparticles was compared with plain
gamma-oryzanol and standard anti-hyperlipidemic drug,
atorvastatin.
MATERIALS AND METHODS
Materials
Sodium tripolyphosphate (TPP), Tween 80, sodium
sulfate, and sodium metabisulte were procured from the
Central drug house, New Delhi. Glutaraldehyde was
obtained from S.D. Fine Chemical Ltd., Mumbai. Atorva-
statin tablets (5 mg) were procured from Troikaa Pharma-
ceuticals Limited, India. Gamma-oryzanol was purchased
from Tokyo Chemical Company. Poloxamer-407 (Pluronic
RF-127) was obtained from Sigma-Aldrich, USA. Diagnos-
tic kits for lipid prole were procured from Lab Care
Diagnostics Pvt. Ltd., India. Diagnostic kits for activated
partial thromboplastin time (APTT) and plasma prothrom-
bin time (PT) were obtained from Diagnostic Stago, France.
Chitosan (degree of deacetylation ratio of 7585%) and
dialysis bags (MWCO 1214 kDa) were obtained from
Himedia, Laboratory Pvt. Ltd., India. All other chemicals
and reagents used were of analytical grade.
Pre-formulation Studies
Drug-Excipient Compatibility Studies by Fourier Transform
Infrared Spectroscopy
The compatibility studies were carried out by FTIR
spectroscopy (Jasco FT-IR 6100, Japan). All the samples were
stored for 1 month at room temperature and analyzed further
for drug excipient compatibility. FTIR spectra of pure drug
and physical mixture of drug and excipient were studied by
making KBr mixture pellet. The absorption peaks of gamma-
oryzanol, excipients, and combination of gamma-oryzanol
and excipients were obtained at different wave numbers.
Formulation Development
Preparation of Chitosan Nanoparticles
The chitosan nanoparticles can be prepared by several
methods such as thermal/emulsion cross-linking, ionic gela-
tion, coacervation precipitation, and coalescence.The ionic
gelation method is the simplest and a viable one. Sodium
tripolyphosphate (TPP), a polyanion, interacts with positively
charged chitosan via electrostatic forces. This reversible
physical cross-linking via electrostatic interaction avoids the
toxicity of several reagents. The oppositely charged particles
undergo complexation and precipitated out to form spherical
particles [19]. The nanoparticles were formulated via ionic
gelation method, and chitosan was dissolved in an aqueous
solution containing acetic acid and Tween 80. TPP and drug
were dissolved in water and added dropwise into chitosan
solution with stirring. The nanoparticles were separated by
centrifugation at 15,000 rpm for 15 min at 10 °C. The particles
were suspended in phosphate buffer (PBS) pH 7.4 and
analyzed [2023].
Preliminary Batches
Many preliminary batches were prepared to evaluate the
effect of various formulation parameters, including drug to
polymer ratio (1:1, 1:1.5, and 1:2), polymer to TPP ratio (2:1,
4:1, 6:1, and 8:1), concentration of acetic acid solution (1, 2, 4,
and 8%), concentration of Tween 80 (0.25, 0.5, and 0.75%),
as well as process parameters such as stirring speed (200, 400,
and 600 rpm), the size of syringe needle (18G, 22G) on the
particle size, entrapment efciency, drug loading, in vitro drug
release, and permeation.
Optimization of Nanoparticles
A systematic optimization of the polymeric nanoparticles
is required to understand the interaction amongst various
parameters using suitable experimental designs. The data of
the preliminary trials was used to select the high-risk factors
for the optimization. Full factorial design (2
3
) was applied for
the optimization of various formulations using drug/polymer
ratio (X
1
), polymer/cross-linking agent ratio (X
2
), and stirring
speed (X
3
) as the independent variables and particle size, %
entrapment efciency, and in vitro drug release at 2, 12, and
Rawal et al.
24 h were taken as dependent variables. Eight batches were
prepared and evaluated extensively (Table I). The design
expert 7.0.0 software (StatEase Inc., USA) was used for data
treatment and design space generation. The suspension of
selected batch was centrifuged (BIO LAB BL 150 R) at
10,000 rpm, and pellets were collected. The pellets were dried
using 20% w/vlactose as cryo-protectant using freeze-dryer
(DELVAC) for 24 h over the temperature range of 40 °C to
20 °C for 1 h. The freeze-dried powder was then sealed,
stored in a refrigerator, and evaluated.
Characterization of Nanoparticles
Particle Size Distribution
The average particle size of nanoparticles was deter-
mined by differential laser scanning (DLS microtrac software)
taking PBS pH 7.4 as background. The particle size of the
selected batch was measured using zetasizer (ZS 200,
Malvern Instruments, UK), and experiments were done in
triplicate. The average values were employed for the
calculation of response surfaces [24].
Entrapment Efficiency
The supernatant was collected at the nal stage of
separation by centrifugation during the formulation of
nanoparticles. The supernatant was analyzed for the estima-
tion of the amount of free drug present using UV spectro-
photometer (UV 1800 Shimadzu, ScienticInstruments,
Japan) at 327 nm. Amount of the entrapped drug was
calculated using the following formula [25].
Entrapment efficiency %ðÞ¼
initial drugfree drugðÞ
initial drug 100 ð1Þ
In vitro Drug Release Studies
The drug release was measured using the method
reported elsewhere [26]withslightmodications. The
nanoparticles equivalent to 100 mg of gamma-oryzanol were
placed in a dialysis bag (Himedia; molecular weight cutoff
12,00014,000 Da) prepared using cellophane membrane
previously soaked in PBS pH 7.4 for 24 h. The dialysis bag
was kept in 100 ml of intestinal media (PBS pH 7.4) at 37 ±
0.5 °C with continuous stirring at 100 rpm. Subsequently, a
series of 2 ml solutions were withdrawn at specic time
intervals and replaced with the same volume of PBS (pH 7.4),
and absorbance was measured at 327 nm using UV-Visible
spectrophotometer. The drug release was also performed
using PBS in the presence of 20% ethanol to determine the
effect of pH and alcohol.
Measurement of Zeta Potential
The zeta potential of the selected batch was measured
using Malvern Zetasizer ZS 200 at 25 ± 0.5 °C. All the
samples were measured in triplicate. [27]
Morphological Analysis by Scanning Electron Microscopy
The surface morphological characteristics of gamma-
oryzanol-loaded chitosan nanoparticle batch were carried
out using a scanning electron microscope (SEM LEO 1530,
Oberkochen, Germany). The method used was plasma
deposition having a gold coating unit to make the sample
surface conductive to scanning electron beam. SEM of
samples was carried out at high vacuum with specimen
working distance of 6.3 mm and an accelerating voltage of
5kV[28].
Mucoadhesion Studies
Freshly cut, 2-cm-long piece of rat intestinal mucosa was
taken and tied onto the glass slide with the help of rubber
bands. Fifty milligrams of nanoparticles of the selected batch
was accurately weighed and transferred onto mucosa. It was
kept under controlled humidity for 10 min to allow the
anionic mucosa to interact with cationic chitosan. Then, the
slide was hanged at an angle of 45° under the burette tip and
Table I. Composition and Evaluation of the Optimization Batches
Batch no. X
1a
(drug/polymer)
X
2a
(polymer/TPP)
X
3a
(stirring speed)
PS (nm) PDI % EE % In vitro drug release
2h 12h 24h
B1 111 216.0 ± 0.1 0.218 ± 0.02 43.5 ± 3.1 12.6 ± 1.1 40.2 ± 0.9 57.0 ± 3.1
B2 1 11 168.4 ± 0.2 0.333 ± 0.03 26.0 ± 0.9 10.4 ± 2.3 38.4 ± 0.8 55.9 ± 0.6
B3 111 180.2 ± 1.1 0.246 ± 0.01 72.5 ± 0.1 28 ± 0.1 68.0 ± 1.4 77.7 ± 0.9
B4 1 1 1 186.7 ± 8.1 0.261 ± 0.01 68.2 ± 3.6 24 ± 0.1 67.8 ± 1.6 75.6 ± 1.6
B5 11 1 141.6 ± 2.6 0.382 ± 0.02 65.5 ± 0.4 13.2 ± 1.2 43.4 ± 2.1 59.6 ± 5.1
B6 1 1 1 99.10 ± 0.4 0.515 ± 0.04 39.5 ± 0.1 10.9 ± 0.4 39.7 ± 0.3 56.7 ± 2.3
B7 1 1 1 317.0 ± 0.1 0.342 ± 0.01 75.4 ± 0.6 29.7 ± 0.3 69.4 ± 0.1 79.8 ± 0.1
B8 1 1 1 333.0 ± 1.2 0.521 ± 0.02 69.3 ± 1.1 26.0 ± 0.9 67.9 ± 0.5 76.8 ± 0.4
PS: Particle size, PDI: Polydispersity Index, % EE :% entrapment efciency
a
Transformed values for X
1
,X
2
, and X
3
Formulation and Evaluation of Gamma-Oryzanol-Loaded Chitosan Nanoparticles
PBS (pH 7.4) was allowed to ow at the rate of 2 ml/min.
After 3 h, a number of nanoparticles which did not adhere to
the mucosal surface were collected on Whatman lter paper
and weighed. The percentage of mucoadhesion was calcu-
lated using following formula [28,29].
%Mucoadhesion ¼w1w2ðÞ
w1100 ð2Þ
where
w1 weight of gamma-oryzanol-loaded chitosan nanopar-
ticles taken initially.
w2 weight of gamma-oryzanol-loaded chitosan nanopar-
ticles collected on lter paper.
Stability Studies of Formulation
The stability is dened as the extent to which a product
remains within specied limits throughout its period of
storage and use. The selected batch of nanosuspension and
freeze-dried formulation was stored as per ICH guidelines at
25° ±2 °C and 60 ± 5% RH and at refrigerated condition (5 ±
3 °C) in a closed vial for 3 months and characterized with
respect to particle size and entrapment efciency [30].
Animal Studies
In vivo Anti-hyperlipidemic Activity
The animal protocol was approved by the Institutional
Animal Ethics Committee (Protocol no. IP/PCEU/MPH/14-1/
010). The Swiss albino male mice weighing 6040 g were
obtained from the Institute of Pharmacy, Nirma Universitys
animal house. All animals were treated in accordance with
the guidelines of the committee for CPCSEA.
Study Design
Swiss albino mice (n= 30) were randomized into follow-
ing groups:
(a) Normal control (NC): administered with saline for
3 days
(b) Normal induced (P-407): administered with saline for
3 days and on the 3rd day, poloxamer-407 (500 mg/
kg) was injected intraperitoneally
(c) Standard group (STD): administered with aqueous
suspension of atorvastatin (2 mg/kg/day, orally) for
all 3 days and on the 3rd day, animals were given
poloxamer-407 (500 mg/kg) intraperitoneally
(d) Gamma-oryzanol powder-treated group (OZ): admin-
istered with an aqueous suspension ofgamma-oryzanol
powder (100 mg/kg/day, orally in 0.5% CMC) for
3 days and on the 3rd day, animals were treated with
poloxamer-407 (500 mg/kg) intraperitoneally
(e) Gamma-oryzanol-loaded chitosan nanoparticle-
treated group (OZF): administered with an aqueous
suspension of drug-loaded freeze-dried powder of
nanoparticles (100 mg/kg/day, orally) for 3 days and
on the 3rd day, animals were treated with poloxamer-
407 (500 mg/kg, i.p.).
The blood was collected after 24 h of poloxamer-407
treatment, and animals were euthanized to extract the livers
[3134].
Assessment of Various Biochemical Parameters
Determination of Body Weight
Hyperlipidemia is dened as the elevations in the
fasting total cholesterol levels in the body [1]. Hence, it
becomes imperative to determine the body weight to
determine anti-hyperlipidemic activity of formulation. The
body weights were determined before and after the therapy.
Lipid Profile
The lipid prole is to quantify the body lipids and
atherogenic index (AI) to dene the cardiac risk [35]. The
increased lipid levels and AI represent the hyperlipidemic
conditions. It therefore becomes inevitable to study the
said effects in order to evaluate the anti-hyperlipidemic
activity. The blood was collected by puncture of retro-
orbital plexus of mice and allowed to stand for 10 min
without the addition of anti-coagulant followed by centri-
fugation at 4000 rpm for 10 min. The collected serum was
stored at 4 °C, and total cholesterol (TC), triglycerides
(TG), and high-density lipoprotein (HDL) were estimated
by procedure following the manufacturers recommenda-
tions (Lab Care Diagnostics, India). The very low-density
lipoprotein (VLDL) and low-density lipoprotein (LDL)
were determined using Friedewalds formula.
VLDLcholesterol ¼TG
5ð3Þ
LDLC¼TCHDLVLDLðÞ ð4Þ
In the AI, HDL ratio was measured using the following
equation:
Atherogenic index AIðÞ¼
TCHDLðÞ
HDL ð5Þ
HDL ratio ¼HDLC
TC ð6Þ
The LDL-C/HDL-C ratio was calculated as well.
Rawal et al.
Blood Coagulation Parameters
Patients with hyperlipidemia exhibit elevated serum
cholesterol levels and have a high risk of thrombosis.
Thrombosis is a condition which may result in either local
coagulation or blood clotting in the circulatory system
pathway. Thus, this necessitates the study of blood coagula-
tion [36]. The APTT and PT are the coagulation assays widely
used to diagnose the general state of coagulation system [37].
The APTT is a general coagulation screening test which
measures the time required to produce brin from initiation
of intrinsic pathways. The APTT measurement involves the
recalcication of plasma in the presence of a standardized
amount of coagulation factor XII activator (kaolin) and
cephalin (platelet substitute). PT is a screening test of
extrinsic pathways (coagulation factors II, V, VII, and X).
The PT measurement involves the use of calcium thrombo-
plastin to induce plasma coagulation. The plasma was
collected by addition of anti-coagulant, sodium citrate, in
the blood followed by centrifugation at 4000 rpm for 10 min
and stored at 4 °C. APTT and PT were measured in the
plasma using commercially available diagnostic kits (C.K.
PREST®, Diagnostica Stago, France).
Oxidative Stress Parameters
The isolated liver was washed by ice-cold buffer; tissues
were minced and homogenized in ice-cold PBS with 25
strokes of a tight Teon pestle of glass homogenizer (Remi
Motors Pvt. Ltd.) at 2500 rpm. The clear supernatant was
taken to estimate catalase (CAT), superoxide dismutase
(SOD), and reduced glutathione (GSH) levels, and tissue
homogenate were used to estimate malondialdehyde (MDA)
levels. All parameters were measured following the proce-
dures as reported elsewhere [38].
Histopathology of Carotid Artery
The heart removed from anesthetized animals was
washed by physiological saline and xed with 10% formalin.
After opening the right atrium, the heart was immediately
transferred to 10% formalin for 48 h for xation. Next, the
left coronary artery was dissected from its proximal part and
processed by liquid parafn for sectioning. The sections were
stained by hematoxylin-eosin stain and observed under light
microscope.
Statistical Analysis
Each experiment was performed in triplicates. The
statistical signicance was calculated using ANOVA. The
mean difference was considered signicant (p< 0.05).
RESULTS AND DISCUSSION
Hyperlipidemia is a major health issue and established as
one of the major reasons for cerebrovascular diseases,
coronary artery diseases, and peripheral vascular diseases.
Lipid accumulation promotes endothelial injury due to the
formation of Oxi-LDL and initiates atherosclerotic events.
Therefore, dyslipidemia and thrombosis along with oxidative
stress play a vital role in this whole process. Despite the
availability of various rst-line drugs such as statins and
brates, the adverse effects limit their use and beget the need
to explore the use of various novel herbal nutrients. The
gamma-oryzanol, extracted from rice bran oil, shows the
potential of improving serum lipid prole and decreases
coronary risk factors. Therefore, chitosan nanoparticles of
gamma-oryzanol were prepared in order to augment its
bioavailability and good anti-hyperlipidemic activity.
Drug-Excipient Compatibility Study
The characteristic peaks of gamma-oryzanol were iden-
tied with pure drug and combination of drug-excipient
spectrum by FTIR after the storage for 1 month at room
temperature. FTIR spectra of gamma-oryzanol and its
mixture with chitosan and TPP (Fig. 1) show the presence
of identical peaks (2954.00, 1602.00, 3298.68, and 1689.34/cm)
in both gamma-oryzanol and its mixture with chitosan and
TPP (2950.55, 1606.41, 3299.61, and 1689/cm) showing their
compatibility. The peaks represent the following groups
present in gamma-oryzanol: 2954/cm of alkanes (C-H
stretching), 1602/cm of aromatic hydrocarbons (C=C
stretching), 3298.68/cm of alcohol (intermolecular H bonded
OH stretching), and 1689.34/cm of ester (C=O stretching
enolic) groups.
Formulation of Nanoparticles
Preliminary Trials
Of all the factors studied, the drug to polymer ratio, the
polymer to TPP ratio, and stirring speed were identied as
important factors and selected for optimization. The concen-
tration of acetic acid, the concentration of Tween 80, and size
of the syringe needle were xed at 4%, 0.5%, and 22 gauge,
respectively. In all, eight batches were prepared at two levels
of each factor: drug/polymer, polymer/TPP, and stirring
speed.
Optimization of Nanoparticles
The 2
3
full factorial design was applied for the
optimization of the nanoparticles. Eight batches were
prepared using drug/polymer (X
1
), polymer/cross-linking
agent ratio (X
2
), and stirring speed (X
3
) as independent
variables (Table I).
Characterization of Nanoparticles
Particle Size
Nanoparticle size plays a crucial role in determining their
penetration throughout the intestinal mucosal layers. Red-
head et al. [39] reported penetration of nanoparticles of
100 nm size through the sub-mucosal layers of a rat intestinal
loop model than that seen with the microparticles which got
Formulation and Evaluation of Gamma-Oryzanol-Loaded Chitosan Nanoparticles
localized in the epithelial linings. As reported elsewhere [40,
41], nanoparticles within size range 200400 nm were seen
absorbed by the intestinal lining through macrophage uptake.
Hence, the particles with the size range within 100400 nm
were taken into consideration for proper absorption across
the intestinal mucosa. All the batches showed particle size in
the range of 99.1 ± 0.4 to 333 ± 1.2 nm [42]. Thus, it can be
said that the fabricated nanoparticles might get easily
absorbed through the intestinal linings for facilitated uptake
via macrophages. The polydispersity index (PDI) values
represent the average uniformity of a particle solution. Large
PDI values represent the large size distribution of the
particles. The PDI values ranged between 0.218 ± 0.02 and
0.521 ± 0.02. PDI, an important determinant of particle size
distribution as well as the stability of the nanoparticles,
presented well below 0.3 at lower speeds. However, at higher
speeds, the nanoparticles became polydisperse with PDI
values greater than 0.3 [42].
The polynomial equation obtained for the two-level
three-factor factorial design using Design Expert 7.0.0
software for the particle size was as follows:
YPS ¼2:0258:45X1þ48:97X217:425X3þ14:075X1X2
þ53:35X2X3þ1:825X1X3ð7Þ
The equation indicates that drug/polymer ratio had a
negative impact on the particle size. This may be explained by
the fact that the reduced chitosan concentration led to the less
cross-linking leading to small matrix formation. This in turn
may have driven the smaller particle size, whereas the
polymer/TPP and the stirring speed had a positive impact
on the particle size. The particle size was increased with
increasing polymer/TPP ratio. This is probably due to
increased cross-linking prompts large matrix formation be-
cause of increase in the polymer concentration. Further, the
increase in the stirring speed leads to the decrease in the
particle size due to higher shear forces.
Entrapment Efficiency
The entrapment efciency is an important parameter to
determine whether the drug is getting entrapped within the
nanoparticles or not. The polynomial equation obtained for
the entrapment efciency was as follows:
Y%EE ¼57:486:73X1þ13:86X2þ4:93X3
þ4:13X1X23:93X2X31:28X1X3ð8Þ
The drug to polymer ratio had a negative impact on the
entrapment efciency due to the decreased entrapment of the
drug within the same polymer concentration matrix upon
increasing the drug concentration. On the contrary, polymer/
TPP ratio and stirring speed leave a positive impact. This
could be explained due to the presence of the greater amount
of polymer leading to complete entrapment of drug. More-
over, increased stirring speed led to the formation of compact
nanoparticles resulting into the greater drug entrapment.
Fig. 1. Overlay FTIR spectra of gamma-oryzanol and its mixture with the excipients (chitosan and TPP)
showing compatibility of gamma-oryzanol with chitosan and TPP
Rawal et al.
In vitro Drug Release
The polynomial equation to calculate the in vitro drug
release at 2, 12, and 24 h:
Y2h¼19:351:525X1þ7:57X2þ0:6X30:4X1X2
þ0:325X2X3þ0:025X1X3ð9Þ
Y12h¼54:350:9X1þ13:925X2þ0:75X3þ0:475X1X2
þ0:375X2X3þ0:04X1X3ð10Þ
Y24h¼67:381:13X1þ10:08X2
þ0:83X30:13X1X20:01X2X30:33X1X3ð11Þ
The coefcient values calculated by the equation of
all three factors reveal that the selected factors did not
show any signicant effect on the drug release. The Food
and Drug Administration (FDA) has enforced the testing
of oral dosage forms in release media containing ethanol.
The FDA has mentioned that the potential interaction
between the oral formulation and alcohol may result in
the impairment of the formulation and dose dumping.
Hence, it becomes essential to carry out the in vitro
release studies in the presence of alcohol. However, no
dose dumping effect was observed in any of our batches
on the inclusion of 20% ethanol in PBS pH 7.4 [43].
In order to obtain the overlay plot, the particle size
between 100 and 200 nm and maximum drug entrapment
were selected as constraints. The desirability graph
showed the desirability of 0.922 for the composition of
Fig. 2. Contour plots and the overlay plot showing design space (range of desired product)
Formulation and Evaluation of Gamma-Oryzanol-Loaded Chitosan Nanoparticles
batch B5 and was selected as the Boptimized batch.^The
contour plots and overlay plot (Fig. 2) showed the yellow
acceptable region, within which the formulated batches
would fulll the criteria. To identify the signicance of the
effects and their interactions, ANOVA was run for each
parameter. The ANOVA analysis indicates that the
estimated responses are well described by the model, as
evinced by pand R
2
values. The signicant pvalues (<
0.05) for the particle size and the entrapment efciency
were 0.0135 and 0.0120, and R
2
values were 0.999 for both
the responses. However, the pvalue was greater than 0.05
(R
2
=0.997) in case of in vitro drug release, thus showing
the insignicant effect of the factors on the response
variables.
Zeta Potential of the Optimized Batch
Zeta potential of batch B5 was + 6.45 mV indicating
that the nanoparticles have a little cationic charge due to
the presence of chitosan that would help in the better
interaction of these nanoparticles to that with anionic
intestinal linings. However, the lower positive value of
zeta potential might result in lower repulsive forces
between particles which may lead to the agglomeration.
Fig. 3. Particle size and zeta potential of the optimized batch
Rawal et al.
The particle size and zeta potential of the optimized batch
are shown in Fig. 3.
Morphological Analysis by Scanning Electron Microscopy
SEM analysis (Fig. 4) showed the spherical shape of
spherical gamma-oryzanol-loaded nanoparticles. The polydis-
persity index value of the optimized batch (0.281 ± 0.01)
revealed the uniform distribution of nanoparticles.
Mucoadhesion Studies
Chitosan is well-known polymer for its mucoadhesive
properties. The higher the mucoadhesion, the better is the
effect. Sakloetsakun et al. [44] reported 44% of
mucoadhesion of the chitosan nanoparticles to the intestinal
mucosa. Bhosale et al. [45] investigated 67.3% mucoadhesion
of poly (lactic-co-glycolic acid) nanoparticles to the intestinal
mucosa. Our chitosan nanoparticle formulation exhibited a
high mucoadhesion of 83 ± 0.02%. The high mucoadhesion
value reveals good adherence property of prepared nanopar-
ticles to the intestinal mucosa which would help drug
absorption from the gastrointestinal (GI) tract.
Stability Studies
Freeze-drying has been reported as an effective method
by many researchers for converting liquid formulation into
solid to increase its shelf-life [46].Batch B5 after freeze-drying
showed similar results of zeta potential, particle size, and
entrapment(Table II). However, the freeze-dried product was
stable when stored in a tightly closed vial in refrigerated
condition for 3 months whereas the liquid suspension showed
an increase in particle size due to agglomeration. The stability
study should be further continued to label storage condition
and expiry date on the product.
Animal Studies
In vivo Anti-hyperlipidemic Study
The hyperlipidemia has been estimated by the various
models developed: high cholesterol diet-induced hyperlipid-
emia, triton-induced hyperlipidemia, poloxamer-407-induced
hyperlipidemia. The intraperitoneal administration of
poloxamer-407 was seen better to induce hypertriglyc-
eridemia within 24 h. It deviates normal lipoprotein level
from its regular range by lipoprotein lipase inhibition and
upregulation of 3-hydroxy-3-methylgluteryl coenzyme A
reductase (HMG-CoA reductase), regulatory enzyme for
cholesterol biosynthesis pathway.
Determination of Body Weight
As P-407 is a hyperlipidemia-inducing agent [47], group
animals exhibited increased body weight. Moreover, OZ- and
OZF-treated animals exhibited comparatively normal body
weight. The estimation of the changes in body weight is
shown in Fig. 5.
Table II. Stability Studies of the Optimized Nanoparticle Batch Before and After Lyophilization
Condition Time Average particle
size (nm)
Drug entrapment
efciency (%)
25 °C/60% RH Liquid nanoparticles Initial 141.6 ± 2.6 65.5 ± 0.4
1 month 242.8 ± 6.8 56.1 ± 3.6
3 months 466.0 ± 3.2 44.3 ± 2.1
Refrigerated condition (5 °C ± 3 °C) Initial 141.6 ± 2.6 65.5 ± 0.4
1 month 167.0 ± 3.9 62.8 ± 1.3
3 months 208.3 ± 3.2 49.6 ± 2.7
25 °C/60% RH Freeze-dried powder Initial 157.0 ± 2.1 65.5 ± 0.8
1 month 199.9 ± 0.8 64.2 ± 0.6
3 months 309.0 ± 2.4 53.2 ± 0.9
Refrigerated condition (5 °C ± 3 °C) Initial 157.0 ± 2.1 65.5 ± 0.8
1 month 159.2 ± 0.5 65.0 ± 0.3
3 months 168.0 ± 1.2 59.6 ± 1.2
Fig. 4. SEM image of the optimized design batch showing the size of
the nanoparticles
Formulation and Evaluation of Gamma-Oryzanol-Loaded Chitosan Nanoparticles
Lipid Profile
Hyperlipidemia is characterized by increased levels of
serum TC, TG, and LDL resulting in increased risk of
coronary heart disease. The LDL is bad cholesterol
whereas HDL is good cholesterol as it conveys extra
cholesterol to the liver for the removal from the body.
The following effects were observed in lipid prole: P-407
is reported to increase TC, TG, LDL, and VLDL and
reduce the HDL levels [48,49]. In our study, P-407 group
animals showed elevated TC, TG, VLDL, and LDL levels
by the upregulation of HMG-Co-A reductase enzyme and
decreased HDL levels, whereas STD, OZ, and OZF
groups showed a signicant reduction in TC, TG, VLDL,
and LDL levels and signicant elevation in HDL levels as
compared to that seen with the P-407-induced group.
However, the insignicant difference in lipid prole was
observed between OZ and OZF groups.
It has been reported that P-407 increases AI which is a
representative marker of atherosclerosis (plaque or lipid
deposition in aorta and liver) [50]. In our study, P-407 group
exhibited an elevation in AI which was seen decreased in
STD, OZ, and OZF animal group. The decrease in AI
represents reduced cardiac risk and therefore revealing the
good anti-hyperlipidemic activity of nanoparticle
formulation.
The low LDL/HDL ratio is an indicator of lower risk
for coronary diseases. P-407 group animals showed
elevated LDL/HDL ratio whereas OZ and OZF showed
asignicant decrease in LDL/HDL ratio as compared to
P-407-induced group and representing their good anti-
atherogenic property. However, again, the insignicant
difference was seen in the effects of OZ and OZF
(Table III;Fig.6). All these values were seen normalized
in STD, OZ, and OZF animals.
Blood Coagulation Parameters
The plasma levels of APTT and PT were measured as
the intrinsic, extrinsic, and common coagulation pathways.
APTT and PT were seen reduced in the P-407 group. OZ
and OZF animals showed elevated APTT and PT. OZF-
treated animals showed a signicant increase in APTT and
PT when compared with all other groups. The pretreat-
ment with OZF prolonged the APTT and PT and showed
its positive effect on both intrinsic and extrinsic coagula-
tion pathways (Table IV)[51]. The elevations in the
APTT and PT values for an extended period of time
might be observed due to the sustained release of gamma-
oryzanol from the nanoparticles. The elevations in APTT
and PT present the reduced risk of thrombosis and
advocate the good anti-hyperlipidemic activity of the
nanoparticle formulation.
Oxidative Stress Parameters
The oxidative stress occurs when MDA is the by-
product of lipid peroxidation. MDA is an indicator for the
index of lipid peroxidation in liver homogenate. P-407 is
reported to aggravate oxidative damage and formation of
oxi-LDL [51]. The anti-oxidant enzymes (SOD, CAT, and
GSH) inhibit the MDA levels and reduce the risk of
atherogenesis. This study shows that the P-407 group
exhibited a marked rise in MDA levels and reduces the
levels of antioxidant enzymes. A signicant reduction in
MDA levels and increase in SOD, CAT, and GSH levels
were seen in STD-, OZ-, and OZF-treated animals in
comparison to that seen with the P-407-induced group. A
signicant rise in SOD was observed in OZF-treated
animals when compared with the OZ-treated animals
indicating a preventive effect of OZF against
atherothrombotic complications and oxidative stresses
(Fig. 7). The levels of CAT and GSH levels were the
same in both OZ and OZF animals.
Table III. Results of Coronary Risk Factors
Groups Atherogenic index (AI) HDL ratio LDL/HDL
NC 0.549 ± 0.09 0.66 ± 0.04 0.36 ± 0.09
P-407 9.432 ± 0.97 0.10 ± 0.01 7.18 ± 0.78
STD (2 mg/kg) 1.808 ± 0.13 0.36 ± 0.02 1.42 ± 0.12
OZ (100 mg/kg) 1.582 ± 0.14 0.39 ± 0.02 0.89 ± 0.11
OZF (100 mg/kg) 1.209 ± 0.44 0.45 ± 0.01 0.76 ± 0.04
Fig. 5. Comparative estimation of body weight of normal control and
the treatment groups. Each group contains six animals; values
expressed as mean ± SEM. NC normal control group, P-407
poloxamer-407-induced group, STD standard drug atorvastatin
(2 mg/kg), OZ gamma-oryzanol drug powder (175 mg/kg) in 0.5%
CMC, OZF gamma-oryzanol-loaded chitosan nanoparticles (175 mg/
kg); asterisk indicates signicant difference from normal control
group; number sign indicates signicant difference from P-407-
induced group (p< 0.05)
Rawal et al.
Histopathology of Coronary Artery
The reduction in the damage to endothelium and
lipid core was seen on the histological samples of the
carotid artery of animals treated with our prepared
formulation (Fig. 8). It can be clearly seen that P-407
induced thrombus formation in the artery due to lipid
accumulation and formation of reactive oxygen species
(Fig. 8b). The STD and OZ led to the reduced lipid
accumulation (Fig. 8c, d). The complete clearance of
accumulated lipids was observed with OZF-treated group
(Fig. 8e) and therefore indicating its benecial effect
against thrombotic risk in the body.
CONCLUSION
In this study, we encapsulated gamma-oryzanol into
chitosan-TPP nanoparticles to develop a polymeric
nanocarrier with improved anti-hyperlipidemic activity.
The optimized nanoparticle formulation had a particle
size of 141.6 ± 2.6 nm falling well within the desired range
(100200 nm). The formulation exhibited well in vivo
anti-hyperlipidemic activity similar to that of marketed
formulation (atorvastatin). Although insignicant differ-
ence was observed in the lipid prole exhibited by plain
gamma-oryzanol and gamma-oryzanol nanoparticles, the
nanoparticulate formulation manifested a prominent effect
on blood coagulation and oxidative stress parameters.
The histopathological examination of carotid artery also
revealed the complete clearance of accumulated lipids in
the artery via gamma-oryzanol nanoparticles in compari-
son to atorvastatin and plain gamma-oryzanol. Therefore,
it can be stated that chitosan-loaded gamma-oryzanol
Fig. 6. Comparative estimation of lipid prole (A: total cholesterol; B: triglyceride; C: very low-density lipoproteins; D : high-density
lipoprotein) of normal control and the treatment groups. Each group contains six animals; values expressed as mean ± SEM. NC normal
control group, P-407 poloxamer-407-induced group, STD standard drug atorvastatin (2 mg/kg), OZ gamma-oryzanol drug powder (175 mg/kg)
in 0.5% CMC, OZF gamma-oryzanol-loaded chitosan nanoparticles (175 mg/kg); asterisk indicates signicant difference from normal control
group; number sign indicates signicant difference from P-407-induced group (p< 0.05)
Table IV. Coagulation Parameters of All Animal Groups
Groups APTT (s) PT (s)
NC 20.33 ± 0.92 11.83 ± 0.48
P-407 (500 mg/kg) 18.65 ± 0.37 10.83 ± 0.60
STD (2 mg/kg) 25.98 ± 0.77 18.50 ± 0.76
OZ (100 mg/kg) 30.08 ± 0.73 22.17 ± 0.79
OZF (100 mg/kg) 32.00 ± 1.58 26.10 ± 1.12
Formulation and Evaluation of Gamma-Oryzanol-Loaded Chitosan Nanoparticles
Fig. 8. Histopathological images of the carotid artery of groups. aNormal control. bP-407 treated. cSTD treated. dOZ
(175 mg/kg) treated. eOZF (175 mg/kg) treated
Fig. 7. Comparative estimation of oxidative stress parameters (A: malondialdehyde; B: superoxide dismutase; C: catalase; D: glutathione) of
normal control and the treatment groups. Each group contains six animals; values expressed as mean ± SEM. NC normal control group, P-407
poloxamer-407-induced group, STD standard drug atorvastatin (2 mg/kg), OZ gamma-oryzanol drug powder (175 mg/kg) in 0.5% CMC, OZF
gamma-oryzanol-loaded chitosan nanoparticles (175 mg/kg); asterisk indicates signicant difference from normal control group; number sign
indicates signicant difference from P-407-induced group (p< 0.05)
Rawal et al.
nanoparticles can serve as a better carrier with improved
anti-hyperlipidemic activity.
ACKNOWLEDGEMENTS
The authors would like to thank Institute of Pharmacy,
Nirma University, Ahmedabad for providing us all the
facilities to carry out our research work.
COMPLIANCE WITH ETHICAL STANDARDS
The animal protocol was approved by the Institutional
Animal Ethics Committee (Protocol no. IP/PCEU/MPH/14-1/
010). All animals were treated in accordance with the
guidelines of the committee for CPCSEA.
Conict of Interest The authors declare that they have no
conict of interest.
REFERENCES
1. Nelson RH. Hyperlipidemia as a risk factor for cardiovascular
disease. Primary Care: Clinics in Ofce Practice [Internet]. 2013
[cited 2017 Aug 23];40:195211. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S0095454312000991
2. Baker WL, Tercius A, Anglade M, White CM, Coleman CI. A
meta-analysis evaluating the impact of chitosan on serum
lipids in hypercholesterolemic patients. Ann NutrMetab.
2009;55:36874.
3. Zhang J, Zhang W, Mamadouba B, Xia W. A comparative study
on hypolipidemic activities of high and low molecular weight
chitosan in rats. Int J Biol Macromolecules [Internet]. 2012
[cited 2017 Nov 20];51:5048. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S0141813012002346
4. Zavoshy R, Noroozi M, Jahanihashemi H. Effect of low calorie
diet with rice bran oil on cardiovascular risk factors in
hyperlipidemic patients. J Res Med Sci. 2012;17:62631.
5. Cicero AF, Gaddi A. Rice bran oil and gamma-oryzanol in the
treatment of hyperlipoproteinaemias and other conditions.
Phytother Res. 2001;15:27789.
6. Ghatak SB, Panchal SJ. Anti-hyperlipidemic activity of
oryzanol, isolated from crude rice bran oil, on Triton WR-
1339-induced acute hyperlipidemia in rats. RevistaBrasileira de
Farmacognosia [Internet]. 2012 [cited 2017 Nov 20];22:6428.
Ava i lab l e from : http://www.scielo.br/
scielo.php?script=sci_arttext&pid=S0102-
695X2012000300024&lng=en&nrm=iso&tlng=en
7. Cho J-Y, Lee HJ, Kim GA, Kim GD, Lee YS, Shin SC, et al.
Quantitative analyses of individual γ-Oryzanol (Steryl
Ferulates) in conventional and organic brown rice (Oryza sativa
L.). J Cereal Sci [Internet]. 2012 [cited 2017 Aug 23];55:33743.
Available from: http://linkinghub.elsevier.com/retrieve/pii/
S0733521012000173
8. Plapied L, Duhem N, des Rieux A, Préat V. Fate of polymeric
nanocarriers for oral drug delivery. Curr Opin Colloid Interface
Sci [Internet]. 2011 [cited 2017 Aug 23];16:22837. Available
from: http://linkinghub.elsevier.com/retrieve/pii/
S1359029411000033
9. Letchford K, Burt H. A review of the formation and classica-
tion of amphiphilic block copolymer nanoparticulate structures:
micelles, nanospheres, nanocapsules and polymersomes. Eur J
Pharm Biopharm [Internet]. 2007 [cited 2017 Aug 23];65:259
69. Available from: http://linkinghub.elsevier.com/retrieve/pii/
S0939641106003316
10. Ghaderi S, Ghanbarzadeh S, Mohammadhassani Z,
Hamishehkar H. Formulation of gammaoryzanol-loaded nano-
particles for potential application in fortifying food products.
Tabriz University of Medical Sciences; 2014 p.
11. Seetapan N, Bejrapha P, Srinuanchai W, Ruktanonchai UR.
Rheological and morphological characterizations on physical
stability of gamma-oryzanol-loaded solid lipid nanoparticles
(SLNs). Micron [Internet]. 2010 [cited 2018 Feb 6];41:518.
Available from: http://linkinghub.elsevier.com/retrieve/pii/
S096843280900122X
12. Viriyaroj A, Ngawhirunpat T, Sukma M, Akkaramongkolporn
P, Ruktanonchai U, Opanasopit P. Physicochemical properties
and antioxidant activity of gamma-oryzanol-loaded liposome
formulations for topical use. Pharm Dev Technol [Internet].
2009 [cited 2018 Feb 6];14:66571. Available from: http://
www.tandfonline.com/doi/full/10.3109/10837450902911937,14,
665, 671
13. Manosroi A, Chutoprapat R, Abe M, Manosroi W, Manosroi J.
Transdermal absorption enhancement of rice bran bioactive
compounds entrapped in niosomes. AAPS Pharm Sci Tech
[Internet]. 2012 [cited 2018 Feb 6];13:32335. Available from:
http://www.springerlink.com/index/10.1208/s12249-012-9751-1
14. Sapino S, Carlotti ME, Cavalli R, Ugazio E, Berlier G, Gastaldi
L, et al. Photochemical and antioxidant properties of gamma-
oryzanol in beta-cyclodextrin-based nanosponges. J Incl Phe-
nom Macrocycl Chem [Internet]. 2013 [cited 2018 Feb 6];75:69
76. Available from: http://link.springer.com/10.1007/s10847-012-
0147-3
15. Gelperina S, Kisich K, Iseman MD, Heifets L. The potential
advantages of nanoparticle drug delivery systems in chemother-
apy of tuberculosis. Am J Respir Crit Care Med [Internet]. 2005
[cited 2018 Feb 6];172:148790. Available from: http://
www.atsjournals.org/doi/abs/10.1164/rccm.200504-613PP
16. Ghosh PK. Hydrophilic polymeric nanoparticles as drug car-
riers. Indian J Biochem Biophys. 2000;37:27382. Available
from:http://nopr.niscair.res.in/bitstream/123456789/19833/1/
IJBB%2037%285%29%20273-282.pdf
17. QiL,XuZ,JiangX,HuC,ZouX.Preparationand
antibacterial activity of chitosan nanoparticles. Carbohydr Res
[Internet]. 2004 [cited 2018 Feb 6];339:2693700. Available
from: http://linkinghub.elsevier.com/retrieve/pii/
S000862150400388X
18. Nagpal K, Singh SK, Mishra DN. Chitosan nanoparticles: a
promising system in novel drug delivery. Chem Pharm Bull.
2010;58:142330.
19. Fan W, Yan W, Xu Z, Ni H. Formation mechanism of
monodisperse, low molecular weight chitosan nanoparticles by
ionic gelation technique. Colloids Surf B: Biointerfaces [Inter-
net]. 2012 [cited 2017 Nov 20];90:217. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S0927776511005650
20. Yadav R, Kumar D, Kumari A, Yadav SK. Encapsulation of
podophyllotoxin and etoposide in biodegradable poly-D,L-
lactide nanoparticles improved their anticancer activity. J
Microencapsul [Internet]. 2014 [cited 2017 Nov 20];31:2119.
Available from: http://www.tandfonline.com/doi/full/10.3109/
02652048.2013.834988, 31, 211, 219
21. Rajitha P, Gopinath D, Biswas R, Sabitha M, Jayakumar R.
Chitosan nanoparticles in drug therapy of infectious and
inammatory diseases. Expert Opin Drug Deliv [Internet].
2016 [cited 2017 Nov 20];13:117794. Available from: http://
www.tandfonline.com/doi/full/10.1080/17425247.2016.1178232,
13, 1177, 1194
22. Paolicelli P, de la Fuente M, Sánchez A, Seijo B, Alonso MJ.
Chitosan nanoparticles for drug delivery to the eye. Expert
Opin Drug Deliv [Internet]. 2009 [cited 2017 Nov 20];6:23953.
Available from: http://www.tandfonline.com/doi/full/10.1517/
17425240902762818, 6, 239, 253
23. Agnihotri SA, Mallikarjuna NN, Aminabhavi TM. Recent
advances on chitosan-based micro- and nanoparticles in drug
delivery. J Control Release [Internet]. 2004 [cited 2017
Formulation and Evaluation of Gamma-Oryzanol-Loaded Chitosan Nanoparticles
Nov 20];100:528. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S0168365904003803
24. Bucci R, Magrì AD, Magrì AL, Marini F. Comparison of three
spectrophotometric methods for the determination of γ-
oryzanol in rice bran oil. Anal Bioanal Chem [Internet]. 2003
[cited 2017 Nov 20];375:12549. Available from: http://
link.springer.com/10.1007/s00216-002-1700-5, 375, 1254, 1259
25. Guan J, Cheng P, Huang SJ, Wu JM, Li ZH, You XD, et al.
Optimized preparation of levooxacin-loaded chitosan nano-
particles by ionotropic gelation. Physics Procedia [Internet].
2011 [cited 2017 Nov 20];22:1639. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S1875389211006808
26. Sahu BP, Das MK. Nanosuspension for enhancement of oral
bioavailability of felodipine. Appl Nanosci [Internet]. 2014
[cited 2017 Nov 20];4:18997. Available from: http://
link.springer.com/10.1007/s13204-012-0188-3, 4, 189, 197
27. Dong Y, Ng WK, Hu J, Shen S, Tan RBH. A continuous and
highly effective static mixing process for antisolvent precipita-
tion of nanoparticles of poorly water-soluble drugs. Int J Pharm
[Internet]. 2010 [cited 2017 Nov 20];386:25661. Available from:
http://linkinghub.elsevier.com/retrieve/pii/S0378517309008072
28. Rawal T, Parmar R, Tyagi RK, Butani S. Rifampicin loaded
chitosan nanoparticle dry powder presents an improved thera-
peutic approach for alveolar tuberculosis. Colloids Surf B:
Biointerfaces [Internet]. 2017 [cited 2017 Nov 29];154:32130.
Available from: http://linkinghub.elsevier.com/retrieve/pii/
S0927776517301625
29. Cory AH, Owen TC, Barltrop JA, Cory JG. Use of an aqueous
soluble tetrazolium/formazan assay for cell growth assays in
culture. Cancer Commun. 1991;3:20712.
30. Semete B, Booysen L, Lemmer Y, Kalombo L, Katata L,
Verschoor J, et al. In vivo evaluation of the biodistribution and
safety of PLGA nanoparticles as drug delivery systems.
Nanomedicine: Nanotechnol, Biol Med [Internet]. 2010 [cited
2017 Nov 20];6:66271. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S1549963410000961
31. Araujo FB, Barbosa DS, Hsin CY, Maranhão RC, Abdalla DS.
Evaluation of oxidative stress in patients with hyperlipidemia.
Atherosclerosis. 1995;117:6171.
32. Acay A, Ulu MS, Ahsen A, Ozkececi G, Demir K, Ozuguz U,
et al. Atherogenic index as a predictor of atherosclerosis in
subjects with familial Mediterranean fever. Medicina [Internet].
2014 [cited 2017 Nov 20];50:32933. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S1010660X14001153
33. Johnston TP, Zhou X. Oxidation of low-density lipoprotein cholesterol
following administration of poloxamer 407 to mice results from an
indirect effect: J Cardiovasc Pharmacol [Internet]. 2007 [cited 2017
Nov 20];49:24652. Available from: http://content.wkhealth.com/
linkback/openurl?sid=WKPTLP:landingpage&an=00005344-
200704000-00009
34. Yasuda T, Johnston TP, Shinohara M, Inoue M, Ishida T. The
effect of poloxamer 407 on the functional properties of HDL in
mice: P-407 mouse model of dysfunctional HDL. J Pharm
Pharmacol [Internet]. 2012 [cited 2017 Nov 20];64:67787.
Available from: http://doi.wiley.com/10.1111/j.2042-
7158.2011.01444.x
35. Swapnil SA, Anup BT, Ashok B, Ravishankar B, Shukla V.
Evaluation of anti-hyperlipidemic activity of LekhanaBasti in
albino rats. AYU (An International Quarterly Journal of
Research in Ayurveda) [Internet]. 2013 [cited 2018
Feb 9];34:220. Available from: http://www.ayujournal.org/
text.asp?2013/34/2/220/119687
36. Owens AP, Byrnes JR, Mackman N. Hyperlipidemia, tissue
factor, coagulation, and simvastatin. Trends in Cardiovascular
Medicine [Internet]. 2014 [cited 2018 Feb 9];24:958. Available
from: http://linkinghub.elsevier.com/retrieve/pii/
S105017381300114X
37. Kim J-A, Kim J-E, Song SH, Kim HK. Inuence of blood lipids
on global coagulation test results. Annals of Laboratory
Medicine [Internet]. 2015 [cited 2018 Feb 9];35:15. Available
from: https://synapse.koreamed.org/DOIx.php?id=10.3343/
alm.2015.35.1.15, 35, 15, 21
38. Lundquist P, Artursson P. Oral absorption of peptides and
nanoparticles across the human intestine: opportunities, limita-
tions and studies in human tissues. Advanced Drug Delivery
Reviews [Internet]. 2016 [cited 2017 Nov 20];106:25676.
Available from: http://linkinghub.elsevier.com/retrieve/pii/
S0169409X16302277
39. Redhead HM, Davis SS, Illum L. Drug delivery in poly(lactide-co-
glycolide) nanoparticles surface modied with poloxamer 407 and
poloxamine 908: in vitro characterisation and in vivo evaluation. J
Control Release [Internet]. 2001 [cited 2018 Feb 9];70:35363.
Available from: http://linkinghub.elsevier.com/retrieve/pii/
S0168365900003679
40. Chen J, Liu C, Shan W, Xiao Z, Guo H, Huang Y. Enhanced
stability of oral insulin in targeted peptide ligand trimethyl
chitosan nanoparticles against trypsin. J Microencapsul [Inter-
net]. 2015 [cited 2018 Feb 6];32:63241. Available from: http://
www.tandfonline.com/doi/full/10.3109/02652048.2015.1065920,
32, 632, 641
41. Fan T, Chen C, Guo H, Xu J, Zhang J, Zhu X, et al.Design
and evaluation of solid lipid nanoparticles modied with
peptide ligand for oral delivery of protein drugs. Eur J Pharm
Biopharm [Internet]. 2014 [cited 2018 Feb 6];88:51828.
Ava ila b le fr om: http://linkinghub.elsevier.com/retrieve/pii/
S0939641114002033
42. Pathak L, Kanwal A, Agrawal Y. Curcumin loaded self
assembled lipid-biopolymer nanoparticles for functional food
applications. J Food Sci Technol [Internet]. 2015 [cited 2018
Feb 14];52:614356. Available from: http://link.springer.com/
10.1007/s13197-015-1742-2, 52, 6143, 6156
43. Gohel MC, Bariya SH. Fabrication of triple-layer matrix tablets
of venlafaxine hydrochloride using xanthan gum. AAPS Pharm
Sci Tech [Internet]. 2009 [cited 2018 Feb 14];10:62430. Avail-
able from: http://www.springerlink.com/index/10.1208/s12249-
009-9244-z
44. Sakloetsakun D, Perera G, Hombach J, Millotti G, Bernkop-
Schnürch A. The impact of vehicles on the mucoadhesive
properties of orally administrated nanoparticles: a case study
with chitosan-4-thiobutylamidine conjugate. AAPS
PharmSciTech [Internet]. 2010 [cited 2018 Feb 7];11:118592.
Available from: http://www.springerlink.com/index/10.1208/
s12249-010-9479-8
45. Bhosale UV, Kusum Devi V, Jain N. Formulation and optimi-
zation of mucoadhesive nanodrug delivery system of acyclovir. J
Young Pharmacists [Internet]. 2011 [cited 2018 Feb 7];3:27583.
Available from: http://www.jyoungpharm.org/article/559
46. Abdelwahed W, Degobert G, Stainmesse S, Fessi H. Freeze-
drying of nanoparticles: formulation, process and storage
considerations. Adv Drug Deliv Rev [Internet]. 2006 [cited
2017 Nov 20];58:1688713. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S0169409X06001840
47. Chaudhary HR, Brocks DR. The single dose poloxamer 407
model of hyperlipidemia; systemic effects on lipids assessed
using pharmacokinetic methods, and its effects on adipokines. J
Pharm Pharm Sci. 2013;16:6573.
48. Sarker M, Mahmud ZA, Saha SK, Tithi NS, Ali MS, Bachar SC.
Anti hyperlipidemic activity of owers of Punica granatum in
poloxamer-407 induced hyperlipidemic mice model.
Pharmacogn J [Internet]. 2012 [cited 2017 Nov 20];4:6670.
Available from: http://linkinghub.elsevier.com/retrieve/pii/
S0975357512800241
49. Korolenko T, Johnston TP, Dubrovina NI, Kisarova YA,
Zhanaeva SY, Cherkanova MS, et al. Effect of poloxamer 407
administration on the serum lipids prole, anxiety level and
protease activity in the heart and liver of mice. Interdisc Toxicol
[Internet]. 2013 [cited 2017 Nov 20];6. Available from: http://
www.degruyter.com/view/j/intox.2013.6.issue-1/intox-2013-0004/
intox-2013-0004.xml
50. Omari-Siaw E, Wang Q, Sun C, Gu Z, Zhu Y, Cao X, et al. Tissue
distribution and enhanced in vivo anti-hyperlipidemic-antioxidant
effects of perillaldehyde-loaded liposomal nanoformulation
against Poloxamer 407-induced hyperlipidemia. Int J Pharm
[Internet]. 2016 [cited 2017 Nov 20];513:6877. Available from:
http://linkinghub.elsevier.com/retrieve/pii/S0378517316307839
51. Ghatak SB, Dhamecha PS, Bhadada SV, Panchal SJ. Investiga-
tion of the potential effects of metformin on atherothrombotic
risk factors in hyperlipidemic rats. Eur J Pharmacol [Internet].
2011 [cited 2017 Nov 20];659:21323. Available from: http://
linkinghub.elsevier.com/retrieve/pii/S0014299911003414
Rawal et al.
... Moreover, these two elements have been identified as compounds that increase antioxidant levels such as those of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) (9)(10)(11)(12). However, studies have found that the beneficial effects of curcumin and γ-oryzanol are limited by their low water-solubility, leading to low bioavailability (13)(14)(15)(16). Therefore, curcumin and γ-oryzanol were prepared in a solid dispersion to enhance their water solubility. ...
... Curcumin and γ-oryzanol compounds exhibit a wide range of pharmacological properties, including antioxidant, anti-inflammatory, anti-tumor, and anti-diabetic properties (9)(10)(11)(12)30). Nevertheless, both compounds have been reported to be limited with regard to their use as a nutritional product given their low water-solubility, resulting in low absorption and bioavailability (13)(14)(15)(16). The present study prepared curcumin and γ-oryzanol as a solid dispersion to enhance their water solubility. ...
Article
Full-text available
Oxidative stress is one of the major factors that contributes to brain deterioration in the elderly. Oxidation causes molecular alterations, structural damage, and brain dysfunction, which includes cognitive impairment. Memory loss can begin in middle-aged individuals, so prevention of brain deterioration before aging is important. Several studies have reported that curcumin and γ-oryzanol exhibits anti-oxidant and anti-inflammatory properties. However, curcumin and γ-oryzanol exhibit low aqueous solubility. Thus, a solid dispersion technique was used to prepare curcumin and γ-oryzanol to enhance their solubility and stability. This study aims to evaluate the effects and mechanisms of γ-oryzanol solid dispersion (GOSD) and curcumin solid dispersion (CURSD) on learning and memory in six groups of male rats (n=5/group). Group one was the adult control consisting of 6-week old male rats, and the remaining five groups consisted of 42-week (middle-aged) male rats. The groups were labeled as the control group, the GO group (GOSD 10 mg/kg·BW), the Cur group (CURSD 50 mg/kg·BW), the GO-LCur group (GOSD 10 mg/kg·BW plus CURSD 25 mg/kg·BW), and the GO-HCur group (GOSD 10 mg/kg·BW plus CURSD 50 mg/kg·BW). Substances were administrated by oral gavage once daily for 42 consecutive days. The GO-HCur group exhibited significantly increased learning and memory performance in a Morris water maze and in reacting to a spontaneous tendency novel object test. The rats also exhibited decreased levels of lipid peroxidation, increased superoxide dismutase levels, glutathione peroxidase levels, catalase activity, and enhanced c-Fos expression both in the hippocampus and prefrontal cortex. The results indicated that GOSD 10 mg/kg plus CURSD 50 mg/kg was able to enhance learning and memory performance in the middle-aged rats.
... 52,53 In addition, antioxidant materials incorporated in the liposomal structure have also been reported to alleviate oxidative stress and protect cells from damage. 54,55 The results of this study are consistent with previous research and provide further support for the antioxidant effects of γ oryzanol and liposome NCs. This antioxidant effect is evidenced by the reduction of lipid peroxidation (reducing the MDA activity), the elevation of catalase activity, and the increase in GSH levels. ...
Article
Full-text available
The present study aimed to develop and characterize liposome nanocarriers based on γ oryzanol and evaluate their potential in vitro and in vivo toxicity and antioxidant effects. The liposomes were physicochemically characterized using various techniques, including dynamic light scattering (DLS) for size and polydispersity index (PDI) measurements and ζ-potential analysis. The in vitro toxicity assessments were performed using hemolysis and MTT assays on the HS5 cell line. In vivo, acute oral toxicity was evaluated by using LD 50 assays in mice. Additionally, antioxidant activity was assessed through biochemical analysis of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and liver tissue catalase, malondialdehyde (MDA), and glutathione (GSH) levels. The results revealed that the liposomes exhibited a uniform and spherical morphology with suitable physicochemical properties for drug delivery applications. The in vitro cytotoxicity and hemolysis assays and the in vivo LD 50 experiment indicated the potential safety of γ oryzanol liposomes, especially at lower concentrations. In addition, the assessment of liver enzymes, i.e., ALT and AST, and the antioxidant markers further revealed the safety of the formulation, particularly for the liver as a highly sensitive soft organ. Overall, the liposome nanocarriers based on γ oryzanol were successfully formulated and expressed potential safety, supporting their application for the purposes of drug delivery and therapeutic interventions, particularly for hepatocellular and antioxidant therapies; however, further investigations for preclinical and clinical studies could be the future prospects for liposome nanocarriers based on γ oryzanol to explore the safety and efficacy of these nanocarriers in various disease models and clinical settings.
... Moreover, preserving the typical shape of polyhedrons as a result of using the NPs is another benefit of TPP [24,25,40]. TEM and DLS were performed to determine the size range of NPs and the obtained results reported the size of NPs around 150 ± 50 nm, as confirmed in past studies on drug delivery uses [41]. It is notable that, as discussed above, EGF has a positive charge while it is coated with the negative charges of chitosan. ...
Article
Full-text available
The chronic lesion has become a major biological difficulty. Using nanoparticles as drug delivery systems is remarkable nowadays. The unique properties of chitosan in combination with epidermal growth factor (EGF) can accelerate the wound‐healing process. In this study, Chitosan‐EGF (CS‐EGF) nanoparticles were manipulated and characterized by Transmission Electron Microscopy (TEM) microscopy, dynamic light scattering (DLS) method, and Fourier Transform Infrared Spectroscopy (FTIR) radiation. The antibacterial effect was estimated by minimum inhibitory concentration (MIC)/minimum bactericidal concentration (MBC) methods and the proliferation assay was measured on the HFF‐1 (human fibroblast cell line). Then, migration assay was accomplished and the gene expression analysis for transforming growth factor‐beta (TGF‐β), vascular endothelial growth factor (VEGF), and platelets‐derived growth factor (PDGF) was manipulated by the real‐time‐PCR method. The obtained results were considered statistically significant with P < 0.05. Obtained results illustrated no toxic effect on the HFF‐1 cell line treated with CS‐EGF. In cellular proliferation and migration assays, CS‐EGF nanoparticles demonstrated 2‐folds higher than the control. In the duration of 72 h of the experiment, and concentration of 10 µM, 90 ±10% of cells were migrated and the whole scratch was covered by fibroblasts. The real‐time‐PCR analysis also showed 7.5, 4.5‐, and 7‐fold upregulation of all TGF‐β, VEGF, and PDGF genes in comparison with the control group. This study confirmed that using chitosan nanoparticles as a carrier for EGF can accelerate cellular remodelling and annihilate bacterial infection in the process of treatment. As CS‐EGF nanoparticles in the acceleration of the skin remodelling process showed promising results, subsequent studies might be useful.
... Several studies have been conducted to improve the efficacy and delivery of GO either in topical or oral routes. Recently, chitosan-based nanoparticles of GO were formulated for improved anti-hyperlipidemic activity (Rawal et al. 2018). GO-loaded liposomes using phosphatidylcholine and cholesterol or sodium taurocholate as the encapsulating agents were also prepared to enhance the antioxidant activity for topical use (Viriyaroj et al., 2009). ...
Article
The current work presents the development of a nanostructured lipid carrier (NLC) as a colloidal encapsulation and controlled release system for bioactive γ-oryzanol (GO) from rice bran oil. GO-loaded NLC (GO-NLC) were prepared using rice bran oil GO via a hot homogenisation method with sunflower oil (liquid lipid) and stearic acid (solid lipid) as the encapsulating lipid matrix, and rice bran phospholipids and Tween 80 as stabilisers. Fabricated GO-NLC had a 95% encapsulation efficiency with a spherical ‘yolk–shell’ morphology and a 143 nm mean particle diameter. A 60-day storage stability study showed that GO-NLC had high GO retention (>90%) at ambient temperature in light and dark conditions. In vitro release studies demonstrated that GO can be released from GO-NLC via different modes: slow controlled release into simulated intestinal fluid, following the Korsmeyer-Peppas model, and rapidly into a surfactant solution (Tween 80), following first-order release kinetics. Lastly, in vitro studies showed encapsulation of GO in NLCs afforded an 18-fold increase in free radical-scavenging activity, a two-fold increase in hyaluronidase-inhibitory, and a 200-fold improvement in anti-inflammatory activity. These suggest that GO-NLC is a suitable GO delivery system, which can either perform as an oral extended-release formulation or a rapid-release, surfactant triggered system, with improved health-promoting bioactivities.
... Moreover, preserving the typical shape of polyhedrons as a result of using the NPs is another benefit of TPP (24,25,39). TEM and DLS were performed to determine the size range of NPs and the obtained results reported the size of NPs around 150±50nm, as confirmed in past studies on drug delivery uses (40). It is notable that, as discussed above, EGF has a positive charge while it is coated with the negative charges of chitosan. ...
Preprint
Chronic lesion has become a major biological burden for individual patients and health organizations. Using nanoparticles as drug delivery systems is remarkable nowadays. The unique properties of chitosan without any toxicity for living creations make it a suitable option for drug delivery. Epidermal growth factor (EGF) is one of the important agents for wound healing, cellular proliferation, extracellular matrix formation, and skin remodeling. A combination of these properties can accelerate the wound healing process. In this study, rh-EGF is embedded into the chitosan nanoparticles by the Ion-gelation method. Nanoparticles are characterized by TEM microscopy and the DLS method and conjugation efficacy is measured by FT-IR radiation. The antibacterial effect of manipulated nanoparticles was estimated by MIC/MBC methods. The cytotoxicity and proliferation were measured by MTT assay on the HFF-1 human fibroblast cell line. Migration assay was accomplished by in vitro scratch model and the gene expression analysis for TGF-β, VEGF, and PDGF were manipulated by the real time-PCR method. The obtained results were considered statistically significant with P < 0.05. Obtained results illustrated no toxic effect on the HFF-1 cell line treated with Chitosan-EGF (CS-EGF). In cellular proliferation and migration assays, CS-EGF nanoparticles demonstrated a better effect than free rh-EGF. For the duration of 72h of the experiment, the whole scratch was covered by fibroblasts. The real time-PCR analysis also showed upregulation of all TGF-β, VEGF, and PDGF genes. As CS-EGF nanoparticles in the acceleration of the skin remodeling process showed promising results, subsequent studies might be useful.
Article
A novel functional food for hyperlipidemia named icariin (ICA) pro-glycymicelles (ICA-PGs) using glycyrrhizin as a phytonanomaterial was easily prepared with improved storage, pH, and salt stabilities. ICA-PGs can easily dissolve in water to self-assemble into a clear glycymicelle solution with high ICA encapsulation efficiency. The ICA in ICA-PGs exhibits significantly increased aqueous solubility, faster in vitro release, and higher bioaccessibility than bare ICA. The ICA-PGs exhibited improved in vitro activities including antioxidant, anti-α-glucosidase, anti-lipase, and anti-cholesterol esterase activities. The ICA-PG also demonstrated improved antioxidant activity in cells. In vivo evaluation confirmed that the ICA-PG demonstrated a significant protective effect against experimental hyperlipidemia in mice, exhibiting decreasing levels of triglycerides (TGs), total cholesterol (TC), and low-density lipoprotein-cholesterol (LDL-C) in the serum, and restoring the hepatic morphology to the normal state. These results indicated that the ICA-PG could improve in vitro/in vivo profiles of ICA, providing a new concept and a promising functional food for hyperlipidemia.
Article
Gamma oryzanol (ORZ) is a nutraceutical that is poorly water soluble with poor intestinal absorption. In the current work, ORZ was nanoformulated into uncoated and chitosan coated micelles based on methoxy-poly(ethylene glycol)-b-poly(ε-caprolactone) (mPEG-PCL) and poly(ε-caprolactone)-b-methoxy-poly(ethylene glycol)-b-poly(ε-caprolactone) (PCL-PEG-PCL) copolymers for augmenting ORZ oral delivery. The physicochemical properties, morphological study, in-vitro release and safety of the nanoplaforms were determined. Importantly, the nephroprotective competence of the nanoplaforms was analyzed against acute kidney injury (AKI) rat model and the sirtuin-1 associated machineries were assessed. The results revealed that the micelles exerted particle size (PS) from 97.9 to 117.8 nm that was markedly increased after chitosan coating. The reversal of zeta potential from negative to highly positive further confirmed efficient coating. In vitro release profiles demonstrated prolonged release pattern. The nanoforms conferred higher cell viability values than free ORZ on Vero cell line. The designed micelles displayed augmented nephroprotection compared to free ORZ with the supremacy of CS coated micelles over uncoated ones in restoring kidney parameters to normal levels. The attenuated AKI was fulfilled via the modulation of sirtuin-1 signaling pathways translated by restoring the histological features, increasing renal antioxidant states, renal autophagy and decreasing renal inflammation and renal apoptosis. These outcomes confirmed that surface modification with chitosan had a considerable leverage on micelles safety, release behavior and in vivo performance.
Article
Electrospraying is a technique to improve the application and stability of bioactive compounds in food. Here, electrospraying was applied to fabricate gliadin particles incorporated γ-oryzanol. The round particles were obtained, with an average diameter of 481.56 ± 283.74 nm, from scanning electron microscopy. Simulations demonstrated how γ-oryzanol-loaded gliadin particles were unfolded in acetic acid and culminated in their globular shape under an electric field. The results also revealed that γ-oryzanol was present in gliadin particles. Moreover, there was a successful formation of particles with a homogeneous distribution and an enhanced thermostabilization of γ-oryzanol. In food simulants, γ-oryzanol demonstrated an initial burst release, followed by a subsequent, slower release that occurred gradually. Finally, MTT assays showed concentration- and time-dependent inhibitions of γ-oryzanol-loaded gliadin particles on HT-29 cells, with IC50 values of 0.47 and 0.40 mg/mL for 24 and 48 h, respectively. This study described a protocol for developing γ-oryzanol-loaded gliadin particles with enhanced stability, valuable release-behavior, and decreased HT-29 proliferation.
Article
Full-text available
Oral insulin delivery is often limited by protease degradation. 2-(Dimethylamino)-2-oxoethyl 4-(4-guanidinobenzoyloxy)phenylacetate methanesulphonate (Camostat mesylate) is reported to have the ability to inhibit trypsin activity, which is the main protease responsible for protein degradation. This study attempted to form a novel nanoparticle by covalently conjugating 4-(2-(2-aminoethylamino)-2-oxoethyl)phenyl 4-guanidinobenzoyloxy (FOY-251), an active derivative of camostat mesylate, to the backbone of poly (γ-glutamic acid) (γ-PGA), in order to improve insulin stability against protease. Goblet cell targeting CSKSSDYQC (CSK) peptide was demonstrated to effectively improve the epithelial absorption of insulin. Therefore, the novel nanoparticle was prepared by mixing cationic peptide modified trimethyl chitosan (TMC-CSK) with anionic γPGA-FOY conjugate using multi-ion crosslinked method. Results showed that not only the γPGA-FOY conjugate but also the prepared novel nanoparticle could inhibit trypsin activity both in vitro environment and on the intestinal mucosal surface. This study would be beneficial for peptide modified nanoparticles in oral insulin delivery.
Article
Full-text available
The supramolecular nano-assemblies formed by electrostatic interactions of two oppositely charged lipid and polymer have been made and used as nanocarriers for curcumin to address its bioavailability and solubility issues. These curcumin encapsulated nano-supramolecular assemblies were characterized with respect to their size (dynamic light scattering), morphology (TEM, SEM), zeta potential (Laser Doppler Velocimetry), encapsulation efficiency (EE), curcumin loading (CL) etc. Stability of the nano-assemblies was assessed at different storage times as a function of varying pH and temperature. The physicochemical characterization of nano-assemblies was performed using Fourier Transform Infra Red Spectroscopy (FT-IR) and Differential Scanning Calorimetry (DSC). The in-vitro antioxidant lipid peroxida-tion (TBARS), radical scavenging (DPPH, NO, H 2 O 2 , reducing power) activity assays of powdered curcumin and nano-encapsulated curcumin were performed. It was found that nano-encapsulated curcumin were roughly spherical in shape, presented high positive zeta potential (>30 mV), monodis-perse (polydispersity index <0.3), amorphous in nature, stable in the pH range of 2–6 and have enhanced antioxidant potency in comparison to crystalline curcumin in aqueous media. In conclusion, the curcumin encapsulated nanocarriers system has great potential as functional food ingredient of natural origin.
Article
Full-text available
The field of nanoparticle delivery systems for nutrients and nutraceuticals with poor water solubility has attracted a great attention during the last decades. Ethyl cellulose (EC) based GO-loaded nanoparticles were prepared by solvent evaporation method. The effects of formulation parameters on nanoparticle size, encapsulation efficiency (EE%) and loading efficiency (LE%) were investigated. X-ray diffraction (XRD) and differential scanning calorimetry (DSC) techniques were used to investigate the crystalline behavior of GO and EC after the preparation of nanoparticles. Stability of the prepared nanoparticle was investigated during five weeks of storage. Particle sizes of all formulation were in the range of 70-100 nm with narrow size distribution. Increase in the time of sonication from 1 to 5 minutes decreased the particle size. However, the mean particle size was increased when the sonication time increased from 5 to 7 minutes. The results showed that in the same concentration of PVA, increasing the ratio of EC:GO led to an increase in the GO encapsulation efficiency and decrease in loading efficiency. During five weeks, the mean diameter and size distribution indexes (SPAN values) of nanoparticles did not show significant changes. DSC and XRD studies indicated that crystallinity of GO was decreased in nanoparticles. EC based nanoparticles are promising carriers for addition of GO as a water insoluble antioxidant to fortify liquid food products without any change in quality of products.
Article
A new tetrazolium analog of 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) was evaluated as a substitute for MTT in the microculture screening assay for in vitro cell growth. This new tetrazolium compound, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2- (4-sulfophenyl)-2H-tetrazolium, inner salt (MTS), in the presence of phenazine methosulfate (PMS), gave a water-soluble formazan product that had an absorbance maximum at 490-500 nm in phosphate-buffered saline. The amount of colored product formed was proportional to the number of cells and the time of incubation of the cells with MTS/PMS. MTS/PMS was reactive in all the cell lines tested which included mouse leukemia L1210 cells, mouse Ehrlich tumor cells, mouse 3T3 fibroblasts, and human colon tumor cells (HT-29). HT-29 and 3T3 fibroblasts reduced MTS/PMS more efficiently than they reduced MTT. Comparable to the amount of product formed from MTT, MTS/PMS gave excellent product formation. The IC50 value for pyrazoloimidazole obtained using MTS/PMS was 200 microM; for 5-fluoro-2'-deoxyuridine, the IC50 value was 0.9 nM. These values compared very favorably with the IC50 values obtained by direct cell counts. Further, the same IC50 values were obtained when the absorbances of the formazan product in the 96-well plates were determined after different times of incubation.
Article
Current treatment therapeutic approach for tuberculosis is the administration of first line drugs in the form of tablets and capsules for 4-6 months. However, this approach leads to severe adverse effects. Therefore, present study was designed to achieving local and sustained targeting of anti-tubercular drugs in order to reduce dose and frequency. The nanoparticle based dry powder formulation of rifampicin was developed and analyzed with respect to its direct targeting potential of lungs. Rifampicin loaded nanoparticles were formulated by ionic gelation probe sonication method, and characterized with respect to particle size, zeta potential, entrapment and drug loading efficiency. The range of size and entrapment efficiency of prepared nanoparticles was estimated from 124.1 ± 0.2 to 402.3 ± 2.8 nm and 72.00 ± 0.1%, respectively. The freeze-dried powder of nanoparticle formulation was used to carry out in vitro lung deposition studies through Andersen cascade impactor. The cumulative in vitro drug release studies with developed nanoparticle formulation showed sustained release up to 24 hrs. Our in vitro sustained drug release results were corroborated by the extended residence and slow clearance of rifampicin from the lungs. Furthermore, our results suggest the minimum lung distribution of drug in treated rats which confirms the negligible toxicity rendered by nanoparticle dry powder formulation. Moreover, pharmacokinetic and toxicity studies carried out with prepared NPs dry powder inhalation (DPI) formulations and compared with conventional DPI
Article
An optimized perillaldehyde liposomal nanoformulation (PAH-LNF) was successfully applied to improve the pharmacological effect of perillaldehyde (PAH) in poloxamer 407-induced hyperlipidemia. Oral administration of PAH-LNF (240mg/kg per body weight) in rats significantly enhanced solubility and relative bioavailability (270.7%) compared to the free PAH with about 2.7-, 1.5-, 1.3-, 1.3- and 1.5-fold increase in AUC, T1/2, MRT, Cmax and Tmax, respectively. Tissue distribution study also revealed the accumulation of PAH in the liver, lungs, spleen, kidney, brain and heart in order of decreasing affinity. Moreover, a significant decrease in serum total cholesterol (TC), triglyceride (TG) and low-density lipoprotein cholesterol (LDL-C) with simultaneous increase in high-density lipoprotein cholesterol (HDL-C) level was observed in the chemically-induced hyperlipidemic mice which further confirmed PAH's anti-hyperlipidemic properties. Additionally, PAH-LNF also significantly increased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) with a concurrent decrease in malondialdehyde (MDA) to affirm the antioxidant and hepatoprotective effects of PAH. Thus, liposomal nanoformulation promises to be a useful drug delivery system for the development of PAH.
Article
In this contribution, we review the molecular and physiological barriers to oral delivery of peptides and nanoparticles. We discuss the opportunities and predictivity of various in vitro systems with special emphasis on human intestine in Ussing chambers. First, the molecular constraints to peptide absorption are discussed. Then the physiological barriers to peptide delivery are examined. These include the gastric and intestinal environment, the mucus barrier, tight junction between epithelial cells, the enterocytes of the intestinal epithelium, and the subepithelial tissue. Recent data from human proteome studies are used to provide information about the protein expression profiles of the different physiological barriers to peptide and nanoparticle absorption. Strategies that have been employed to increase peptide absorption across each of the barriers are discussed. Special consideration is given to attempts at utilizing endogenous transcytotic pathways. To reliably translate in vitro data on peptide or nanoparticle permeability to the in vivo situation in a human subject, the in vitro experimental system needs to realistically capture the central aspects of the mentioned barriers. Therefore, characteristics of common in vitro cell culture systems are discussed and compared to those of human intestinal tissues. Attempts to use the cell and tissue models for in vitro-in vivo extrapolation are reviewed.
Article
Introduction: Chitosan, a polymer from the chitin family has diverse pharmaceutical and bio-medical utility because of its easy widespread availability, non-toxicity, biocompatibility, biodegradability, rich functionalities and high drug-loading capacity. Recent pharmaceutical research has examined the use of chitosan-based systems for drug delivery applications in various diseases. The availability of functional groups permits the conjugation of specific ligands and thus helps to target loaded drugs to the site of infection/inflammation. Slow biodegradation of chitosan permits controlled and sustained release of loaded moieties; reduces the dosing frequency and is useful for improving patient compliance in infectious drug therapy. The muco-adhesion offered by chitosan makes it an attractive candidate for anti-inflammatory drug delivery, where rapid clearance of the active moiety due to the increased tissue permeability is the major problem. The pH-dependent swelling and drug release properties of chitosan present a means of passive targeting of active drug moieties to inflammatory sites. Areas covered: Development of chitosan-based nanoparticulate systems for drug delivery applications is reviewed. The current state of chitosan-based nanosystems; with particular emphasis on drug therapy in inflammatory and infectious diseases is also covered. Expert opinion: The authors believe that chitosan-based nanosystems, due to the special and specific advantages, will have a promising role in the management of infectious and inflammatory diseases.
Article
Polymeric hydrophilic nanoparticles have attracted attention because of their potential usefulness as drug-carriers in target specific cells, tissues or nucleus. Applications are expected through injectable, oral, nasal or ocular routes. Hydrophilic nanoparticles have been prepared that are hydrophilic on the surface as well as inside the core; particles have also been made that are hydrophilic on the outer surface but are hydrophobic within the core. External stimuli responsive hydrophilic nanoparticles are yet another new addition to the spectra of new materials in drug delivery. Water soluble as well as insoluble drugs can be entrapped into these polymers by adjusting the hydrophilicity/hydrophobicity of the core of the nanoparticles. The review discusses the requirements that these hydrogel nanoparticles must meet to be effective in drug delivery, and the importance of the preparative methods for controlling their sizes below 100 nm, with narrow size distribution.