You are on page 1of 16

Send Orders for Reprints to reprints@benthamscience.

net 239

Pharmaceutical Nanotechnology, 2020, 8, 239-254


RESEARCH ARTICLE
ISSN: 2211-7385
eISSN: 2211-7393

Eudragit L-100 Capsules Aromatize and Quaternerize Chitosan for


Insulin Nanoparticle Oral Delivery During Toxic Oxidative Stress in
Rat Liver and Kidney
BENTHAM
SCIENCE

Reza Mahjub1, Farzane K. Najafabadi2, Narges Dehkhodaei2, Nejat Kheiripour3, Amir N.


Ahmadabadi1, Sara S. Asl4, Masomeh Taheri-Azandariani2 and Akram Ranjbar2,5,*

1
Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran;
2
Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal Plants and Natural Products Re-
search Center, Hamadan University of Medical Sciences, Hamadan, Iran; 3Research Center for Biochemistry and
Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran; 4Anatomy Department,
School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; 5Research Center for Molecular
Medicine, Hamadan University of Medical Sciences, Hamadan, Iran

Abstract: Background: Insulin, like most peptides, is classified as a hydrophilic and macromolecular
drug that is considered as a low permeable and unstable compound in the gastrointestinal (GI) tract.
The acidic condition of the stomach can degrade insulin molecules. Moreover, the presence of proteo-
Pharmaceutical Nanotechnology

lytic activities of some enzymes such as trypsin and chymotrypsin can hydrolyze amide-bonds be-
tween various amino-acids in the structures of peptides and proteins. However, due to its simplicity
and high patient compliance, oral administration is the most preferred route of systemic drug delivery,
and for the development of an oral delivery system, some obstacles in oral administration of peptides
and proteins including low permeability and low stability of the proteins in GI should be overcome.
Objective: In this study, the effects of orally insulin nanoparticles (INPs) prepared from quaternerized
N-aryl derivatives of chitosan on the biochemical factors of the liver in diabetic rats were studied.
Methods: INPs composed of methylated (amino benzyl) chitosan were prepared by the PEC method.
Lyophilized INPs were filled in pre-clinical capsules, and the capsules were enteric-coated with Eu-
ARTICLE HISTORY dragit L100. Twenty Male Wistar rats were randomly divided into four groups: group1: normal control
rats, group 2: diabetic rats, group 3: diabetic rats received capsules INPs(30 U/kg/day, orally), group 4:
Received: January 17, 2020 the diabetic rats received regular insulin (5 U/kg/day, subcutaneously). At the end of the treatment, se-
Revised: April 01, 2020
Accepted: June 02, 2020 rum, liver and kidney tissues were collected. Biochemical parameters in serum were measured using
spectrophotometric methods. Also, oxidative stress was measured in plasma, liver and kidney. Histo-
DOI:
10.2174/2211738508666200628033442 logical studies were performed using H and E staining .
Results: Biochemical parameters, and liver and kidney injury markers in serum of the diabetic rats that
received INPs improved significantly compared with the diabetic group. INPs reduced oxidative toxic
stress biomarkers in serum, liver and kidney of the diabetic treated group. Furthermore, a histopatho-
logical change was developed in the treated groups.
Conclusion: Capsulated INPs can prevent diabetic liver and oxidative kidney damages (similar regu-
lar insulin). Therefore oral administration of INPs appears to be safe.
Lay Summary: Although oral route is the most preferred route of administration, but oral delivery of
peptides and proteins is still a challenging issue. Diabetes Mellitus may lead to severe complications,
which most of them are life-threatening. In this study, we are testing the toxicity of oral insulin na-
noparticles in kidney and liver of rats. For this investigation, we will prepare insulin nanoparticles
composed of a quaternized derivative of chitosan. The nanoparticles will be administered orally to
rats and the level of oxidative stress in their liver and kidney will be determined. The data will be
compared to the subcutaneous injection of insulin.

Keywords: Chitosan, diabetes, Eudragit L100, insulin nanoparticles, kidney, liver.

*Address correspondence to this author at the Research Center for Molecular Medicine, Hamadan University of Medical
Sciences, Hamadan, Iran; E-mail: akranjbar2015@gmail.com

2211-7393/20 $65.00+.00 © 2020 Bentham Science Publishers


240 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

1. INTRODUCTION fore, these nano-carriers are regarded as excellent


candidates for the oral delivery of peptides and
Type 1 and type 2 diabetes mellitus (T1DM and
proteins. Chitosan, which is a polymer with bio-
T2DM) are metabolic diseases characterized by a
compatible and biodegradable features, can be
progressive decrease in β-cell function and, when
used in the pharmaceutical industry as an absorp-
not or inappropriately treated, they may lead to
tion enhancer, dissolution improver, and sustained
severe complications, which are life-threatening
release factor in oral drug formulation. In this re-
and very costly to be controlled. Insulin is usually
gard, the mucoadhesive properties of chitosan are
administered by the subcutaneous route, which
introduced as the results of ionic interaction be-
significantly reduces morbidity and mortality
tween negatively charged functional groups in the
rates; however, approximately 60% of patients fail
surface of epithelial cells and positively charged
to achieve long-term glycemic control [1]. There-
amino groups in chitosan. Recently, aromatic de-
fore, the maintenance of blood glucose levels at
rivatives of chitosan received attention of many
near-normal levels reduces the risk of long-term
researchers, and the characterization of the synthe-
complications related to diabetes, such as cardio-
sized methylated chitosan has also been reported,
vascular diseases, non traumatic amputation, and
including an aromatic ring known as methylated
diabetic nephropathy. To overcome these prob-
N-(4-N,N-dimethyl aminobenzyl) chitosan. Ac-
lems, different routes of insulin administration
cordingly, it has been demonstrated that this deriv-
have been tested. The oral way still remains the
ative of chitosan can be applied as a capable gene
preferred choice for drug administration, because
carrier, which poses a greater transfection
of its noninvasive nature, simplicity, and high pa-
efficiency compared with trimethylated derivative
tient compliance [2]. Also, the significant obstruc-
and chitosan [5]. Also, the imbalance between the
tions for oral delivery of insulin are summarized as
production and the elimination of reactive oxy-
follows: hydrolytic enzymatic activity in the dif-
gen/nitrogen species (ROS/RNS) indicates Oxida-
ferent areas of gastrointestinal (GI) such as stom-
tive/Nitrosative stress and the decreased genera-
ach; intestinal lumen; and intestinal brush borders
tion of antioxidants leading to oxidative stress [6].
that cause decay of the peptides, unpleasant acidic
Under the hyperglycemic conditions, oxidative
environment of the stomach, the presence of tight
stress affects macromolecules in the kidney, liver,
junctions in the intestinal epithelium that act as a
and other tissues [7]. Although the relationship
hindrance for paracellular permeation, as well as
between oxidative stress inductions in diabetes has
the low mean residence time (MRT) of chemicals
been demonstrated, studying the effects of
through intestinal lumen that reduces the time of
nanoinsulin on various tissues in the body is still
exposure of insulin to the absorption site. There-
essential [8]. Therefore, the aim of this study was
fore, a wide variety of procedures have been eval-
to investigate the effects of oral administration of
uated to overcome these obstacles including the
Eudragit L-100 coated capsules containing insulin
application of permeation enhancers, niosomes,
nanoparticles (INPs) composed of a quaternized
chemical modification, mucoadhesive polymers,
aromatic derivative of chitosan on liver, kidney,
and development of lipid- based carriers including
and serum biomarkers oxidative damage compared
liposomes, enzyme inhibitors, microparticles, and
with subcutaneous insulin injection in male rats
solid lipid nanoparticles [3]. Entrapment of pro-
with type1 diabetes.
teins and peptides to mucoadhesive polymeric na-
noparticles is still regarded as the most useful ap- 2. MATERIALS AND METHODS
proach for the inhibition of drug instability against
degradation in GI as well as prolongation of pep- 2.1. Materials
tide permeability across the intestinal epithelium. In this study, Chitoclear™ chitosan [viscosity,
Notably, the preparation of a mucoadhesive sys- 1% (w/v) solution in acetic acid, 22 mPa s, with a
tem can improve the residence time of the thera- molecular weight of 120 KDa] was purchased
peutic compounds at the region of absorption with- from Primex (Siglufjordur, Iceland). Crystalline
in GI, and therefore, the bioavailability of an oral recombinant human insulin was provided by Exir
drug delivery system can be increased [4]. There- pharmaceutical company (Iran-Denmark). Manni-
Effect of Insulin Nanoparticles on Diabetes Pathogenesis Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 241

tol was obtained from Helm AG (Hamburg, Ger- 2.2.3. HPLC Analysis
many).
For HPLC analysis, 20 microliters of the sam-
Simulated intestinal fluid (SIF) was also ples were injected into a Schimadzu™ LC-20AD
prepared using potassium dihydrogen phosphate HPLC system (Kyoto, Japan) equipped with SPD-
(0.05 M) and sodium hydroxide (0.2 M) was ad- 10A vp UV-Visible detector set at the wavelength
justed to a pH value of 6.8. Moreover, phosphate- of 214 nm. Also, the Shimadzu ODS C18 column
buffered saline (PBS) was prepared by mixing so- (250 mm*4.6 mm) was applied as the solid sta-
dium chloride (137 mM), potassium chloride (2.7 tionary phase. The mobile phase consisted of ace-
mM), disodium hydrogen phosphate (10mM), and tonitrile/0.1% (v/v) trifluoroacetic acid (30:70),
potassium dihydrogen phosphate (2mM) (pH=7.4). and the pump flow was set at 1 ml/ min. The peaks
The degree of N-quaternization was determined were automatically integrated using the lab solu-
using 1H-NMR Spectroscopy, which was reported tion® software.
to be 43%. All other reagents were of pharmaceu-
tical or analytical grade and were used as received. 2.2.4. Determination of Entrapment Efficiency
(EE%) and Loading Efficiency (LE%)
2.2. Methods The direct procedure was used to determine the
2.2.1. Preparation and Characterization of EE% and LE%. In this method, the insulin value in
Nanoparticles nanoparticles was calculated according to the fol-
lowing equations 1 and 2:
INPs were developed by polyelectrolyte com-
plexation (PEC) procedure. Previously, Mahjub 

   (Eq. 1)

et al. [9] and Kheiripour et al. [10] in their studies
prepared insulin nanoparticles using the PEC 

   (Eq. 2)

method. Firstly, a quaternized aromatic derivative
of chitosan (i.e., Methylated 4-N,N-dimethyl ami- 2.2.5. Lyophilization of the Nanoparticles
no benzyl chitosan) was synthesized and then
characterized according to previous reports [9]. Before lyophilization, nanoparticles were fro-
Afterward, 5ml of insulin solution (1 mg/ml, zen at -20°C for a night. Lyophilization was pre-
pH=8.0) was added to an equal volume of the so- ceded using Lyotrap Plus (LTE Scientific Ltd.,
lution containing the newly synthesized polymer, Oldham, UK). Based on previous reports on
under gentle magnetic stirring via dropwise pro- freeze-drying, the desired samples were dried for
cess. For the improved curing of the particles, the 48 h, at a working pressure of 0.07 mbar, and a
obtained opalescent colloidal nano-suspension was condenser temperature of -46°C. The process of
continually stirred for 20 min., subsequently, the freeze-drying was carried out using mannitol 5%
obtained suspension was centrifuged at 17,000 rpm as the cryoprotectant.
for 20 min at 4°C. The separated nanoparticles 2.2.6. In Vitro Release Studies
were then reconstituted in the freshly prepared dis-
tilled water, and the supernatant was discarded. In this study, to investigate the release profile
Finally, the obtained nanoparticles were character- of nanoparticles in acidic and neutral media, both
ized for their size, PDI, and zeta potential using a of the simulated gastric fluid (SGF) and simulated
Zeta sizer ZEN3600 (Malvern instrument, UK) at intestinal fluid (SIF) were used for in vitro release
25°C. studies. To prepare the simulated gastric fluid
(SGF), hydrochloric acid solution (0.2 N, 39 ml)
2.2.2. Insulin Content in Nanoparticles was added to sodium chloride solution (0.2 N, 250
The restructured NPs were submerged in HCl ml), and the volume was then adjusted to 1000 ml
(1N) aqueous solution for 7 min. The particles with deionized water, while the pH was main-
were degraded, and their total contents of insulin tained at 2.2. Besides, the simulated intestinal fluid
was released to the acidic aqueous media. The (SIF) was prepared by dissolving KH2PO4 (6.8 g)
whole insulin content of the particles was deter- in 250 ml of deionized water. The obtained solu-
mined by the detection of insulin using HPLC. tion was mixed with NaOH (0.2 N, 77 ml), and
242 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

[+log]

7.1050

4.6924
4.5452
7.7399
7.5445

3.5709
3.4102
3.2386
3.0216
2.8457
2.7410
2.3376

1.2207
-N+(CH3)2
Ar-N(CH3)2
2

-N(CH3)2
Aromatic
1

Ar-N+(CH3)3
0

15.8236
0.2500
1.3551

1.5595

7.0671
7.1268
1.8566
4.7380
5.2231

9.7350

3.2931
10 8 6 4 2 [ppm]

Fig. (1). Proton nuclear magnetic resonance (H NMR) spectra of insulin nanoparticles. (A higher resolution / col-
our version of this figure is available in the electronic copy of the article).

finally, the volume was adjusted to 1000 ml using of sodium hydroxide (NaOH) and potassium dihy-
deionized water, while the pH was maintained at drogen phosphate (KH2PO4). Subsequently, the
6.8. The proper amount of lyophilized powder (i.e. pH was ascertained to 6.8 using a Sartorious® pH-
414.75 mg) equivalent to 70 mg of insulin was meter, and the samples were taken at 150, 195,
weighted and dispersed in the SGF by considering 240, 300, and 360 min (Fig. 1) [11].
the loading efficiency (LE %) and the amount of
2.2.7. Preparation of the Enteric-coated Capsules
cryoprotectant (i.e. 25 mg) for freeze-drying.
Throughout the study, the temperature was kept Before performing in vivo studies, the nanopar-
constant at 37 ± 1.0°C, and the release medium ticles were filled in the Capsugle® preclinical cap-
was gently agitated at 50 rpm using the USP bas- sules, and the capsules were then enteric-coated
ket apparatus I. Moreover, the experimental condi- using Eudragit-L100. Firstly, Eudragit L-100® was
tion was adjusted in a manner to ensure that the dissolved in double distilled water to produce a
sink condition is established. At the predetermined concentration of 5 mg/ ml. After filling each cap-
time intervals of 15, 30, 45, 60, 75, 90, and 120 sule with a proper amount of lyophilized powder,
min, one ml of the solution was sampled and re- the capsules were immersed in the Eudragit® solu-
placed by a freshly prepared and preheated medi- tion and then remained there for 2 min. Afterward,
um. Afterward, the samples were centrifuged at the capsules were removed and dried using hot air.
12,000 rpm for 20 min using a Beckman-Coulter
2.2.8. In Vivo Studies
Optima™ XPN-100 ultracentrifuge (Georgia,
United States). The supernatant was also collected In this study, 20 Male Wistar rats (220-250 g)
for insulin determination using the HPLC method. were housed under the controlled temperature
120 min post-incubation, the release medium was conditions and fed in terms of the laboratory ani-
changed to SIF by the addition of suitable amounts mal standard diet with water provided ad libitum.
Effect of Insulin Nanoparticles on Diabetes Pathogenesis Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 243

These animals were then divided into four groups Total antioxidant capacity (TAC) was measured
as follows (n=5): group1: normal control rats, using ferric reducing ability of plasma (FRAP)
group 2: diabetic rats, group 3: diabetic rats re- method. This procedure was performed based on
ceived pre-clinical capsules INPs (30U/kg/day, the strength of plasma in reducing Fe3+ to Fe2+ in
orally), group 4: the diabetic rats received regular the presence of TPTZ. The reaction of Fe2+ and
insulin (5U/kg/day, subcutaneously). TPTZ provided a complex with blue color and
maximum absorbance at 593 nm [14].
Before the study, the rats were rendered diabet-
ic by a single intraperitoneal (IP) injection of To evaluate the total thiol groups (TTG),
60 mg/kg of streptozocin (STZ) in an isotonic cit- DTNB was used as a reagent. In this regard, it is
rate buffer. The development of type 1 diabetes in noteworthy that DTNB reacts with thiol molecules
rats was confirmed by the estimation of fasting and creates a yellow complex with a good absorb-
blood glucose seven days after the STZ injection. ance at 412 nm [15].
The animals with fasting blood glucose levels
2.2.10. Estimation of Biochemical Parameters
above 250 mg/dl were considered diabetic [12].
This study design was approved by the Ethics Liver damage biomarkers (ALP, AST, ALT,
Committee Guidelines of Hamadan University of and total and direct bilirubin), glucose level, creat-
Medical Sciences (IR.UMSHA.REC.1395.440). inine, gamma glutamine transferase (GGT), and
Lyophilized nanoparticles were orally administered urea were measured in terms of the standard pro-
to rats using pre-clinical capsules (Chitoclear™ cedure of kits (Pars Azemon kit, Iran).
Austria). Also, the capsules used had been previ-
2.2.11. Liver and Kidney Histology
ously enteric-coated using Eduragit L-100” aque-
ous enteric system (Colorcon, Idstein, Germany). For histopathological assessments, a small
Subsequently, freeze-dried nanoparticles prepared piece of tissues was fixed with 10% formalin and
from amino benzyl chitosan were administrated at then embedded in paraffin. Moreover, the serial
an equivalent dose of insulin. After 28 days, the sections (thickness of 5 μm) were processed and
fasted rats were anesthetized with ketamine (50 stained with hematoxylin and eosin (H&E), ac-
mg/kg), and serum samples for biochemical anal- cording to the standard pathology laboratory pro-
yses were collected and then stored at -20°C. Also, tocols. Afterward, the stained liver and kidney
the liver and kidney tissues were separated and slices were evaluated under light microscopy.
cleaned with ice-cold saline solution, then some of 2.2.12. Statistical Analysis
their parts were transferred into cryotubes, kept in
liquid nitrogen for an hour, and finally stored at - All values are expressed as Mean ± SE. Statisti-
70°C until analysis. Notably, the remaining parts cal analysis was done using one-way ANOVA,
of the tissues were fixed in 10% neutral buffer and post hoc comparisons were carried out using
formalin for histopathological evaluation. the Tukey test. P>0.05 was considered as non-
significant difference, while P<0.05 was consid-
2.2.9. Estimation of Oxidative Stress Parameters ered as statistically significant.
To measure the rate of lipid peroxidation
3. RESULTS
(LPO), TBA was used, which reacts with lipid
peroxide molecules. The plasma samples were also The H-NMR spectra of the quaternized and
blended with TCA (20%), and the precipitate was aromatized chitosan polymer synthesized are
dispersed in H2SO4 (0.05 M). Subsequently, TBA shown in Fig. (1). Accordingly, the sharp peaks in
(0.2% in sodium sulphate 2M) was added and the regions of 2.3 and 2.7 ppm represent chitosan
heated for 30 min in a boiling water bath. TBARS methyl (-N (CH3)2) and (-N + (CH3)3) protons of
adducts were extracted using n-butanol, and the methyl aliphatic amines, respectively. Also, the
absorbance was measured as 532 nm. This reac- sharp peaks in the 3.4 and 3.6 ppm regions repre-
tion occurred in acidic pH on high temperature, sent the methyl (-N (CH3) 2) and (-N + (CH3)3) pro-
and the maximum absorption was observed for a tons of the aromatic amine attached to the benzyl
pink complex at 532 nm [13]. ring, respectively.
244 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

<Chromatogram>
mAU

PDA Multi 1 214nm,4nm

4.021
12.5

10.0

7.5

2.691
5.0

2.287
2.031
2.165
2.5

2.488
0.0

0 1 2 3 4 5
min
Fig. (2). Chromatogram of insulin with 5 μgr/ml. Insulin eluted from the column with an approximate retention time
of 4 minutes. (A higher resolution / colour version of this figure is available in the electronic copy of the article).

Table 1. The loading efficiency (LE%) and entrapment efficiency (EE%) of HPLC injection.

LE% Mean ± EE% Mean ±


LE1% LE2% LE3% EE1% EE2% EE3%
SD SD

18% 18.40% 17.50% 17.96% ± 0.45 62% 62.28% 61.83% 62.03% ± 0.22

Table 2. Physico-chemical characteristics of nanoparticles, before and after lyophilization.

- Before Lyophilization After Lyophilization

Size (nm) 138.2 ± 31 324.3 ± 22.94


Zeta potential (mV) +9.31 ± 1.18 +13.01 ± 1.1
Polydispersity 0.243 ± 0.07 0.345 ± 0.05

The HPLC chromatogram of insulin is depicted The in vitro release of insulin from the enteric-
in Fig. (2). As shown in the figure, insulin was coated capsules was studied in the Simulated Gas-
eluted from the column with an approximate reten- tric Fluid (SGF) adjusted to pH value of 2.2 and
tion time of 4 minutes. the Simulated Intestinal Fluid (SIF) adjusted to pH
value of 6.8. The release profile of different types
The LE and EE of INPs were 17.96% ± 0.45 of prepared nanoparticles is shown in Fig. (4).
and 62.03 ± 0.22, respectively. Also, the evalua- From minute 0 to 120, enteric-coated capsules
tion of shape by TEM in different magnifications with Eudragit L100 and containing INPs in SGF
confirmed the spherical shape of INPs (Table 1). were released with deficiency and only 8.35% of
The size, zeta potential, and polydispersity of insulin was released into the SGF after 120
INPs before and after lyophilization are presented minutes. Then, the capsules were transferred to
in Table 1. The results show that after lyophiliza- SIF, and 84.81% of insulin was released into the
tion, these parameters were increased further com- SIF after 240 minutes.
pared to before lyophilization; however, this in- The results of the size of quaternized INPs,
crease was not significant (p>0.05) (Fig. 3) (Table 2). their zeta characteristics, and the zeta potential
Effect of Insulin Nanoparticles on Diabetes Pathogenesis Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 245

100

80

Cumulative Release (%)


60

40

20

0
0 100 200 300 400

Time (min)
Fig. (3). Release of insulin from nanoparticles. The release of insulin in simulated gastric fluid (SGF) (0-120 min)
was 8.35% and this amount was 84.81% in simulated intestinal fluid (SIF) (120-360 min). (A higher resolution /
colour version of this figure is available in the electronic copy of the article).

Size (d.nm): % Intensity: St Dev (d.nm):

Z-Average (d.nm): 159.0 Peak 1: 191.1 100.0 44.17

PdI: 0.221 Peak 2: 0.00 0.00 0.00


Intercept: 0.737 Peak 3: 0.00 0.00 0.00

Size Distribution by Intensity

20
Intensity (Percent)

15

10

0
0.1 1 10 100 1000 10000
Size (d.nm)

Record 6: 94/12/15 s2 1

Fig. (4). Size distribution of insulin nanoparticles. (A higher resolution / colour version of this figure is available in
the electronic copy of the article).

indicate the average size of the insulin nanoparti- chitosan nanoparticles (20.000×, 80,000×, and
cles to be 159 diameter nanometer (d.nm) and pol- 125,000×, respectively).
ydispersity index (PDI) equal to 0.221 (Fig. 4).
As summarized in Table 3, diabetes caused sig-
Moreover, as shown in Fig. (5), the zeta potential
nificant weight loss in this study. The final body
for the insulin nanoparticles was +8.31 mV.
weight significantly decreased in the diabetic
Fig. (6) indicates the Transmission electron mi- group compared to the control group (p<0.05).
croscopy (TEM) of the insulin-loaded trimethyl However, the oral administration of the coated
246 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

Results
Mean (mV) Area (%) width (mV)
Zeta Potential (mV): 8.31 Peak 1: 8.31 100.0 4.27
Zeta Deviation (mV): 4.27 Peak 2: 0.00 0.0 0.00
Conductivity (ms/cm): 0.420 Peak 3: 0.00 0.0 0.00
Result quality: Good

Zeta Potential Distribution

500000

400000
Total Counts

300000

200000

100000

0
-200 -100 0 100 200
Zeta Potential (mV)

Record 85: 95/07/18 insu 1

Fig. (5). Monomodal graph of zeta potential of insulin nanoparticles. (A higher resolution / colour version of this
figure is available in the electronic copy of the article).

Fig. (6). Transmissionelectronmicroscopy (TEM) of insulin-loaded trimethyl chitosan nanoparticles. (20.000×,


80,000×, 125,000× respectively). (A higher resolution / colour version of this figure is available in the electronic
copy of the article).
Effect of Insulin Nanoparticles on Diabetes Pathogenesis Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 247

Table 3. Weight of Rat (g) in the studied groups.

Variable/Group C D D+N D+I

Initial body weight (g) 228.04 ± 9.3 230.3 ± 10.4 227.2 ± 9.2 224.2 ± 3.2
Final body weight (g) 274.8 ± 13.9 214.4 ± 11.32a 229.3 ± 6.7a,b 222.1 ± 8.3b
Mean weight level during treatment in different studied groups. Data are shown as Mean ± SE. C: healthy control. D: diabetic control. N: encapsulated insulin
nanoparticle (30U/kg). I: injectable insulin (5U/kg). a Significantly different compared to the control groups. b Significantly different compared to diabetic con-
trol groups. p<0.05.

compared to the healthy control rats (p<0.01).


Blood Glucose Level (mg/dl)

600 day1
a day7 While STZ enhances the level of MDA, the oral
a day28 treatment with the coated form (in serum p<0.01,
400
b kidney p<0.01 and liver p<0.05) as well as the
subcutaneous treatment with the non-coated form
200 of insulin (in serum p<0.01) could remarkably re-
a duce MDA (p<0.05). There were no substantial
differences between diabetic groups and diabetic
0 groups treated by the non-coated form of insulin in
C D D+N D+I the kidney and liver MDA levels (p>0.05)
Fig. (7). Fasting blood sugar level during treatment in (Fig. 8A).
different studied groups. Data are shown as Mean ±
SE. C: healthy control. D: diabetic control. N: encapsu- The comparisons of the TAC levels among all
lated insulin nanoparticle (30U/kg). I: injectable insulin groups are summarized in Fig. (8B). As shown,
(5U/kg). aSignificantly different compared to the con- diabetes resulted in a notable decrease in the TAC
trol groups. bSignificantly different compared to diabet- levels in serum, kidney, and liver tissues (p< 0.01).
ic control groups. p<0.05. (A higher resolution / colour The coated (in serum p<0.01, kidney p<0.01, and
version of this figure is available in the electronic copy liver p<0.05) and non-coated forms of insulin (in
of the article). serum p<0.01, kidney p<0.05, and liver p<0.05)
could relieve these oxidative markers with a sig-
insulin could significantly compensate for these nificant increase in the TAC level (p<0.05).
weight losses compared to the diabetic group
(p<0.05). Furthermore, STZ-induced T1D significantly
increased TOS levels in the treatment groups com-
FBS of the control group did not change up to pared to the control group (p<0.01), but treatment
the end of the treatment. On the other hand, after by encapsulated INP (in serum p<0.01, and kidney
the STZ injection, FBS in three diabetic groups p<0.05) and insulin (in serum p<0.05) remarkably
significantly increased compared to the healthy modulated the levels of TOS (Fig. 8C).
control group (p<0.05). Also, a significant
decrease was observed in blood glucose levels of In addition, the levels of OSI were significantly
diabetic groups receiving oral capsules containing higher in diabetic groups compared to the control
nanoparticles insulin and insulin-treated diabetic group (p<0.01). Also, treatment with encapsulated
group on day 28, compared to the control group INP (in serum p<0.01, kidney p<0.01, and liver
(Fig. 7). Notably, insulin treatment in a non-coated p<0.01) and insulin (in serum p<0.01, kidney
way, similar to the coated form with a quaternized p<0.01, and liver p<0.01) had a significant effect
INPs chitosan-based nanoparticle, could enhance on this level (Fig. 8D).
the blood glucose level (p<0.05). However, a sig- The levels of TTG decreased in diabetic group
nificant difference was observed between the in- compared to the control groups (p<0.01). How-
jectable insulin and the control rat (p<0.05). ever, encapsulated INPs significantly improved
The diabetic rats indicated an elevated lipid pe- TTG concentrations in serum, kidney, and liver
roxidation level in kidney, serum, and liver tissues (p<0.05), and insulin also showed a significant
248 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

(A) 4
(B) (C) Serum
Kidney

MDA (nmol / L serum or mg protein)


400 80 Liver
a#

TOS (nmol / L serum or mg protein)


TAC (nmol / L serum or mg protein)
a#
3 60
300
b* b* a#
b# b# b* a#
2 b# a# 40 b*
a# 200
a# b#
a# b* b#
b# b#
b#
1 100 20
a#

0 0 0

(D) (E) (F)

I
C
1.0

+
+

D
80

D
TTM (nmol / L serum or mg protein)
a#
0.8 b* 40 a#
60

Serum 8-oxo-dG (ng/ml)


OST (TOS/TAC)

0.6 30
a# b# b#
40 b*
0.4 a# b*
b# b* b* 20
a# b#
20 a#a#
0.2 b# b#
b# b# 10

0.0 0
0

I
N
D
D

+
I

C
C

I
N
+

D
+

C
D
+

+
D

+
D
D

D
D
Fig. (8). A: Lipid peroxidation, B: Total antioxidant capacity, C: Total oxidant status, D: oxidative stress index,
E: Total thiol groups in serum, kidney and liver in rats and F: Serum DNA damage. (A higher resolution / colour
version of this figure is available in the electronic copy of the article).
Data are presented as Mean ± SE.C: healthy control. D: diabetic control. N: encapsulated insulin nanoparticle
(30U/kg). I: injectable insulin (5U/kg),a Significant differences with the control group, b Significant differences with
the diabetic group. *p < 0.05, #p < 0.01.

effect on TTG level (in serum p<0.05 and kidney amelioration of these markers. The benefits of
p<0.05) (Fig. 8E). ALP and total and direct bilirubin reached the val-
ues of the healthy control group by subcutaneous
The results of DNA damage in serum are pre-
treatment with non-coated insulin.
sented in Fig. (8F). Induction of diabetes by STZ
caused a significant increase in DNA damage As shown in Fig. (10), the levels of creatinine,
(p<0.01), and treatment of diabetic group with in- urea, and GGT activity were significantly higher
sulin as well as encapsulated INPs significantly in the diabetic non-treatment group compared to
improved the serum DNA damage (p<0.01). the control group (p<0.05). Besides, it was found
that the treatment with insulin-coated trimethyl
As depicted in Fig. (9), the levels of ALP, AST,
chitosan-based nanoparticles and injectable insulin
ALT, and total and direct bilirubin were signifi-
could better ameliorate creatinine, urea, and GGT
cantly higher in the STZ non-treatment group
activity compared to the diabetic group (p<0.05).
compared to the healthy control group (p<0.05).
Moreover, it was found that the treatment with in- The diabetic rats showed a decreased urine cre-
sulin-coated trimethyl chitosan-based nanoparti- atinine level compared to the healthy control ani-
cles could significantly ameliorate ALT, ALP, and mals (p<0.05). The oral treatment with coated
total and direct bilirubin (p<0.05). However, no form and the subcutaneous treatment with a non-
remarkable differences were observed between the coated form of insulin could more increase urine
coated and the non-coated forms of insulin in the creatinine compared to the diabetic animals
Effect of Insulin Nanoparticles on Diabetes Pathogenesis Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 249

(A) (B) 250


(C) 3000 a
150
200 a

serum AST (U/L)

serum ALP (U/L)


a
serum ALT (U/L)
2000
100 150
b
b 100
1000 b
50
50

0 0
0

D
.I

.N
.N
C

.I
D

D
.N
C

.I
D

D
D
D
D
(D) 1.5
(E)
0.20
a
Total bilirubin mg/dl

Direct bilirubin mg/dl


1.0 a
0.15
b
b b
b 0.10
0.5
0.05

0.0 0.00
C

.I
.N

.I
.N
D

D
D

D
Fig. (9). Liver function factors in different studied groups. Data are shown as Mean ± SE. (A) Serum Alanine ami-
notransferase level, (B) Aspartate aminotransferase level, (C) Alkaline phosphatase level, (D) Total bilirubin, (D)
Direct bilirubin. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
C: healthy control. D: diabetic control. N: encapsulated insulin nanoparticle (30 U/kg). I: injectable insulin (5 U/kg),
a
Significantly different compared to the control groups. bSignificantly different compared to diabetic control groups.
p < 0.05.
(A) (B) (C)
1.0 40
5
a a a
0.8 b
CGT activity (unit/L)
Cratinine(mg/dl)

30 4
b
urea (mg/dl)

0.6
b b 3
20
0.4 2
b b
10 1
0.2

0
0.0 0
C D D+N D+I C D D+N D+I C D D+N D+I
(D) 80 (E)
400
b Healthy Control
Urin cratinin (mg/dl)

60 a
a 300 Diabetic
b
Alb (mg/dl)

40 200 b Diabetic + Insulin Nanoparticle


b
a
Diabetic + Injectable Insulin
20 100

0 0
C D D+N D+I C D D+N D+I

Fig. (10). Kidney function factors in different studied groups. Data are shown as Mean ± SE. (A) Urea level, (B)
Creatinine level, (C) γ-Glutamyl transferase (GGT) activity in serum and (D) Creatinine, and (E) Albumin (Alb) in
the urine. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
C: healthy control. D: diabetic control. N: encapsulated insulin nanoparticle (30 U/kg). I: injectable insulin (5 U/kg),
a
Significantly different compared to the control groups. bSignificantly different compared to diabetic control groups.
p < 0.05.
250 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

(p<0.05). However, significant differences were significant increase compared to the diabetic con-
seen in the urine creatinine levels between the in- trol group. There was no significant difference in
jectable insulin and the control rats (p<0.05). glomerular and proximal convoluted sizes among
the diabetic group and the healthy controls and the
Also, the levels of urine albumin showed an el-
other groups.
evation in the diabetic animals compared to the
healthy control animals (p<0.05). It was indicated In the diabetic group, distal tubule wide and
that the treatment with INPs and injectable insulin length have significantly decreased compared to
could decrease urine albumin compared to diabetic the healthy control group. It should also be noted
animals (p<0.05). that there was no substantial difference between
the other study groups and the healthy control
Liver tissues of all the control rats showed typi-
group; however, there was a significant increase in
cal lobular architecture with central veins, radiat-
the distal tubule wide and length compared to the
ing hepatic cords, the portal triads containing por-
diabetic control group (Figs. 11 and 12).
tal vein, hepatic artery, and bile duct. Notably, the
sinusoids are naturally in the rows of hepatocytes
4. DISCUSSION
with no dilatation, and the Kupffer cells are at-
tached to the sinusoidal wall. Diabetes mellitus is one of the main challenges
in the world that burdens society. Currently, in-
In the diabetic control group, dilated sinusoids
jectable insulin is the best choice for the treatment
were grown. Hepatocytes have been removed from
of diabetes mellitus. However, in addition to sev-
the radial cord, and the number and size of hepato-
eral side effects, this method of therapy has many
cytes decreased. Besides, the number of macro-
psychological consequences for diabetic patients.
phages and lymphocytes has significantly in-
Nowadays, nanotechnology plays an essential role
creased compared to the healthy control group. In
in the development of pharmaceutical strategies.
the diabetic group receiving capsules containing
The current study showed that encapsulated insu-
insulin nanoparticles, dilated sinusoids have in-
lin nanoparticles, as well as insulin, have a consid-
creased; however, hepatocytes maintained the ra-
erable effect on the maintenance of the body
dial cord state. Furthermore, there was a signifi-
weight, glucose homeostasis, liver and kidney tox-
cant decrease in the number of macrophages in
icity and oxidative stress. However, encapsulated
this group compared to the diabetic control group.
insulin nanoparticles were more effective in the
Moreover, there was a substantial increase in correction of diabetes pathogenesis.
the number of lymphocytes in this group compared
In recent years, ionic gelation and polyelectro-
to the healthy control group. In the diabetic group
lyte complexation (PEC) has been extensively
receiving injectable insulin, dilated sinusoids have
used to prepare the nanoparticles entrapping sensi-
increased. Also, hepatocytes were released from
tive molecules such as peptides and proteins. In
the radial cord. The number of macrophages and
these methods, procedures such as sonication and
lymphocytes counted in this group has decreased
the presence of organic solvents can be avoided,
compared to the diabetic control group and in-
thus reducing the possible damage to peptides and
creased compared to the healthy control group.
proteins. The PEC method can prepare nanoparti-
In the kidney tissues of the control group, the cles with a high EE%, zeta potential, and ionic ge-
proximal and distal convoluted tubules, renal cor- lation that can provide drug retention and eventu-
puscles, glomerulus, and glomerular capsule ally, higher bioavailability. Methylated N-(4-N,N-
showed their typical structures. Additionally, the dimethyl aminobenzyl) chitosan are considered a
urinary pole had a rational size. In addition, the polycation and a polyanion, respectively, which
urinary pole significantly decreased in the diabetic can form a polyelectrolyte complex and take part
group compared to the healthy control group. It in peptide and protein delivery [9]. Traditional in-
should also be noted that there was no considera- jectable insulin is the current treatment of diabetes;
ble difference between the other study groups and this procedure has caused a heavy burden on the
the healthy control group; however, there was a patient. Today, attempts are being made to use
Effect of Insulin Nanoparticles on Diabetes Pathogenesis Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 251

(A) (B) hp

hp Dilated
sinosoid

cv sn
cv kup

sn

kup

lym
20 pixels 20 pixels
lym
(C) (D)

hp

cv
sn
sn kup

cv
kup

hp

lym
20 pixels 20 pixels

Fig. (11). Histopathologic changes of the liver. Normal control rats (A), diabetic rats (B), diabetic rats that received
encapsulated insulin nanoparticle (30U/kg) (C), diabetic rats that received injectable insulin (5U/kg) (D) (40×).
(A higher resolution / colour version of this figure is available in the electronic copy of the article).

(A) (B) Distal


convoluted
tubules
Distal
Proximal convolated
convoluted Urinary pole
tubule
tubule

Glomerulus

Glomerular Urinary pole Progximal


convoluted
tubule

2.5 inches 2.5 inches

Progximal
(C) convoluted (D)
tubule
Necrotic Urinary pole

cell debris

Glomerular

Distal urinary pole


convoluted
Distal
tubule
Proximal convoluted
Necrotic
convoluted tubule
cell debris
tubule

Glomerular

2.5 inches 2.5 inches

Fig. (12). Histopathological changes in the kidney. A: Healthy control, B: Diabetic + encapsulated insulin nanopar-
ticles (30 U/kg), C: Diabetic group, D: Diabetic + injectable insulin (5 U/kg). (A higher resolution / colour version
of this figure is available in the electronic copy of the article).
252 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

alternatives for the treatment. One of these ways is indicated that INPs directly affect glucose metabo-
the oral administration of Eudragit 100 insulin lism, and increase the expression of glucokinase
[16]. In this work, Eudragit-coated quaternized and pyruvate kinase genes. These enzymes are the
aromatic derivatives of chitosan used for INPs key rate-limiting enzymes mediating the oxidation
preparation. The Eudragit 100 INPs, which were of glucose and generating ATP [8].
applied in the present study, were designed on the
In diabetic condition, hyperglycemia can also
basis of biocompatibility, insulin stability, and in-
induce toxic oxidative stress through various
testinal epithelial permeability. Therefore, to
mechanisms and decline antioxidant capacity. A
achieve this purpose and to increase its intestinal
high level of glucose inhibits mitochondrial respir-
absorption, the oral administration of insulin ne-
atory complex III and leads to the production of
cessitated the design of the nanoparticles, which
reactive oxygen species in the liver and other or-
would protect and facilitate its intestinal absorp-
gans. Oxidative stress also has a vital role in the
tion, and mimic its physiological rate [17]. It
pathology of diabetes [22]. In the present study,
seems that by the increased intestinal mucosal ab-
the oxidative stress due to diabetic rats was ap-
sorption of Eudragit 100 insulin, the hepatic glu-
proved via the significant increase in oxidative
coneogenesis, and the glycogenolysis effect of this
markers such as LPO, TOS, OSI and DNA dam-
hormone in the liver could notably progress [18].
age, and decrease in the antioxidant factors such as
In our study, the efficacy of the insulin ab- TTG and TAC level. Similar results were obtained
sorbed from the GI was evaluated by its effect on in the studies on animal models and human studies
the body weight, the FBS level, and the biochemi- [8, 20]. These results show that Eudragit L-100
cal and histopathological changes in the liver and capsules aromatize and quaternerize chitosan for
kidney tissues, and the oxidative biomarkers. Stud- insulin nanoparticle coating that can stimulate the
ies have shown that STZ decreased weight due to bioavailability of insulin and reduce oxidative
an impact on the pancreas and suppresses the pro- stress toxicity.
duction of insulin. Insulin promotes glycogen and
Hyperglycemia in the STZ groups induced liver
lipid synthesis in muscle cells. Therefore; reduc-
injury causing changes in liver enzyme levels and
tion in insulin production causes reduced entry of
secretion of soluble cytosolic enzymes into the cir-
glucose into muscle and increasing lipolysis and culation. Aminotransferase (ALT, AST and ALP),
gluconeogenesis can be causing muscle wasting
direct and total bilirubin, are the most sensitive
and weight loss [19]. The results of our study dem- and the most widely used enzymes to detect any
onstrate that treatment with INPS, induced weight liver injuries. INPs inhibit liver injury by its effect
gain in diabetic rats compared to the untreated di- on the improvement in liver histopathology. Our
abetic rat. Given that injectable insulin keeps the result validated the previous study that used INPs
concentration of insulin up for a short time, it has a for improving liver injury [8].
much lower effect on weight gain than oral INPs.
In line with our results, Heidarisasan et al., Diabetes mellitus is generally accompanied by
showed that INPs remarkably induced weight gain significant changes in kidney function, including
more effectively than injectable insulin [20]. increased creatinine, urea and GGT in serum and
decreasing the renal filtration of creatinine, urea in
STZ penetrates the pancreas and induces diabe- urine. In this study, INPs improved renal histo-
tes moderately by destroying the pancreatic beta pathology in diabetic rats. The results reveal that
cells, inducing DNA alkylation, reducing the pro- creatinine and urea and GGT serum in the diabetic
duction of insulin and increasing concentration of group have shown significant changes and in-
plasma glucose level. Our results showed that the creased the values of diabetic groups who received
fasting blood sugar of diabetic control rats was insulin (oral and SC) at normal levels. Also, in the
significantly increased, but in diabetic treatment diabetic group, the level of albumin is increased
with INPs, it was significantly decreased. Our result and they show a degree of proteinuria. On the oth-
validated the previous study that used insulin-incor- er hand, a few studies strongly support the out-
porated nanoparticles of chitosan and alginates to come of our study. In line with these findings, an-
decrease the glucose level [21]. Jamshidi et al. other study showed that treatment with insulin
Effect of Insulin Nanoparticles on Diabetes Pathogenesis Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 253

could reduce urea and creatinine concentration in with the animal Ethics Committee Guidelines of
the serum of STZ-diabetic rats. To support this Hamadan University of Medical Sciences, Ap-
notion, it has been reported that the use of Eu- proval no. IR.UMSHA.REC.1395.440.
dragit® RS 100 microparticles (ERS) containing
insulin (ILNP) in the alloxan-induced diabetic CONSENT FOR PUBLICATION
rabbits and sheep model [23] and the biopolymer-
ic- based delivery nanoparticulate system im- Not applicable.
proved disorders caused by type 1 and type 2 dia-
betes mellitus [24]. AVAILABILITY OF DATA AND MATERI-
ALS
CONCLUSION The authors confirm that the data supporting
Diabetes type 1 and type 2 cause kidney and the findings of this research are available within
liver complications. The results of this study the article.
showed that Eudragit L-100 capsules aromatize
and quaternerize chitosan for insulin nanoparticle FUNDING
oral delivery that can be effective in kidney and This study was financially supported by the
liver toxicity in diabetic rats. Results showed that Deputy of Research, Hamadan University of Med-
INPs reduced the liver and kidney injury and ame- ical Sciences (No. 9510146103).
liorated oxidative toxic stress in these tissues.
However, we need further studies to examine and CONFLICT OF INTEREST
verify the molecular pathways underlying the ef-
fects of insulin nanoparticles (INPs) in the liver The authors declare no conflict of interest, fi-
and kidney tissues of diabetic animal models. The- nancial or otherwise.
se data may contribute to the management and
treatment strategies of diabetic patients. ACKNOWLEDGEMENTS
Declared none.
AUTHORS’ CONTRIBUTIONS
In this study, all authors have contributed: Reza REFERENCES
Mahjub: analysis & revising of the manuscript, [1] Fröde TS, Medeiros YS. Animal models to test drugs
Farzane Kazemi Najafabadi: design & performance, with potential antidiabetic activity. J Ethnopharmacol
Narges Dehkhodaei: design & performance Nejat 2008; 115(2): 173-83.
http://dx.doi.org/10.1016/j.jep.2007.10.038
Kheiripour: analysis & performance, Amir Nili PMID: 18068921
Ahmadabadi analysis & drafting of manuscript, [2] Fonte P, Araújo F, Reis S, Sarmento B. Oral insulin
Sara Soleymani Asl: report of work & histology delivery: how far are we? J Diabetes Sci Technol
analysis, Masomeh Taheri -Azandariani, perfor- 2013; 7(2): 520-31.
mance of animal treatment, Akram Ranjbar de- http://dx.doi.org/10.1177/193229681300700228
PMID: 23567010
sign, analysis, report work and drafting and revis- [3] Mahjub R, Radmehr M, Dorkoosh FA, Ostad SN,
ing of the manuscript, must also be acknowledged. Rafiee-Tehrani M. Lyophilized insulin nanoparticles
prepared from quaternized N-aryl derivatives of chi-
ETHICS APPROVAL AND CONSENT TO tosan as a new strategy for oral delivery of insulin: in
PARTICIPATE vitro, ex vivo and in vivo characterizations. Drug Dev
Ind Pharm 2014; 40(12): 1645-59.
This research was approved by Animal Ethics http://dx.doi.org/10.3109/03639045.2013.841187
Committee Guidelines of Hamadan University of PMID: 24093431
[4] Cao SJ, Xu S, Wang HM, et al. Nanoparticles: oral
Medical Sciences, Hamadan, Iran.
delivery for protein and peptide drugs. AAPS
PharmSciTech 2019; 20(5): 190.
HUMAN AND ANIMAL RIGHTS http://dx.doi.org/10.1208/s12249-019-1325-z PMID:
31111296
No human were used in this study. Research [5] Prego C, Torres D, Fernandez-Megia E, Novoa-
work on animals was carried out in accordance Carballal R, Quiñoá E, Alonso MJ. Chitosan-PEG
254 Pharmaceutical Nanotechnology, 2020, Vol. 8, No. 3 Mahjub et al.

nanocapsules as new carriers for oral peptide delivery. http://dx.doi.org/10.1016/S0076-6879(94)33044-1


Effect of chitosan pegylation degree. J Control Re- PMID: 8015473
lease 2006; 111(3): 299-308. [15] Gergel’ D, Cederbaum AI. Interaction of nitric oxide
http://dx.doi.org/10.1016/j.jconrel.2005.12.015 with 2-thio-5-nitrobenzoic acid: implications for the
PMID: 16481062 determination of free sulfhydryl groups by Ellman’s
[6] Shateri H, Ranjbar A, Kheiripour N, et al. Tempol reagent. Arch Biochem Biophys 1997; 347(2): 282-8.
improves oxidant/antioxidant parameters in testicular http://dx.doi.org/10.1006/abbi.1997.0352
tissues of diabetic rats. Life Sci 2019; 221: 65-71. PMID: 9367537
http://dx.doi.org/10.1016/j.lfs.2019.02.016 PMID: [16] Chen S, Guo F, Deng T, et al. Eudragit S100-Coated
30738867 Chitosan Nanoparticles Co-loading Tat for Enhanced
[7] Ranjbar A, Kheiripour N, Ghasemi H, Seif Rabiei oral colon absorption of insulin. AAPS PharmSciTech
MA, Dadras F, Khoshjou F. Antioxidative effects of 2017; 18(4): 1277-87.
tempol on mitochondrial dysfunction in diabetic http://dx.doi.org/10.1208/s12249-016-0594-z PMID:
nephropathy. Iran J Kidney Dis 2018; 12(2): 84-90. 27480441
PMID: 29507270 [17] Reis CP, Ribeiro AJ, Veiga F, Neufeld RJ, Damgé C.
[8] Jamshidi M, Ziamajidi N, Khodadadi I, Dehghan A, Polyelectrolyte biomaterial interactions provide na-
Kalantarian G, Abbasalipourkabir R. The effect of in- noparticulate carrier for oral insulin delivery. Drug
sulin-loaded trimethylchitosan nanoparticles on rats Deliv 2008; 15(2): 127-39.
with diabetes type I. Biomed Pharmacother 2018; 97: http://dx.doi.org/10.1080/10717540801905165
729-35. PMID: 18293199
http://dx.doi.org/10.1016/j.biopha.2017.10.097 [18] Sharma S, Jyoti K, Sinha R, Katyal A, Jain UK,
PMID: 29102916 Madan J. Protamine coated proliposomes of recombi-
[9] Mahjub R, Dorkoosh FA, Amini M, Khoshayand MR, nant human insulin encased in Eudragit S100 coated
Rafiee-Tehrani M. Preparation, statistical optimiza- capsule offered improved peptide delivery and per-
tion, and in vitro characterization of insulin nanopar- meation across Caco-2 cells. Mater Sci Eng C 2016;
ticles composed of quaternized aromatic derivatives 67: 378-85.
of chitosan. AAPS PharmSciTech 2011; 12(4): 1407- http://dx.doi.org/10.1016/j.msec.2016.05.010 PMID:
19. 27287134
http://dx.doi.org/10.1208/s12249-011-9716-9 PMID: [19] Wilcox G. Insulin and insulin resistance. The Clinical
22033812 biochemist. Reviews 2005; 26(2): 19-39.
[10] Keiripour N, et al. The effects of synthetic orally ad- [20] Heidarisasan S, Ziamajidi N, Karimi J, Abbasali-
ministrated insulin nanoparticles in comparison to in- pourkabir R. Effects of insulin-loaded chitosan-
jectable insulin on the renal function markers of type alginate nanoparticles on RAGE expression and oxi-
1-diabetic rats. Iran J Basic Med Sci 2020; 23(6): dative stress status in the kidney tissue of rats with
810-8. type 1 diabetes. Iran J Basic Med Sci 2018; 21(10):
[11] Mahjub R, Dorkoosh FA, Amini M, et al. In vitro 1035-42.
characterization and cell cytotoxicity of insulin nano- PMID: 30524677
particles composed of quaternized aromatic deriva- [21] Sahu SK. Development and evaluation of insulin in-
tives of chitosan. J Control Release 2013; 172(1): corporated nanoparticles for oral administration.
e112. ISRN Nanotechnology 2013; 2013: 1-6.
http://dx.doi.org/10.1016/j.jconrel.2013.08.270 [22] Kheiripour N, Karimi J, Khodadadi I, Tavilani H,
[12] Qinna NA, Badwan AA. Impact of streptozotocin on Taghi Goodarzi M, Hashemnia M. Hepatoprotective
altering normal glucose homeostasis during insulin effects of silymarin on liver injury via irisin upregula-
testing in diabetic rats compared to normoglycemic tion and oxidative stress reduction in rats with type 2
rats. Drug Des Devel Ther 2015; 9: 2515-25. diabetes. Iran J Med Sci 2019; 44(2): 108-17.
http://dx.doi.org/10.2147/DDDT.S79885 PMID: 30936597
PMID: 26005328 [23] Olya S, et al. Oral delivery of insulin-loaded nanopar-
[13] Benzie IF, Strain JJ. Ferric reducing/antioxidant pow- ticles in diabetic rabbits and in sheep. Czech J Anim
er assay: direct measure of total antioxidant activity Sci 2014; 59: 251-6.
of biological fluids and modified version for simulta- http://dx.doi.org/10.17221/7495-CJAS
neous measurement of total antioxidant power and [24] Lopes M, Aniceto D, Abrantes M, et al. In vivo bio-
ascorbic acid concentration. Methods Enzymol 1999; distribution of antihyperglycemic biopolymer-based
299: 15-27. nanoparticles for the treatment of type 1 and type 2
http://dx.doi.org/10.1016/S0076-6879(99)99005-5 diabetes. Eur J Pharm Biopharm 2017; 113: 88-96.
PMID: 9916193 http://dx.doi.org/10.1016/j.ejpb.2016.11.037 PMID:
[14] Hu ML. Measurement of protein thiol groups and 28007370
glutathione in plasma. Methods Enzymol 1994; 233:
380-5.

You might also like