SlideShare a Scribd company logo
1 of 11
Download to read offline
Carbohydrate Polymers 157 (2017) 1065–1075
Contents lists available at ScienceDirect
Carbohydrate Polymers
journal homepage: www.elsevier.com/locate/carbpol
Hydrophilic polymeric nanoparticles prepared from Delonix
galactomannan with low cytotoxicity for ocular drug delivery
Abayomi T. Ogunjimia,b
, Shaiani M.G. Meloa
, Carem G. Vargas-Rechiaa
, Flávio S. Emerya
,
Renata F.V. Lopeza,∗
a
University of São Paulo, School of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto, SP, Brazil
b
Department of Pharmaceutics, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
a r t i c l e i n f o
Article history:
Received 12 July 2016
Received in revised form 20 October 2016
Accepted 24 October 2016
Available online 25 October 2016
Keywords:
Cellular uptake
Cytotoxicity
Delonix polymeric nanoparticles
Ocular delivery
Quality by design
a b s t r a c t
Delonix is a galactomannan polysaccharide extracted from the endosperm of Delonix regia plant. This
study aims at the development of Delonix nanoparticle and assesses its potential for ocular delivery by
evaluating its in-vitro stability, toxicity and cellular uptake. Fluorescent nanoparticles (BODIPY-loaded
nanoparticles) were prepared by a Quality-by-Design modified nanoprecipitation technique. Optimized
nanoparticles had mean sizes <240 nm, PdI < 0.2 and zeta potential of <−30 mV. Mixture of surfactants
with different hydrophilic-lipophilic balance controlled nanoparticle swelling. Nanoparticles, which were
stable in the presence of simulated lachrymal fluid and lysozyme also sustained the release of BODIPY.
In-vitro studies suggest no toxicity of the nanoparticles in concentration range of 100–1483.3 ␮g/mL
on retinal and corneal epithelial cells. Flow cytometry and confocal microscopy techniques showed that
retinal cells but not corneal cells, uptake 18% of the nanoparticles. Therefore, Delonix nanoparticles could
be a safe and promising tool for ocular drug delivery.
© 2016 Elsevier Ltd. All rights reserved.
1. Introduction
Drug delivery for the treatment of various eye diseases has been
a challenge due to the critical micro-environment that exists in the
eye. Nanoparticulate drug delivery has shown promising results in
ocular drug delivery to both the anterior and posterior parts of the
eye in the last two decades. However, the quest for drug delivery
tools that could target diseases of the eye, improve bioavailability
and particularly minimize non-specific toxicity has not abated (Dey
& Mitra, 2005; Sahoo, Dilnawaz, & Krishnakumar, 2008; Achouri,
Alhanout, Piccerelle, & Andrieu, 2013).
Interest in biodegradable polymeric nanoparticles as potential
ocular drug delivery tools has increased due to their applications as
DNA carriers for gene therapy and their ability to deliver proteins,
peptides and genes (Zuo & Lee, 2008). Synthetic and semi-synthetic
polymers such as poly lactic acid (PLA), poly glycolic acid (PGA),
poly lactic acid-co-glycolic acid (PLGA), ethyl cellulose and hydrox-
ypropyl methyl cellulose (HPMC) have been widely studied as
∗ Corresponding author at: School of Pharmaceutical Sciences of Ribeirão Preto,
University of São Paulo, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil.
E-mail address: rvianna@fcfrp.usp.br (R.F.V. Lopez).
delivery agents via the ocular route (Kumari, Yadav, & Yadav, 2010;
Elzoghby, El-Fotoh, & Elgindy, 2011; Anderson & Shive, 2012).
However, attracting more interest in nano-delivery are the nat-
ural hydrophilic polymers due to their accomplished properties
of biocompatibility, affordability, little or no toxicity, availability
and non irritant nature (Huang & Fu, 2010). In addition to mucoad-
hesion, hydrophilic polymers generally swells and form a gel-like
layer on their particle surface which retards fluid ingress into the
polymer core thereby sustaining drug release(Sarmah, Mahanta,
Bhattacharjee, Mahanta, & Biswas, 2011).
Several natural polymers including alginate, guar gum and
chitosan have been studied as ocular nano delivery agents
(Nagarwal, Kant, Singh, Maiti, & Pandit, 2009; Bhowmik et al., 2013;
Gratieri et al., 2010). Nanoparticle preparation with some of these
hydrophilic polymers involve the use of toxic cross-linking agents
such as glutaraldehyde that could pose toxicity concerns to liv-
ing cells and tissues. Chitosan, a cationic polysaccharide and its
derivatives with favorable biological properties has been exten-
sively studied in topical ocular delivery of drugs either individually
or as a surface modifier for other polymers (Gratieri et al., 2010;
de la Fuente et al., 2010; Nagarwal, Kumar, & Pandit, 2012). How-
ever, there exist other natural polysaccharides with little or no
information in literature, but with potentials for ocular delivery.
http://dx.doi.org/10.1016/j.carbpol.2016.10.076
0144-8617/© 2016 Elsevier Ltd. All rights reserved.
1066 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075
One of such polymer is Delonix polymer (DLX), a galactomannan
polysaccharide (Supplementary material, Fig. S1.1) extracted from
the endosperm of Delonix regia (Bojer ex Hook.) plant (Kapoor,
1972; Tamaki, Teruya, & Tako, 2010). Delonix regia or flamboyant
is a leguminous plant of the family Fabaceae native to Madagascar
but freely growing in diverse countries around the world today. The
plant is widely used as an ornamental while its seed, highly rich in
a polymeric galactomannan are sometimes used as beads. The use
of the seed polymer as a drug delivery tool, in addition to being a
green approach, could also be of economic benefit for low income
communities, where the plant is inhabited.
Delonix and its modified derivatives have previously been
studied as a sustained release polymer for tablets (Adetogun
& Alebiowu, 2007; Adetogun & Alebiowu, 2009), mucoadhesive
(Devkar, Tekade, & Khandelwal, 2014) and microencapsulation
agent (Betancur-Ancona, Pacheco-Aguirre, Castellanos-Ruelas, &
Chel-Guerrero, 2011). Our laboratory recently commenced studies
on DLX as a controlled nano-delivery agent with ocular deliv-
ery prospects. Unlike some natural polymeric nanoparticles whose
process of production involve the use of toxic cross-linking agents
like glutaraldehyde (Soumya, Ghosh, & Abraham, 2010), we have
designed a simple method for the production of stable DLX
nanoparticles without the use of such toxic cross-linking agents.
Thus, the present study aims at developing a method for the
preparation and characterization of a DLX nanoparticle formula-
tion for ocular delivery. The study also evaluates the encapsulation
prowess of DLX nanoparticle using hydrophilic and lipophilic probe
molecules, while assessing its potential for ocular delivery by eval-
uating its stability in simulated lachrymal tear and enzymes, in vitro
cellular toxicity and uptake.
2. Materials and methods
2.1. Materials
DLX extracted and purified in our laboratory; absolute ethanol
and neutral red (NRU) dye from Merck, Germany; acetone, dimethyl
sulfoxide, polyoxyethylene (20) sorbitan monooleate (tween
80) and [2-(3,4-dihydroxyoxolan-2-yl)-2-hydroxyethyl] octadec-
9-enoate (span 80) from Synth, Brazil; mucin from porcine stomach
Type III, ITS liquid media supplement, hydrocortisone, cholera toxin
A, lysozyme from chicken egg white and 3-(4,5-dimethylthiazol-
2-yl)-2,5-diphenyltetrazolium bromide (MTT) dye from Sigma-
Aldrich, USA; fetal bovine serum (FBS) from Invitrogen, USA;
Dulbecco’s modified Eagles medium (DMEM), Hams F12 (F12), gen-
tamycin and DAPi molecular probe from Life technologies, New
Zealand; Supplemented Hormonal Epidermal Medium (SHEM)
prepared in our laboratory, hydrophilic and lipophilic BODIPY syn-
thesized by our collaborators (Supplementary material, Figs. S2.1,
S2.2, S2.3, S2.4), Retina Pigmented Epithelium cell line (ARPE-19,
BCJR code: 0041) from the Cell Bank in Rio de Janeiro, Brazil and
Immortalized Human Corneal Epithelial Cell line (HCE SV-40) were
all the materials used in this work.
2.2. Collection and purification of polymer
DLX was collected manually from mature and ripe pods (fruit)
of Delonix regia plant within the vicinity of University of Sao Paulo,
Ribeirao Preto campus. The seeds from the pods were split open
and the polymeric endosperm removed, dried (50 ◦C) in oven and
milled. The milled unpurified DLX mass was redispersed in suffi-
cient distilled water for 48 h, filtered under negative pressure with a
Buchner funnel to remove all undissolved materials and then pre-
cipitated with absolute ethanol. The precipitate was filtered and
washed twice with acetone, oven dried at 50 ◦C, pulverized, sieved
(250 ␮m sieve-size) and stored until needed.
2.3. Characterization of DLX polymer
2.3.1. General analysis
Total carbohydrate content of the DLX polymer was performed
by the phenol-sulfuric acid assay method (Dubois, Gilles, Hamilton,
Rebers, & Smith, 1956) while protein content was performed
by Bradford assay (Bradford & Williams, 1977) method using
Coomassie Brilliant Blue G 250.
2.3.2. Monosaccharide composition
Monosaccharide composition was determined using DLX poly-
mer obtained in Section 2.2 (DLXp), DLX polymer recovered from
sonicated polymer solution described in Section 2.4.1 (DLXs) and
DLX polymer recovered from polymer solution described in Section
2.4.1 but without sonication (DLXu). The DLX polymer was recov-
ered by precipitation using ethanol, followed by filtration, drying
at 50 ◦C and pulverization.
The DLXp, DLXs and DLXu were completely hydrolyzed with
2 mol/L trifluoroacetic acid (TFA, 5 h, 100 ◦C). The hydrolyzed
polymers were reduced with sodium borohydride for 2 h and
the alditols were acetylated with pyridine:acetic anhydride
(1:1 v/v, 18 h) at room temperature. The resulting alditol acetates
were analyzed by gas-liquid chromatography/mass spectrometry
(GCMS-QP2010 Ultra − SHIMADZU) using ZB-5MS (30 m x 0.25 mm
x 0.25um) Zebron-Phenomenex column.
2.3.3. Methylation analysis: DLXs and DLXu
The polysaccharides (5 mg) were methylated according to
the established methods (Ciucanu & Kerek, 1984). Each per-O-
methylated polysaccharide was pre-treated with 72% v/v H2SO4
(1 h, 4 ◦C) and water was added to a final acid concentration of 8%
(18 h) at room temperature. The hydrolyzed polymer were reduced
with acetylated borohydride and analyzed by CGMS as described
above (section 2.3.2).
2.3.4. 13C and 1H spectroscopy
All NMR spectra were obtained using a Bruker 400 MHz Avance
III 14.1 T spectrometer equipped with an inverse 5-mm pentanu-
clear resonance probe head (QXI) at 303 K. Chemical shifts were
expressed in ␦ (ppm) relative to the resonance of DSS (sodium
4,4-dimethyl-4-silapentane-l-sulphonate) and methanol used as
an internal standard (∼ = 0) for 1H and for 13C respectively.
2.3.5. Determination of Molecular weight (Mw)
The average Mw of DLXs and DLXu polysaccharides were
determined by size exclusion chromatography (GPC), with a
Sepharose CL–4 B chromatographic column (43 × 1.8 cm, Mw range
of 3 × 104–5 × 106) and water as the mobile phase set at a flow rate
of 0.6 mLmin−1. Eluted samples were monitored with a differen-
tial refractometer (model R401, Waters). Sugars were identified by
comparing the retention times shown in the peaks of DLXs and
DLXu samples with those of dextran standard samples.
2.4. Nanoparticles preparation
2.4.1. Preparation of DLX polymer solution
DLX polymer solution was prepared by dispersing required
amount of DLX polymer (Table 1) in 0.5 M NaOH for 24 h to ascer-
tain complete dissolution/dispersion. This dispersion medium was
chosen based on results of some preliminary experiments (Sup-
plementary material, Section S3). The solubilized polymer was
centrifuged and filtered (15000 x g, 20 min; 0.8 ␮m micropore fil-
ter) to remove undissolved matter. The mildly viscous polymer
A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1067
Table 1
Box Behnken design for DLX polymeric nanoparticle formulation.
Coded variables
Formulation X1 X2 X3
F1 −1 0 1
F2 1 1 0
F3 1 0 1
F4 −1 1 0
F5 1 −1 0
F6 0 0 0
F7 −1 0 −1
F8 0 1 1
F9 0 0 0
F10 −1 −1 0
F11 1 0 −1
F12 0 −1 −1
F13 0 0 0
F14 0 −1 1
F15 0 1 −1
Coded variables Uncoded variables −1 0 1
X1 Polymer concentration (%) 1.0 1.5 2.0
X2 Surfactant mix ratio (tween 80:span 80) 1:0 4:1 3:2
X3 Non-solvent: solvent ratio 10 15 20
filtrate was sonicated (20 Hz, 40% amplitude; VCX 500, Sonic &
Materials, Inc, USA) for 30 min and the solution stored in refrig-
erator for at least 24 h before use.
2.4.2. Design of Experiment (DoE)
To adequately control critical parameters that could influence
the DLX nanoparticle properties, a Quality-by-Design, modified
nanoprecipitation approach was employed. The potential factors
that could impart the desired quality of the DLX nanoparticle were
assessed by deploying a Fishbone diagram (Supplementary mate-
rial, Fig. S3.1) in creating a structure that could identify such factors.
Based on carefully researched knowledge space and some initial
basic experiments, we arrived at three probable factors − poly-
mer concentration, surfactant mix ratio and non-solvent/solvent
ratio − that could influence the desired DLX nanoparticle prop-
erties. For example, polymer concentration was chosen based on
the knowledge space available; increase in polymer concentration
generally lead to increase in viscosity, which consequently produce
nanoparticles with larger sizes. In addition, inclusion of tween 20
led to the formation of a viscous dispersion which was difficult
to manipulate while surfactant concentration had no significant
influence on DLX nanoparticle size in initial experiments leading
to the choice of 5% surfactant concentration for all experiments.
Surfactant mix ratio was selected based on some initial experi-
ments which showed that the use of only tween 80 produced DLX
nanoparticle sizes in the range of 300 nm with PdI > 0.3, while the
use of only span 80 produced DLX nanoparticles with sizes ranging
between 600 and 700 nm but PdI < 0.2. The inclusion of poloxamer
(P407) only produced DLX nanoparticles sizes in range of 250 nm
but with PdI > 0.4. Nonsolvent-solvent ratio is mostly important in
terms of economics of solvent, however, it was included as a fac-
tor because most studies that discussed nonsolvent-solvent ratio
influence on nanoparticle size as far as we know involved the use
of synthetic polymers. As such, our polymer being hydrophilic and
actively swelling might produce a different response (Supplemen-
tary material, Section S3). Each of these factors were tested at three
levels using a completely randomized 15-experiment Box Behnken
statistical design (Table 1). The influence of these process variables
on selected DLX nanoparticle properties of hydrodynamic diame-
ter, polydispersibility index (PdI) and zeta potential was evaluated.
Analysis of Variance (ANOVA) was performed to indentify statis-
tically significant factors while general linear model was used for
parameter estimate.
2.4.3. Preparation of blank DLX nanoparticle
DLX nanoparticles were prepared by a modified nanoprecip-
itation process using absolute ethanol as the non-solvent. The
non-solvent solution was first prepared by adding necessary vol-
ume of 5% surfactant mix (tween 80 and/or span 80) to absolute
ethanol based on a Box Behnken statistical design (Table 1) and
mixing for 10 min under moderate magnetic stirring. Exactly 1 mL
of DLX solution was added to the non-solvent mix under mag-
netic stirring (1000 rpm, TECNAL magnetic mixer, Brazil) and at
a polymer flow rate of approximately 1 mL/min. The mildly tur-
bid nanoparticle dispersion was allowed to stand for about 30 min
under reduced magnetic stirring speed. The mixture was then cen-
trifuged at 15,000g for 10 min (Thermo Scientific Heraeus Megafuge
16R, ThermoFisher Scientific, USA), the supernatant decanted and
the surface of the nanoparticle washed twice with distilled water.
The nanoparticles were redispersed in distilled water and passed
through a 0.45 ␮m syringe filter before evaluation. The nanopar-
ticles were evaluated by hydrodynamic diameter, PdI and zeta
potential. Experiments were performed in triplicate.
2.5. DLX nanoparticle characterization
2.5.1. Microscopy, size distribution and zeta potential analysis
Nanoparticle size and distribution were measured using 1 mL
of syringe-filtered DLX nanodispersion by dynamic light scatter-
ing (DLS) using a Nano ZS (Malvern Zetasizer nano ZS90 series)
at 25 ◦C and a scattering angle of 90◦. Zeta potential was deter-
mined by laser doppler micro-electrophoresis technique with the
same instrument (Malvern Zetasizer nano ZS90 series). Results are
reported as average of triplicate determinations. The DLX nanopar-
ticles were also characterized by transmission electron microscopy
(TEM), using a JEM-100CXII transmission electron microscope
(JEOL, Japan) with an accelerating voltage ranging from 100 kV to
200 kV.
2.5.2. Surfactant mix influence on DLX nanoparticle size
Based on results obtained from Box Behnken experimental
design, three separate DLX nanoparticle formulations were pre-
pared using 1% (w/v) DLX polymer solution, non-solvent/solvent
ratio of 10, but varying surfactant mix ratio (tween 80:span 80;
1:0, 4:1, 3:2). DLX nanoparticle size, PdI and zeta potential were
evaluated and average of quadruplicate experiments reported.
2.5.3. Surfactant mix influence on DLX nanoparticle
swelling/stability
The formulations described in Section 2.5.2 above were sub-
jected to swelling/stability analysis by measuring the nanoparticles
size, PdI and zeta potential of the nanoparticle formulation daily
for six days. Results were recorded as percent size increase while
experiment were performed in quadruplicates.
2.5.4. Statistical analysis
Results obtained were subjected to appropriate statistical anal-
ysis including Response Surface Regression (Statistica 10, StatSoft,
USA), One-Way and Two-Way ANOVA (GraphPad Prism 5.0, Graph-
Pad, USA).
2.6. 2.6. In vitro DLX nanoparticle stability in simulated ocular
fluids
2.6.1. Incubation with lysozyme
The stability of optimized DLX nanoparticles was analyzed
according to established methods (de Campos, Diebold, Carbalho,
1068 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075
Sánchez, & José Alonso, 2005) following their incubation at 37 ◦C
in a solution of lysozyme in purified water (1 mg/mL) under mod-
erate stirring. The mean particle size, PdI and zeta potential were
monitored during the incubation process. Average of quadruplicate
determinations were recorded.
2.6.2. Incubation in simulated tear fluid (STF)
The stability of optimized DLX nanoparticles was analyzed in STF
containing 0.67% w/v NaCl, 0.2% w/v NaHCO3 and 0.008% w/v CaCl2
2H2O (Miyazaki et al., 2001). The mean particle size, PdI and zeta
potential were monitored during the incubation process at 35 ◦C.
Results recorded were average of quadruplicate determinations.
The osmolarity of DLX nanoparticle dispersion was also determined
using an osmometer (Semi-Micro Osmometer K-7400, KNAUER).
2.6.3. Incubation with mucin
The zeta potential of DLX nanoparticle was also evaluated in
mucin (0.4 mg/mL) dispersion. Mucin, like lysozyme is a major
component of the precorneal fluid. The zeta potential of mucin
dispersion with and without DLX nanoparticles at 35 ◦C was evalu-
ated. Values reported are average of quadruplicate determinations.
Results obtained were subjected to ANOVA (GraphPad Prism 5.0,
GraphPad, USA).
2.7. BODIPY-loaded DLX nanoparticle
Hydrophilic and lipophilic BODIPY dyes (Supplementary mate-
rial, section S2) were used as test molecules. To prepare loaded
DLX nanoparticle, 10 mg of lipophilic or hydrophilic BODIPY were
initially dissolved in the non-solvent medium or DLX solution
respectively before nanoparticle preparation as described in sec-
tion 2.4.3 above. The preparation was based on the selected optimal
DLX nanoparticle formulation determined from the Box Behnken
statistical design (DLX nanoparticle containing tween-span ratio
3:2). The nanoparticles were evaluated by size, PdI and zeta poten-
tial. Results are average of triplicate determinations.
The entrapment efficiency (EE) of BODIPY-loaded DLX nanopar-
ticles was determined by ultracentrifugation (15,000g, 10 min,
Thermo Scientific Heraeus Megafuge 16R, ThermoFisher Scientific,
USA) and the amount of free BODIPY in the supernatant was mea-
sured spectrophotometrically at an absorbance of 495 nm. The EE
was calculated indirectly as described in Eq. (1).
EE (%) =
Theoretical BODIPY amount − BODIPY in supernatant
Theoretical BODIPY amount
× 100 (1)
2.8. In vitro release studies
In vitro release studies of BODIPY-loaded DLX nanoparticles
were carried out in Franz diffusion cells under magnetic stirring
using dialysis membrane (12000 − 14000 Mw) as the diffusion bar-
rier/membrane and distilled water as the receiver medium. Exactly
1 mL of BODIPY-loaded DLX nanoparticles (containing ≈ 550 ␮g/mL
of BODIPY) was added to the donor compartment respectively
while BODIPY (≈550 ␮g/mL) in DLX solution was used as control.
All experiments were performed protected under dark condition at
room temperature. At specific times, 1 mL of the receiver medium
was removed and immediately replaced with 1 mL of fresh dis-
tilled water. Amount of BODIPY at each evaluation time in the
receiver medium was quantified using UV spectroscopy (wave-
length = 495 nm). For this study, only the hydrophilic BODIPY was
used while experiment was performed in triplicate. Kinetic mod-
eling was performed using DDSolver Excel Add-in software (Zhang
et al., 2010).
2.9. In vitro cell toxicity studies
In vitro cytotoxicity assay was carried out using two ocular
cell lines namely Retinal Pigment Epithelial (ARPE-19) and Human
Corneal Epithelial (HCE) cells.
The ARPE-19 cells were routinely cultured in DMEM/F12
medium (1:1) supplemented with 10% FBS and antibiotics while
the HCE cells were cultured in SHEM made of DMEM:F12 (1:1) and
supplemented with 5% FBS, amphotericin B, ITS, DMSO, hydrocorti-
sone, EGF, cholera toxin and gentamycin. The cells were incubated
in standard culture conditions (incubation at 37 ◦C in 5% CO2 atmo-
sphere) and the medium was change every 2 − 3 days while
cell growth was monitored daily by phase-contrast microscopy.
Cells were allowed to reach at least 80% confluence before each
tripsinization process.
2.9.1. Cytotoxicity assay
Cytotocixity was carried by MTT and NRU assay techniques.
The ARPE-19 and HCE cells were seeded at a density of 1.5 × 105
cells/well in 100 ␮L of their respective complete growth medium
into 96-well plates and incubated (37 ◦C in a 5% CO2 atmosphere)
for 24 h for adherence. The growth medium was thereafter removed
and replaced with 100 ␮L blank DLX nanoparticle dispersed in
DMEM:F12 (1:1) supplemented with 1% FBS in concentration range
of 100 − 1483.3 ␮g/mL and were incubated for 24, 48 and 72 h. After
the specified hours of incubation, the test nanoparticle dispersion
was removed, cells were washed with 150 ␮L of PBS and incubated
with 100 ␮L of NRU (final concentration of 0.05 mg/mL) or 100 ␮L
of MTT (final concentration of 5 ␮g/mL) in incomplete DMEM:F12
(1:1) respectively for 3 h. The cells were washed with 150 ␮L of
PBS and completely decanted before addition of 100 ␮L DMSO, for
MTT assay or 150 ␮L of neutral red desorb solution (1:50:49 v/v of
glacial acetic acid/ethanol/water) for NRU assay. The plates were
agitated for 10 min before absorbance was read at 570 nm (MTT)
and 540 nm (NRU) in a microplate reader (Cary 50 MPR, VARIAN
Inc., USA). Cell viability (%) was calculated as described in Eq. (2)
Cellviability (%) =
Abssample
Abscontrol
× 100% (2)
where Abssample represent measurements from wells treated with
DLX nanoparticles and Abscontrol measurement from untreated
wells. Results presented as mean ± SD were from quadruplicate
determinations. Results obtained were subjected to statistical anal-
ysis to determine significant differences between controls and
treated wells using ANOVA (GraphPad Prism 5.0, GraphPad, USA).
2.10. In vitro cellular uptake studies
Cellular uptake studies of BODIPY-loaded DLX nanoparticle
were performed in ARPE-19 and HCE cell lines using the hydrophilic
BODIPY. Flow cytometry, a laser based microfluidic cell analysis
technology for cell counting and biomarker detection was deployed
to evaluate/quantify the cellular uptake of the labeled DLX nanopar-
ticles by the ARPE-19 and HCE cells while confocal microscopy
technique was further used to affirm the results obtained from the
flow cytometry.
2.10.1. Flow cytometry
The ARPE-19 and HCE cells were seeded into a 6-well plate at
a density of 3 × 105 cells/well and 4 × 105 cells/well in respective
DMEM:F12 (1:1) complete medium and incubated (37 ◦C in a 5%
CO2 atmosphere) for 24 h for the cells to adhere. The cells were
thereafter washed with PBS and incubated with 1 mL of BODIPY-
loaded DLX nanoparticle (corresponding to 5 mg/mL of BODIPY)
dispersed in incomplete DMEM:F12 (1:1) medium for 24 h. After
24 h, the cells were washed twice with PBS, tripsinized with 300 ␮L
A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1069
of tripsin (0.25%), neutralized with respective DMEM:F12 (1:1)
complete medium, centrifuged and resuspended in PBS. Prior to
analysis in the flow cytometer (BD FACSCanto I, USA), 5 ␮L of pro-
pidium iodide was added to the samples. Analysis were performed
at excitation wavelengths of 495 nm and 488 nm with emission
wavelengths at 525 nm and 670 nm respectively for BODIPY and
propidium iodide. Percent uptake were calculated based on the dot
plots (BD-FACSDiva Software) obtained from the flow cytometry
result. Results reported were average of quadruplicate determina-
tions.
2.10.2. Confocal microscopy
The ARPE-19 and HCE cells were seeded into a 6 well plates
containing sterilized glass slips (22 mm x 22 mm) at a density of
3 × 105 cells/well and 4 × 105 cells/well in respective DMEM:F12
(1:1) complete medium and incubated (37 ◦C in a 5% CO2 atmo-
sphere) for 24 h for adherence. The cells were washed with PBS
and incubated for 24 h with 1 mL BODIPY-loaded DLX nanopar-
ticle (corresponding to 5 mg/mL of BODIPY) or BODIPY solution
(5 mg/mL) dispersed in DMEM:F12 (1:1) incomplete medium. Con-
trol wells were treated with blank DLX nanoparticle dispersion.
After treatment, the cells were washed twice with PBS, fixed with
2 mL paraformaldehyde (1%) per well and stored at 4 ◦C overnight
protected from light. The cells were washed again with PBS and the
glass slips were carefully lifted and placed inverted on glass slides
containing a drop of DAPi. The slides were kept for 24 h protected
from light before visualization in confocal microscope (Leica TCS
SP8 Confocal microscope, Leica, Germany). It should be noted that
the HCE cells were also incubated with loaded nanoparticles and
BODIPY solution for 48 h in addition to 24 h experiment.
3. Results and discussion
3.1. DLX polymer characterizaiton
The result shows that the carbohydrate content of DLXp was
about 54% with trace amount of protein. However, DLXs used in
the preparation of DLX nanoparticles and DLXu contains 99 and
98% sugars respectively; the sugars were predominantly mannose
(Man) and galactose (Gal) (Supplementary material, Table S4.1).
The Man:Gal ratio of DLXp, DLXs and DLXu was 4.6:1, 6.3:1 and
5.3:1 respectively (Supplementary material, Table S4.1); this result
is in accordance with previous studies on DLX polymer (Bento et al.,
2013). The difference between the amount of galactose present in
DLXp and DLXs may be due to a beta elimination process in the
presence of alkali (NaOH), a known phenomenon associated with
alkali treatment of galactomannans (Bento et al., 2013). While the
difference between the Man:Gal ratio of DLXs and DLXu may be
attributed to the sonication process (Prajapat & Gogate, 2015), frac-
tions of DLXs and DLXu analyzed by GPC showed a polydispersed
polymer system with close molecular weight of ∼170 and 185 kDa
respectively.
Methylation analysis of the galactomannan from DLXs and
DLXu were performed by a sequence of procedures in order
to obtain fully O-methylated derivatives (Sassaki, Gorin, Souza,
Czelusniak, & Iacomini, 2005). Hydrolysis of per-O-methylated
derivatives produced mixtures of partially methylated monosac-
charides: 2,3,4,6-Me,-Man, 2,3,4,6-Me,-Gal, 2,3,6-Me3-Man and
2,3-Me2-Man (Supplementary material, Fig. S4.6). These results are
consistent with a main chain of (1–4)-linked mannopyranosyl units
substituted at O6 of galactopyranosyl units. The galactomannans 1H
NMR (Supplementary material, Figs. S4.2 and S4.3) and 13C NMR
(Supplementary material, Figs. S4.4 and S4.5) spectroscopy anal-
ysis showed that resonances in the 13C NMR spectra are in close
agreement with those reported for other galactomannans (Bento
Table 2
Hydrodynamic diameter, PdI and zeta potential of DLX polymeric nanoparticles.
Data shown are mean ± SD of 3 replicates.
Formulation Hydrodynamic
diameter (nm)
PdI Zeta potential (mV)
F1 215.4 ± 3.8 0.192 ± 0.022 −38.5 ± 6.1
F2 315.2 ± 9.6 0.189 ± 0.004 −40.3 ± 8.1
F3 321.9 ± 11.2 0.215 ± 0.026 −62.5 ± 4.1
F4 252.7 ± 8.8 0.150 ± 0.031 −37.9 ± 4.3
F5 357.7 ± 8.5 0.214 ± 0.006 −65.2 ± 5.0
F6 276.3 ± 13.1 0.185 ± 0.008 −67.7 ± 6.4
F7 231.3 ± 5.8 0.140 ± 0.006 −39.9 ± 4.1
F8 266.7 ± 4.4 0.149 ± 0.020 −37.2 ± 5.9
F9 276.3 ± 13.1 0.185 ± 0.008 −67.7 ± 6.4
F10 225.7 ± 3.9 0.162 ± 0.009 −59.7 ± 8.5
F11 318.8 ± 10.6 0.175 ± 0.007 −68.8 ± 8.2
F12 260.1 ± 10.0 0.157 ± 0.016 −31.8 ± 3.1
F13 276.3 ± 13.1 0.185 ± 0.008 −67.7 ± 6.4
F14 261.1 ± 15.2 0.225 ± 0.021 −39.8 ± 2.4
F15 270.3 ± 12.5 0.176 ± 0.006 −49.1 ± 8.0
et al., 2013). The 1H- and 13C NMR spectral also demonstrated a
close relationship in the structure of DLXs and DLXu, signifying
minimal alteration due to sonication. The signals at the anomeric
region of 1H NMR (Supplementary material, Figs. S4.2 and S4.3)
indicates that the mannopyranosyl residues are ␤-linked while the
galactopyranosyl residues are ␣-linked. The DLXu O-substituted
resonances at C4 showed two clear peaks. The more abundant peak
at ı 76.3 can be attributed to two contiguous unsubstituted O-
mannopyranosyl residues while the peak at ı 77.59 may be ascribed
to the superimposition of signals from diads, where only one of
the two mannose residues is substituted. The same results were
observed for DLXs, indicating an irregular distribution of Gal in
the side chain of DLX galactomannan (Supplementary material, Fig.
S4.6).
3.2. DLX nanoparticle physicochemical properties
From the perspective of formulation design, various process and
formulation variables can impart the product outcome of a nano-
precipitation process to different extents, depending particularly
on the interaction of the variables. Thus, the effects of the selected
variables on the quality attributes of the DLX nanoparticle were
examined. Table 2 shows the nanoparticle size, PdI and zeta poten-
tial of the DLX nanoparticle formulations based on the DoE (Table 1).
The DLX nanoparticle formulations showed mean particle size
between 215 and 360 nm (Table 2) which is adequate for oph-
thalmic applications (Vega et al., 2008). Particle size distribution
was narrow in all cases as PdI was less than 0.230 indicating a
system that is not very polydispersed. Zeta potential result shows
that all nanoparticle formulations carry a negative charge smaller
than −30 mV, indicating the presence of an adequate particle stabil-
ity due to repulsive forces existing between particles (Veronovski,
Andreozzi, La Mesa, & Sfiligoj-Smole, 2010).
Neutral galactomannans like DLX in solution tends to have
slightly negative values confirming their neutrality (Carneiro-da-
Cunha, Cerqueira, Souza, Teixeira, & Vicente, 2011). The highly
negative zeta potential of our DLX nanoparticles dispersion may
be due to the addition of NaOH in DLX solution used to prepare the
nanoparticles. Increase in the pH of a solution have been shown to
have a significant influence on the zeta potential of nanoparticles
(Carneiro-da-Cunha et al., 2011). DLX solution containing NaOH
resulted in a dispersion with a pH higher than 7 which imparts a
negative zeta potential on the nanoparticle.
Thus, we successfully prepared nanoparticles of hydrophilic
Delonix polymer using a simple and innovative nanoprecipitation
process by using surfactant mix to control nanoparticle properties
and without the use of toxic cross-linking agents such as glutaralde-
1070 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075
hyde. This simple method could also be deployed in the preparation
of other hydrophilic polymers of natural origin.
Regression analysis and general linear modeling were per-
formed on DLX nanoparticle size result with a view to
understanding the relationship existing between the variables and
nanoparticle size. With a 15-run Box Behnken design, all individ-
ual variable effects and two-way interactions among the effects are
estimable while the three-way interaction is used to estimate the
error. Thus, the statistical model in Eq. (3) was used
y = ˇo + ˇ1X1 + ˇ2X2 + ˇ3X3 + ˇ4X1X2 + ˇ5X1X3 + ˇ5X2X3 + ε (3)
where y is the response (DLX nanoparticle size), X1, X2 and X3
(predictors) are polymer concentration, tween-span ratio and non-
solvent/solvent ratio respectively, ␤o is the mean response when
all variables are zero and ␤1-5 represent change in mean response
per unit change in predictors respectively.
From the regression analysis result (Supplementary material,
Table S5.1), a general linear model for predicting the DLX nanopar-
ticle size was derived from significant predictors as in Eq. (4).
y = 275.0533 + 48.5625X1 − 17.3750X1X2 (4)
Based on Eq. (4), it is easy to see that polymer concentra-
tion significantly influenced the nanoparticle size (Supplementary
material, Table S5.1). The parameter ␤1 estimated is also posi-
tive, indicating that a higher polymer concentration will produce
larger nanoparticle size. This result is in agreement with other
studies as higher polymer concentration produce highly viscous
solution which hampers the diffusion of solvent towards the non-
solvent in a nanoprecipitation process (Bilati, Allemann, & Doelker,
2005; Dong, Chang, Wang, Tong, & Zhou, 2015). Nanoprecipitation
involves the diffusion of one solvent into the other. It should be
noted that the nanoprecipitation process in this study is a reverse
of nanoprecipitation involving synthetic polymers such as PLGA as
the DLX polymer was solubilized in the aqueous phase while the
nanoparticles were formed in a less polar (ethanol) phase.
The surfactant mix ratio and the non-solvent/solvent ratio did
not significantly influence the DLX nanoparticle size as previously
described for other nanoparticles prepared by the nanoprecipi-
tation method (Ali & Lamprecht, 2013; Bilati et al., 2005). The
importance of non-solvent/solvent proportions in nanoprecipita-
tion processes is in solvent volume control (economics) (Bilati et al.,
2005). However, there was an interacting influence of polymer con-
centration and surfactant mix ratio on the DLX nanoparticle size
(Eq. (4)). This effect trend is elucidated in the surface response plot
in Fig. 1A.
The surface plot shows that smaller DLX nanoparticle size
are achieved at lower polymer concentration and surfactant mix
ratio containing higher span 80 proportion. This result could be
attributed partly to the low viscosity of polymer solution at lower
concentrations and probably the orientation and proportions of the
tween 80 and span 80 in the nanoparticle dispersion.
Based on this result, we propose an hypothesis based on the
orientation of tween and span in the nanoparticle dispersions.
Hydrophilic polymers such as DLX polymer have the tendency to
swell when in contact with aqueous medium. Thus, in the pres-
ence of only or higher amounts of tween 80 (hydrophilic-lipophilic
balance (HLB) = 15) in the surfactant mix, DLX nanoparticle could
have direct contact with the aqueous medium, leading to ‘minute’
swellings that could increase size; however, with increase in span
80 (HLB = 4.3) ratio, the orientation of span 80 could reduce or
prevent DLX nanoparticle contact with the dispersion medium,
leading to reduction in swelling and consequently its size. A prob-
able schematic representation of DLX nanoparticle prepared with
a mixture of tween80:span80 is shown in Fig. S6.1 (Supplementary
material, Section S6). In addition, other authors also proposed that
changes in nanocapsule diameter with respect to tween and span
Table 3
Surfactant mix influence on size of DLX nanoparticle containing different tween-
span ratios. Data shown are the mean ± SD of 4 replicates.
Tween 80:Span 80 Hydrodynamic
diameter (nm)
PdI Zeta Potential
1:0a,b
232.8 ± 3.6 0.185 ± 0.005 −64.6 ± 8.7
4:1b,c
215.4 ± 3.6 0.193 ± 0.003 −66.5 ± 3.9
3:2a,c
203.6 ± 6.1 0.156 ± 0.006 −58.9 ± 4.4
One-Way ANOVA with Tukey’s posttest:
a
p < 0.001.
b
p < 0.01.
c
p < 0.05.
concentration could be dependent on surfactant packing parame-
ters (Khoee & Yaghoobian, 2009; Mitchell & Ninham, 1981).
To affirm the influence of tween-span ratio on DLX nanoparticles
size and proposed swelling hypothesis for differences in nanopar-
ticle size, DLX nanoparticles were prepared and swelling/stability
study was performed. Table 3 shows the size of the DLX nanoparti-
cles as a function of 3 different tween-span ratios. Results subjected
to ANOVA indicate significant differences in nanoparticle size of
the three formulations; with the formulation containing higher
proportion of span 80 having the smallest nanoparticle size.
Fig. 1B shows surfactant mix influence on DLX nanoparticle size
increase as a function of time. It can be observed that DLX nanopar-
ticle containing higher amount of span 80 (tween-span ratio 3:2)
has a lower percent size increase as compared to the other nanopar-
ticle formulations. A 2-Way ANOVA further indicate a significant
difference in the percent size increase of the nanoparticle formula-
tion, with nanoparticles containing tween-span ratio of 3:2 having
the lowest percent size increase after 6 days. Therefore, the DLX
nanoparticle formulation containing 3:2 tween-span ratio was cho-
sen to prepare the DLX nanoparticles for all subsequent studies.
Nanoparticle size (tween 80:span80, 3:2) based on TEM photo
(Fig. 1C) analyzed with Fiji (ImageJ, USA) shows that DLX nanopar-
ticle size ranges between 28 and 38 nm in size. The difference in
the DLS and TEM sizes may be attributed partly to the DLS eval-
uation being based on intensity determination of hydrodynamic
diameter while TEM based on numbers. Also the hydrophilic and
swelling nature of the DLX polymer might have influenced the size
difference observed.
3.3. Stability of DLX nanoparticle in the presence of simulated
lachrymal components
The osmolarity of tear fluid is very important as it dictates the
integrity of the cornea. The normal osmolarity of normal tears
varies between 290 and 310 mOsm/kg (Marques, Loebenberg, &
Almukainzi, 2011). The osmolarity of DLX nanoparticle dispersion
determined using an osmometer (Semi-Micro Osmometer K-7400,
KNAUER) was 220 mOsm/L, which is not hyperosmolar.
The stability of colloidal biomaterials in biological fluids con-
taining ample amount of enzymes and proteins is very crucial. Also,
size of particles could play an important role in interaction with
mucosal surfaces including ocular mucosa. To evaluate the stabil-
ity of DLX nanoparticle in the presence of ocular fluid components,
DLX nanoparticles were incubated with lysozyme, SLF and mucin.
Lysozyme and mucin are two major components of the precorneal
fluid.
The particle size and zeta potential of DLX nanoparticle upon
incubation with or without lysozyme was evaluated as a function
of time (Fig. 2).
Fig. 2 shows that nanoparticle size reduced upon incubation
with lysozyme as compared to control without lysozyme. The size
reduction could be attributed to the partial hydrolysis of the gly-
cosidic bond in DLX polysaccharide structure which could result
A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1071
Fig. 1. (A) Surface Plot of hydrodynamic diameter against DLX concentration and tween span ratio (B) Surfactant mix (tween 80:span 80) influence on DLX nanoparticle% size
increase in function of time (2-Way ANOVA with Bonferroni multiple comparison test: a
p < 0.001; b
p < 0.01; c,d
p < 0.05) (C) TEM photo of DLX nanoparticle (mag: 200,000).
Fig. 2. Influence of lysozyme on DLX nanoparticle size and zeta potential incubated
in lysozyme as a function of time.
in minute break-off on the surface of the nanoparticle (de Campos
et al., 2005; Hu, Chen, & Zhang, 2010). This result also suggests
the biodegradability of the polymer; however, there was minimal
nanoparticle size reduction (13%) over the period of 6 h. The size
reduction of DLX nanoparticles in the presence of lysozyme can
also be due to some electrostatic interactions between the negative
nanoparticles and the cationic enzyme. Indeed, there was a signif-
icant reduction in the zeta potential of the nanoparticles (Fig. 2),
however the reduction would not adversely affect the nanoparti-
cle stability as the values were larger than −30 mV. This stability
is an advance in comparison to poly-␧-caprolactone nanocapsules
which aggregate upon contact with lysozyme (Calvo, Vila-Jato, &
Table 4
Physicochemical properties of DLX nanoparticle incubated with simulated lachry-
mal fluid.
DLX nanoparticle size (nm) Zeta potential (mV)
Time (h) Control In SLF Control In SLF
0 209.6 ± 13.5 211.4 ± 15.5 −45.0 ± 2.2 −5.4 ± 0.5
1 206.6 ± 9.2 204.5 ± 14.6 −47.5 ± 3.4 −5.9 ± 0.3
2 203.0 ± 12.3 205.0 ± 8.9 −42.0 ± 0.8 −6.3 ± 1.3
3 207.5 ± 8.9 206.2 ± 8.5 −48.6 ± 1.7 −6.0 ± 0.7
4 205.1 ± 10.1 205.4 ± 13.3 −43.7 ± 1.9 −6.2 ± 0.4
5 204.8 ± 11.1 206.9 ± 10.0 −43.0 ± 0.9 −6.2 ± 0.9
6 203.4 ± 8.0 205.4 ± 7.7 −45.9 ± 2.7 −6.1 ± 0.7
Alonso, 1997; de Campos et al., 2005). Furthermore, the PdI was
not affected by the presence of lysozyme.
Table 4 shows the influence of SLF on DLX nanoparticles size
and zeta potential as a function of time. The SLF has no signifi-
cant effect on the nanoparticle size and PdI, however there was a
significant reduction in the zeta potential of the DLX nanoparticle.
This is understandable as the presence of mono and multivalent
ions in the SLF could lead to compression of the electrical dou-
ble layer and consequently a reduction in the negative charge on
the nanoparticle surface (McConaughy, Stroud, Boudreaux, Hester,
& McCormick, 2008; Carneiro-da-Cunha et al., 2011). Despite the
reduction in zeta potential, no alterations in the size and in the
PdI of nanoparticles were observed, confirming their stability in a
physiological medium. This is an important advantage compared
to chitosan nanoparticles, that are instable in phosphate buffer pH
7.4 (de Campos et al., 2005).
The addition of mucin to DLX nanoparticle dispersion did not
significantly change (p > 0.05) the zeta potential of the dispersion
(DLX nanoparticles −42.3 ± 2.8 mV; DLX nanoparticle dispersion
with mucin −38.1 ± 0.3 mV); however, there was a significant
difference (p < 0.05) between the zeta potential of the mucin
1072 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075
dispersion (-15.5 ± 0.5 mV) as compared to that containing DLX
nanoparticle. Interactions between nanoparticles and mucin in
terms of surface charge could be an indirect way of predict-
ing mucoadhesion. Although, there was no significant interaction
between the DLX nanoparticle and mucin, biopolymers mucoad-
hesion to surfaces is a multi-mechanism process which does not
only involve interactions between surface charges. Adsorption
and diffusion have also been proposed as possible mechanism
for mucoadhesion. While it might be interesting to conduct an
in vivo ocular study to evaluate mucoadhesion prowess of DLX
nanoparticles in addition to its probable mechanism of mucoadhe-
sion (Smart, 2005; Serra, Domenech, & Peppas, 2009), study have
already reported its use as a mucoadhesion enhancer (Devkar et al.,
2014).
3.4. BODIPY-loaded DLX nanoparticle
Table 5 shows properties of BODIPY-loaded DLX nanoparticle
demonstrating the possibility of loading hydrophilic and lipophilic
molecules into DLX nanoparticles. Nanoparticle size was < 240 nm,
PdI was < 0.2 while zeta potential values was < −30 mV affirm-
ing stability. However, blank nanoparticles (215.5 ± 4.6 nm) were
smaller than the loaded DLX nanoparticles (hydrophilic BODIPY:
232.8 ± 11.1 nm; lipophilic BODIPY: 222.8 ± 9.1 nm). Entrapment
efficiency of lipophilic BODIPY into DLX nanoparticle was signif-
icantly low (≈ 6%) as compared to the hydrophilic BODIPY (≈ 69%).
The entrapment efficiency can be explained by the partition coef-
ficient (Log Poctanol/water) of the hydrophilic (-4.93) and lipophilic
(-0.19) BODIPY. The Log Poctanol/water was calculated using a free
MarvinSketch software (ChemAxon, Hungary). Although, the Mar-
vinSketch software used in determining the LogPocatanol/water do
have some limitations, particularly in relation to the structure of the
BODIPYs as it did not take into account the core of the structure,
thereby, making the values calculated only relative and ‘illustra-
tive’; however, we have demonstrated that the lipophilic BODIPY
is insoluble in water as shown in section 7 (Fig. S7.1) of the Supple-
mentary material.
In comparison to free lipophilic BODIPY which is obviously
insoluble in aqueous medium, lipophilic BODIPY-loaded DLX
nanoparticle homogeneously dispersed in aqueous medium (Sup-
plementary material, Fig. S7.1). Thus, despite the encapsulation
efficiency of lipophilic BODIPY in DLX nanoparticles being consid-
erably low, the loaded DLX nanoparticles homogenously dispersed
in aqueous medium.
In vitro release from BODIPY-loaded DLX nanoparticles and
BODIPY-DLX solution was studied. BODIPY-loaded DLX nanopar-
ticles showed a burst release of 48.2 ± 6.84% while free BODIPY
in DLX solution had a 97.4 ± 17.5% release within the first 30 min
respectively. The BODIPY-loaded DLX nanoparticles released
69.5 ± 13.9% of BODIPY after 24 h as compared to the BODIPY solu-
tion release of 97.4 ± 17.5% within the first 30 min. Hydrophilic
polymers are known to generally swell and form a gel-like layer
on their particle surface which retards fluid ingress into the poly-
mer core thereby sustaining drug release (Sarmah et al., 2011). This
result suggests that DLX nanoparticle exhibit significant sustained
release (Supplementary material, Fig. S7.2) in comparison to BOD-
IPY solution. The result also suggest that BODIPY release from DLX
nanoparticles followed a Higuchi model as indicated by r2 value of
0.9745 (DDSolver software).
3.5. In vitro cell toxicity
Determination of cell viability is a well recognized assay to eval-
uate in vitro cytotoxicity of nano-materials. Cell viability in this
study was assessed using neutral red and MTT uptake assays. The
MTT assay is a rapid colorimetric and quantitative method based
on the conversion of a yellow tetrazolium salt to insoluble purple
formazan crystals in the mitochondrial of viable cells while neu-
tral red is a dye taken up by viable cells and internalized within
the lysosomes (Bouallagui, Han, Isoda, & Sayadi, 2011). The combi-
nation of these two assay methods could have a benefit of adding
reliability to cytotoxicity results of tested materials.
A nanoparticle system with prospect for ocular use must be
capable of delivering optimum amount of active agents without
compromising the viability of the host cells. The result of MTT
and NRU assays (Supplementary material, Fig. S8.1) suggest that
DLX nanoparticles are non-toxic on both ARPE-19 and HCE cells at
all tested concentrations (100–1483.3 ␮g/mL) within 24 h. ARPE-
19 showed reduction in cell viability after 48 and 72 h, but the
reduction was only significant (p < 0.05) at DLX nanoparticle con-
centration of 1483.3 ␮g/mL. Both MTT and NRU assay produced
comparable results, giving reliability to the study. The HCE showed
reduced cell viability after 48 and 72 h but the reduction was sig-
nificantly different at 1483.3 ␮g/mL (p < 0.05) after 48 h; after 72 h,
there was significant difference in cell viability at 1009 (p < 0.05)
and 1483.3 ␮g/mL (p < 0.001). The MTT and NRU assay on HCE cells
suggest same results.
Based on the result, it could be said that in vitro DLX nanoparticle
use on ARPE-19 and HCE cells did not show obvious toxic effects
within the concentration limits in this study, however, more studies
would be needed to ascertain its in vivo toxicity.
3.6. Cell uptake
Cellular uptake is an important prerequisite for nanoparticle
effectiveness and physicochemical characteristics such as size,
surface charge, hydrophobicity dictates the extent of uptake of
nanoparticles by cells. For uptake to occur, it is considered that
there is an adhesion process followed by internalization. Cellular
uptake of nanoparticle apart from dictating effectiveness, could
also give insight on the toxicity propensity of nanoparticles (Zhao
et al., 2011). Two methods were employed to assess the uptake of
BODIPY-loaded DLX nanoparticles by ARPE-19 and HCE cell lines;
flow cytometry and confocal microscopy. Fig. 3A shows flow cytom-
etry results for hydrophilic BODIPY-loaded DLX nanoparticles in
contact with ARPE-19 and HCE cells.
Flow cytometry result as depicted by the dot plots in Fig. 3A2, Q4,
suggests a 17.45 ± 1.14% uptake of BODIPY-loaded DLX nanoparti-
cles by the ARPE-19 cells after 24 h of incubation while there is
almost no uptake of the nanoparticles by the HCE cells. The low or
no cellular uptake by the HCE cells could be due to the morpholog-
ical tight junctions conformation of the cells after adherence and
confluence (Yi, Wang, & Fu-Shin, 2000; Imayasu, Shiraishi, Ohashi,
Shimada, & Cavanagh, 2008; Kimura, Teranishi, Fukuda, Kawamoto,
& Nishida, 2008).
Fig. 3B shows confocal micrographs of ARPE-19 cells incubated
with BODIPY solution, blank DLX nanoparticle and BODIPY-loaded
DLX nanoparticle. Confocal micrographs of HCE cells treated with
these same formulations are shown in Supplementary material,
Figs. S9.1 and S9.2.
Confocal micrographs show a minimal uptake of BODIPY (faint
green) by ARPE-19 or HCE cells treated with BODIPY solution
(Fig. 3B5 and Supplementary material, Fig. S6.2 B2) as compared
to the ARPE-19 cells treated with BODIPY-loaded DLX nanoparticle
which showed bright color (Fig. 3B8) of the BODIPY after 24 h. Con-
focal result of BODIPY-loaded DLX nanoparticle uptake by ARPE-19
did corroborate the results obtained from the flow cytometry
experiment (Fig. 3A). Studies have also suggested nanoparticles
possess effective cellular uptake when compared with their free
drug solutions (Ahn, Seo, Kim, & Lee, 2013). Thus, the DLX nanopar-
ticle improved the uptake of BODIPY by the ARPE-19 cells in
comparison to control BODIPY solution, implying a possibility of
A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1073
Table 5
Properties of BODIPY-loaded DLX nanoparticle containing tween-span ratio 3:2. Data shown are mean ± SD of 3 determinations.
DLX nanoparticle Hydrodynamic diameter (nm) PdI Zeta Potential (mV) Encapsulation efficiency (%)
Blank 215.5 ± 4.6 0.178 ± 0,046 −61.7 ± 8.2 –
Loaded with hydrophilic BODIPY 232.8 ± 11.1 0.185 ± 0.009 −69.7 ± 7.5 68.6 ± 5.7
Loaded with lipophilic BODIPY 222.8 ± 9.1 0.132 ± 0.013 −65.6 ± 3.2 6.04 ± 1.62
Fig. 3. (A) Flow cytometry analysis: Dot plots showing uptake by ARPE-19 and HCE cells incubated for 24 h with BODIPY-loaded DLX nanoparticles (2 and 4) and blank DLX
nanoparticles (1 and 3). (B) Confocal micrographs of ARPE-19 cells incubated for 24 h with blank DLX nanoparticles (1–3), 5 mg/mL BODIPY solution (4–6) and BODIPY-loaded
DLX nanoparticles (7–9). Cell nuclei are stained blue with DAPi; BODIPY solution or BODIPY-loaded DLX nanoparticles appears green. (x 63 mag with immersion oil). Scale
bar: 20 ␮m.(For better interpretation of this figure legend in colour, the reader is referred to the web version of the this article).
using DLX nanoparticle as a potential drug carrier for retina related
ocular disorders. Furthermore, differences between the uptake of
BODIPY in solution and in DLX nanoparticles suggest that the dye
visualized inside the cells are not due to only passive diffusion
of BODIPY from the carrier. It is also important to consider the
hydrophilicity of the BODIPY used in this experiments, which can
hamper its passive redistribution. Therefore, there is a high possi-
bility for the dye observed inside the cells being a result of the DLX
nanoparticle uptake. This uptake noticed even after 24 h of experi-
ment suggests that DLX nanoparticles may be useful for a sustained
delivery of hydrophilic drugs in the posterior region of the eye.
Confocal result also shows no uptake of BODIPY-loaded DLX
nanoparticle by HCE cells after 24 h (Supplementary material, Fig.
S9.1 B1). Treating the cells with BODIPY-loaded DLX nanoparticle
for 48 h (Supplementary material, Fig. S9.2 B3) did not improve
uptake either, thus corroborating the result of flow cytometry per-
formed with the HCE cells. However, there was a minute uptake
of the BODIPY solution by the cells after 48 h treatment (Fig. S9.2
1074 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075
B2). The low uptake of BODIPY from solution and no uptake from
BODIPY-loaded nanoparticles could be due to the morphological
tight junctions of the HCE cells as previously mentioned (Imayasu
et al., 2008; Kimura et al., 2008; Yi et al., 2000). It could also be due
to BODIPY metabolism as a function of the longtime of experiment.
Therefore, if nanoparticles were taken up after 5 to 15 min incuba-
tion, as shown in other study using HCE cells (Pretor et al., 2015),
the probability of BODIPY degradation might be higher after cellular
uptake. Although, there was no significant uptake by the HCE cells,
in vitro release study in this work previously demonstrated the abil-
ity of DLX nanoparticle to sustain the release of an encapsulated
drug molecule. Thus it can be said that DLX polymer’s mucoadhe-
sive (Devkar et al., 2014) and sustained release ability could favor
its use topically in the eye, thereby increasing residence time and
also sustaining drug release.
4. Conclusion
This study demonstrated the possibility of obtaining nanopar-
ticles from a natural hydrophilic polymer, DLX, using a simple
and innovative modified nanoprecipitation technique. A gen-
eral linear model for predicting the DLX nanoparticle size was
proposed and showed that polymer concentration and the inter-
action between polymer concentration and tween-span mix ratio
influence the nanoparticle size. Based on the predicted model,
nanoparticles composed of optimized concentration of DLX and
surfactant mix ratio were prepared. These DLX nanoparticles were
stable when incubated in SLF, lysozyme solution and mucin dis-
persion, all components being present in the precorneal region of
the eye. Nanoparticles were able to encapsulate both lipophilic and
hydrophilic test molecules. In vitro release studies showed that
DLX nanoparticle sustained the release of hydrophilic BODIPY. The
ARPE-19 and HCE cells did not show obvious cellular toxicity to
DLX nanoparticles after 24 h of treatment. While there was no visi-
ble uptake of hydrophilic BODIPY-loaded DLX nanoparticles by HCE
cells in the time the experiment was performed, DLX nanoparticle
improved uptake of BODIPY by ARPE-19 cells. This study concluded
that DLX nanoparticle could be a promising tool for ocular drug
delivery.
Competing financial interests
The authors declare no competing financial interests.
Acknowledgements
This work was supported by the São Paulo Research Founda-
tion (FAPESP grant #2014/22451-7) and The World Academy of
Science-Brazilian National Council for Scientific and Technological
Development (TWAS-CNPq grant #190007/2013-3).
We also thank and acknowledge Guilherme L. Sassaki (PhD)
from the NMR Center of the Universidade Federal do Parana, Parana,
Brazil for analytical support.
Appendix A. Supplementary data
Supplementary data associated with this article can be found, in
the online version, at http://dx.doi.org/10.1016/j.carbpol.2016.10.
076.
References
Achouri, D., Alhanout, K., Piccerelle, P., & Andrieu, V. (2013). Recent advances in
ocular drug delivery. Drug Development and Industrial Pharmacy, 39(11),
1599–1617. http://dx.doi.org/10.3109/03639045.2012.736515
Adetogun, G. E., & Alebiowu, G. (2007). Influence of Delonix regia seed gum on the
compressional characteristics of paracetamol tablet formulations. Journal of
Drug Delivery Science and Technology, 17(6), 443–445. http://dx.doi.org/10.
1016/s1773-2247(07)50086-9
Adetogun, G. E., & Alebiowu, G. (2009). Properties of Delonix regia seed gum as a
novel tablet binder. Acta Poloniae Pharmaceutica, 66(4), 433–438.
Ahn, S., Seo, E., Kim, K., & Lee, S. J. (2013). Controlled cellular uptake and drug
efficacy of nanotherapeutics. Scientific reports, 3, 1997. http://dx.doi.org/10.
1038/srep01997
Ali, M. E., & Lamprecht, A. (2013). Polyethylene glycol as an alternative polymer
solvent for nanoparticle preparation. International Journal of Pharmaceutics,
456(1), 135–142. http://dx.doi.org/10.1016/j.ijpharm.2013.07.077
Anderson, J. M., & Shive, M. S. (2012). Biodegradation and biocompatibility of PLA
and PLGA microspheres. Advanced Drug Delivery Reviews, 64, 72–82. http://dx.
doi.org/10.1016/j.addr.2012.09.004
Bento, J. F., Mazzaro, I., de Almeida Silva, L. M., de Azevedo Moreira, R., Ferreira, M.
L. C., Reicher, F., & de Oliveira Petkowicz, C. L. (2013). Diverse patterns of cell
wall mannan/galactomannan occurrence in seeds of the Leguminosae.
Carbohydrate Polymers, 92(1), 192–199.
Betancur-Ancona, D., Pacheco-Aguirre, J., Castellanos-Ruelas, A., & Chel-Guerrero,
L. (2011). Microencapsulation of papain using carboxymethylated flamboyant
(Delonix regia) seed gum. Innovative Food Science & Emerging Technologies,
12(1), 67–72. http://dx.doi.org/10.1016/j.ifset.2010.11.002
Bhowmik, M., Kumari, P., Sarkar, G., Bain, M. K., Bhowmick, B., Mollick, M. M., &
Chattopadhyay, D. (2013). Effect of xanthan gum and guar gum on in situ
gelling ophthalmic drug delivery system based on poloxamer-407.
International Journal of Biological Macromolecules, 62, 117–123. http://dx.doi.
org/10.1016/j.ijbiomac.2013.08.024
Bilati, U., Allemann, E., & Doelker, E. (2005). Development of a nanoprecipitation
method intended for the entrapment of hydrophilic drugs into nanoparticles.
European Journal of Pharmaceutical Sciences, 24(1), 67–75. http://dx.doi.org/10.
1016/j.ejps.2004.09.011
Bouallagui, Z., Han, J., Isoda, H., & Sayadi, S. (2011). Hydroxytyrosol rich extract
from olive leaves modulates cell cycle progression in MCF-7 human breast
cancer cells. Food Chemical Toxicology, 49(1), 179–184. http://dx.doi.org/10.
1016/j.fct.2010.10.014
Bradford, M. M., Williams, W. L., 1977. Protein-assay reagent and method: Google
Patents.
Calvo, P., Vila-Jato, J., & Alonso, M. (1997). Effect of lysozyme on the stability of
polyester nanocapsules and nanoparticles: Stabilization approaches.
Biomaterials, 18(19), 1305–1310.
Carneiro-da-Cunha, M. G., Cerqueira, M. A., Souza, B. W. S., Teixeira, J. A., & Vicente,
A. A. (2011). Influence of concentration, ionic strength and pH on zeta potential
and mean hydrodynamic diameter of edible polysaccharide solutions
envisaged for multinanolayered films production. Carbohydrate Polymers,
85(3), 522–528. http://dx.doi.org/10.1016/j.carbpol.2011.03.001
Ciucanu, I., & Kerek, F. (1984). A simple and rapid method for the permethylation
of carbohydrates. Carbohydrate Research, 131(2), 209–217.
Devkar, T. B., Tekade, A. R., & Khandelwal, K. R. (2014). Surface engineered
nanostructured lipid carriers for efficient nose to brain delivery of ondansetron
HCl using Delonix regia gum as a natural mucoadhesive polymer. Colloids
Surface B Biointerfaces, 122, 143–150. http://dx.doi.org/10.1016/j.colsurfb.2014.
06.037
Dey, S., & Mitra, A. K. (2005). Transporters and receptors in ocular drug delivery:
Opportunities and challenges. Expert Opinion on Drug Delivery, 2(2), 201–204.
http://dx.doi.org/10.1517/17425247.2.2.201
Dong, Y., Chang, Y., Wang, Q., Tong, J., & Zhou, J. (2015). Effect of operating
conditions on size and morphology of amylose nanoparticles prepared by
precipitation. Starch − Stärke, 67(3–4), 365–372. http://dx.doi.org/10.1002/
star.201400182
Dubois, M., Gilles, K. A., Hamilton, J. K., Rebers, P., & Smith, F. (1956). Colorimetric
method for determination of sugars and related substances. Analytical
Chemistry, 28(3), 350–356.
Elzoghby, A. O., El-Fotoh, W. S., & Elgindy, N. A. (2011). Casein-based formulations
as promising controlled release drug delivery systems. Journal of Controlled
Release, 153(3), 206–216. http://dx.doi.org/10.1016/j.jconrel.2011.02.010
Gratieri, T., Gelfuso, G. M., Rocha, E. M., Sarmento, V. H., de Freitas, O., & Lopez, R. F.
(2010). A poloxamer/chitosan in situ forming gel with prolonged retention
time for ocular delivery. European Journal of Pharmaceutics and
Biopharmaceutics, 75(2), 186–193. http://dx.doi.org/10.1016/j.ejpb.2010.02.011
Hu, R., Chen, Y. Y., & Zhang, L. M. (2010). Synthesis and characterization of in situ
photogelable polysaccharide derivative for drug delivery. International Journal
of Pharmaceutics, 393(1–2), 96–103. http://dx.doi.org/10.1016/j.ijpharm.2010.
04.011
Huang, S., & Fu, X. (2010). Naturally derived materials-based cell and drug delivery
systems in skin regeneration. Journal of Controlled Release, 142(2), 149–159.
http://dx.doi.org/10.1016/j.jconrel.2009.10.018
Imayasu, M., Shiraishi, A., Ohashi, Y., Shimada, S., & Cavanagh, H. D. (2008). Effects
of multipurpose solutions on corneal epithelial tight junctions. Eye & Contact
Lens, 34(1), 50–55. http://dx.doi.org/10.1097/ICL.0b013e318073cbdb
Kapoor, V. (1972). A galactomannan from the seeds of Delonix regia.
Phytochemistry, 11(3), 1129–1132.
Khoee, S., & Yaghoobian, M. (2009). An investigation into the role of surfactants in
controlling particle size of polymeric nanocapsules containing penicillin-G in
double emulsion. European Journal of Medicine Chemistry, 44(6), 2392–2399.
http://dx.doi.org/10.1016/j.ejmech.2008.09.045
Kimura, K., Teranishi, S., Fukuda, K., Kawamoto, K., & Nishida, T. (2008). Delayed
disruption of barrier function in cultured human corneal epithelial cells
A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1075
induced by tumor necrosis factor-alpha in a manner dependent on NF-kappaB.
Investigative Ophthalmology and Visual Science, 49(2), 565–571. http://dx.doi.
org/10.1167/iovs.07-0419
Kumari, A., Yadav, S. K., & Yadav, S. C. (2010). Biodegradable polymeric
nanoparticles based drug delivery systems. Colloids Surface B Biointerfaces,
75(1), 1–18. http://dx.doi.org/10.1016/j.colsurfb.2009.09.001
Marques, M. R., Loebenberg, R., & Almukainzi, M. (2011). Simulated biological
fluids with possible application in dissolution testing. Dissolution Technology,
18(3), 15–28.
McConaughy, S. D., Stroud, P. A., Boudreaux, B., Hester, R. D., & McCormick, C. L.
(2008). Structural characterization and solution properties of a galacturonate
polysaccharide derived from aloe vera capable of in situ gelation .
Biomacromolecules, 9(2), 472–480.
Mitchell, D. J., & Ninham, B. W. (1981). Micelles, vesicles and microemulsions.
Journal of the Chemical Society, Faraday Transactions 2: Molecular and Chemical
Physics, 77(4), 601–629. http://dx.doi.org/10.1039/F29817700601
Miyazaki, S., Suzuki, S., Kawasaki, N., Endo, K., Takahashi, A., & Attwood, D. (2001).
In situ gelling xyloglucan formulations for sustained release ocular delivery of
pilocarpine hydrochloride. International Journal of Pharmaceutics, 229(1),
29–36.
Nagarwal, R. C., Kant, S., Singh, P. N., Maiti, P., & Pandit, J. K. (2009). Polymeric
nanoparticulate system: A potential approach for ocular drug delivery. Journal
of Controlled Release, 136(1), 2–13. http://dx.doi.org/10.1016/j.jconrel.2008.12.
018
Nagarwal, R. C., Kumar, R., & Pandit, J. K. (2012). Chitosan coated sodium
alginate-chitosan nanoparticles loaded with 5-FU for ocular delivery: In vitro
characterization and in vivo study in rabbit eye. European Journal of
Pharmaceutical Sciences, 47(4), 678–685. http://dx.doi.org/10.1016/j.ejps.2012.
08.008
Prajapat, A. L., & Gogate, P. R. (2015). Depolymerization of guar gum solution using
different approaches based on ultrasound and microwave irradiations.
Chemical Engineering and Processing: Process Intensification, 88, 1–9.
Pretor, S., Bartels, J., Lorenz, T., Dahl, K., Finke, J. H., Peterat, G., . . . &
Muller-Goymann, C. C. (2015). Cellular uptake of coumarin-6 under
microfluidic conditions into HCE-T cells from nanoscale formulations.
Molecular Pharmaceutics, 12(1), 34–45. http://dx.doi.org/10.1021/mp500401t
Sahoo, S. K., Dilnawaz, F., & Krishnakumar, S. (2008). Nanotechnology in ocular
drug delivery. Drug Discovery Today, 13(3–4), 144–151. http://dx.doi.org/10.
1016/j.drudis.2007.10.021
Sarmah, J. K., Mahanta, R., Bhattacharjee, S. K., Mahanta, R., & Biswas, A. (2011).
Controlled release of tamoxifen citrate encapsulated in cross-linked guar gum
nanoparticles. International Journal of Biological Macromolecules, 49(3),
390–396. http://dx.doi.org/10.1016/j.ijbiomac.2011.05.020
Sassaki, G. L., Gorin, P. A., Souza, L. M., Czelusniak, P. A., & Iacomini, M. (2005).
Rapid synthesis of partially O-methylated alditol acetate standards for GC–MS:
Some relative activities of hydroxyl groups of methyl glycopyranosides on
Purdie methylation. Carbohydrate Research, 340(4), 731–739.
Serra, L., Domenech, J., & Peppas, N. A. (2009). Engineering design and molecular
dynamics of mucoadhesive drug delivery systems as targeting agents.
European Journal of Pharmaceutics and Biopharmaceutics, 71(3), 519–528.
http://dx.doi.org/10.1016/j.ejpb.2008.09.022
Smart, J. D. (2005). The basics and underlying mechanisms of mucoadhesion.
Advance Drug Delivery Review, 57(11), 1556–1568. http://dx.doi.org/10.1016/j.
addr.2005.07.001
Soumya, R. S., Ghosh, S., & Abraham, E. T. (2010). Preparation and characterization
of guar gum nanoparticles. International Journal of Biological Macromolecules,
46(2), 267–269. http://dx.doi.org/10.1016/j.ijbiomac.2009.11.003
Tamaki, Y., Teruya, T., & Tako, M. (2010). The chemical structure of galactomannan
isolated from seeds of Delonix regia. Bioscience Biotechnology Biochemistry,
74(5), 1110–1112. http://dx.doi.org/10.1271/bbb.90935
Vega, E., Gamisans, F., Garcia, M. L., Chauvet, A., Lacoulonche, F., & Egea, M. A.
(2008). PLGA nanospheres for the ocular delivery of flurbiprofen: Drug release
and interactions. Journal of Pharmaceutical Sciences, 97(12), 5306–5317. http://
dx.doi.org/10.1002/jps.21383
Veronovski, N., Andreozzi, P., La Mesa, C., & Sfiligoj-Smole, M. (2010). Stable TiO2
dispersions for nanocoating preparation. Surface and Coatings Technology,
204(9–10), 1445–1451. http://dx.doi.org/10.1016/j.surfcoat.2009.09.041
Yi, X.-, Wang, Y., & Fu-Shin, X. Y. (2000). Corneal epithelial tight junctions and their
response to lipopolysaccharide challenge. Investigative Ophthalmology and
Visual Science, 41(13), 4093–4100.
Zhang, Y., Huo, M., Zhou, J., Zou, A., Li, W., Yao, C., & Xie, S. (2010). DDSolver: An
add-in program for modeling and comparison of drug dissolution profiles. The
AAPS Journal, 12(3), 263–271. http://dx.doi.org/10.1208/s12248-010-9185-1
Zhao, F., Zhao, Y., Liu, Y., Chang, X., Chen, C., & Zhao, Y. (2011). Cellular uptake,
intracellular trafficking, and cytotoxicity of nanomaterials. Small, 7(10),
1322–1337. http://dx.doi.org/10.1002/smll.201100001
Zuo, Z., Lee, V. H. L., Controlled Drug Delivery: Pharmacokinetic Considerations,
Methods and Systems (2008);10.1002/9780470048672. wecb099.
de Campos, A. M., Diebold, Y., Carbalho, E. L., Sánchez, A., & José Alonso, M. (2005).
Chitosan nanoparticles as new ocular drug delivery systems: In vitro stability,
in vivo fate, and cellular toxicity. Pharmaceutical Research, 22(6), 1007.
de la Fuente, M., Ravina, M., Paolicelli, P., Sanchez, A., Seijo, B., & Alonso, M. J.
(2010). Chitosan-based nanostructures: A delivery platform for ocular
therapeutics. Advance Drug Delivery Review, 62(1), 100–117. http://dx.doi.org/
10.1016/j.addr.2009.11.026

More Related Content

What's hot

Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...
Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...
Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...Smruti Chaudhari, Ph.D.
 
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...Smruti Chaudhari, Ph.D.
 
Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...
Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...
Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...ijsrd.com
 
7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...
7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...
7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...BIOLOGICAL FORUM
 
Chitin chitosan properties
Chitin chitosan propertiesChitin chitosan properties
Chitin chitosan propertiesNgcon35
 
Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...
Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...
Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...rkkoiri
 
A comparative study of biopolymers and alum in the separation and recovery of...
A comparative study of biopolymers and alum in the separation and recovery of...A comparative study of biopolymers and alum in the separation and recovery of...
A comparative study of biopolymers and alum in the separation and recovery of...Soumyadeep Mukherjee
 
Preparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivatives
Preparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivativesPreparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivatives
Preparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivativeselshimaa eid
 
EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.
EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.
EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.DAWN V TOMY
 
Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...
Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...
Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...BRNSS Publication Hub
 
Determination of cefadroxil antibiotic by an analytical method
Determination of cefadroxil antibiotic by an analytical methodDetermination of cefadroxil antibiotic by an analytical method
Determination of cefadroxil antibiotic by an analytical methodAlexander Decker
 
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...pharmaindexing
 
IOSR Journal of Pharmacy (IOSRPHR)
IOSR Journal of Pharmacy (IOSRPHR)IOSR Journal of Pharmacy (IOSRPHR)
IOSR Journal of Pharmacy (IOSRPHR)iosrphr_editor
 
Article wjpps 1449127501
Article wjpps 1449127501Article wjpps 1449127501
Article wjpps 1449127501Princy Rana
 

What's hot (18)

Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...
Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...
Investigating the Effect of Molecular Weight of Polyvinylpyrrolidone and Hydr...
 
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
 
Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...
Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...
Comparative Study on Phytochemical Screening and HPLC Analysis of Daucus Caro...
 
7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...
7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...
7 synthesis, characterisation and antimicrobial activity of schiff base of 7 ...
 
Chitin chitosan properties
Chitin chitosan propertiesChitin chitosan properties
Chitin chitosan properties
 
Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...
Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...
Structural characterization and cytotoxicity studies of ruthenium(ii)–dmso–ch...
 
A comparative study of biopolymers and alum in the separation and recovery of...
A comparative study of biopolymers and alum in the separation and recovery of...A comparative study of biopolymers and alum in the separation and recovery of...
A comparative study of biopolymers and alum in the separation and recovery of...
 
journal published article
journal published articlejournal published article
journal published article
 
Jcpr 2014-6-4-1225-1231
Jcpr 2014-6-4-1225-1231Jcpr 2014-6-4-1225-1231
Jcpr 2014-6-4-1225-1231
 
Preparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivatives
Preparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivativesPreparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivatives
Preparation of pyrimido[4,5 b][1,6]naphthyridin-4(1 h)-one derivatives
 
EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.
EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.
EVALUATION OF ANTI-OXIDANT AND ANTI-TUMOUR ACTIVITIES OF CROTON LAEVIGATUS Vahl.
 
Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...
Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...
Structural Characterization and Antimicrobial Activity of Cocculus hirsutus L...
 
Determination of cefadroxil antibiotic by an analytical method
Determination of cefadroxil antibiotic by an analytical methodDetermination of cefadroxil antibiotic by an analytical method
Determination of cefadroxil antibiotic by an analytical method
 
Ijms 13-11681-v2
Ijms 13-11681-v2Ijms 13-11681-v2
Ijms 13-11681-v2
 
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
317 - In-vitro Antioxidant studies on ethanolic extracts of Boswellia ovalifo...
 
IOSR Journal of Pharmacy (IOSRPHR)
IOSR Journal of Pharmacy (IOSRPHR)IOSR Journal of Pharmacy (IOSRPHR)
IOSR Journal of Pharmacy (IOSRPHR)
 
Article wjpps 1449127501
Article wjpps 1449127501Article wjpps 1449127501
Article wjpps 1449127501
 
Ijms 13-11681-v2
Ijms 13-11681-v2Ijms 13-11681-v2
Ijms 13-11681-v2
 

Similar to carbohydrate polymer

Development and characterization of anionic liposaccharides for enhanced oral
Development and characterization of anionic liposaccharides for enhanced oralDevelopment and characterization of anionic liposaccharides for enhanced oral
Development and characterization of anionic liposaccharides for enhanced oralAdel Abdelrahim, PhD
 
Quality by Design Approach for the Development of UHPLC Method indicating the...
Quality by Design Approach for the Development of UHPLC Method indicating the...Quality by Design Approach for the Development of UHPLC Method indicating the...
Quality by Design Approach for the Development of UHPLC Method indicating the...IRJET Journal
 
Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...
Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...
Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...inventionjournals
 
Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...
Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...
Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...Madiha Mushtaque
 
Research Highlights
Research HighlightsResearch Highlights
Research HighlightsEmily Scott
 
Mini review of polysaccharide nanoparticles and drug delivery process
Mini review of polysaccharide nanoparticles and drug delivery process Mini review of polysaccharide nanoparticles and drug delivery process
Mini review of polysaccharide nanoparticles and drug delivery process AANBTJournal
 
ZQL-journal of material chemistry B校稿前版
ZQL-journal of material chemistry B校稿前版ZQL-journal of material chemistry B校稿前版
ZQL-journal of material chemistry B校稿前版Quanlei Zhu
 
MA-Phenol-Paper_JECE
MA-Phenol-Paper_JECEMA-Phenol-Paper_JECE
MA-Phenol-Paper_JECEMustafa Nabil
 
Melton_Stephanie_PosterPresentation
Melton_Stephanie_PosterPresentationMelton_Stephanie_PosterPresentation
Melton_Stephanie_PosterPresentationStephanie Melton
 
Dna hydrogel microspheres and their potential applications for protein delive...
Dna hydrogel microspheres and their potential applications for protein delive...Dna hydrogel microspheres and their potential applications for protein delive...
Dna hydrogel microspheres and their potential applications for protein delive...Giovanne Delechiave
 
1-s2.0-S0014305718318718-am.pdf
1-s2.0-S0014305718318718-am.pdf1-s2.0-S0014305718318718-am.pdf
1-s2.0-S0014305718318718-am.pdfHuascarFernandez2
 
JBEI Research Highlights - May 2019
JBEI Research Highlights - May 2019JBEI Research Highlights - May 2019
JBEI Research Highlights - May 2019Irina Silva
 
Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...
Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...
Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...IJERA Editor
 

Similar to carbohydrate polymer (20)

Development and characterization of anionic liposaccharides for enhanced oral
Development and characterization of anionic liposaccharides for enhanced oralDevelopment and characterization of anionic liposaccharides for enhanced oral
Development and characterization of anionic liposaccharides for enhanced oral
 
(2102 2112) nd14
(2102 2112) nd14(2102 2112) nd14
(2102 2112) nd14
 
9 Mehra and Jain JDT
9 Mehra and Jain JDT9 Mehra and Jain JDT
9 Mehra and Jain JDT
 
Polysaccharide
PolysaccharidePolysaccharide
Polysaccharide
 
Quality by Design Approach for the Development of UHPLC Method indicating the...
Quality by Design Approach for the Development of UHPLC Method indicating the...Quality by Design Approach for the Development of UHPLC Method indicating the...
Quality by Design Approach for the Development of UHPLC Method indicating the...
 
Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...
Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...
Design and Evaluation of Ion Induced in Situ Gel formulation For Levofloxacin...
 
Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...
Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...
Development and Pharmaceutical Evaluation of Clotrimazole Loaded Topical Hydr...
 
Research Highlights
Research HighlightsResearch Highlights
Research Highlights
 
Mini review of polysaccharide nanoparticles and drug delivery process
Mini review of polysaccharide nanoparticles and drug delivery process Mini review of polysaccharide nanoparticles and drug delivery process
Mini review of polysaccharide nanoparticles and drug delivery process
 
ZQL-journal of material chemistry B校稿前版
ZQL-journal of material chemistry B校稿前版ZQL-journal of material chemistry B校稿前版
ZQL-journal of material chemistry B校稿前版
 
Synthesis of bioplastics
Synthesis of bioplasticsSynthesis of bioplastics
Synthesis of bioplastics
 
MA-Phenol-Paper_JECE
MA-Phenol-Paper_JECEMA-Phenol-Paper_JECE
MA-Phenol-Paper_JECE
 
Melton_Stephanie_PosterPresentation
Melton_Stephanie_PosterPresentationMelton_Stephanie_PosterPresentation
Melton_Stephanie_PosterPresentation
 
Dna hydrogel microspheres and their potential applications for protein delive...
Dna hydrogel microspheres and their potential applications for protein delive...Dna hydrogel microspheres and their potential applications for protein delive...
Dna hydrogel microspheres and their potential applications for protein delive...
 
ausama PP.PDF
ausama PP.PDFausama PP.PDF
ausama PP.PDF
 
6. arun patel article.pdf
6. arun patel article.pdf6. arun patel article.pdf
6. arun patel article.pdf
 
1-s2.0-S0014305718318718-am.pdf
1-s2.0-S0014305718318718-am.pdf1-s2.0-S0014305718318718-am.pdf
1-s2.0-S0014305718318718-am.pdf
 
Ijms 13-11681-v2
Ijms 13-11681-v2Ijms 13-11681-v2
Ijms 13-11681-v2
 
JBEI Research Highlights - May 2019
JBEI Research Highlights - May 2019JBEI Research Highlights - May 2019
JBEI Research Highlights - May 2019
 
Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...
Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...
Biocompatibility of Poly (L-Lactic Acid) Synthesized In Polymerization Unit B...
 

Recently uploaded

Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Dipal Arora
 
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service AvailableDipal Arora
 
O898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
O898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In AhmedabadO898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
O898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In AhmedabadGENUINE ESCORT AGENCY
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...narwatsonia7
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Call Girls in Nagpur High Profile
 
Call Girls Varanasi Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 8250077686 Top Class Call Girl Service AvailableDipal Arora
 
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...Ishani Gupta
 
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...Dipal Arora
 
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any TimeTop Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any TimeCall Girls Delhi
 
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...perfect solution
 
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...chandars293
 
Call Girls Bareilly Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Bareilly Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 8250077686 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...Taniya Sharma
 
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service AvailableDipal Arora
 
(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...
(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...
(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...parulsinha
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 

Recently uploaded (20)

Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
 
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
 
O898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
O898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In AhmedabadO898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
O898O367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
 
Call Girls Varanasi Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Varanasi Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Varanasi Just Call 8250077686 Top Class Call Girl Service Available
 
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
 
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 9907093804 Top Class Call Girl Service Ava...
 
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any TimeTop Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
 
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
College Call Girls in Haridwar 9667172968 Short 4000 Night 10000 Best call gi...
 
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
 
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Nagpur Just Call 9907093804 Top Class Call Girl Service Available
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 9332606886 ⟟ Call Me For Genuine ...
 
Call Girls Bareilly Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Bareilly Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bareilly Just Call 8250077686 Top Class Call Girl Service Available
 
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
 
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
 
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
 
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
 
(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...
(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...
(Low Rate RASHMI ) Rate Of Call Girls Jaipur ❣ 8445551418 ❣ Elite Models & Ce...
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
 

carbohydrate polymer

  • 1. Carbohydrate Polymers 157 (2017) 1065–1075 Contents lists available at ScienceDirect Carbohydrate Polymers journal homepage: www.elsevier.com/locate/carbpol Hydrophilic polymeric nanoparticles prepared from Delonix galactomannan with low cytotoxicity for ocular drug delivery Abayomi T. Ogunjimia,b , Shaiani M.G. Meloa , Carem G. Vargas-Rechiaa , Flávio S. Emerya , Renata F.V. Lopeza,∗ a University of São Paulo, School of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto, SP, Brazil b Department of Pharmaceutics, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria a r t i c l e i n f o Article history: Received 12 July 2016 Received in revised form 20 October 2016 Accepted 24 October 2016 Available online 25 October 2016 Keywords: Cellular uptake Cytotoxicity Delonix polymeric nanoparticles Ocular delivery Quality by design a b s t r a c t Delonix is a galactomannan polysaccharide extracted from the endosperm of Delonix regia plant. This study aims at the development of Delonix nanoparticle and assesses its potential for ocular delivery by evaluating its in-vitro stability, toxicity and cellular uptake. Fluorescent nanoparticles (BODIPY-loaded nanoparticles) were prepared by a Quality-by-Design modified nanoprecipitation technique. Optimized nanoparticles had mean sizes <240 nm, PdI < 0.2 and zeta potential of <−30 mV. Mixture of surfactants with different hydrophilic-lipophilic balance controlled nanoparticle swelling. Nanoparticles, which were stable in the presence of simulated lachrymal fluid and lysozyme also sustained the release of BODIPY. In-vitro studies suggest no toxicity of the nanoparticles in concentration range of 100–1483.3 ␮g/mL on retinal and corneal epithelial cells. Flow cytometry and confocal microscopy techniques showed that retinal cells but not corneal cells, uptake 18% of the nanoparticles. Therefore, Delonix nanoparticles could be a safe and promising tool for ocular drug delivery. © 2016 Elsevier Ltd. All rights reserved. 1. Introduction Drug delivery for the treatment of various eye diseases has been a challenge due to the critical micro-environment that exists in the eye. Nanoparticulate drug delivery has shown promising results in ocular drug delivery to both the anterior and posterior parts of the eye in the last two decades. However, the quest for drug delivery tools that could target diseases of the eye, improve bioavailability and particularly minimize non-specific toxicity has not abated (Dey & Mitra, 2005; Sahoo, Dilnawaz, & Krishnakumar, 2008; Achouri, Alhanout, Piccerelle, & Andrieu, 2013). Interest in biodegradable polymeric nanoparticles as potential ocular drug delivery tools has increased due to their applications as DNA carriers for gene therapy and their ability to deliver proteins, peptides and genes (Zuo & Lee, 2008). Synthetic and semi-synthetic polymers such as poly lactic acid (PLA), poly glycolic acid (PGA), poly lactic acid-co-glycolic acid (PLGA), ethyl cellulose and hydrox- ypropyl methyl cellulose (HPMC) have been widely studied as ∗ Corresponding author at: School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil. E-mail address: rvianna@fcfrp.usp.br (R.F.V. Lopez). delivery agents via the ocular route (Kumari, Yadav, & Yadav, 2010; Elzoghby, El-Fotoh, & Elgindy, 2011; Anderson & Shive, 2012). However, attracting more interest in nano-delivery are the nat- ural hydrophilic polymers due to their accomplished properties of biocompatibility, affordability, little or no toxicity, availability and non irritant nature (Huang & Fu, 2010). In addition to mucoad- hesion, hydrophilic polymers generally swells and form a gel-like layer on their particle surface which retards fluid ingress into the polymer core thereby sustaining drug release(Sarmah, Mahanta, Bhattacharjee, Mahanta, & Biswas, 2011). Several natural polymers including alginate, guar gum and chitosan have been studied as ocular nano delivery agents (Nagarwal, Kant, Singh, Maiti, & Pandit, 2009; Bhowmik et al., 2013; Gratieri et al., 2010). Nanoparticle preparation with some of these hydrophilic polymers involve the use of toxic cross-linking agents such as glutaraldehyde that could pose toxicity concerns to liv- ing cells and tissues. Chitosan, a cationic polysaccharide and its derivatives with favorable biological properties has been exten- sively studied in topical ocular delivery of drugs either individually or as a surface modifier for other polymers (Gratieri et al., 2010; de la Fuente et al., 2010; Nagarwal, Kumar, & Pandit, 2012). How- ever, there exist other natural polysaccharides with little or no information in literature, but with potentials for ocular delivery. http://dx.doi.org/10.1016/j.carbpol.2016.10.076 0144-8617/© 2016 Elsevier Ltd. All rights reserved.
  • 2. 1066 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 One of such polymer is Delonix polymer (DLX), a galactomannan polysaccharide (Supplementary material, Fig. S1.1) extracted from the endosperm of Delonix regia (Bojer ex Hook.) plant (Kapoor, 1972; Tamaki, Teruya, & Tako, 2010). Delonix regia or flamboyant is a leguminous plant of the family Fabaceae native to Madagascar but freely growing in diverse countries around the world today. The plant is widely used as an ornamental while its seed, highly rich in a polymeric galactomannan are sometimes used as beads. The use of the seed polymer as a drug delivery tool, in addition to being a green approach, could also be of economic benefit for low income communities, where the plant is inhabited. Delonix and its modified derivatives have previously been studied as a sustained release polymer for tablets (Adetogun & Alebiowu, 2007; Adetogun & Alebiowu, 2009), mucoadhesive (Devkar, Tekade, & Khandelwal, 2014) and microencapsulation agent (Betancur-Ancona, Pacheco-Aguirre, Castellanos-Ruelas, & Chel-Guerrero, 2011). Our laboratory recently commenced studies on DLX as a controlled nano-delivery agent with ocular deliv- ery prospects. Unlike some natural polymeric nanoparticles whose process of production involve the use of toxic cross-linking agents like glutaraldehyde (Soumya, Ghosh, & Abraham, 2010), we have designed a simple method for the production of stable DLX nanoparticles without the use of such toxic cross-linking agents. Thus, the present study aims at developing a method for the preparation and characterization of a DLX nanoparticle formula- tion for ocular delivery. The study also evaluates the encapsulation prowess of DLX nanoparticle using hydrophilic and lipophilic probe molecules, while assessing its potential for ocular delivery by eval- uating its stability in simulated lachrymal tear and enzymes, in vitro cellular toxicity and uptake. 2. Materials and methods 2.1. Materials DLX extracted and purified in our laboratory; absolute ethanol and neutral red (NRU) dye from Merck, Germany; acetone, dimethyl sulfoxide, polyoxyethylene (20) sorbitan monooleate (tween 80) and [2-(3,4-dihydroxyoxolan-2-yl)-2-hydroxyethyl] octadec- 9-enoate (span 80) from Synth, Brazil; mucin from porcine stomach Type III, ITS liquid media supplement, hydrocortisone, cholera toxin A, lysozyme from chicken egg white and 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) dye from Sigma- Aldrich, USA; fetal bovine serum (FBS) from Invitrogen, USA; Dulbecco’s modified Eagles medium (DMEM), Hams F12 (F12), gen- tamycin and DAPi molecular probe from Life technologies, New Zealand; Supplemented Hormonal Epidermal Medium (SHEM) prepared in our laboratory, hydrophilic and lipophilic BODIPY syn- thesized by our collaborators (Supplementary material, Figs. S2.1, S2.2, S2.3, S2.4), Retina Pigmented Epithelium cell line (ARPE-19, BCJR code: 0041) from the Cell Bank in Rio de Janeiro, Brazil and Immortalized Human Corneal Epithelial Cell line (HCE SV-40) were all the materials used in this work. 2.2. Collection and purification of polymer DLX was collected manually from mature and ripe pods (fruit) of Delonix regia plant within the vicinity of University of Sao Paulo, Ribeirao Preto campus. The seeds from the pods were split open and the polymeric endosperm removed, dried (50 ◦C) in oven and milled. The milled unpurified DLX mass was redispersed in suffi- cient distilled water for 48 h, filtered under negative pressure with a Buchner funnel to remove all undissolved materials and then pre- cipitated with absolute ethanol. The precipitate was filtered and washed twice with acetone, oven dried at 50 ◦C, pulverized, sieved (250 ␮m sieve-size) and stored until needed. 2.3. Characterization of DLX polymer 2.3.1. General analysis Total carbohydrate content of the DLX polymer was performed by the phenol-sulfuric acid assay method (Dubois, Gilles, Hamilton, Rebers, & Smith, 1956) while protein content was performed by Bradford assay (Bradford & Williams, 1977) method using Coomassie Brilliant Blue G 250. 2.3.2. Monosaccharide composition Monosaccharide composition was determined using DLX poly- mer obtained in Section 2.2 (DLXp), DLX polymer recovered from sonicated polymer solution described in Section 2.4.1 (DLXs) and DLX polymer recovered from polymer solution described in Section 2.4.1 but without sonication (DLXu). The DLX polymer was recov- ered by precipitation using ethanol, followed by filtration, drying at 50 ◦C and pulverization. The DLXp, DLXs and DLXu were completely hydrolyzed with 2 mol/L trifluoroacetic acid (TFA, 5 h, 100 ◦C). The hydrolyzed polymers were reduced with sodium borohydride for 2 h and the alditols were acetylated with pyridine:acetic anhydride (1:1 v/v, 18 h) at room temperature. The resulting alditol acetates were analyzed by gas-liquid chromatography/mass spectrometry (GCMS-QP2010 Ultra − SHIMADZU) using ZB-5MS (30 m x 0.25 mm x 0.25um) Zebron-Phenomenex column. 2.3.3. Methylation analysis: DLXs and DLXu The polysaccharides (5 mg) were methylated according to the established methods (Ciucanu & Kerek, 1984). Each per-O- methylated polysaccharide was pre-treated with 72% v/v H2SO4 (1 h, 4 ◦C) and water was added to a final acid concentration of 8% (18 h) at room temperature. The hydrolyzed polymer were reduced with acetylated borohydride and analyzed by CGMS as described above (section 2.3.2). 2.3.4. 13C and 1H spectroscopy All NMR spectra were obtained using a Bruker 400 MHz Avance III 14.1 T spectrometer equipped with an inverse 5-mm pentanu- clear resonance probe head (QXI) at 303 K. Chemical shifts were expressed in ␦ (ppm) relative to the resonance of DSS (sodium 4,4-dimethyl-4-silapentane-l-sulphonate) and methanol used as an internal standard (∼ = 0) for 1H and for 13C respectively. 2.3.5. Determination of Molecular weight (Mw) The average Mw of DLXs and DLXu polysaccharides were determined by size exclusion chromatography (GPC), with a Sepharose CL–4 B chromatographic column (43 × 1.8 cm, Mw range of 3 × 104–5 × 106) and water as the mobile phase set at a flow rate of 0.6 mLmin−1. Eluted samples were monitored with a differen- tial refractometer (model R401, Waters). Sugars were identified by comparing the retention times shown in the peaks of DLXs and DLXu samples with those of dextran standard samples. 2.4. Nanoparticles preparation 2.4.1. Preparation of DLX polymer solution DLX polymer solution was prepared by dispersing required amount of DLX polymer (Table 1) in 0.5 M NaOH for 24 h to ascer- tain complete dissolution/dispersion. This dispersion medium was chosen based on results of some preliminary experiments (Sup- plementary material, Section S3). The solubilized polymer was centrifuged and filtered (15000 x g, 20 min; 0.8 ␮m micropore fil- ter) to remove undissolved matter. The mildly viscous polymer
  • 3. A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1067 Table 1 Box Behnken design for DLX polymeric nanoparticle formulation. Coded variables Formulation X1 X2 X3 F1 −1 0 1 F2 1 1 0 F3 1 0 1 F4 −1 1 0 F5 1 −1 0 F6 0 0 0 F7 −1 0 −1 F8 0 1 1 F9 0 0 0 F10 −1 −1 0 F11 1 0 −1 F12 0 −1 −1 F13 0 0 0 F14 0 −1 1 F15 0 1 −1 Coded variables Uncoded variables −1 0 1 X1 Polymer concentration (%) 1.0 1.5 2.0 X2 Surfactant mix ratio (tween 80:span 80) 1:0 4:1 3:2 X3 Non-solvent: solvent ratio 10 15 20 filtrate was sonicated (20 Hz, 40% amplitude; VCX 500, Sonic & Materials, Inc, USA) for 30 min and the solution stored in refrig- erator for at least 24 h before use. 2.4.2. Design of Experiment (DoE) To adequately control critical parameters that could influence the DLX nanoparticle properties, a Quality-by-Design, modified nanoprecipitation approach was employed. The potential factors that could impart the desired quality of the DLX nanoparticle were assessed by deploying a Fishbone diagram (Supplementary mate- rial, Fig. S3.1) in creating a structure that could identify such factors. Based on carefully researched knowledge space and some initial basic experiments, we arrived at three probable factors − poly- mer concentration, surfactant mix ratio and non-solvent/solvent ratio − that could influence the desired DLX nanoparticle prop- erties. For example, polymer concentration was chosen based on the knowledge space available; increase in polymer concentration generally lead to increase in viscosity, which consequently produce nanoparticles with larger sizes. In addition, inclusion of tween 20 led to the formation of a viscous dispersion which was difficult to manipulate while surfactant concentration had no significant influence on DLX nanoparticle size in initial experiments leading to the choice of 5% surfactant concentration for all experiments. Surfactant mix ratio was selected based on some initial experi- ments which showed that the use of only tween 80 produced DLX nanoparticle sizes in the range of 300 nm with PdI > 0.3, while the use of only span 80 produced DLX nanoparticles with sizes ranging between 600 and 700 nm but PdI < 0.2. The inclusion of poloxamer (P407) only produced DLX nanoparticles sizes in range of 250 nm but with PdI > 0.4. Nonsolvent-solvent ratio is mostly important in terms of economics of solvent, however, it was included as a fac- tor because most studies that discussed nonsolvent-solvent ratio influence on nanoparticle size as far as we know involved the use of synthetic polymers. As such, our polymer being hydrophilic and actively swelling might produce a different response (Supplemen- tary material, Section S3). Each of these factors were tested at three levels using a completely randomized 15-experiment Box Behnken statistical design (Table 1). The influence of these process variables on selected DLX nanoparticle properties of hydrodynamic diame- ter, polydispersibility index (PdI) and zeta potential was evaluated. Analysis of Variance (ANOVA) was performed to indentify statis- tically significant factors while general linear model was used for parameter estimate. 2.4.3. Preparation of blank DLX nanoparticle DLX nanoparticles were prepared by a modified nanoprecip- itation process using absolute ethanol as the non-solvent. The non-solvent solution was first prepared by adding necessary vol- ume of 5% surfactant mix (tween 80 and/or span 80) to absolute ethanol based on a Box Behnken statistical design (Table 1) and mixing for 10 min under moderate magnetic stirring. Exactly 1 mL of DLX solution was added to the non-solvent mix under mag- netic stirring (1000 rpm, TECNAL magnetic mixer, Brazil) and at a polymer flow rate of approximately 1 mL/min. The mildly tur- bid nanoparticle dispersion was allowed to stand for about 30 min under reduced magnetic stirring speed. The mixture was then cen- trifuged at 15,000g for 10 min (Thermo Scientific Heraeus Megafuge 16R, ThermoFisher Scientific, USA), the supernatant decanted and the surface of the nanoparticle washed twice with distilled water. The nanoparticles were redispersed in distilled water and passed through a 0.45 ␮m syringe filter before evaluation. The nanopar- ticles were evaluated by hydrodynamic diameter, PdI and zeta potential. Experiments were performed in triplicate. 2.5. DLX nanoparticle characterization 2.5.1. Microscopy, size distribution and zeta potential analysis Nanoparticle size and distribution were measured using 1 mL of syringe-filtered DLX nanodispersion by dynamic light scatter- ing (DLS) using a Nano ZS (Malvern Zetasizer nano ZS90 series) at 25 ◦C and a scattering angle of 90◦. Zeta potential was deter- mined by laser doppler micro-electrophoresis technique with the same instrument (Malvern Zetasizer nano ZS90 series). Results are reported as average of triplicate determinations. The DLX nanopar- ticles were also characterized by transmission electron microscopy (TEM), using a JEM-100CXII transmission electron microscope (JEOL, Japan) with an accelerating voltage ranging from 100 kV to 200 kV. 2.5.2. Surfactant mix influence on DLX nanoparticle size Based on results obtained from Box Behnken experimental design, three separate DLX nanoparticle formulations were pre- pared using 1% (w/v) DLX polymer solution, non-solvent/solvent ratio of 10, but varying surfactant mix ratio (tween 80:span 80; 1:0, 4:1, 3:2). DLX nanoparticle size, PdI and zeta potential were evaluated and average of quadruplicate experiments reported. 2.5.3. Surfactant mix influence on DLX nanoparticle swelling/stability The formulations described in Section 2.5.2 above were sub- jected to swelling/stability analysis by measuring the nanoparticles size, PdI and zeta potential of the nanoparticle formulation daily for six days. Results were recorded as percent size increase while experiment were performed in quadruplicates. 2.5.4. Statistical analysis Results obtained were subjected to appropriate statistical anal- ysis including Response Surface Regression (Statistica 10, StatSoft, USA), One-Way and Two-Way ANOVA (GraphPad Prism 5.0, Graph- Pad, USA). 2.6. 2.6. In vitro DLX nanoparticle stability in simulated ocular fluids 2.6.1. Incubation with lysozyme The stability of optimized DLX nanoparticles was analyzed according to established methods (de Campos, Diebold, Carbalho,
  • 4. 1068 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 Sánchez, & José Alonso, 2005) following their incubation at 37 ◦C in a solution of lysozyme in purified water (1 mg/mL) under mod- erate stirring. The mean particle size, PdI and zeta potential were monitored during the incubation process. Average of quadruplicate determinations were recorded. 2.6.2. Incubation in simulated tear fluid (STF) The stability of optimized DLX nanoparticles was analyzed in STF containing 0.67% w/v NaCl, 0.2% w/v NaHCO3 and 0.008% w/v CaCl2 2H2O (Miyazaki et al., 2001). The mean particle size, PdI and zeta potential were monitored during the incubation process at 35 ◦C. Results recorded were average of quadruplicate determinations. The osmolarity of DLX nanoparticle dispersion was also determined using an osmometer (Semi-Micro Osmometer K-7400, KNAUER). 2.6.3. Incubation with mucin The zeta potential of DLX nanoparticle was also evaluated in mucin (0.4 mg/mL) dispersion. Mucin, like lysozyme is a major component of the precorneal fluid. The zeta potential of mucin dispersion with and without DLX nanoparticles at 35 ◦C was evalu- ated. Values reported are average of quadruplicate determinations. Results obtained were subjected to ANOVA (GraphPad Prism 5.0, GraphPad, USA). 2.7. BODIPY-loaded DLX nanoparticle Hydrophilic and lipophilic BODIPY dyes (Supplementary mate- rial, section S2) were used as test molecules. To prepare loaded DLX nanoparticle, 10 mg of lipophilic or hydrophilic BODIPY were initially dissolved in the non-solvent medium or DLX solution respectively before nanoparticle preparation as described in sec- tion 2.4.3 above. The preparation was based on the selected optimal DLX nanoparticle formulation determined from the Box Behnken statistical design (DLX nanoparticle containing tween-span ratio 3:2). The nanoparticles were evaluated by size, PdI and zeta poten- tial. Results are average of triplicate determinations. The entrapment efficiency (EE) of BODIPY-loaded DLX nanopar- ticles was determined by ultracentrifugation (15,000g, 10 min, Thermo Scientific Heraeus Megafuge 16R, ThermoFisher Scientific, USA) and the amount of free BODIPY in the supernatant was mea- sured spectrophotometrically at an absorbance of 495 nm. The EE was calculated indirectly as described in Eq. (1). EE (%) = Theoretical BODIPY amount − BODIPY in supernatant Theoretical BODIPY amount × 100 (1) 2.8. In vitro release studies In vitro release studies of BODIPY-loaded DLX nanoparticles were carried out in Franz diffusion cells under magnetic stirring using dialysis membrane (12000 − 14000 Mw) as the diffusion bar- rier/membrane and distilled water as the receiver medium. Exactly 1 mL of BODIPY-loaded DLX nanoparticles (containing ≈ 550 ␮g/mL of BODIPY) was added to the donor compartment respectively while BODIPY (≈550 ␮g/mL) in DLX solution was used as control. All experiments were performed protected under dark condition at room temperature. At specific times, 1 mL of the receiver medium was removed and immediately replaced with 1 mL of fresh dis- tilled water. Amount of BODIPY at each evaluation time in the receiver medium was quantified using UV spectroscopy (wave- length = 495 nm). For this study, only the hydrophilic BODIPY was used while experiment was performed in triplicate. Kinetic mod- eling was performed using DDSolver Excel Add-in software (Zhang et al., 2010). 2.9. In vitro cell toxicity studies In vitro cytotoxicity assay was carried out using two ocular cell lines namely Retinal Pigment Epithelial (ARPE-19) and Human Corneal Epithelial (HCE) cells. The ARPE-19 cells were routinely cultured in DMEM/F12 medium (1:1) supplemented with 10% FBS and antibiotics while the HCE cells were cultured in SHEM made of DMEM:F12 (1:1) and supplemented with 5% FBS, amphotericin B, ITS, DMSO, hydrocorti- sone, EGF, cholera toxin and gentamycin. The cells were incubated in standard culture conditions (incubation at 37 ◦C in 5% CO2 atmo- sphere) and the medium was change every 2 − 3 days while cell growth was monitored daily by phase-contrast microscopy. Cells were allowed to reach at least 80% confluence before each tripsinization process. 2.9.1. Cytotoxicity assay Cytotocixity was carried by MTT and NRU assay techniques. The ARPE-19 and HCE cells were seeded at a density of 1.5 × 105 cells/well in 100 ␮L of their respective complete growth medium into 96-well plates and incubated (37 ◦C in a 5% CO2 atmosphere) for 24 h for adherence. The growth medium was thereafter removed and replaced with 100 ␮L blank DLX nanoparticle dispersed in DMEM:F12 (1:1) supplemented with 1% FBS in concentration range of 100 − 1483.3 ␮g/mL and were incubated for 24, 48 and 72 h. After the specified hours of incubation, the test nanoparticle dispersion was removed, cells were washed with 150 ␮L of PBS and incubated with 100 ␮L of NRU (final concentration of 0.05 mg/mL) or 100 ␮L of MTT (final concentration of 5 ␮g/mL) in incomplete DMEM:F12 (1:1) respectively for 3 h. The cells were washed with 150 ␮L of PBS and completely decanted before addition of 100 ␮L DMSO, for MTT assay or 150 ␮L of neutral red desorb solution (1:50:49 v/v of glacial acetic acid/ethanol/water) for NRU assay. The plates were agitated for 10 min before absorbance was read at 570 nm (MTT) and 540 nm (NRU) in a microplate reader (Cary 50 MPR, VARIAN Inc., USA). Cell viability (%) was calculated as described in Eq. (2) Cellviability (%) = Abssample Abscontrol × 100% (2) where Abssample represent measurements from wells treated with DLX nanoparticles and Abscontrol measurement from untreated wells. Results presented as mean ± SD were from quadruplicate determinations. Results obtained were subjected to statistical anal- ysis to determine significant differences between controls and treated wells using ANOVA (GraphPad Prism 5.0, GraphPad, USA). 2.10. In vitro cellular uptake studies Cellular uptake studies of BODIPY-loaded DLX nanoparticle were performed in ARPE-19 and HCE cell lines using the hydrophilic BODIPY. Flow cytometry, a laser based microfluidic cell analysis technology for cell counting and biomarker detection was deployed to evaluate/quantify the cellular uptake of the labeled DLX nanopar- ticles by the ARPE-19 and HCE cells while confocal microscopy technique was further used to affirm the results obtained from the flow cytometry. 2.10.1. Flow cytometry The ARPE-19 and HCE cells were seeded into a 6-well plate at a density of 3 × 105 cells/well and 4 × 105 cells/well in respective DMEM:F12 (1:1) complete medium and incubated (37 ◦C in a 5% CO2 atmosphere) for 24 h for the cells to adhere. The cells were thereafter washed with PBS and incubated with 1 mL of BODIPY- loaded DLX nanoparticle (corresponding to 5 mg/mL of BODIPY) dispersed in incomplete DMEM:F12 (1:1) medium for 24 h. After 24 h, the cells were washed twice with PBS, tripsinized with 300 ␮L
  • 5. A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1069 of tripsin (0.25%), neutralized with respective DMEM:F12 (1:1) complete medium, centrifuged and resuspended in PBS. Prior to analysis in the flow cytometer (BD FACSCanto I, USA), 5 ␮L of pro- pidium iodide was added to the samples. Analysis were performed at excitation wavelengths of 495 nm and 488 nm with emission wavelengths at 525 nm and 670 nm respectively for BODIPY and propidium iodide. Percent uptake were calculated based on the dot plots (BD-FACSDiva Software) obtained from the flow cytometry result. Results reported were average of quadruplicate determina- tions. 2.10.2. Confocal microscopy The ARPE-19 and HCE cells were seeded into a 6 well plates containing sterilized glass slips (22 mm x 22 mm) at a density of 3 × 105 cells/well and 4 × 105 cells/well in respective DMEM:F12 (1:1) complete medium and incubated (37 ◦C in a 5% CO2 atmo- sphere) for 24 h for adherence. The cells were washed with PBS and incubated for 24 h with 1 mL BODIPY-loaded DLX nanopar- ticle (corresponding to 5 mg/mL of BODIPY) or BODIPY solution (5 mg/mL) dispersed in DMEM:F12 (1:1) incomplete medium. Con- trol wells were treated with blank DLX nanoparticle dispersion. After treatment, the cells were washed twice with PBS, fixed with 2 mL paraformaldehyde (1%) per well and stored at 4 ◦C overnight protected from light. The cells were washed again with PBS and the glass slips were carefully lifted and placed inverted on glass slides containing a drop of DAPi. The slides were kept for 24 h protected from light before visualization in confocal microscope (Leica TCS SP8 Confocal microscope, Leica, Germany). It should be noted that the HCE cells were also incubated with loaded nanoparticles and BODIPY solution for 48 h in addition to 24 h experiment. 3. Results and discussion 3.1. DLX polymer characterizaiton The result shows that the carbohydrate content of DLXp was about 54% with trace amount of protein. However, DLXs used in the preparation of DLX nanoparticles and DLXu contains 99 and 98% sugars respectively; the sugars were predominantly mannose (Man) and galactose (Gal) (Supplementary material, Table S4.1). The Man:Gal ratio of DLXp, DLXs and DLXu was 4.6:1, 6.3:1 and 5.3:1 respectively (Supplementary material, Table S4.1); this result is in accordance with previous studies on DLX polymer (Bento et al., 2013). The difference between the amount of galactose present in DLXp and DLXs may be due to a beta elimination process in the presence of alkali (NaOH), a known phenomenon associated with alkali treatment of galactomannans (Bento et al., 2013). While the difference between the Man:Gal ratio of DLXs and DLXu may be attributed to the sonication process (Prajapat & Gogate, 2015), frac- tions of DLXs and DLXu analyzed by GPC showed a polydispersed polymer system with close molecular weight of ∼170 and 185 kDa respectively. Methylation analysis of the galactomannan from DLXs and DLXu were performed by a sequence of procedures in order to obtain fully O-methylated derivatives (Sassaki, Gorin, Souza, Czelusniak, & Iacomini, 2005). Hydrolysis of per-O-methylated derivatives produced mixtures of partially methylated monosac- charides: 2,3,4,6-Me,-Man, 2,3,4,6-Me,-Gal, 2,3,6-Me3-Man and 2,3-Me2-Man (Supplementary material, Fig. S4.6). These results are consistent with a main chain of (1–4)-linked mannopyranosyl units substituted at O6 of galactopyranosyl units. The galactomannans 1H NMR (Supplementary material, Figs. S4.2 and S4.3) and 13C NMR (Supplementary material, Figs. S4.4 and S4.5) spectroscopy anal- ysis showed that resonances in the 13C NMR spectra are in close agreement with those reported for other galactomannans (Bento Table 2 Hydrodynamic diameter, PdI and zeta potential of DLX polymeric nanoparticles. Data shown are mean ± SD of 3 replicates. Formulation Hydrodynamic diameter (nm) PdI Zeta potential (mV) F1 215.4 ± 3.8 0.192 ± 0.022 −38.5 ± 6.1 F2 315.2 ± 9.6 0.189 ± 0.004 −40.3 ± 8.1 F3 321.9 ± 11.2 0.215 ± 0.026 −62.5 ± 4.1 F4 252.7 ± 8.8 0.150 ± 0.031 −37.9 ± 4.3 F5 357.7 ± 8.5 0.214 ± 0.006 −65.2 ± 5.0 F6 276.3 ± 13.1 0.185 ± 0.008 −67.7 ± 6.4 F7 231.3 ± 5.8 0.140 ± 0.006 −39.9 ± 4.1 F8 266.7 ± 4.4 0.149 ± 0.020 −37.2 ± 5.9 F9 276.3 ± 13.1 0.185 ± 0.008 −67.7 ± 6.4 F10 225.7 ± 3.9 0.162 ± 0.009 −59.7 ± 8.5 F11 318.8 ± 10.6 0.175 ± 0.007 −68.8 ± 8.2 F12 260.1 ± 10.0 0.157 ± 0.016 −31.8 ± 3.1 F13 276.3 ± 13.1 0.185 ± 0.008 −67.7 ± 6.4 F14 261.1 ± 15.2 0.225 ± 0.021 −39.8 ± 2.4 F15 270.3 ± 12.5 0.176 ± 0.006 −49.1 ± 8.0 et al., 2013). The 1H- and 13C NMR spectral also demonstrated a close relationship in the structure of DLXs and DLXu, signifying minimal alteration due to sonication. The signals at the anomeric region of 1H NMR (Supplementary material, Figs. S4.2 and S4.3) indicates that the mannopyranosyl residues are ␤-linked while the galactopyranosyl residues are ␣-linked. The DLXu O-substituted resonances at C4 showed two clear peaks. The more abundant peak at ı 76.3 can be attributed to two contiguous unsubstituted O- mannopyranosyl residues while the peak at ı 77.59 may be ascribed to the superimposition of signals from diads, where only one of the two mannose residues is substituted. The same results were observed for DLXs, indicating an irregular distribution of Gal in the side chain of DLX galactomannan (Supplementary material, Fig. S4.6). 3.2. DLX nanoparticle physicochemical properties From the perspective of formulation design, various process and formulation variables can impart the product outcome of a nano- precipitation process to different extents, depending particularly on the interaction of the variables. Thus, the effects of the selected variables on the quality attributes of the DLX nanoparticle were examined. Table 2 shows the nanoparticle size, PdI and zeta poten- tial of the DLX nanoparticle formulations based on the DoE (Table 1). The DLX nanoparticle formulations showed mean particle size between 215 and 360 nm (Table 2) which is adequate for oph- thalmic applications (Vega et al., 2008). Particle size distribution was narrow in all cases as PdI was less than 0.230 indicating a system that is not very polydispersed. Zeta potential result shows that all nanoparticle formulations carry a negative charge smaller than −30 mV, indicating the presence of an adequate particle stabil- ity due to repulsive forces existing between particles (Veronovski, Andreozzi, La Mesa, & Sfiligoj-Smole, 2010). Neutral galactomannans like DLX in solution tends to have slightly negative values confirming their neutrality (Carneiro-da- Cunha, Cerqueira, Souza, Teixeira, & Vicente, 2011). The highly negative zeta potential of our DLX nanoparticles dispersion may be due to the addition of NaOH in DLX solution used to prepare the nanoparticles. Increase in the pH of a solution have been shown to have a significant influence on the zeta potential of nanoparticles (Carneiro-da-Cunha et al., 2011). DLX solution containing NaOH resulted in a dispersion with a pH higher than 7 which imparts a negative zeta potential on the nanoparticle. Thus, we successfully prepared nanoparticles of hydrophilic Delonix polymer using a simple and innovative nanoprecipitation process by using surfactant mix to control nanoparticle properties and without the use of toxic cross-linking agents such as glutaralde-
  • 6. 1070 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 hyde. This simple method could also be deployed in the preparation of other hydrophilic polymers of natural origin. Regression analysis and general linear modeling were per- formed on DLX nanoparticle size result with a view to understanding the relationship existing between the variables and nanoparticle size. With a 15-run Box Behnken design, all individ- ual variable effects and two-way interactions among the effects are estimable while the three-way interaction is used to estimate the error. Thus, the statistical model in Eq. (3) was used y = ˇo + ˇ1X1 + ˇ2X2 + ˇ3X3 + ˇ4X1X2 + ˇ5X1X3 + ˇ5X2X3 + ε (3) where y is the response (DLX nanoparticle size), X1, X2 and X3 (predictors) are polymer concentration, tween-span ratio and non- solvent/solvent ratio respectively, ␤o is the mean response when all variables are zero and ␤1-5 represent change in mean response per unit change in predictors respectively. From the regression analysis result (Supplementary material, Table S5.1), a general linear model for predicting the DLX nanopar- ticle size was derived from significant predictors as in Eq. (4). y = 275.0533 + 48.5625X1 − 17.3750X1X2 (4) Based on Eq. (4), it is easy to see that polymer concentra- tion significantly influenced the nanoparticle size (Supplementary material, Table S5.1). The parameter ␤1 estimated is also posi- tive, indicating that a higher polymer concentration will produce larger nanoparticle size. This result is in agreement with other studies as higher polymer concentration produce highly viscous solution which hampers the diffusion of solvent towards the non- solvent in a nanoprecipitation process (Bilati, Allemann, & Doelker, 2005; Dong, Chang, Wang, Tong, & Zhou, 2015). Nanoprecipitation involves the diffusion of one solvent into the other. It should be noted that the nanoprecipitation process in this study is a reverse of nanoprecipitation involving synthetic polymers such as PLGA as the DLX polymer was solubilized in the aqueous phase while the nanoparticles were formed in a less polar (ethanol) phase. The surfactant mix ratio and the non-solvent/solvent ratio did not significantly influence the DLX nanoparticle size as previously described for other nanoparticles prepared by the nanoprecipi- tation method (Ali & Lamprecht, 2013; Bilati et al., 2005). The importance of non-solvent/solvent proportions in nanoprecipita- tion processes is in solvent volume control (economics) (Bilati et al., 2005). However, there was an interacting influence of polymer con- centration and surfactant mix ratio on the DLX nanoparticle size (Eq. (4)). This effect trend is elucidated in the surface response plot in Fig. 1A. The surface plot shows that smaller DLX nanoparticle size are achieved at lower polymer concentration and surfactant mix ratio containing higher span 80 proportion. This result could be attributed partly to the low viscosity of polymer solution at lower concentrations and probably the orientation and proportions of the tween 80 and span 80 in the nanoparticle dispersion. Based on this result, we propose an hypothesis based on the orientation of tween and span in the nanoparticle dispersions. Hydrophilic polymers such as DLX polymer have the tendency to swell when in contact with aqueous medium. Thus, in the pres- ence of only or higher amounts of tween 80 (hydrophilic-lipophilic balance (HLB) = 15) in the surfactant mix, DLX nanoparticle could have direct contact with the aqueous medium, leading to ‘minute’ swellings that could increase size; however, with increase in span 80 (HLB = 4.3) ratio, the orientation of span 80 could reduce or prevent DLX nanoparticle contact with the dispersion medium, leading to reduction in swelling and consequently its size. A prob- able schematic representation of DLX nanoparticle prepared with a mixture of tween80:span80 is shown in Fig. S6.1 (Supplementary material, Section S6). In addition, other authors also proposed that changes in nanocapsule diameter with respect to tween and span Table 3 Surfactant mix influence on size of DLX nanoparticle containing different tween- span ratios. Data shown are the mean ± SD of 4 replicates. Tween 80:Span 80 Hydrodynamic diameter (nm) PdI Zeta Potential 1:0a,b 232.8 ± 3.6 0.185 ± 0.005 −64.6 ± 8.7 4:1b,c 215.4 ± 3.6 0.193 ± 0.003 −66.5 ± 3.9 3:2a,c 203.6 ± 6.1 0.156 ± 0.006 −58.9 ± 4.4 One-Way ANOVA with Tukey’s posttest: a p < 0.001. b p < 0.01. c p < 0.05. concentration could be dependent on surfactant packing parame- ters (Khoee & Yaghoobian, 2009; Mitchell & Ninham, 1981). To affirm the influence of tween-span ratio on DLX nanoparticles size and proposed swelling hypothesis for differences in nanopar- ticle size, DLX nanoparticles were prepared and swelling/stability study was performed. Table 3 shows the size of the DLX nanoparti- cles as a function of 3 different tween-span ratios. Results subjected to ANOVA indicate significant differences in nanoparticle size of the three formulations; with the formulation containing higher proportion of span 80 having the smallest nanoparticle size. Fig. 1B shows surfactant mix influence on DLX nanoparticle size increase as a function of time. It can be observed that DLX nanopar- ticle containing higher amount of span 80 (tween-span ratio 3:2) has a lower percent size increase as compared to the other nanopar- ticle formulations. A 2-Way ANOVA further indicate a significant difference in the percent size increase of the nanoparticle formula- tion, with nanoparticles containing tween-span ratio of 3:2 having the lowest percent size increase after 6 days. Therefore, the DLX nanoparticle formulation containing 3:2 tween-span ratio was cho- sen to prepare the DLX nanoparticles for all subsequent studies. Nanoparticle size (tween 80:span80, 3:2) based on TEM photo (Fig. 1C) analyzed with Fiji (ImageJ, USA) shows that DLX nanopar- ticle size ranges between 28 and 38 nm in size. The difference in the DLS and TEM sizes may be attributed partly to the DLS eval- uation being based on intensity determination of hydrodynamic diameter while TEM based on numbers. Also the hydrophilic and swelling nature of the DLX polymer might have influenced the size difference observed. 3.3. Stability of DLX nanoparticle in the presence of simulated lachrymal components The osmolarity of tear fluid is very important as it dictates the integrity of the cornea. The normal osmolarity of normal tears varies between 290 and 310 mOsm/kg (Marques, Loebenberg, & Almukainzi, 2011). The osmolarity of DLX nanoparticle dispersion determined using an osmometer (Semi-Micro Osmometer K-7400, KNAUER) was 220 mOsm/L, which is not hyperosmolar. The stability of colloidal biomaterials in biological fluids con- taining ample amount of enzymes and proteins is very crucial. Also, size of particles could play an important role in interaction with mucosal surfaces including ocular mucosa. To evaluate the stabil- ity of DLX nanoparticle in the presence of ocular fluid components, DLX nanoparticles were incubated with lysozyme, SLF and mucin. Lysozyme and mucin are two major components of the precorneal fluid. The particle size and zeta potential of DLX nanoparticle upon incubation with or without lysozyme was evaluated as a function of time (Fig. 2). Fig. 2 shows that nanoparticle size reduced upon incubation with lysozyme as compared to control without lysozyme. The size reduction could be attributed to the partial hydrolysis of the gly- cosidic bond in DLX polysaccharide structure which could result
  • 7. A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1071 Fig. 1. (A) Surface Plot of hydrodynamic diameter against DLX concentration and tween span ratio (B) Surfactant mix (tween 80:span 80) influence on DLX nanoparticle% size increase in function of time (2-Way ANOVA with Bonferroni multiple comparison test: a p < 0.001; b p < 0.01; c,d p < 0.05) (C) TEM photo of DLX nanoparticle (mag: 200,000). Fig. 2. Influence of lysozyme on DLX nanoparticle size and zeta potential incubated in lysozyme as a function of time. in minute break-off on the surface of the nanoparticle (de Campos et al., 2005; Hu, Chen, & Zhang, 2010). This result also suggests the biodegradability of the polymer; however, there was minimal nanoparticle size reduction (13%) over the period of 6 h. The size reduction of DLX nanoparticles in the presence of lysozyme can also be due to some electrostatic interactions between the negative nanoparticles and the cationic enzyme. Indeed, there was a signif- icant reduction in the zeta potential of the nanoparticles (Fig. 2), however the reduction would not adversely affect the nanoparti- cle stability as the values were larger than −30 mV. This stability is an advance in comparison to poly-␧-caprolactone nanocapsules which aggregate upon contact with lysozyme (Calvo, Vila-Jato, & Table 4 Physicochemical properties of DLX nanoparticle incubated with simulated lachry- mal fluid. DLX nanoparticle size (nm) Zeta potential (mV) Time (h) Control In SLF Control In SLF 0 209.6 ± 13.5 211.4 ± 15.5 −45.0 ± 2.2 −5.4 ± 0.5 1 206.6 ± 9.2 204.5 ± 14.6 −47.5 ± 3.4 −5.9 ± 0.3 2 203.0 ± 12.3 205.0 ± 8.9 −42.0 ± 0.8 −6.3 ± 1.3 3 207.5 ± 8.9 206.2 ± 8.5 −48.6 ± 1.7 −6.0 ± 0.7 4 205.1 ± 10.1 205.4 ± 13.3 −43.7 ± 1.9 −6.2 ± 0.4 5 204.8 ± 11.1 206.9 ± 10.0 −43.0 ± 0.9 −6.2 ± 0.9 6 203.4 ± 8.0 205.4 ± 7.7 −45.9 ± 2.7 −6.1 ± 0.7 Alonso, 1997; de Campos et al., 2005). Furthermore, the PdI was not affected by the presence of lysozyme. Table 4 shows the influence of SLF on DLX nanoparticles size and zeta potential as a function of time. The SLF has no signifi- cant effect on the nanoparticle size and PdI, however there was a significant reduction in the zeta potential of the DLX nanoparticle. This is understandable as the presence of mono and multivalent ions in the SLF could lead to compression of the electrical dou- ble layer and consequently a reduction in the negative charge on the nanoparticle surface (McConaughy, Stroud, Boudreaux, Hester, & McCormick, 2008; Carneiro-da-Cunha et al., 2011). Despite the reduction in zeta potential, no alterations in the size and in the PdI of nanoparticles were observed, confirming their stability in a physiological medium. This is an important advantage compared to chitosan nanoparticles, that are instable in phosphate buffer pH 7.4 (de Campos et al., 2005). The addition of mucin to DLX nanoparticle dispersion did not significantly change (p > 0.05) the zeta potential of the dispersion (DLX nanoparticles −42.3 ± 2.8 mV; DLX nanoparticle dispersion with mucin −38.1 ± 0.3 mV); however, there was a significant difference (p < 0.05) between the zeta potential of the mucin
  • 8. 1072 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 dispersion (-15.5 ± 0.5 mV) as compared to that containing DLX nanoparticle. Interactions between nanoparticles and mucin in terms of surface charge could be an indirect way of predict- ing mucoadhesion. Although, there was no significant interaction between the DLX nanoparticle and mucin, biopolymers mucoad- hesion to surfaces is a multi-mechanism process which does not only involve interactions between surface charges. Adsorption and diffusion have also been proposed as possible mechanism for mucoadhesion. While it might be interesting to conduct an in vivo ocular study to evaluate mucoadhesion prowess of DLX nanoparticles in addition to its probable mechanism of mucoadhe- sion (Smart, 2005; Serra, Domenech, & Peppas, 2009), study have already reported its use as a mucoadhesion enhancer (Devkar et al., 2014). 3.4. BODIPY-loaded DLX nanoparticle Table 5 shows properties of BODIPY-loaded DLX nanoparticle demonstrating the possibility of loading hydrophilic and lipophilic molecules into DLX nanoparticles. Nanoparticle size was < 240 nm, PdI was < 0.2 while zeta potential values was < −30 mV affirm- ing stability. However, blank nanoparticles (215.5 ± 4.6 nm) were smaller than the loaded DLX nanoparticles (hydrophilic BODIPY: 232.8 ± 11.1 nm; lipophilic BODIPY: 222.8 ± 9.1 nm). Entrapment efficiency of lipophilic BODIPY into DLX nanoparticle was signif- icantly low (≈ 6%) as compared to the hydrophilic BODIPY (≈ 69%). The entrapment efficiency can be explained by the partition coef- ficient (Log Poctanol/water) of the hydrophilic (-4.93) and lipophilic (-0.19) BODIPY. The Log Poctanol/water was calculated using a free MarvinSketch software (ChemAxon, Hungary). Although, the Mar- vinSketch software used in determining the LogPocatanol/water do have some limitations, particularly in relation to the structure of the BODIPYs as it did not take into account the core of the structure, thereby, making the values calculated only relative and ‘illustra- tive’; however, we have demonstrated that the lipophilic BODIPY is insoluble in water as shown in section 7 (Fig. S7.1) of the Supple- mentary material. In comparison to free lipophilic BODIPY which is obviously insoluble in aqueous medium, lipophilic BODIPY-loaded DLX nanoparticle homogeneously dispersed in aqueous medium (Sup- plementary material, Fig. S7.1). Thus, despite the encapsulation efficiency of lipophilic BODIPY in DLX nanoparticles being consid- erably low, the loaded DLX nanoparticles homogenously dispersed in aqueous medium. In vitro release from BODIPY-loaded DLX nanoparticles and BODIPY-DLX solution was studied. BODIPY-loaded DLX nanopar- ticles showed a burst release of 48.2 ± 6.84% while free BODIPY in DLX solution had a 97.4 ± 17.5% release within the first 30 min respectively. The BODIPY-loaded DLX nanoparticles released 69.5 ± 13.9% of BODIPY after 24 h as compared to the BODIPY solu- tion release of 97.4 ± 17.5% within the first 30 min. Hydrophilic polymers are known to generally swell and form a gel-like layer on their particle surface which retards fluid ingress into the poly- mer core thereby sustaining drug release (Sarmah et al., 2011). This result suggests that DLX nanoparticle exhibit significant sustained release (Supplementary material, Fig. S7.2) in comparison to BOD- IPY solution. The result also suggest that BODIPY release from DLX nanoparticles followed a Higuchi model as indicated by r2 value of 0.9745 (DDSolver software). 3.5. In vitro cell toxicity Determination of cell viability is a well recognized assay to eval- uate in vitro cytotoxicity of nano-materials. Cell viability in this study was assessed using neutral red and MTT uptake assays. The MTT assay is a rapid colorimetric and quantitative method based on the conversion of a yellow tetrazolium salt to insoluble purple formazan crystals in the mitochondrial of viable cells while neu- tral red is a dye taken up by viable cells and internalized within the lysosomes (Bouallagui, Han, Isoda, & Sayadi, 2011). The combi- nation of these two assay methods could have a benefit of adding reliability to cytotoxicity results of tested materials. A nanoparticle system with prospect for ocular use must be capable of delivering optimum amount of active agents without compromising the viability of the host cells. The result of MTT and NRU assays (Supplementary material, Fig. S8.1) suggest that DLX nanoparticles are non-toxic on both ARPE-19 and HCE cells at all tested concentrations (100–1483.3 ␮g/mL) within 24 h. ARPE- 19 showed reduction in cell viability after 48 and 72 h, but the reduction was only significant (p < 0.05) at DLX nanoparticle con- centration of 1483.3 ␮g/mL. Both MTT and NRU assay produced comparable results, giving reliability to the study. The HCE showed reduced cell viability after 48 and 72 h but the reduction was sig- nificantly different at 1483.3 ␮g/mL (p < 0.05) after 48 h; after 72 h, there was significant difference in cell viability at 1009 (p < 0.05) and 1483.3 ␮g/mL (p < 0.001). The MTT and NRU assay on HCE cells suggest same results. Based on the result, it could be said that in vitro DLX nanoparticle use on ARPE-19 and HCE cells did not show obvious toxic effects within the concentration limits in this study, however, more studies would be needed to ascertain its in vivo toxicity. 3.6. Cell uptake Cellular uptake is an important prerequisite for nanoparticle effectiveness and physicochemical characteristics such as size, surface charge, hydrophobicity dictates the extent of uptake of nanoparticles by cells. For uptake to occur, it is considered that there is an adhesion process followed by internalization. Cellular uptake of nanoparticle apart from dictating effectiveness, could also give insight on the toxicity propensity of nanoparticles (Zhao et al., 2011). Two methods were employed to assess the uptake of BODIPY-loaded DLX nanoparticles by ARPE-19 and HCE cell lines; flow cytometry and confocal microscopy. Fig. 3A shows flow cytom- etry results for hydrophilic BODIPY-loaded DLX nanoparticles in contact with ARPE-19 and HCE cells. Flow cytometry result as depicted by the dot plots in Fig. 3A2, Q4, suggests a 17.45 ± 1.14% uptake of BODIPY-loaded DLX nanoparti- cles by the ARPE-19 cells after 24 h of incubation while there is almost no uptake of the nanoparticles by the HCE cells. The low or no cellular uptake by the HCE cells could be due to the morpholog- ical tight junctions conformation of the cells after adherence and confluence (Yi, Wang, & Fu-Shin, 2000; Imayasu, Shiraishi, Ohashi, Shimada, & Cavanagh, 2008; Kimura, Teranishi, Fukuda, Kawamoto, & Nishida, 2008). Fig. 3B shows confocal micrographs of ARPE-19 cells incubated with BODIPY solution, blank DLX nanoparticle and BODIPY-loaded DLX nanoparticle. Confocal micrographs of HCE cells treated with these same formulations are shown in Supplementary material, Figs. S9.1 and S9.2. Confocal micrographs show a minimal uptake of BODIPY (faint green) by ARPE-19 or HCE cells treated with BODIPY solution (Fig. 3B5 and Supplementary material, Fig. S6.2 B2) as compared to the ARPE-19 cells treated with BODIPY-loaded DLX nanoparticle which showed bright color (Fig. 3B8) of the BODIPY after 24 h. Con- focal result of BODIPY-loaded DLX nanoparticle uptake by ARPE-19 did corroborate the results obtained from the flow cytometry experiment (Fig. 3A). Studies have also suggested nanoparticles possess effective cellular uptake when compared with their free drug solutions (Ahn, Seo, Kim, & Lee, 2013). Thus, the DLX nanopar- ticle improved the uptake of BODIPY by the ARPE-19 cells in comparison to control BODIPY solution, implying a possibility of
  • 9. A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1073 Table 5 Properties of BODIPY-loaded DLX nanoparticle containing tween-span ratio 3:2. Data shown are mean ± SD of 3 determinations. DLX nanoparticle Hydrodynamic diameter (nm) PdI Zeta Potential (mV) Encapsulation efficiency (%) Blank 215.5 ± 4.6 0.178 ± 0,046 −61.7 ± 8.2 – Loaded with hydrophilic BODIPY 232.8 ± 11.1 0.185 ± 0.009 −69.7 ± 7.5 68.6 ± 5.7 Loaded with lipophilic BODIPY 222.8 ± 9.1 0.132 ± 0.013 −65.6 ± 3.2 6.04 ± 1.62 Fig. 3. (A) Flow cytometry analysis: Dot plots showing uptake by ARPE-19 and HCE cells incubated for 24 h with BODIPY-loaded DLX nanoparticles (2 and 4) and blank DLX nanoparticles (1 and 3). (B) Confocal micrographs of ARPE-19 cells incubated for 24 h with blank DLX nanoparticles (1–3), 5 mg/mL BODIPY solution (4–6) and BODIPY-loaded DLX nanoparticles (7–9). Cell nuclei are stained blue with DAPi; BODIPY solution or BODIPY-loaded DLX nanoparticles appears green. (x 63 mag with immersion oil). Scale bar: 20 ␮m.(For better interpretation of this figure legend in colour, the reader is referred to the web version of the this article). using DLX nanoparticle as a potential drug carrier for retina related ocular disorders. Furthermore, differences between the uptake of BODIPY in solution and in DLX nanoparticles suggest that the dye visualized inside the cells are not due to only passive diffusion of BODIPY from the carrier. It is also important to consider the hydrophilicity of the BODIPY used in this experiments, which can hamper its passive redistribution. Therefore, there is a high possi- bility for the dye observed inside the cells being a result of the DLX nanoparticle uptake. This uptake noticed even after 24 h of experi- ment suggests that DLX nanoparticles may be useful for a sustained delivery of hydrophilic drugs in the posterior region of the eye. Confocal result also shows no uptake of BODIPY-loaded DLX nanoparticle by HCE cells after 24 h (Supplementary material, Fig. S9.1 B1). Treating the cells with BODIPY-loaded DLX nanoparticle for 48 h (Supplementary material, Fig. S9.2 B3) did not improve uptake either, thus corroborating the result of flow cytometry per- formed with the HCE cells. However, there was a minute uptake of the BODIPY solution by the cells after 48 h treatment (Fig. S9.2
  • 10. 1074 A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 B2). The low uptake of BODIPY from solution and no uptake from BODIPY-loaded nanoparticles could be due to the morphological tight junctions of the HCE cells as previously mentioned (Imayasu et al., 2008; Kimura et al., 2008; Yi et al., 2000). It could also be due to BODIPY metabolism as a function of the longtime of experiment. Therefore, if nanoparticles were taken up after 5 to 15 min incuba- tion, as shown in other study using HCE cells (Pretor et al., 2015), the probability of BODIPY degradation might be higher after cellular uptake. Although, there was no significant uptake by the HCE cells, in vitro release study in this work previously demonstrated the abil- ity of DLX nanoparticle to sustain the release of an encapsulated drug molecule. Thus it can be said that DLX polymer’s mucoadhe- sive (Devkar et al., 2014) and sustained release ability could favor its use topically in the eye, thereby increasing residence time and also sustaining drug release. 4. Conclusion This study demonstrated the possibility of obtaining nanopar- ticles from a natural hydrophilic polymer, DLX, using a simple and innovative modified nanoprecipitation technique. A gen- eral linear model for predicting the DLX nanoparticle size was proposed and showed that polymer concentration and the inter- action between polymer concentration and tween-span mix ratio influence the nanoparticle size. Based on the predicted model, nanoparticles composed of optimized concentration of DLX and surfactant mix ratio were prepared. These DLX nanoparticles were stable when incubated in SLF, lysozyme solution and mucin dis- persion, all components being present in the precorneal region of the eye. Nanoparticles were able to encapsulate both lipophilic and hydrophilic test molecules. In vitro release studies showed that DLX nanoparticle sustained the release of hydrophilic BODIPY. The ARPE-19 and HCE cells did not show obvious cellular toxicity to DLX nanoparticles after 24 h of treatment. While there was no visi- ble uptake of hydrophilic BODIPY-loaded DLX nanoparticles by HCE cells in the time the experiment was performed, DLX nanoparticle improved uptake of BODIPY by ARPE-19 cells. This study concluded that DLX nanoparticle could be a promising tool for ocular drug delivery. Competing financial interests The authors declare no competing financial interests. Acknowledgements This work was supported by the São Paulo Research Founda- tion (FAPESP grant #2014/22451-7) and The World Academy of Science-Brazilian National Council for Scientific and Technological Development (TWAS-CNPq grant #190007/2013-3). We also thank and acknowledge Guilherme L. Sassaki (PhD) from the NMR Center of the Universidade Federal do Parana, Parana, Brazil for analytical support. Appendix A. Supplementary data Supplementary data associated with this article can be found, in the online version, at http://dx.doi.org/10.1016/j.carbpol.2016.10. 076. References Achouri, D., Alhanout, K., Piccerelle, P., & Andrieu, V. (2013). Recent advances in ocular drug delivery. Drug Development and Industrial Pharmacy, 39(11), 1599–1617. http://dx.doi.org/10.3109/03639045.2012.736515 Adetogun, G. E., & Alebiowu, G. (2007). Influence of Delonix regia seed gum on the compressional characteristics of paracetamol tablet formulations. Journal of Drug Delivery Science and Technology, 17(6), 443–445. http://dx.doi.org/10. 1016/s1773-2247(07)50086-9 Adetogun, G. E., & Alebiowu, G. (2009). Properties of Delonix regia seed gum as a novel tablet binder. Acta Poloniae Pharmaceutica, 66(4), 433–438. Ahn, S., Seo, E., Kim, K., & Lee, S. J. (2013). Controlled cellular uptake and drug efficacy of nanotherapeutics. Scientific reports, 3, 1997. http://dx.doi.org/10. 1038/srep01997 Ali, M. E., & Lamprecht, A. (2013). Polyethylene glycol as an alternative polymer solvent for nanoparticle preparation. International Journal of Pharmaceutics, 456(1), 135–142. http://dx.doi.org/10.1016/j.ijpharm.2013.07.077 Anderson, J. M., & Shive, M. S. (2012). Biodegradation and biocompatibility of PLA and PLGA microspheres. Advanced Drug Delivery Reviews, 64, 72–82. http://dx. doi.org/10.1016/j.addr.2012.09.004 Bento, J. F., Mazzaro, I., de Almeida Silva, L. M., de Azevedo Moreira, R., Ferreira, M. L. C., Reicher, F., & de Oliveira Petkowicz, C. L. (2013). Diverse patterns of cell wall mannan/galactomannan occurrence in seeds of the Leguminosae. Carbohydrate Polymers, 92(1), 192–199. Betancur-Ancona, D., Pacheco-Aguirre, J., Castellanos-Ruelas, A., & Chel-Guerrero, L. (2011). Microencapsulation of papain using carboxymethylated flamboyant (Delonix regia) seed gum. Innovative Food Science & Emerging Technologies, 12(1), 67–72. http://dx.doi.org/10.1016/j.ifset.2010.11.002 Bhowmik, M., Kumari, P., Sarkar, G., Bain, M. K., Bhowmick, B., Mollick, M. M., & Chattopadhyay, D. (2013). Effect of xanthan gum and guar gum on in situ gelling ophthalmic drug delivery system based on poloxamer-407. International Journal of Biological Macromolecules, 62, 117–123. http://dx.doi. org/10.1016/j.ijbiomac.2013.08.024 Bilati, U., Allemann, E., & Doelker, E. (2005). Development of a nanoprecipitation method intended for the entrapment of hydrophilic drugs into nanoparticles. European Journal of Pharmaceutical Sciences, 24(1), 67–75. http://dx.doi.org/10. 1016/j.ejps.2004.09.011 Bouallagui, Z., Han, J., Isoda, H., & Sayadi, S. (2011). Hydroxytyrosol rich extract from olive leaves modulates cell cycle progression in MCF-7 human breast cancer cells. Food Chemical Toxicology, 49(1), 179–184. http://dx.doi.org/10. 1016/j.fct.2010.10.014 Bradford, M. M., Williams, W. L., 1977. Protein-assay reagent and method: Google Patents. Calvo, P., Vila-Jato, J., & Alonso, M. (1997). Effect of lysozyme on the stability of polyester nanocapsules and nanoparticles: Stabilization approaches. Biomaterials, 18(19), 1305–1310. Carneiro-da-Cunha, M. G., Cerqueira, M. A., Souza, B. W. S., Teixeira, J. A., & Vicente, A. A. (2011). Influence of concentration, ionic strength and pH on zeta potential and mean hydrodynamic diameter of edible polysaccharide solutions envisaged for multinanolayered films production. Carbohydrate Polymers, 85(3), 522–528. http://dx.doi.org/10.1016/j.carbpol.2011.03.001 Ciucanu, I., & Kerek, F. (1984). A simple and rapid method for the permethylation of carbohydrates. Carbohydrate Research, 131(2), 209–217. Devkar, T. B., Tekade, A. R., & Khandelwal, K. R. (2014). Surface engineered nanostructured lipid carriers for efficient nose to brain delivery of ondansetron HCl using Delonix regia gum as a natural mucoadhesive polymer. Colloids Surface B Biointerfaces, 122, 143–150. http://dx.doi.org/10.1016/j.colsurfb.2014. 06.037 Dey, S., & Mitra, A. K. (2005). Transporters and receptors in ocular drug delivery: Opportunities and challenges. Expert Opinion on Drug Delivery, 2(2), 201–204. http://dx.doi.org/10.1517/17425247.2.2.201 Dong, Y., Chang, Y., Wang, Q., Tong, J., & Zhou, J. (2015). Effect of operating conditions on size and morphology of amylose nanoparticles prepared by precipitation. Starch − Stärke, 67(3–4), 365–372. http://dx.doi.org/10.1002/ star.201400182 Dubois, M., Gilles, K. A., Hamilton, J. K., Rebers, P., & Smith, F. (1956). Colorimetric method for determination of sugars and related substances. Analytical Chemistry, 28(3), 350–356. Elzoghby, A. O., El-Fotoh, W. S., & Elgindy, N. A. (2011). Casein-based formulations as promising controlled release drug delivery systems. Journal of Controlled Release, 153(3), 206–216. http://dx.doi.org/10.1016/j.jconrel.2011.02.010 Gratieri, T., Gelfuso, G. M., Rocha, E. M., Sarmento, V. H., de Freitas, O., & Lopez, R. F. (2010). A poloxamer/chitosan in situ forming gel with prolonged retention time for ocular delivery. European Journal of Pharmaceutics and Biopharmaceutics, 75(2), 186–193. http://dx.doi.org/10.1016/j.ejpb.2010.02.011 Hu, R., Chen, Y. Y., & Zhang, L. M. (2010). Synthesis and characterization of in situ photogelable polysaccharide derivative for drug delivery. International Journal of Pharmaceutics, 393(1–2), 96–103. http://dx.doi.org/10.1016/j.ijpharm.2010. 04.011 Huang, S., & Fu, X. (2010). Naturally derived materials-based cell and drug delivery systems in skin regeneration. Journal of Controlled Release, 142(2), 149–159. http://dx.doi.org/10.1016/j.jconrel.2009.10.018 Imayasu, M., Shiraishi, A., Ohashi, Y., Shimada, S., & Cavanagh, H. D. (2008). Effects of multipurpose solutions on corneal epithelial tight junctions. Eye & Contact Lens, 34(1), 50–55. http://dx.doi.org/10.1097/ICL.0b013e318073cbdb Kapoor, V. (1972). A galactomannan from the seeds of Delonix regia. Phytochemistry, 11(3), 1129–1132. Khoee, S., & Yaghoobian, M. (2009). An investigation into the role of surfactants in controlling particle size of polymeric nanocapsules containing penicillin-G in double emulsion. European Journal of Medicine Chemistry, 44(6), 2392–2399. http://dx.doi.org/10.1016/j.ejmech.2008.09.045 Kimura, K., Teranishi, S., Fukuda, K., Kawamoto, K., & Nishida, T. (2008). Delayed disruption of barrier function in cultured human corneal epithelial cells
  • 11. A.T. Ogunjimi et al. / Carbohydrate Polymers 157 (2017) 1065–1075 1075 induced by tumor necrosis factor-alpha in a manner dependent on NF-kappaB. Investigative Ophthalmology and Visual Science, 49(2), 565–571. http://dx.doi. org/10.1167/iovs.07-0419 Kumari, A., Yadav, S. K., & Yadav, S. C. (2010). Biodegradable polymeric nanoparticles based drug delivery systems. Colloids Surface B Biointerfaces, 75(1), 1–18. http://dx.doi.org/10.1016/j.colsurfb.2009.09.001 Marques, M. R., Loebenberg, R., & Almukainzi, M. (2011). Simulated biological fluids with possible application in dissolution testing. Dissolution Technology, 18(3), 15–28. McConaughy, S. D., Stroud, P. A., Boudreaux, B., Hester, R. D., & McCormick, C. L. (2008). Structural characterization and solution properties of a galacturonate polysaccharide derived from aloe vera capable of in situ gelation . Biomacromolecules, 9(2), 472–480. Mitchell, D. J., & Ninham, B. W. (1981). Micelles, vesicles and microemulsions. Journal of the Chemical Society, Faraday Transactions 2: Molecular and Chemical Physics, 77(4), 601–629. http://dx.doi.org/10.1039/F29817700601 Miyazaki, S., Suzuki, S., Kawasaki, N., Endo, K., Takahashi, A., & Attwood, D. (2001). In situ gelling xyloglucan formulations for sustained release ocular delivery of pilocarpine hydrochloride. International Journal of Pharmaceutics, 229(1), 29–36. Nagarwal, R. C., Kant, S., Singh, P. N., Maiti, P., & Pandit, J. K. (2009). Polymeric nanoparticulate system: A potential approach for ocular drug delivery. Journal of Controlled Release, 136(1), 2–13. http://dx.doi.org/10.1016/j.jconrel.2008.12. 018 Nagarwal, R. C., Kumar, R., & Pandit, J. K. (2012). Chitosan coated sodium alginate-chitosan nanoparticles loaded with 5-FU for ocular delivery: In vitro characterization and in vivo study in rabbit eye. European Journal of Pharmaceutical Sciences, 47(4), 678–685. http://dx.doi.org/10.1016/j.ejps.2012. 08.008 Prajapat, A. L., & Gogate, P. R. (2015). Depolymerization of guar gum solution using different approaches based on ultrasound and microwave irradiations. Chemical Engineering and Processing: Process Intensification, 88, 1–9. Pretor, S., Bartels, J., Lorenz, T., Dahl, K., Finke, J. H., Peterat, G., . . . & Muller-Goymann, C. C. (2015). Cellular uptake of coumarin-6 under microfluidic conditions into HCE-T cells from nanoscale formulations. Molecular Pharmaceutics, 12(1), 34–45. http://dx.doi.org/10.1021/mp500401t Sahoo, S. K., Dilnawaz, F., & Krishnakumar, S. (2008). Nanotechnology in ocular drug delivery. Drug Discovery Today, 13(3–4), 144–151. http://dx.doi.org/10. 1016/j.drudis.2007.10.021 Sarmah, J. K., Mahanta, R., Bhattacharjee, S. K., Mahanta, R., & Biswas, A. (2011). Controlled release of tamoxifen citrate encapsulated in cross-linked guar gum nanoparticles. International Journal of Biological Macromolecules, 49(3), 390–396. http://dx.doi.org/10.1016/j.ijbiomac.2011.05.020 Sassaki, G. L., Gorin, P. A., Souza, L. M., Czelusniak, P. A., & Iacomini, M. (2005). Rapid synthesis of partially O-methylated alditol acetate standards for GC–MS: Some relative activities of hydroxyl groups of methyl glycopyranosides on Purdie methylation. Carbohydrate Research, 340(4), 731–739. Serra, L., Domenech, J., & Peppas, N. A. (2009). Engineering design and molecular dynamics of mucoadhesive drug delivery systems as targeting agents. European Journal of Pharmaceutics and Biopharmaceutics, 71(3), 519–528. http://dx.doi.org/10.1016/j.ejpb.2008.09.022 Smart, J. D. (2005). The basics and underlying mechanisms of mucoadhesion. Advance Drug Delivery Review, 57(11), 1556–1568. http://dx.doi.org/10.1016/j. addr.2005.07.001 Soumya, R. S., Ghosh, S., & Abraham, E. T. (2010). Preparation and characterization of guar gum nanoparticles. International Journal of Biological Macromolecules, 46(2), 267–269. http://dx.doi.org/10.1016/j.ijbiomac.2009.11.003 Tamaki, Y., Teruya, T., & Tako, M. (2010). The chemical structure of galactomannan isolated from seeds of Delonix regia. Bioscience Biotechnology Biochemistry, 74(5), 1110–1112. http://dx.doi.org/10.1271/bbb.90935 Vega, E., Gamisans, F., Garcia, M. L., Chauvet, A., Lacoulonche, F., & Egea, M. A. (2008). PLGA nanospheres for the ocular delivery of flurbiprofen: Drug release and interactions. Journal of Pharmaceutical Sciences, 97(12), 5306–5317. http:// dx.doi.org/10.1002/jps.21383 Veronovski, N., Andreozzi, P., La Mesa, C., & Sfiligoj-Smole, M. (2010). Stable TiO2 dispersions for nanocoating preparation. Surface and Coatings Technology, 204(9–10), 1445–1451. http://dx.doi.org/10.1016/j.surfcoat.2009.09.041 Yi, X.-, Wang, Y., & Fu-Shin, X. Y. (2000). Corneal epithelial tight junctions and their response to lipopolysaccharide challenge. Investigative Ophthalmology and Visual Science, 41(13), 4093–4100. Zhang, Y., Huo, M., Zhou, J., Zou, A., Li, W., Yao, C., & Xie, S. (2010). DDSolver: An add-in program for modeling and comparison of drug dissolution profiles. The AAPS Journal, 12(3), 263–271. http://dx.doi.org/10.1208/s12248-010-9185-1 Zhao, F., Zhao, Y., Liu, Y., Chang, X., Chen, C., & Zhao, Y. (2011). Cellular uptake, intracellular trafficking, and cytotoxicity of nanomaterials. Small, 7(10), 1322–1337. http://dx.doi.org/10.1002/smll.201100001 Zuo, Z., Lee, V. H. L., Controlled Drug Delivery: Pharmacokinetic Considerations, Methods and Systems (2008);10.1002/9780470048672. wecb099. de Campos, A. M., Diebold, Y., Carbalho, E. L., Sánchez, A., & José Alonso, M. (2005). Chitosan nanoparticles as new ocular drug delivery systems: In vitro stability, in vivo fate, and cellular toxicity. Pharmaceutical Research, 22(6), 1007. de la Fuente, M., Ravina, M., Paolicelli, P., Sanchez, A., Seijo, B., & Alonso, M. J. (2010). Chitosan-based nanostructures: A delivery platform for ocular therapeutics. Advance Drug Delivery Review, 62(1), 100–117. http://dx.doi.org/ 10.1016/j.addr.2009.11.026