You are on page 1of 9

Materials Science and Engineering C 68 (2016) 594–602

Contents lists available at ScienceDirect

Materials Science and Engineering C

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

Nanoencapsulation of dietary flavonoid fisetin: Formulation and in vitro


antioxidant and α-glucosidase inhibition activities
Mario Sechi a,b, Deeba N. Syed c, Nicolino Pala a, Alberto Mariani a, Salvatore Marceddu d, Antonio Brunetti e,
Hasan Mukhtar c, Vanna Sanna a,b,⁎
a
Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy
b
Laboratory of Nanomedicine, Department of Chemistry and Pharmacy, University of Sassari, c/o Porto Conte Ricerche, Tramariglio, 07041 Alghero, Italy
c
Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, USA
d
CNR – Istituto Scienze delle Produzioni Alimentari, Traversa La Crucca, 3 – Località Baldinca, 07040 Li Punti, Sassari, Italy
e
POLCOMING Department, Section of Information Engineering, University of Sassari, via Piandanna 4, 07100 Sassari, Italy

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

Article history: The bioactive flavonoid fisetin (FS) is a diet-derived antioxidant that is being increasingly investigated for its
Received 13 April 2016 health-promoting effects. Unfortunately, the poor physicochemical and pharmacokinetic properties affect and
Received in revised form 31 May 2016 limit the clinical application.
Accepted 12 June 2016
In this study, novel polymeric nanoparticles (NPs), based on Poly-(ε-caprolactone) (PCL) and PLGA-PEG-COOH,
Available online 14 June 2016
encapsulating FS were formulated as suitable oral controlled release systems. Results showed NPs having a mean
Keywords:
diameter of 140–200 nm, and a percent loading of FS ranging from 70 to 82%. In vitro release studies revealed that
Fisetin NPs are able to protect and preserve the release of FS in gastric simulated conditions, also controlling the release
Nanoparticles in the intestinal medium. Moreover, the DPPH and ABTS scavenging capacity of FS, as well as α-glucosidase inhi-
Controlled release bition activity, that resulted about 20-fold higher than commercial Acarbose, were retained during
Antioxidant activity nanoencapsulation process.
α-glucosidase inhibition In summary, our developed NPs can be proposed as an attractive delivery system to control the release of antiox-
idant and anti-hyperglycemic FS for nutraceutical and/or therapeutic application.
© 2016 Elsevier B.V. All rights reserved.

1. Introduction gluconeogenesis in vitro [10–12], mainly attributed to its antioxidant


capacities.
Flavonoids are a broadly distributed class of plant pigments that are In spite of the beneficial effects of FS in preventing and treating dis-
regularly consumed in the human diet. Due to their abundance in die- eases, its low water solubility, chemical instability, poor absorption, and
tary products and the potential pharmacological and nutritional effects, extensive and rapid metabolism, dramatically contribute to the low oral
the flavonoids have attracted considerable interest for drug develop- bioavailability, thus restraining its clinical application [13,14].
ment as well as nutraceutical application [1]. In this context, nanoencapsulation is one of the drug/nutraceutical
One such flavonoid, fisetin (FS; 3,3′,4′,7-tetrahydroxyflavone) (Fig. delivery processes that can help in overcoming these physicochemical
1), is found in various fruits and vegetables, such as strawberry, apple, and pharmacokinetic limitations.
persimmon, grape, onion, and cucumber [2]. To date, very few reports are available in the literature on nanosys-
It has been demonstrated that FS possesses multiple pharmacologi- tems for the encapsulation of FS. For example, nanoemulsion and lipo-
cal activities including anti-inflammatory [3,4], antihyperlipidemic [5], somal formulations have been developed by Chabot's group to
neuroprotective [6], antineoplastic, and chemopreventive properties improve the FS bioavailability and the antitumor efficacy in Lewis lung
[7,8]. carcinoma bearing mice [15–17]. It has also been reported that the en-
More recently, FS supplementation has been reported to decrease capsulation of FS into monomethyl poly (ethylene glycol)-poly (ε-
cardiovascular risks [9] and downregulate both glycogenolysis and caprolactone) copolymer micelles improved the therapeutic effect in
colon cancer [18].
In this scenario, the present study was aimed to formulate FS into
⁎ Corresponding author at: Department of Chemistry and Pharmacy, University of
novel polymeric nanoparticles (NPs), based on poly (ε-caprolactone)
Sassari, Via Vienna 2, 07100 Sassari, Italy. (PCL) and poly(D,L-lactic-co-glycolic acid)-block-poly(ethylene glycol)
E-mail address: vsanna@uniss.it (V. Sanna). carboxylic acid (PLGA-PEG-COOH), suitable as oral controlled release

http://dx.doi.org/10.1016/j.msec.2016.06.042
0928-4931/© 2016 Elsevier B.V. All rights reserved.
M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602 595

used as comparison. The obtained NPs suspension was used immediate-


ly for assay or lyophilized for storage at −50 °C.

2.3. Characterization of nanoparticles

2.3.1. Measurement of particle size (PS) and Polydispersity Index (PI)


PS and PI of the NPs were measured by using photon correlation
spectroscopy with a Zetasizer Nano ZS (Malvern Instruments,
Worcestershire, UK) at 25 °C, and a scattering angle of 90°. The samples
were diluted with deionized water and sonicated for 5 min before mea-
surement. Each analysis was carried out in triplicate.

2.3.2. Scanning Electron Microscopy (SEM) analysis


Fig. 1. Chemical structure of fisetin (FS). Morphological examination of NPs was performed by Scanning Elec-
tron Microscopy (SEM) (model DSM 962; Carl Zeiss Inc., Germany). A
drop of NPs aqueous suspension was placed on aluminum stub and
systems. NPs were characterized for morphology, size, FS loading, FTIR dried under vacuum for 12 h. The samples were then analyzed at
and XRD analyses, and in vitro release studies in gastrointestinal simu- 20 kV acceleration voltage after gold sputtering, under an argon
lated fluids. In order to evaluate the retention of FS activity after encap- atmosphere.
sulation, the DPPH and ABTS radical scavenging activities of
encapsulated FS were assessed in comparison to those of free FS. Fur- 2.3.3. FS loading (DLC%), entrapment efficiency (EE%), and yield of NPs
thermore, the α-glucosidase inhibition capacity of FS and FS-loaded (YP%)
NPs was evaluated and compared to commercial drug Acarbose. The amount of FS encapsulated was determined by dissolving an al-
iquot of NPs (1.5 mg) in 1 mL of acetonitrile: EtOH mixture (7:3, v/v).
The solution was analyzed by UV–vis spectroscopy (Cary 3, Varian) at
2. Materials and methods
350 nm, and calculated by referring to the calibration curve (standard
solutions in the range of 1.25–50 μg/mL; R2 = 0.999). The DLC%, EE%,
2.1. Materials
and YP% were presented by the following equations, respectively:
Poly-(ε-caprolactone) (PCL), 2,2-diphenyl-1-picryl hydrazyl DLC% ¼ ðweight of drug in NPs=weight of NPsÞ  100
(DPPH), 2,20-azinobis(3-ethylbenzothiazoline-6-sulphonic acid-)
diammonium salt (ABTS), sodium persulfate, α-glucosidase from Sac- EE% ¼ ðactual RSV content=theoretical RSV contentÞ  100
charomyces cerevisiae (type I; 10 U/mg protein), p-nitrophenyl α-D-
glucopyranoside (pNPG), and trolox (6-hydroxy-2,5,7,8-
YP% ¼ ðweight of NPs recovered=weight of polymer and RSV fed initiallyÞ
tetramethylchromane-2-carboxylic acid) were obtained from Sigma-  100
Aldrich (Steinheim, Germany). Fisetin (FS), from leaves of Rhus succeda-
nea L. (N 98% purity), was purchased from Shaanxi Taiji Huaqing Tech-
nology Co., Ltd. (Shaanxi, People's Republic of China). Acarbose was
2.3.4. Fourier transform infrared spectroscopy (FTIR)
obtained from Carbosynth Limited (Berkshire, UK). The PLGA-PEG-
The chemical composition of FS, unloaded, and FS-loaded NPs was
COOH block copolymer was prepared and characterized as previously
analyzed by FTIR spectroscopy using a Vertex 70 Bruker spectropho-
reported [19]. All solvents and other chemicals were obtained from
tometer equipped with a RT-DTGS detector and a KBr beam splitter.
Sigma-Aldrich, were analytical grade and were used without further
The spectra were recorded in the 400–4000 cm−1 range with a resolu-
purification.
tion of 4 cm−1.

2.2. Preparation of nanoparticles 2.3.5. X-ray diffraction (XRD)


The physical state of the pure FS, PCL and PLGA-PEG-COOH poly-
Fisetin from leaves of Rhus succedanea L. was loaded into NPs by mers, empty and loaded NPs, as well as the FS-polymers physical mix-
modification of a nanoprecipitation method previously described [19]. tures was analyzed by XRD (Bruker — Mod. D2 Phaser), using CuK α-
Briefly, polymers at different weight ratio and FS were dissolved in radiation (λ = 1.5418 Å) and operating at 30 kV and 10 mA. The X-
3 mL of acetonitrile. The organic solution was then added dropwise ray scans were performed in open angle 2θ in the range from 10 to
under magnetic stirring to an aqueous solution of Pluronic F-127 100°, with the scanning rate at 1°/min.
(0.1%, wt.%), giving a final polymer concentration of 5.0 mg/mL.
Table 1 reports the total amount of solid materials used for the prep- 2.4. In vitro release studies
aration of FS-loaded NPs.
The resulting suspension was stirred at room temperature to remove FS-loaded NPs were characterized by in vitro release kinetic at differ-
the organic solvent, and NPs were centrifuged at 5000 rpm for 5 min, ent pH conditions: at pH 1.2 for 2 h, followed by pH 7.4, to simulate gas-
washed three times with water and concentrated. Unloaded NPs tric and intestinal fluids, respectively. About 2.0 mg of FS-loaded NPs
batches (F01, F02, and F03) were produced in a similar manner and were placed into dialysis bags and suspended in 15 mL of release

Table 1
Theoretical composition (wt.%) of prepared loaded NPs.

Batch PCL PLGA-PEG-COOH FS

F1 95.0 – 5.0
F2 66.5 28.5 5.0
F3 47.5 47.5 5.0
596 M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602

media, then incubated at 37 °C and stirred at 200 rpm. At predetermined (3.0 mM) was prepared in 20 mM phosphate buffer, pH 6.9. One hun-
time intervals, 1 mL of samples were withdrawn and replaced with dred μL of α-glucosidase (0.1 U/mL) was mixed with 50 μL of the differ-
equal volume of the corresponding fresh media to maintain a constant ent concentrations of the free and extracted FS (15.6–250 μg/mL), and
volume. Samples were filtered, and EtOH (4%, v/v) was added in order the mixture was incubated at 37 °C for 10 min. After pre-incubation,
to ensure the complete solubilization of FS released. 50 μL of pNPG solution was added to start the reaction. The reaction
FS concentration was detected spectrophotometrically at 350 nm, mixture was incubated at 37 °C for additional 10 min and stopped by
thus calculated by referring to the respective calibration curve with adding 2 mL of 0.1 M Na2CO3. The α-glucosidase activity was deter-
standard solutions in the range of 1.25–50 μg/mL, prepared by using mined by measuring the yellow colored para-nitrophenol released
0.1 N HCl:EtOH (96:4, v/v) for pH 1.2 conditions (R2 = 0.9992), and from pNPG at 405 nm. The α-glucosidase inhibition activity was calcu-
PBS:EtOH (96:4, v/v), for pH 7.4 conditions (R2 = 0.9991). Each exper- lated as follow:
iment was performed in triplicate.
Glucosidase inhibition ð%Þ ¼ ½ðAc −As Þ=Ac   100
2.5. Determination of antioxidant activity
where Ac is the absorbance of control and As is the absorbance of sam-
In order to evaluate the antioxidant activity of FS and its retention ples, respectively.
after encapsulation, the DPPH and ABTS radical scavenging activities Commercial inhibitor Acarbose (1.25–20 mg/mL) was used as a pos-
were investigated on free FS and on FS after extraction from NPs (FS- itive control and distilled water as a negative control. All samples were
NPs) by dissolving a known amount of loaded-NPs in a mixture of assayed in triplicate. As a measure of potency of the inhibitors tested,
ACN:EtOH (4:1, v/v), and diluting them to give different final concentra- IC50 values were calculated from the enzyme activity data and defined
tions of FS. as the concentration of an inhibitor required for reducing 50% of the en-
zyme activity obtained from activity vs concentration plot.
2.5.1. DPPH radical scavenging activity
DPPH scavenging activity of FS before and after encapsulation was 2.7. Statistical analysis
determined according to the methods previously reported, with minor
changes [20–22]. A solution of DPPH (0.1 mM, 1.4 mL) in 80% aqueous All data were subjected to one-way analysis of variance (ANOVA)
methanol and 0.1 mL of sample (free FS and FS-NPs) at different concen- (GraphPadPrism, version 5.03). Individual differences were evaluated
trations (1.9–40 μg/mL) were mixed in a cell. The absorbance was mea- using a nonparametric post hoc test (Tukey's test) and considered sta-
sured at 517 nm, and then the solution has been allowed to stand in the tistically significant at P b 0.05.
dark for 30 min. Trolox, a water-soluble derivative of vitamin E, in the
concentration range of 5–200 μg/mL, was tested as a positive control, 3. Results and discussion
and distilled water as negative control. Additionally, the corresponding
empty NPs were evaluated considering the same concentration of the 3.1. Formulation and characterization of nanoparticles
polymers as in FS-loaded NPs.
The scavenging percentage was calculated using the following for- In this study, FS-loaded NPs were prepared by a nanoprecipitation
mula: technique using PCL and PLGA-PEG-COOH as carriers. These biodegrad-
able and biocompatible polymers have been commonly used to encap-
DPPH scavenging effect ð%Þ ¼ ½ðAc −As Þ=Ac   100 sulate hydrophobic drugs, which often display poor oral bioavailability
[27].
where Ac and As are absorption of control (distilled water) and sample In our previous studies, NPs based of PCL alone or in combination
applied DPPH at 517 nm, respectively. with other polymers (PLGA-PEG-COOH, Alginate) have been developed
as carriers for the encapsulation of resveratrol for prostate cancer treat-
2.5.2. ABTS radical cation scavenging activity ment [19], and white tea extract for nutraceutical application [22].
The radical scavenging activity of samples against the ABTS radical As shown in Table 2, the mean hydrodynamic diameters range from
cation was measured using the previously reported methods with 140 to 200 nm, depending on the composition of the NPs. The larger
some modifications [23,24]. A sodium persulfate stock solution particle size observed for batches containing PLGA-PEG-COOH could
(6.89 × 10− 3 M, 1.0 mL) was added to ABTS stock solution be related to the presence on NPs surface of hydrophilic PEG chains
(5.00 × 10−4 M, 99.0 mL) and stored in the dark for 16 h before use. that orient themselves towards the external aqueous phase, thus deter-
Fifty microlitres of samples at different concentrations (2.5–30 μg/mL) mining an increased of hydrodynamic diameter [28]. Furthermore, all
was mixed with 1.45 mL of ABTS solution. After reaction in darkness NPs were characterized by PI values b0.3, indicating a narrow and
at 37 °C for 10 min, the absorbance at 734 nm was measured. unimodal distribution of particles, as also depicted in Fig. 2.
Trolox (3.1–50 μg/mL) and distilled water were used as a positive The SEM images (Fig. 3) revealed that the NPs are fine dispersed
and negative control, respectively. Furthermore, empty NPs were tested with a well-defined spherical shape. Additionally, not significant differ-
considering the same concentration of the polymers as in FS-NPs. ences were observed for the size and morphology of blank NPs (F01-
The scavenging activity was calculated as follows: F03) with respect to corresponding FS-loaded batches (data not
shown).
ABTS scavenging effect ð%Þ ¼ ½ðAc −As Þ=Ac   100 The encapsulation efficiencies values (Table 2) were found to be 82%,
75%, and 70%, for F1, F2, and F3, respectively.
where Ac is absorbance of a control (distilled water) lacking any radical These findings indicated that composition significantly influences
scavenger, and As is absorbance of the remaining ABTS•+ in the pres- the loading capacity of FS into NPs (4.1%, 3.74%, and 3.49%, for F1, F2,
ence of a scavenger [25]. and F3, respectively). In particular, the PCL ensures a better encapsula-
tion due to high affinity of hydrophobic FS, which decreased with a pro-
2.6. α-Glucosidase inhibition assay gressive increasing of more hydrophilic PLGA-PEG-COOH and the
reduction of PCL content in the formulations.
The effect of the free and FS-NPs on α-glucosidase activity was de- Furthermore, the nanoprecipitation technique provided good yields
termined according to the previously described procedure [26]. The of production that resulted of about 70%, without significant differences
substrate solution p-nitropheynyl-α-D-glucopyranoside (pNPG) within different NPs batches.
M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602 597

Table 2
Particle size (PS), Polydispersity Index (PI), encapsulation efficiencies (EE), Drug (FS) loading content (DLC), and yields of production (YP) of formulated NPs. *Significant differences (P b
0.05) between loaded NPs. Data are means ± SD, n = 3.

Batch PS (nm) PI EE (%) DLC (%) YP (%)

F01 143.2 ± 4.3 0.142 ± 0.03 – – 73.32 ± 3.2


F1 146.2 ± 2.3* 0.123 ± 0.05 81.96 ± 3.8* 4.10 ± 0.2* 71.10 ± 2.0
F02 203.21 ± 5.2 0.123 ± 0.04 – – 76.44 ± 4.5
F2 198.7 ± 6.0* 0.158 ± 0.02 74.78 ± 1.9* 3.74 ± 0.1 74.76 ± 2.4
F03 170.22 ± 3.9 0.112 ± 0.01 – – 70.32 ± 3.8
F3 165.4 ± 3.3* 0.148 ± 0.02 69.76 ± 2.8* 3.49 ± 0.1* 72.49 ± 2.7

The qualitative composition of NPs was investigated by FT-IR spec- C–C stretching (ring C), C–O stretching (ring A, B), C–O–H and C–C–H
troscopy (Fig. 4). bending modes (ring A, B) have been observed, and correlated to the
According to the literature [29], the most intense bands of the FS frequency 1163 cm−1. Finally, the bands at 1110 and 1017 cm−1 are
spectrum have been found at 1610 cm−1 region, attributed to combina- assigned to C–C–H, C–O–H, and C–C–C bending vibrations of the ring B.
tion of C_O (ring C), C–C (ring A, B), and O–C (ring B) stretching modes,
and at 1575 cm− 1, due to C–C (ring A, C), C2_C3 (ring C), and C_O
(ring C) stretching vibration. The bands at 1514 and 1448 cm−1 involve
O–C and C–C stretching of ring B and ring A, respectively. Medium in-
tensity bands at 1385 cm−1 are assigned to C–C–O and C–O–H bending
vibrations of the ring C. The signals centered at 1326 cm−1 are attribut-
able to C–C stretching, C3′–OH, and C4′–OH bending vibrations of the
ring B. Again, the band at 1265 cm−1 corresponds to C–C and C–O
stretching, while band at 1206 cm−1 overally involve C–O stretching
(ring A, C) and C–O–H bending of the ring A (7-OH). Furthermore, the

Fig. 2. Particle size distributions of F1 (A), F2 (B), and F3 (C) NPs. Fig. 3. SEM photographs of F1 (A), F2 (B), and F3 (C) NPs.
598 M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602

For F01 NPs, containing only PCL, it was possible to detect strong The XRD pattern of PLGA-PEG-COOH copolymer displays two peaks
bands such as the C_O stretching mode near 1725 cm−1, and bands at angles of 19.06 and 23.01, and two diffuse broad bands in the 2θ re-
at 2949–2865, 1293, 1240, and 1190 cm−1 corresponding to CH2, C–C, gion at 29.87° and 39.95°.
asymmetric C–O–C, and O–C–O stretching vibrations, respectively The diagram of empty NPs (F01) resulted similar to the pure PCL,
[30]. The spectrum of F03 NPs, containing PCL and PLGA-PEG-COOH confirming that the crystalline structure of PCL is retained and not
mixture, in addition to F0 spectrum, shows the strong signal at changed into amorphous phase during the preparation of NPs. On the
1757 cm−1, assigned to C_O stretching vibrations. As previously re- other hand, disappearance of characteristic FS peaks in the XRD patterns
ported [19], the FTIR spectrum of PLGA-PEG-COOH shows an absorption of the FS-loaded NPs (F1 and F3) further suggest that FS crystalline na-
band at 3300–3500 cm−1, due to the terminal OH groups, and bands ture is converted into amorphous or disordered crystalline form, be-
around 3000 cm−1 and 2860 cm−1, due to C–H stretching of CH3 and cause of the formation of an amorphous complex with the
CH2, respectively. The strong signal observed at 1757 cm−1 is assigned intermolecular interaction between FS and polymers (Fig. 5A and B).
to C–O stretching, the absorption peaks at 1640 and 1556 cm−1 are as- The amorphous state of FS in the polymer matrix is more soluble than
sociated to C–O and C–N of amide bond, while the band centered at crystalline form, due to free energies involved in the dissolution process,
1190 cm−1 is due to C–O stretch (data not shown). leading to improved absorption of FS. Furthermore, the absence of crys-
In F1- and F3-loaded NPs spectra, most of the absorption peaks from talline peak in loaded NPs further confirms that NPs are free from sur-
F01 and F03-unloaded NPs and bands at 1650–1580 cm−1 of pure FS o- face adsorbed FS [33].
verlapped, suggesting that no strong chemical interaction occurred be- Conversely, as also depicted in Fig. 5B, the patterns of physical mix-
tween FS and polymers. tures of PCL-FS (mix 1) and PCL-PLGA-PEG-COOH-FS (mix 2) revealed a
To investigate the crystal transformation of the nanoparticle system superimposition of the patterns of raw materials with both sharp crys-
and to get more information about the interactions between FS and talline peaks of FS and peaks of PCL, thus indicating no change in FS crys-
polymers, a series of powder XRD analyses were performed. Fig. 5A dis- talline state during the mixing process.
play the XRD patterns of pure PCL, pure PLGA-PEG-COOH, pure FS, NPs
(F01, F1 and F3), and those of the physical mixtures of PCL-FS and PCL- 3.2. In vitro release studies
PLGA-PEG-COOH-FS.
The diagram of PCL shows two distinctive peaks at angles of 21.81 Conventional oral administration of flavonoids appears to be ineffi-
and 24.17, and two small additional peaks at 16.08 and 30.37° degrees cient, based on various issues such as low dissolution rate from solid
2θ [31]. dosage forms, partial degradation in the harsh pH conditions of the gas-
The characteristic peaks of pure FS appeared at a diffraction angle of tric environment, poor permeability, and extensive first pass metabo-
2θ 11.54°, 12.89°, 14.40°, 15.75°, 16.70°, 17.72°, 18.44°, 19.38°, 20.73°, lism before reaching systemic circulation [34].
21.49°, 22.44°, 23.05°, 23.90°, 24.26°, 24.78°, 26.53°, and 28.71°, thus Several evidences have revealed that polymeric NPs are suitable car-
suggesting that the FS is present as a highly crystalline structure [32]. riers for the oral administration of bioactive flavonoids because improve
the solubilization and physico-chemical stability, enhance the bioavail-
ability by increasing the absorption from enterocytes, and maintain the
therapeutic levels in blood and plasma, with a significant improvement
in mean residence time as well as bioavailability [35,36].
To evaluate the potential of the prepared NPs for oral administration
of FS, we performed the in vitro release test in simulated gastro-intesti-
nal conditions. As reported in Fig. 6, during the first two hours at acidic
pH the FS released from NPs was b 15% in all cases. This suggests that
most of the FS in the nanoprototypes would be available to be absorbed
within the intestinal tract, and protected from the harsh gastric fluid. No
difference was found in the FS release from all NPs. However, at pH 7.4, a
slight and gradual increase in the FS release was observed in all formu-
lations. In particular, F2 and F3, containing PLGA-PEG-COOH, showed a
similar behavior, with about 70% of FS released after 7 h, and an almost
complete release after 24 h. On the other hand, in the F1 formulation,
based on PCL alone, a lower percentage of FS released was found, with
54% dissolved after 7 h incubation, and about 70% after 24 h. The high
release from F2 and F3 batches can be attributed to the presence of
more hydrophilic PLGA-PEG-COOH, which ease the diffusion of the re-
lease medium into the NPs, compared to F1, containing only hydropho-
bic PCL.

3.3. Antioxidant activity

The diet-derived antioxidant FS is now being increasingly investigat-


ed for its health-promoting effects [2]. Due to specific structural fea-
tures, FS exhibits strong antioxidant properties, mainly due to its
hydroxyl groups at C-3, C-3′, C-4′ and C-7 positions, with the influence
of the carbonyl group at C-4. The presence of double bond between C-2
and C-3 conjugated with the 4-oxo group also facilitates higher electron
delocalization [37].
As far as the radical-scavenging activity of flavonoids is concerned, it
is related to the presence of phenolic hydrogens, which are potentially
Fig. 4. Comparison of FT-IR spectra of pure FS and F01, F1, F03, and F3 NPs, chosen as able to quench free radicals by forming resonance-stabilized phenoxyl
examples. radicals [38].
M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602 599

Fig. 5. Comparison of XRD diagrams of pure PCL, pure PLGA-PEG-COOH, pure FS, empty NPs (F01), loaded NPs (F1 and F3), physical mixtures of PCL-FS (mix 1) and PCL-PLGA-PEG-COOH-
FS (mix 2) (A). Magnified XRD patterns of pure PCL, pure FS, F1 NPs, and mix 1, chosen as examples, to show detail peaks from 10° to 20° of 2θ (B).

In particular, the DPPH molecules are characterized as a stable free free radical scavenging efficiency after encapsulation into NPs. Percent-
radicals due to the delocalisation of the spare electron over the molecule age inhibition of DPPH ranged from 65% at maximum FS concentration
as a whole, that also gives rise to the deep violet colour. Mixing DPPH so- (40 μg/mL) to about 5% at a lower concentration (2.5 μg/mL), with sim-
lution with a substance that can donate a hydrogen atom (like flavo- ilar IC50 values (30.4 ± 0.35 and 31.2 ± 0.52 μg/mL, P N 0.05) for free FS
noids) leading to the reduced form, with the loss of violet colour [39]. and FS-NPs, respectively.
Similarly, the decolorization of the blue-green ABTS radical cation, The DPPH• scavenging effect of positive control Trolox at different
generated by reacting a strong oxidizing potassium persulfate with concentrations (5–200 μg/mL ) is reported in Fig. 8. The percentage in-
the ABTS salt, reflects the capacity of an antioxidant species to donate hibition of DPPH ranged from 90%, detected at higher doses of Trolox
electrons or hydrogen atoms, which are needed to inactivate this radical to 3.5% (the lower doses), with IC50 values of 112.08 ± 3.4 μg/mL,
species [40]. which is approximately 3.5 times lower than that of free and
The radical scavenging activity of FS before and after encapsulation nanoencapsulated FS.
was evaluated by DPPH and ABTS free radicals by using F1 NPs, chosen The data derived from the comparison on the ABTS•+ scavenging
as model. The comparison on the DPPH scavenging ability of native FS ability for free FS and FS-NPs, detected from 30 to 2.5 μg/mL concentra-
and FS extracted from NPs (FS-NPs), from 40 to 2.5 μg/mL concentra- tions, are shown in Fig. 9. Similarly to DPPH inhibition, the ABTS scav-
tions, is presented in Fig. 7. enging effect resulted concentration dependent, with a percentage
The results clearly show that FS scavenged DPPH radicals in a dose- inhibition of about 75% at 30 μg/mL FS concentration that linearly de-
dependent manner, also sharing a good linear correlation between the creased to 7% for lowest concentration, and without significant differ-
DPPH scavenging activity and the FS concentration. It is worth noting ences between free and encapsulated FS. The calculated IC50 values
that no statistical significance between free FS and FS-NPs was observed were found to be 18.6 ± 0.54 and 20.1 ± 0.24 μg/mL, P N 0.05) for
at all tested concentrations, suggesting that FS retained the same DPPH free FS and FS-NPs, respectively.

Fig. 6. In vitro release profiles of F1, F2, and F3 NPs performed in 0.1 N HCl (pH 1.2) for 2 h, Fig. 7. Comparison of DPPH• scavenging effect of FS before (free FS) and after
followed by PBS (pH 7.4). Data are means ± SD, n = 3. encapsulation (FS-NPs) at different concentrations. Data are means ± SD, n = 3.
600 M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602

As presented in Fig. 10, the standard Trolox shows a maximum ABTS


inhibition percentage of about 70%, and a minimum inhibition of 4.5% at
50 and 3.1 μg/mL, respectively.
The calculated IC50 values resulted 36.80 ± 1.6 μg/mL, significantly
lower (about 2-fold) with respect to that of the FS, clearly confirming
that FS is a potent scavenger of free radicals in vitro.
Furthermore, in order to evaluate the influence of polymers on the
DPPH and ABTS assays, further tests were performed using empty NPs
(F01, F02, and F03) at the same concentration of the polymers present
in FS-loaded NPs. At all tested concentrations the absorbance values ob-
served for empty NPs are identical to those of the negative control (dis-
tilled water), indicating that the polymeric materials used did not cause
interference with the antioxidant tests.

3.4. α-Glucosidase inhibition

The α-glucosidase inhibitor Acarbose is widely used in the treatment


Fig. 9. Comparison of ABTS•+ scavenging effect of FS before (free FS) and after
of patients with type 2 diabetes. This drug inhibits the upper gastrointes- encapsulation (FS-NPs) at different concentrations. Data are means ± SD, n = 3.
tinal glucosidases enzymes that convert complex polysaccharide carbo-
hydrates into monosaccharides in a dose-dependent fashion, resulting flavonoids, structurally related to FS, are crucial in engaging direct bind-
in a delayed glucose absorption and a lowering of postprandial hypergly- ing within the active-site residues. Additionally, the 3-OH of C ring is
cemia. However, gastrointestinal side effects, including mainly flatulence predicted to play a role in maintaining the proper binding orientation
and sometimes soft stools or abdominal discomfort, have been reported of flavonoid molecules, whereas the hydroxyl groups located on B ring
as a limiting factor for patients treatment [41,42]. would tightly interact in a hydrophobic pocket of the enzyme [44].
Herein, the α-glucosidase inhibitory activity of free and encapsulat-
ed FS was compared to that of Acarbose, used as positive control. 4. Conclusions
As reported in Fig. 11A, the highest concentration of Acarbose
(20 mg/mL) showed a maximum inhibition percentage of 95%, while In summary, in this work, the natural flavonoid FS was efficiently en-
the lowest concentration of 1.0 mg/mL showed a minimum inhibition capsulated into polymeric NPs by a nanoprecipitation method. NPs pro-
of 46.7%. Furthermore, similar to literature data [43], the half maximal vide a high FS loading capacity and are able to control the FS release in
inhibitory concentration (IC50) resulted of nearly 1.25 mg/mL equiva- simulated gastro-intestinal conditions, thus offering the possibility of
lent to 1.94 mM. On the other hand, FS exerted higher α-glucosidase in- the oral administration of nanosystems. Moreover, the results demon-
hibitory activity than Acarbose, with strong inhibition percentages strated that the scavenging capacity of FS as well as α-glucosidase inhi-
ranging from 93% to about 40% for the tested concentrations of 250– bition activity could be preserved during nanoencapsulation processing.
15.6 μg/mL (Fig. 11B). Additionally, the IC50 values of FS resulted Furthermore, FS showed a better α-glucosidase inhibition capacity than
0.030 mg/mL, equivalent to 0.1 mM, significantly lower (about 20 Acarbose, supporting the potential of FS for postprandial glycaemic con-
fold) with respect to commercial Acarbose. These findings suggest the trol. These findings indicate that the developed NPs can be proposed as
potential application of FS as dietary means for postprandial glycaemic an attractive delivery system to control the release of antioxidant and
control in type-2 diabetics. anti-hyperglycemic FS, with the goal to improve its pharmacological
Concerning the mechanism of action of FS in inhibiting α-glucosi- properties also for prevention and treatment of various diseases.
dase, we assumed that it would behave similarly to other flavonoids
(i.e. quercetin, myricetin), previously identified as putative inhibitors.
Acknowledgments
The structural determinants involved on the interaction between flavo-
noids and the enzyme have been previously explored using the compu-
The authors gratefully acknowledge the Regione Autonoma della
tational molecular docking approach [44]. Briefly, from the literature
Sardegna for the financial support of Grant CRP 25920, awarded to
analysis it emerges that the 3′,4′-dihydroxyl groups of B ring in
M.S. within the frame of “Legge regionale n. 7/2007 - Annualità 2010”.

Fig. 8. DPPH• scavenging effect of standard Trolox at different concentrations. Data are Fig. 10. ABTS•+ scavenging effect of Trolox at different concentrations. Data are means ±
means ± SD, n = 3. SD, n = 3.
M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602 601

[12] G.S. Prasath, S.I. Pillai, S.P. Subramanian, Fisetin improves glucose homeostasis
through the inhibition of gluconeogenic enzymes in hepatic tissues of
streptozotocin induced diabetic rats, Eur. J. Pharmacol. 740 (2014) 248–254.
[13] C.S. Shia, S.Y. Tsai, S.C. Kuo, Y.C. Hou, P.D. Chao, Metabolism and pharmacokinetics of
3,3′,4′,7-tetrahydroxyflavone (fisetin), 5-hydroxyflavone, and 7-hydroxyflavone
and antihemolysis effects of fisetin and its serum metabolites, J. Agric. Food Chem.
57 (2009) 83–89.
[14] N. Mignet, J. Seguin, G.G. Chabot, Bioavailability of polyphenol liposomes: a chal-
lenge ahead, Pharmaceutics 5 (2013) 457–471.
[15] H. Ragelle, S. Crauste-Manciet, J. Seguin, D. Brossard, D. Scherman, P. Arnaud, G.G.
Chabot, Nanoemulsion formulation of fisetin improves bioavailability and
antitumour activity in mice, Int. J. Pharm. 427 (2012) 452–459.
[16] N. Mignet, J. Seguin, M. Ramos Romano, L. Brullé, Y.S. Touil, D. Scherman, M.
Bessodes, G.G. Chabot, Development of a liposomal formulation of the natural flavo-
noid fisetin, Int. J. Pharm. 423 (2012) 69–76.
[17] J. Seguin, L. Brullé, R. Boyer, Y.M. Lu, M. Ramos Romano, Y.S. Touil, D. Scherman, M.
Bessodes, N. Mignet, G.G. Chabot, Liposomal encapsulation of the natural flavonoid
fisetin improves bioavailability and antitumor efficacy, Int. J. Pharm. 444 (2013)
146–154.
[18] Y. Chen, Q. Wu, L. Song, T. He, Y. Li, L. Li, W. Su, L. Liu, Z. Qian, C. Gong, Polymeric mi-
celles encapsulating fisetin improve the therapeutic effect in colon cancer, ACS Appl.
Mater. Interfaces 7 (2015) 534–542.
[19] V. Sanna, I.A. Siddiqui, M. Sechi, H. Mukhtar, Resveratrol-loaded nanoparticles
based on poly(epsilon-caprolactone) and poly(D , L -lactic-co-glycolic acid)-
poly(ethylene glycol) blend for prostate cancer treatment, Mol. Pharm. 10
(2013) 3871–3881.
[20] I. Gülçin, Antioxidant properties of resveratrol: a structure-activity insight, Innova-
tive Food Sci. Emerg. Technol. 11 (2010) 210–218.
[21] V. Sanna, N. Pala, G. Dessì, P. Manconi, A. Mariani, S. Dedola, M. Rassu, C. Crosio, C.
Iaccarino, M. Sechi, Single-step green synthesis and characterization of gold-conju-
gated polyphenol nanoparticles with antioxidant and biological activities, Int. J.
Nanomedicine 9 (2014) 4935–4951.
[22] V. Sanna, G. Lubinu, P. Madau, N. Pala, S. Nurra, A. Mariani, M. Sechi, Polymeric nano-
particles encapsulating white tea extract for nutraceutical application, J. Agric. Food
Chem. 63 (2015) 2026–2032.
[23] H. Zhao, W. Chen, J. Lu, M. Zhao, Phenolic profiles and antioxidant activities of com-
mercial beers, Food Chem. 119 (2010) 1150–1158.
[24] V. Sanna, L. Pretti, Effect of wine barrel ageing or sapa addition on total polyphenol
content and antioxidant activities of some Italian craft beers, Int. J. Food Sci. Technol.
50 (2015) 700–707.
[25] I. Gülçin, R. Elias, A. Gepdiremen, L. Boyer, Antioxidant activity of lignans from
fringe tree (Chionanthus virginicus L.), Eur. Food Res. Technol. 223 (2006)
759–767.
[26] R. Subramanian, M.Z. Asmawi, A. Sadikun, In vitro alpha-glucosidase and alpha-am-
ylase enzyme inhibitory effects of Andrographis paniculata extract and
andrographolide, Acta Biochim. Pol. 55 (2008) 391–398.
Fig. 11. Comparison of α-glucosidase inhibition percentage of Acarbose (A) and FS (B)
[27] E. Marin, M.I. Briceño, C. Caballero-George, Critical evaluation of biodegradable poly-
before (free FS) and after encapsulation (FS-NPs) at different concentrations. Data are
mers used in nanodrugs, Int. J. Nanomedicine 8 (2013) 3071–3091.
means ± SD, n = 3.
[28] J.V. Jokerst, T. Lobovkina, R.N. Zare, S.S. Gambhir, Nanoparticle PEGylation for imag-
ing and therapy, Nanomedicine (London) 6 (2011) 715–728.
[29] J.M. Dimitrić Marković, Z.S. Marković, D. Milenković, S. Jeremić, Application of com-
parative vibrational spectroscopic and mechanistic studies in analysis of fisetin
References structure, Spectrochim. Acta A Mol. Biomol. Spectrosc. 83 (2011) 120–129.
[30] L. Azouz, F. Dahmoune, F. Rezgui, C. G'Sell, Full factorial design optimization of anti-
[1] G.R. Beecher, Overview of dietary flavonoids: nomenclature, occurrence and intake, inflammatory drug release by PCL-PEG-PCL microspheres, Mater. Sci. Eng. C. Mater.
J. Nutr. 133 (2003) 3248S–3254S. Biol. Appl. 58 (2016) 412–419.
[2] N. Khan, D.N. Syed, N. Ahmad, H. Mukhtar, Fisetin: a dietary antioxidant for health [31] H. Ge, Y. Hu, S. Yang, X. Jiang, C. Yang, Preparation, characterization, and drug release
promotion, Antioxid. Redox Signal. 19 (2013) 151–162. behaviors of drug-loaded ε-caprolactone/L-lactide copolymer nanoparticles, J. Appl.
[3] H.H. Park, S. Lee, J.M. Oh, M.S. Lee, K.H. Yoon, B.H. Park, J.W. Kim, H. Song, S.H. Kim, Polym. Sci. 75 (2000) 874–882.
Anti-inflammatory activity of fisetin in human mast cells (HMC-1), Pharmacol. Res. [32] J.Q. Zhang, K.M. Jiang, K. An, S.H. Ren, X.G. Xie, Y. Jin, J. Lin, Novel water-soluble
55 (2007) 31–37. fisetin/cyclodextrins inclusion complexes: preparation, characterization, molecular
[4] O.L. Woodman, E.C. Chan, Vascular and anti-oxidant actions of flavonols and fla- docking and bioavailability, Carbohydr. Res. 418 (2015) 20–28.
vones, Clin. Exp. Pharmacol. Physiol. 31 (2004) 786–790. [33] J.B. Mendes, M.K. Riekes, V.M. de Oliveira, M.D. Michel, H.K. Stulzer, N.M. Khalil, S.F.
[5] I. Myara, I. Pico, B. Vedie, N. Moatti, A method to screen for the antioxidant effect of Zawadzki, R.M. Mainardes, P.V. Farago, PHBV/PCL microparticles for controlled re-
compounds on low-density lipoprotein (LDL): illustration with flavonoids, J. lease of resveratrol: physicochemical characterization, antioxidant potential, and ef-
Pharmacol. Toxicol. Methods 30 (1993) 69–73. fect on hemolysis of human erythrocytes, Sci. World J. 542937 (2012).
[6] P. Maher, T. Akaishi, K. Abe, Flavonoid fisetin promotes ERK-dependent long-term [34] D.V. Ratnam, D.D. Ankola, V. Bhardwaj, D.K. Sahana, M.N.V. Ravi Kumar, Role of an-
potentiation and enhances memory, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) tioxidants in prophylaxis and therapy: a pharmaceutical perspective, J. Control. Re-
16568–16573. lease 113 (2006) 189–207.
[7] N. Khan, M. Asim, F. Afaq, Z.M. Abu, H. Mukhtar, A novel dietary flavonoid fisetin in- [35] G. Leonarduzzi, G. Testa, B. Sottero, P. Gamba, G. Poli, Design and development of
hibits androgen receptor signaling and tumor growth in athymic nude mice, Cancer nanovehicle-based delivery systems for preventive or therapeutic supplementation
Res. 68 (2008) 8555–8563. with flavonoids, Curr. Med. Chem. 17 (2010) 74–95.
[8] I. Murtaza, V.M. Adhami, B.B. Hafeez, M. Saleem, H. Mukhtar, Fisetin, a natural [36] A. Bilia, B. Isacchi, C. Righeschi, C. Guccione, M. Bergonzi, Flavonoids loaded in
flavonoid, targets chemoresistant human pancreatic cancer AsPC-1 cells nanocarriers: an opportunity to increase oral bioavailability and bioefficacy, Food
through DR3-mediated inhibition of NF-kappa B, Int. J. Cancer 125 (2009) Nutr. Sci. 5 (2014) 1212–1327.
2465–2473. [37] Z.S. Marković, S.V. Mentus, J.M. Dimitrić Marković, Electrochemical and density
[9] Y. Cho, J.H. Chung, H.J. Do, H.J. Jeon, T. Jin, M.J. Shin, Effects of fisetin supplementa- functional theory study on the reactivity of fisetin and its radicals: implications on
tion on hepatic lipogenesis and glucose metabolism in Sprague-Dawley rats fed on in vitro antioxidant activity, J. Phys. Chem. A 113 (2009) 14170–14179.
a high fat diet, Food Chem. 139 (2013) 20–727. [38] S. Dudonné, X. Vitrac, P. Coutière, M. Woillez, J.M. Mérillon, Comparative study of
[10] R.P. Constantin, J. Constantin, C.L. Pagadigorria, E.L. Ishii-Iwamoto, A. Bracht, K. Ono antioxidant properties and total phenolic content of 30 plant extracts of industrial
Mde, N.S. Yamamoto, The actions of fisetin on glucose metabolism in the rat liver, interest using DPPH, ABTS, FRAP, SOD, and ORAC assays, J. Agric. Food Chem. 57
Cell Biochem. Funct. 28 (2010) 149–158. (2009) 1768–1774.
[11] G.S. Prasath, S.P. Subramanian, Modulatory effects of fisetin, a bioflavonoid, on hy- [39] C.A. Rice-Evans, N.J. Miller, G. Paganga, Structure-antioxidant activity relationships
perglycemia by attenuating the key enzymes of carbohydrate metabolism in hepatic of flavonoids and phenolic acids, Free Radic. Biol. Med. 20 (1996) 933–956.
and renal tissues in streptozotocin-induced diabetic rats, Eur. J. Pharmacol. 668 [40] M.N. Alam, N.J. Bristi, M. Rafiquzzaman, Review on in vivo and in vitro methods
(2011) 492–496. evaluation of antioxidant activity, Saudi Pharm. J. 21 (2013) 143–152.
602 M. Sechi et al. / Materials Science and Engineering C 68 (2016) 594–602

[41] H. Laube, Acarbose, Clin. Drug Investig. 22 (2002) 141–156. 50% ethanolic extracts and sinensetin from Orthosiphon stamineus Benth as anti-di-
[42] C. Rosak, G. Mertes, Critical evaluation of the role of acarbose in the treatment of di- abetic mechanism, BMC Complement. Altern. Med. 12 (2012) 176.
abetes: patient considerations, Diabetes Metab. Syndr. Obes. 5 (2012) 357–367. [44] H. Xu, Inhibition kinetics of flavonoids on yeast α-glucosidase merged with docking
[43] E.A. Mohamed, M.J. Siddiqui, L.F. Ang, A. Sadikun, S.H. Chan, S.C. Tan, M.Z. Asmawi, simulations, Protein Pept. Lett. 17 (2010) 1270–1279.
M.F. Yam, Potent α-glucosidase and α-amylase inhibitory activities of standardized

You might also like