You are on page 1of 9

LWT - Food Science and Technology 162 (2022) 113474

Contents lists available at ScienceDirect

LWT
journal homepage: www.elsevier.com/locate/lwt

Digestion stability of curcumin-loaded nanostructured lipid carrier


Ji Eun Hyun a, Hye-Yoon Yi a, Geun-Pyo Hong b, Ji-Yeon Chun a, *
a
Department of Food Bioengineering, Jeju National University, Jeju, 63243, South Korea
b
Department of Food Science and Biotechnology, Sejong University, Seoul, 05006, South Korea

A R T I C L E I N F O A B S T R A C T

Keywords: Curcumin is known as a functional substance with various effects such as antioxidant, anti-inflammatory, and
Nanostructured lipid carrier anticancer. However, it is not easily utilized due to low water solubility and bioavailability. Therefore, this study
Curcumin encapsulated into a nanostructured lipid carrier (NLC) with MCT oil of coconut to improve the solubility of
In vitro digestion
insoluble curcumin. The physicochemical properties and stability of curcumin NLCs were observed, and the in
MCT oil
vitro gastrointestinal stability of curcumin NLCs was also studied. The produced curcumin NLCs had a hydro­
dynamic diameter of 126.9 nm, zeta potential of − 25.9 mV, PDI of 0.246, and encapsulation efficiency of 94.5%.
TGA and DSC analysis were performed to confirm the thermal properties (pyrolysis temperature (◦ C), weight loss
(%), melting temperature (◦ C), and crystallinity, etc.) of the components constituting the curcumin NLCs.
Through this, it was confirmed that curcumin was completely entrapped in the NLC structure. In the digestive
imitation system, NLCs released approximately 31% of curcumin in the imitation saliva step, and curcumin
release gradually increased. This study showed that NLCs were an effective carrier to apply in various fields such
as the functional food industry, because they increased the water solubility of curcumin and could also improve
bioavailability and curcumin storage stability.

1. Introduction intermediate carbon-length saturated fats are bound to the glycerol


backbone (Saarela, 2011b), and they are between six and twelve carbons
Curcumin is a major component of yellow spices contained in the in length. The absorption and transport of MCT differ from long chain
roots of turmeric (Curcumac longae rhizoma) and curcuma roots (Cur­ triglycerides (LCTs), which are over twelve carbons in length. They
cuma Longa Radix) (Jo, Kim, & Kwon, 2016). Curcumin is a type of present different metabolic pathways of absorption and
polyphenol and has a molecular weight of 368.37 g/mol and a melting gastric-intestinal digestion. MCT has been applied and studied as an
point of 183 ◦ C (Araiza-Calahorra, Akhtar, & Sarkar, 2018). It is widely effective lipid type for various studies on weight control (Henry, 2007),
used as a natural pigment, spice, and medicine in India and other Asian obesity (Saarela, 2011b), functional fat (Saarela, 2011a), functional
countries (Araiza-Calahorra et al., 2018). In addition, curcumin has health benefits (Rastall, 2007), etc. MCTs are a thinning fluid,
anti-inflammatory (Selvam, Jachak, Thilagavathi, & Chakraborti, light-yellow color, clearness, non-flavor and they are usually made from
2005), anticancer (Fujisawa, Atsumi, Ishihara, & Kadoma, 2004), anti­ coconut or palm kernel.
oxidant (Ak & Gulcin, 2008), neuropathic pain suppression (Kim, Park, Solid lipid nanoparticles (SLNs), developed in the 1990’s are
& Jeon, 2008), skin sclerosis relief, and psoriasis relief, to prevent skin colloidal particles that maintain the solid-state at room temperature by
cancer (Aggarwal, Surh, & Shishodia, 2007; Balasubramanian & Eckert, replacing the liquid lipid of the emulsion with a solid lipid. SLNs have
2006) and antialcoholism functions (Nanji et al., 2003). Despite having excellent physical stability to protect sensitive drugs (Madane &
various health functionalities, curcumin has poor water solubility (11 Mahajan, 2014), high bioavailability, good biocompatibility and low
ng/mol, at ambient temperature), low oral bioavailability, chemical toxicity (Yuan et al., 2007). However, SLNs have low encapsulation ef­
instability, and photolysis. Furthermore, it has low membrane perme­ ficiency due to lack of space to capture drug materials and encapsulated
ability and has difficulties in rapid metabolism and excretion (Hussain drug is easily released because the crystallinity of solid lipids increases
et al., 2017). during long-term storage (Behbahani, Ghaedi, Abbaspour, Rostamiza­
Medium chain triglycerides (MCTs) are lipids in which three deh, & Dashtian, 2019; Muller, Radtke, & Wissing, 2002).

* Corresponding author. Department of Food Bioengineering, Jeju National University, Jeju-si, 63243, South Korea.
E-mail address: chunjiyeon@jejunu.ac.kr (J.-Y. Chun).

https://doi.org/10.1016/j.lwt.2022.113474
Received 1 October 2021; Received in revised form 8 April 2022; Accepted 18 April 2022
Available online 21 April 2022
0023-6438/© 2022 Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
J.E. Hyun et al. LWT 162 (2022) 113474

Nanostructured lipid carriers (NLCs) were developed to compensate absorbance on y-axis and curcumin concentration on x-axis (R2 =
for the deficiency of SLNs. NLCs form by containing liquid lipids (oil) 0.9998). To remove free curcumin NLCs dispersion was mixed with ethyl
with solid lipids (fat) at room temperature, therefore, NLCs have a lower acetate (1:1, v/v) and then vortexed for a few seconds. The mixture was
melting point than SLNs (Jun, Lim, & Jin, 2015). NLCs are partially centrifuged (3,134×g, 10 min, 25 ◦ C) and the supernatant, which
crystallized and have a less ordered crystalline structure (Tamjidi, included free curcumin was removed. The remained curcumin NLCs
Shahedi, Varshosaz, & Nasirpour, 2013). Therefore, the solubility of the dispersion was mixed with methanol and then centrifuged (3,134×g, 40
active ingredients and the drug loading capacity are increased minimize min, 4 ◦ C). The middle part of curcumin NLCs dispersion was filtered by
the leakage of the active compound during storage due to the amorphous using a syringe filter (0.45 μm), and then the absorbance was deter­
solid structure of NLCs (Tamjidi et al., 2013; Varshosaz, Eskandari, & mined by UV–Vis spectrophotometer (Epoch™, BioTek Instruments, Inc,
Tabakhian, 2010). Therefore, NLCs may increase encapsulation effi­ Winooski, USA) at λmax = 423 nm. Encapsulation efficiency of curcumin
ciency, drug loading and physical stability and may improve the was calculated as follows (Eq. (1)):
chemical stability, bioavailability, and controlled release of functional
EC(g)
lipophilic compounds in food. In addition, NLCs can include hydro­ EE (%) = × 100 (1)
IC (g)
phobic and hydrophilic active substances and various types of NLCs are
manufactured according to the mixing ratio of solid lipids and liquid
where EC is from the middle part of curcumin NLCs dispersion and IC is
lipids in the oil phase of NLCs (Muller et al., 2002; Sezer, 2012).
the initial amount of curcumin at NLCs preparation.
In this study, insoluble curcumin was encapsulated into NLCs with
MCT oil from coconut to improve their solubility and availability. The
physicochemical properties and stability of curcumin NLCs were 2.5. Total phenolic content
observed, and the in vitro gastrointestinal stability of curcumin NLCs was
also studied. This test was performed by modifying the Folin-Ciocalteu method of
Singleton and Rossi (1965). In order to evaluate only the curcumin
2. Material and methods collected in NLCs, free curcumin was removed in the same way as in the
encapsulation efficiency (EE%) experiment and used as a sample.
2.1. Reagents and standard Briefly, 0.2 mL of sample was mixed with a 0.9 mL distilled water, fol­
lowed by the addition of 0.1 mL of 2 M Folin-Ciocalteu’s phenol reagent
The curcumin (≥65%), Tween 80, Span 80, glycerol tristearate, so­ and incubated for 5 min, protected from light. Subsequently, 0.3 mL of
dium hydroxide solution, pancreatin from porcine pancreas, pepsin Na2CO3 (2 g/100 mL) and 0.5 mL of distilled water were added and the
porcine gastric mucosa, monobasic potassium phosphate, uric acid so­ mixture left at protected from light of room temperature for 1 h before
dium salt, sodium DL-lactate and Folin-Ciocalteu reagent were purchased the absorbance was measured at 760 nm. The results were calculated by
from Sigma-Aldrich Chemical Co., Ltd. MCT oil (Palm Kernel oil 99.9%, using a standard curve (R2 = 0.9988) which was built by dissolving
canola oil 0.1%) was provided from Korea Medical Foods Co Ltd., and gallic acid in distilled water at different concentrations (100, 200, 300,
Ethyl acetate purchased Junsei Chemical Co., Ltd (Japan). Hydrochloric 400, 500 and 600 μg/mL) and expressed as μg of gallic acid equivalents
acid (HCl) solution was supplied by Yakuri Pure Chemicals Co., Ltd. per mL sample (μg GAE/mL).
(Kyoto, Japan).
2.6. Thermogravimetric analysis (TGA) and differential scanning
2.2. Preparation of nanostructured lipid carriers (NLCs) calorimetry (DSC)

Aqueous dispersion was Tween 80 300 mg in distilled water 100 mL. Blank NLCs and Curcumin NLCs were used after lyophilization for
Oil dispersion was glycerol tristearate (solid lipid) 600 mg/100 mL, MCT analysis. The thermal degradation of glycerol tristearate, MCT oil, cur­
oil (liquid lipid) 240 mg/100 mL, curcumin 3 mg/100 mL and Span 80 cumin, blank NLCs, and curcumin NLCs were analyzed by TGA Q500
300 mg/100 mL. Each dispersion was mixed in magnetic stirring ho­ (TA Instruments, USA). Samples were heated at a rate of 10 ◦ C/min from
mogenization 80 ◦ C, 350 rpm for 10 min and then oil dispersion was room temperature to 500 ◦ C. Thermograms of the curcumin NLCs
immediately dispersed dropwise into the aqueous dispersion. The components were obtained using a DSC Q20 (TA Instruments, USA).
mixture was kept 80 ◦ C and mixed high speed homogenizer (T-25D, IKA, Samples (2–3 mg) were heated from 0 to 200 ◦ C at the scan rate of 10 ◦ C/
Germany) at 8,000 rpm for 5 min. Finally, the mixture was homogenized min. TA Universal analysis software (v 4.5A, TA Instruments, USA) was
by high pressure homogenizer (Picomax MN400, Micronox, Korea) at used to analyze data for both TGA and DSC.
20,000 psi, 3 cycle and cooled to room temperature. After preparation,
each NLC dispersion was stored at 5 ◦ C, 21 ± 2 ◦ C, 40 ◦ C, 65 ◦ C for 28 2.7. Preparation of simulated gastrointestinal fluids (saliva, gastric, small
days. intestinal fluids)

2.3. Nanostructured lipid carriers particle characteristic The in vitro digestion model (Fig. 1) used was the modification of
those described previously (Mao & McClements, 2012). Simulated saliva
The samples were diluted to distilled water (1:9 v/v). The hydro­ fluid (SSF, pH 6.8) consisted of mucin and various salts (Table 1) and pH
dynamic diameter (nm) and PDI were measured by dynamic light was adjusted by using 1 mol/L sodium hydroxide (NaOH). The SSF was
scattering (DLS) on DelsaMax Pro (Beckman Coulter, USA) at 25 ◦ C. The continuously shaken to heat at 105 rpm and 37 ◦ C for 15 min. The
zeta potential (mV) was measured by electrophoretic light scattering simulated gastric fluid (SGF) conditions were defined by the ‘United
(ELS) on DelsaMax Pro. The samples were measured at least 3 times. States Pharmacopeia and National Formulary 200’ method (United
States Pharmacopeial Convention, 2009). For the SGF condition, 0.7 mL
2.4. Encapsulation efficiency (EE%) of 1 mol/L hydrochloric acid (HCl) was added to 90 mL of deionized
water, and 200 mg of sodium chloride (NaCl) was dissolved in the HCl
The encapsulation efficiency method (Riaz et al., 2019; Yu et al., solution. Then, 302 mg of pepsin from porcine gastric mucosa (≥2,500
2016) was modified. The standard calibration curve of curcumin was Units/mg) was added in the HCl–NaCl solution. The pH of SGF was
obtained by measuring the absorbance of curcumin solution in con­ adjusted to 1.2. The simulated small intestinal fluid (SIF) conditions
centration range prepared from stock solutions in methanol at 423 nm in were defined by the ‘United States Pharmacopeia and National Formu­
triplicate. Calibration curve of curcumin was then plotted with lary 200’ method. For the SIF condition, 680 mg of monobasic potassium

2
J.E. Hyun et al. LWT 162 (2022) 113474

Fig. 1. Preparation of simulated gastrointestinal fluids and protocol of curcumin release in in vitro lipid digestion.

2.9. Statistical analysis


Table 1
Chemical composition of artificially simulated saliva fluid (Mao & McClements,
All experiments were carried out at least three times. The results are
2012).
reported as means ± standard deviation. Data were analyzed by one-
Chemicals Chemical formula Concentration (g/L) way ANOVA followed by Tukey’s multiple range test at p values of
Sodium chloride NaCl 1.594 <0.05 using Minitab ver. 17(PA, USA) statistical tests.
Ammonium nitrate NH4NO3 0.328
Potassium phosphate KHPO4 0.636
3. Results and discussion
Potassium chloride KCl 0.202
Potassium citrate K3C6H5O7•H2O 0.308
Uric acid sodium salt C5H3N4O3•Na 0.021 3.1. Effect of curcumin concentration on curcumin NLC properties
Urea H2NCONH2 0.198
Lactic acid sodium salt C3H5O3Na 0.146 Curcumin NLCs were produced after establishing the blank NLCs
Porcine gastric mucin (type II) – 30
optimal formulation in our previous study (Supplementary material).
Several conditions were confirmed, such as hydrophilic emulsifier con­
phosphate (KH₂PO₄) was dissolved in 75 mL of deionized water and then centration (Tween 80), lipophilic emulsifier concentration (Span 80), oil
mixed with 7.7 mL of 0.2 mol/L NaOH. One g of pancreatin from the type (MCT oil or soybean oil), oil concentration, and high-pressure ho­
porcine pancreas and 240 mg of bile extract was added in KH₂PO₄-NaOH mogenization cycles. To manufacture curcumin NLCs, Tween 80 300
solution. Finally, SIF was adjusted to pH to 6.8 ± 0.1. mg/100 mL, Span 80 300 mg/100 mL, MCT oil 240 mg/100 mL, and 3
cycles of high-pressure homogenization were applied (data not shown).
2.8. Curcumin release in in vitro lipid digestion Various concentrations (1–20 mg/100 mL) of curcumin were applied
to the blank NLCs, and then the appearance, particle properties, and
NLCs dispersion was applied into three simulated gastrointestinal encapsulation efficiency were observed. Particle properties of curcumin
fluids to observe curcumin release in in vitro lipid digestion. In the first NLC dispersion, such as hydrodynamic diameter, zeta potential, poly­
digestive step (mouth condition), each NLC was mixed with preheated dispersity index, and encapsulation efficiency, were estimated (Table 2
SSF (1:1 v/v) and then reacted for 10 min at 37 ◦ C with 105 rpm shaking. and Fig. 2). Hydrodynamic diameter was not significantly different ac­
In the second digestive step (stomach condition), digested NLCs cording to curcumin concentration, and it ranged from 115.2 to 141.4
dispersion was mixed with SGF (1:1 v/v) and then reacted in a shaking nm. Zeta potential was similar to hydrodynamic diameter, and the value
water bath (37 ◦ C, 105 rpm) for 2 h. The sample was collected at 5, 10,
30, 60, 90, and 120 min. In the third digestive step (small intestinal
condition), digested NLCs dispersion mixed with the SIF (1:1 v/v) and Table 2
adjusted pH of intestinal condition sample to pH 7.0 ± 0.1 using 1 mol/L Hydrodynamic diameter and zeta potential of curcumin-loaded nanostructured
NaOH. The mixture was then reacted in a shaking water bath (37 ◦ C, lipid carriers with different curcumin concentrations.
105 rpm) for 2 h. The sample was collected at 5, 10, 30, 60, 90, and 120 Curcumin (mg/100 mL) Hydrodynamic diameter (nm) Zeta potential (mV)
min. The release amount of curcumin was measured at each time of the 1 118.9 ± 1.1cde − 25.4 ± 0.4bc
digestive process, and the particle properties of curcumin NLCs were 2 118.4 ± 1.3de − 26.6 ± 0.4cd
also determined at the same time. The release amount of curcumin was 3 126.9 ± 1.6b − 25.9 ± 0.6bc
4 126.3 ± 2.3b 25.8 ± 0.3bc
measured through the encapsulation efficiency method and then −
5 125.3 ± 1.3bc − 25.3 ± 0.7b
calculated as follows (Eq. (2)): 10 139.8 ± 1.8a − 23.2 ± 0.4a
[ ] 12 122.8 ± 3.3bcd − 29.7 ± 1.1e
EC(g)
Curcumin release (%) = 100 − × 100 (2) 14 137.4 ± 3.0a − 25.9 ± 1.3bc
IC (g) 16 115.2 ± 2.9e − 27.5 ± 0.6d
18 118.2 ± 1.7de − 27.9 ± 0.6 d
where EC is from the middle part of curcumin NLCs dispersion and IC is 20 141.4 ± 8.7a − 25.7 ± 0.4bc
the initial amount of curcumin at NLCs preparation. Data are expressed as the mean ± standard deviation (n = 3).
a-e
The means followed by the same letter are not significant by Tukey’s multiple
range test at p > 0.05.

3
J.E. Hyun et al. LWT 162 (2022) 113474

100 mL curcumin was selected to manufacture the optimum curcumin


NLC dispersion with 300 mg/100 mL Tween 80, 300 mg/100 mL Span
80, and 240 mg/100 mL MCT oil. Moreover, the particle properties of
the optimum curcumin NLC dispersion were 126.9 nm in diameter,
− 25.9 mV zeta potential, 0.246 PDI (monodisperse), and 94.5%
encapsulation efficiency.
NLCs can be manufactured in various formulations and have
different particle properties. Dolatabadi et al. (2021) made curcuminoid
(79.4% curcumin) NLCs using various lipids (glycerol palmitostearate,
MCT oil) and emulsifiers (Na cholate, Span 80, Poloxamer 188). Some
materials were similar to our study, but they produced NLCs using high
shear homogenization and ultrasound. The hydrodynamic diameter was
148.8–225.5 nm, the zeta potential was − 16.7–24.9 mV, and the best
encapsulation efficiency was over 95%. In the Araujo et al. (2020) study,
curcumin NLCs were produced by using specific lipids (MCT oil, castor
oil) and emulsifiers (Tween 20, Tween 80), and they used only
Fig. 2. Encapsulation efficiency and polydispersity index of curcumin-loaded
high-speed homogenization to manufacture curcumin NLCs. The hy­
nanostructured lipid carriers with different curcumin concentrations; gray
drodynamic diameter was 192.6 or 247.7 nm, the PDI was 0.164 or
square: encapsulation efficiency and black square: polydispersity index. a-cThe
means followed by the same letter in the encapsulation efficiency result are not 0.161 according to oil type, and the encapsulation efficiency was 88.34
significant by Tukey’s multiple range test at p > 0.05. Error bars indicate or 94.14%. Rapalli et al. (2020) also produced curcumin NLCs, and
standard deviation. probe sonication was applied to reduce size variation. The hydrody­
namic diameter was 96.2–298.0 nm, and the PDI was 0.257–0.690
was from − 23.2 to − 29.7 mV, which is usually described as unstable depending on sonication time. The encapsulation efficiency was
stability in colloid suspensions. The stable state of colloid solution is 46.3–70.5%, which was not sonication time-dependent. There are many
usually over ±30 mV (Aditya et al., 2014). Even after measuring the formulations to produce optimum NLC. NLC properties, such as hydro­
hydrodynamic diameter and zeta potential, selecting an appropriate dynamic diameter, PDI, zeta potential, encapsulation efficiency, etc., are
curcumin NLC dispersion was not clear. influenced by solid lipids, liquid lipids, emulsifier type, emulsifier HLB,
The results of the polydispersity index and encapsulation efficiency emulsification method, etc.
showed significant differences among various samples. Fig. 2 shows In this study, curcumin NLCs were relatively smaller (nm), had a
monodisperse curcumin NLC dispersions from 1 to 10 mg/100 mL cur­ higher zeta potential (mV), and had a higher encapsulation efficiency
cumin, and the value was below the 0.25 polydispersity index. However, (%) than other curcumin NLCs. The reason may be the emulsification
the polydispersity index increased depending on the curcumin concen­ method among various conditions. High-pressure homogenization is a
tration, especially from 14 to 20 mg/100 mL. Furthermore, curcumin high-energy emulsification. The intensifier pump makes 2,000–30,000
NLC dispersions (14–20 mg/100 mL) were over the 0.3 polydispersity psi, and pressure generates power, such as high shear, impact, and
index, which is polydisperse, and precipitates were also observed. cavitation. Three energy effects on fluid, including oil in water emulsion,
Encapsulation efficiency was also significantly different according to solid in liquid suspension and the energy, break and disperse particles.
concentration (p < 0.05). The encapsulation efficiency of the curcumin Therefore, this energy could reduce the hydrodynamic diameter and
NLCs according to the different curcumin concentrations was polydispersity index of a particle in colloid solution. Moreover, the
80.9–95.9% (Fig. 2), and all samples had high efficiency, but it was number of process cycles usually improves particle properties.
significantly decreased depending on the increase in curcumin concen­
tration (p < 0.05). When the curcumin was over 4 mg/100 mL, pre­ 3.2. Temperature-dependent storage stability
cipitation was observed after 24 h at room temperature (Fig. 3). The
yellow color derived from the original curcumin color increased up to 4 The appearance of the curcumin NLC dispersion according to storage
mg/100 mL curcumin NLC dispersion, but sedimentation was observed temperature is shown in Table 3. There was no phase separation at any
from the 5 mg/100 mL curcumin NLC dispersion. This phenomenon did storage temperatures, but at 65 ◦ C, the color of the curcumin NLC
not affect hydrodynamic diameter or zeta potential, but it seemed that dispersion changed as if curcumin was decomposed by high temperature
encapsulation efficiency influenced the polydispersity index of curcu­ (Table 3). Fig. 5 shows that the encapsulation efficiency of NLCs grad­
min NLC dispersions (Fig. 2). From NLC with 4 mg/100 mL or more of ually decreased; therefore, the released free curcumin might be influ­
curcumin added, the encapsulation efficiency was significantly lower (p enced by high temperature. Curcumin is labile to basic solvents, oxygen,
< 0.05), and as the concentration of curcumin increased, a polydisperse and light and decomposes, eventually yielding volatile phenolic com­
particle distribution was shown. These results show that at the con­ pounds, such as vanillin, guaiacol and isoeugenol, which are relatively
centrations of solid and liquid lipids and emulsifiers set in this experi­ hydrophilic (Kharat, Du, Zhang, & McClements, 2017). It is thought that
ment, up to 3 mg/100 mL of curcumin can be stably entrapped in the during this storage period, the components are hydrolyzed and volatil­
NLC structure. However, at concentrations above that, curcumin ap­ ized (Esatbeyoglu, Ulbrich, Rehberg, Rohn, & Rimbach, 2015), and the
pears to precipitate without being completely dissolved in the lipid curcumin content decreases.
structure. Therefore, in the characterization of particle properties, 3 mg/ The hydrodynamic diameter and zeta potential during the 28-day
storage period obtained by varying the storage temperature of

Fig. 3. Pictorial views of nanostructured lipid carriers with different curcumin concentrations (% w/v). (a) 1 mg/100 mL, (b) 2 mg/100 mL, (c) 3 mg/100 mL, (d) 4
mg/100 mL, (e) 5 mg/100 mL, (f) 20 mg/100 mL.

4
J.E. Hyun et al. LWT 162 (2022) 113474

Table 3 curcumin NLCs are shown in Fig. 4. The hydrodynamic diameter


Pictorial views of curcumin nanostructured lipid carriers at increased significantly to 112.5 nm, 100.8 nm and 118.8 nm when
different temperatures during 28 days of storage. stored at 5 ◦ C, 21 ◦ C and 65 ◦ C, respectively (p < 0.05). However, at
40 ◦ C, the hydrodynamic diameter was maintained at an average of
90–100 nm, and there was no difference during the storage period (p >
0.05). Hydrodynamic diameter might why the emulsifier and lipid that
were used for forming NLCs were structurally changed. NLC was pro­
duced by the hot high shear homogenization (HSH) method (Salvi &
Pawar, 2019). Solid lipids were mixed with liquid lipids and core ma­
terials, such as curcumin, and then emulsifiers and aqueous phases, were
added above the melting temperature of solid lipids. Generally, the
melting point of solid lipid glycerol tristearate is 54.0–72.5 ◦ C. Then,
while emulsification of the lipid phase and aqueous phase occurred by
passing through HPH (high-pressure homogenization) at varying pres­
sures for 3 cycles, the pre-emulsion cooled rapidly, and NLC was formed
(Salvi & Pawar, 2019). Namely, solid lipids easily melt above the
melting point, and the crystallin structure of NLCs breaks down. In
addition, NLCs are usually amorphous at high temperatures (Klucker,
Dalencon, Probeck, & Haensler, 2012); therefore, curcumin might be
easily released at 65 ◦ C. The absolute zeta potential was significantly
decreased at 21, 40, and 65 ◦ C and it was rapidly decreased to − 16.9 mV
after 7 days at 65 ◦ C. This result was due to the release and decompo­
sition of curcumin, and wall materials, such as lipids and emulsifiers,
were also unstable during storage under temperature and time stress.
The encapsulation efficiency (EE%) and total phenolic content were
measured for 28 days at certain times, as shown in Figs. 5 and 6. Storage
temperature affected the release rate of curcumin encapsulated in NLCs.
Significantly, EE% was dramatically reduced from 92% to 24% at 65 ◦ C.
Next, curcumin NLCs were stored at 40 ◦ C, which was a faster release
rate than other samples. Curcumin NLCs were relatively stable when
stored at 5 ◦ C and 21 ◦ C for 28 days after preparation. The content of
curcumin is reduced due to structural instability from high-temperature
exposure, exposure to the water phase of curcumin, oxidation and hy­
drolysis (Esatbeyoglu et al., 2015). The total phenolic content of NLC
also showed a trend similar to that of EE%. At the beginning of storage (7
days), gallic acid (μg/mL) rapidly decreased and then slowly decreased
in all samples (Fig. 6). However, significantly, gallic acid (μg/mL) was
dramatically reduced from 76% to 27% at 65 ◦ C. As the storage period of
the curcumin emulsion increases, the curcumin content gradually de­
creases, and the curcumin content in the emulsion is lower at 55 ◦ C than
at 20 ◦ C (Kharat et al., 2017). Therefore, as a result of this study, the
dramatic reduction in the total phenolic content of curcumin NLCs at
65 ◦ C appears to be the result of a large amount of curcumin being
released at relatively high temperatures, resulting in the loss of many
phenolic components. From this analysis, it was observed NLC could be
stable and maintain functionality at certain storage condition. As we
mentioned in introduction, NLC system is effective to improve entrap­
ment efficiency, loading efficiency, physico-chemical stability and
bioavailability compare with other carriers. Therefore, it is useful to
apply into encapsulation of functional component such as curcumin.
Especially, curcumin is one of health functional materials but it is not
appropriate to apply directly in food production because of its low sol­
ubility, low chemical stability, low bioavailability and so on. For the
food production, food processing efficiency could be improving by using
curcumin NLC instead of pure curcumin. For the human health, around
30 ppm of curcumin which is produced by using nano-encapsulation
technology also have function in cancer drug delivery system (Santad­
kha, Skolpap, & Thitapakorn, 2021). According to the report 30–50 μM
concentration of curcumin affected proliferation and apoptosis of breast
cancer cells (Kim, Yi, Lee, Cho, & Yoon, 2004; Park et al., 2013).
Fig. 4. Hydrodynamic diameter and zeta potential of curcumin nanostructured
Thermogravimetric analysis (TGA) is a useful method for evaluation
lipid carriers at different temperatures during 28 days of storage. Error bars
indicate standard deviation. the thermal stability of compounds by recording the weight loss of a
sample with increasing temperature (Soltanzadeh, Peighambardoust,
Ghanbarzadeh, Mohammadi, & Lorenzo, 2021). The TGA results for the
components constituting the curcumin NLCs are shown in Fig. 7 and
Table 4. The thermal decomposition temperature and weight loss rates

5
J.E. Hyun et al. LWT 162 (2022) 113474

a-d
Fig. 5. Encapsulation efficiency of curcumin nanostructured lipid carriers at different temperatures during 28 days of storage. The means followed by the same
letter are not significant by Tukey’s multiple range test at p > 0.05. Error bars indicate standard deviation.

Fig. 6. Total phenolic content of curcumin nanostructured lipid carriers at different temperatures during 28 days of storage. a-eThe means followed by the same letter
are not significant by Tukey’s multiple range test at p > 0.05. Error bars indicate standard deviation.

of glycerol tristearate and MCT oil, which were used as lipids for NLCs change in temperature and energy in a phase transition. Therefore,
preparation, were 250.6, 243.4 ◦ C and 94.1, 99.8%, respectively. Cur­ through DSC analysis, the melting point of a compound and the crys­
cumin showed the highest thermal decomposition temperature of tallization or amorphous state of a drug can be known (Madane &
251.7 ◦ C and the lowest weight loss rate of 76.4% compared to other Mahajan, 2014). The results of DSC analysis of glycerol tristearate, MCT
materials composing NLC in this experiment, because phenolic com­ oil, curcumin, blank NLCs, and curcumin NLCs are shown in Fig. 7F and
pounds have good thermal stability (Valencia et al., 2021). The thermal Table 4. Glycerol tristearate, used as a solid lipid in the preparation of
decomposition temperatures of blank NLCs and curcumin NLCs did not NLCs showed a sharp melting peak around 55.2 ◦ C, suggesting that this
show a significant difference at 207.8 and 192.4 ◦ C, respectively. In material has crystallinity. And curcumin exhibited the maximum
addition, both samples showed a stable state up to 180 ◦ C. These results endothermic peak near about 177.7 ◦ C, which was found to have a value
may suggest that curcumin is efficiently encapsulated in the amorphous similar to the results of several studies related to curcumin formulation
state. This showed a similar trend to the studies related to (Agrawal, Saraf, Pradhan, Patel, Singhvi, & Alexa et al., 2021; Behba­
carbamazepine-loaded solid lipid nanoparticles and nanostructured hani et al., 2019; Puglia, Frasca, Musumeci, Rizza, Puglisi, & Bonina
lipid carriers (Montoto et al., 2018). et al., 2012). However, the maximum endothermic peak of curcumin
Differential scanning calorimetry (DSC) is a method to determine the NLCs appeared around 56.2 ◦ C, and there was no peak near 177 ◦ C

6
J.E. Hyun et al. LWT 162 (2022) 113474

Fig. 7. Thermal characterization using TGA and DSC: TGA of glycerol tristearate (A), MCT oil (B), curcumin (C), blank NLCs (D), and curcumin NLCs (E); and DSC of
curcumin NLCs components.

3.3. In vitro digestion


Table 4
Parameter extracted from the TGA and DSC of the curcumin NLCs components.
The change in the curcumin NLC particle properties was observed
TGA DSC under in vitro digestion, as shown in Fig. 8A. There was no change in
Samples
Thermal Weight Melting ΔH curcumin NLC hydrodynamic diameter in in vitro model saliva. Hydro­
decomposition loss (%) temperature (J/g) dynamic diameter was increased to 165.3–180.8 nm in simulated gastric
temperature (◦ C) (◦ C)
fluid conditions, but there was no significant difference between the
Glycerol 250.6 94.1 55.2 179.3 original diameter and the hydrodynamic diameter after in vitro model
tristearate saliva (p > 0.05). Notably, there was no significant difference in hy­
MCT oil 243.4 99.8 ND ND
drodynamic diameter according to digestion time (p > 0.05). A nonionic
Curcumin 251.7 76.4 177.7 138.8
Blank NLCs 207.8 99.6 55.7 86.9 surfactant, Tween 80, which surrounded NLCs, was adequate to main­
Curcumin 192.4 99.7 56.2 77.5 tain the stability of NLCs from hydrolysis by pepsin and low pH (Van
NLCs Aken, Bomhof, Zoet, Verbeek, & Oosterveld, 2011). Similarly, the re­
ND: not detected. ported curcumin NLC maintained a hydrodynamic diameter of 200–300
nm, and there was no aggregation in simulated gastric fluid for 2 h
where the peak of curcumin appeared. These results indicate that cur­ (Behbahani et al., 2019). In the simulated small intestinal fluid, the
cumin is completely encapsulated inside the NLCs. In addition, the hydrodynamic diameter was increased by more than 100 nm after 5 min
enthalpy changes of blank NLCs (86.9 J/g) and curcumin NLCs (77.5 of digestion time and increased significantly to 445.1 nm at 120 min of
J/g) was decreased compared to glycerol tristearate (179.3 J/g) and reaction time. The enzymes, and bile extract might hydrolyze the
curcumin (138.8 J/g), indicating that blank NLCs and curcumin NLCs emulsifier and lipid layer as free micelles and monoacylglycerols,
have an amorphous structure. In the study of Behbahani et al. (2019), resulting in micelle formation (Aditya et al., 2014; Park, Garcia, Shin, &
curcumin-loaded NLCs were prepared using stearic acid as a solid lipid Kim, 2017). Simultaneously, NLCs were also treated in an in vitro
and Tween 80 and pluronic F167 as emulsifiers. At this time, the result digestion system without an enzyme. There was no hydrodynamic
of DSC analysis of curcumin NLC showed a small peak near 173 ◦ C, diameter change by adjusting the pH. According to one study related to a
which is the maximum endothermic peak of curcumin. This showed that phloretin-loaded nanostructured lipid carrier, an in vitro digestion model
some curcumin was not enclosed by NLCs, showing a different trend of phloretin NLC initially showed a hydrodynamic diameter of approx­
from the results of this study. imately 100 nm. However, it has been reported that NLC aggregation
occurred during the small intestine stage, which increased the hydro­
dynamic diameter to approximately 800 nm (Gu, Sun, Wang, & Xia,
2022).

7
J.E. Hyun et al. LWT 162 (2022) 113474

influence of pH, but considering the hydrodynamic diameter, it is


thought that the amount of curcumin release increased to 82% by
enzyme and pH as the hydrodynamic diameter of NLCs increased by an
enzyme (Fig. 8). Curcumin is rapidly decomposed under alkaline con­
ditions, so it is not easy to maintain curcumin from the digestive tract to
the intestine.

4. Conclusions

In this study, optimal conditions were established, and various


characteristics were observed to prepare nanostructured lipid carriers
(NLCs), which were included to increase the safety and utilization rate of
the poorly soluble material curcumin. Curcumin NLCs were micronized
into 3 mg/100 mL curcumin, 300 mg/100 mL Tween 80 and 300 mg/
100 mL Span 80. The produced curcumin NLCs had a hydrodynamic
diameter of 126.9 nm, zeta potential of − 25.9 mV, PDI of 0.246, and
encapsulation efficiency of 94.5%. When the curcumin NLC was stored
at various temperatures for 28 days, it was stably maintained at 5 ◦ C and
21 ◦ C, but at 40 ◦ C and 65 ◦ C, the encapsulation efficiency and total
phenolic content decreased dramatically. As a result of thermal char­
acterization, it was confirmed that curcumin NLCs did not show a peak
near 177 ◦ C, the maximum endothermic peak of curcumin, and was
completely captured in the NLC structure. In the digestive imitation
system, NLCs released approximately 31% of curcumin in imitation
saliva, and curcumin release gradually increased. This study showed
that NLC was an effective transporter for the solubilization of curcumin.
Furthermore, NLCs could apply to various fields by increasing water
solubility, bioavailability, and curcumin storage stability. Especially,
curcumin NLC could be applied into functional foods such as beverage,
jelly food, etc. in the food industry.

CRediT authorship contribution statement

Ji Eun Hyun: Investigation, Writing – original draft, Writing – re­


Fig. 8. Changes in properties of curcumin nanostructured lipid carriers ac­ view & editing, Visualization. Hye-Yoon Yi: Validation, Investigation,
cording to simulated gastrointestinal conditions and pH: hydrodynamic diam­ Writing – review & editing, Visualization, Software. Geun-Pyo Hong:
eter (A) and curcumin release (B); Cur-NLC (curcumin nanostructured lipid Conceptualization, Writing – original draft, Resources. Ji-Yeon Chun:
carrier), stomach condition (S), and small intestine condition (I). a-gThe means Conceptualization, Investigation, Writing – original draft, Writing – re­
followed by the same letter are not significant by Tukey’s multiple range test at view & editing, Supervision.
p > 0.05. Error bars indicate standard deviation.

Declaration of competing interest


Regarding curcumin release (Fig. 8B), approximately 31.7% of cur­
cumin was released in in vitro model saliva, and curcumin was released All authors declared that there is no conflict of interest.
up to 37.7% after 2 h in imitation gastric juice. There was no significant
difference in imitation saliva (p > 0.05). In imitation intestinal fluid, Appendix A. Supplementary data
curcumin release dramatically increased to 48.1% in 5 min, and up to
74.5% after 30 min, and the amount of curcumin released increased over Supplementary data to this article can be found online at https://doi.
time and increased to 82% after 2 h, but after 30 min, there was no org/10.1016/j.lwt.2022.113474.
significant difference (p > 0.05). In addition, when measuring the
amount of curcumin released by adjusting the pH and temperature,
References
excluding enzymes, curcumin was released when the pH was 6.8 or
higher, and there was no significant difference from the amount of Aditya, N. P., Macedo, A. S., Doktorovova, S., Souto, E. B., Kim, S., Chang, P. S., et al.
curcumin first released when the pH was acidic (p > 0.05). When the pH (2014). Development and evaluation of lipid nanocarriers for quercetin delivery: A
was 8 or higher, the amount of curcumin released increased significantly comparative study of solid lipid nanoparticles (SLN), nanostructured lipid carriers
(NLC), and lipid nanoemulsions (LNE). Lebensmittel-Wissenschaft und -Technologie-
after 10 min to 30.9–53.6% (p < 0.05). Curcumin is highly affected by Food Science and Technology, 59(1), 115–121. https://doi.org/10.1016/J.
pH. In an alkaline environment, oxidation and hydrolysis reactions LWT.2014.04.058
occur at the interface between the aqueous and oil phases, producing Aggarwal, B. B., Surh, Y. J., & Shishodia, S. (2007). The molecular targets and
therapeutic uses of curcumin in health and disease. In R. L. Thangapazham,
reaction products such as bicyclopentadione, ferulic acid, and feruloyl A. Sharma, & R. K. Maheshwar (Eds.), Beneficial role of curcumin in skin diseases (pp.
methane. Because most of these decomposition products are relatively 343–357). Springer.
hydrophilic, they migrate to the water phase, which is unstable. How­ Agrawal, M., Saraf, S., Pradhan, M., Patel, R. J., Singhvi, G., Ajazuddin, et al. (2021).
Design and optimization of curcumin loaded nano lipid carrier system using Box-
ever, to restore equilibrium, more curcumin molecules will migrate from Behnken design. Biomedicine & Pharmacotherapy, 141, 111919. https://doi.org/
the oil phase to the interface, and the decomposition process will 10.1016/j.biopha.2021.111919
continue (Kharat et al., 2017). At acidic pH, curcumin is predominantly Ak, T., & Gulcin, İ. (2008). Antioxidant and radical scavenging properties of curcumin.
Chemico-Biological Interactions, 174(1), 27–37. https://doi.org/10.1016/j.
in an enolic structure, so the main decomposition mechanism is oxida­
cbi.2008.05.003
tion rather than hydrolysis. Curcumin is also released under the Araiza-Calahorra, A., Akhtar, M., & Sarkar, A. (2018). Recent advances in emulsion-
based delivery approaches for curcumin: From encapsulation to bioaccessibility.

8
J.E. Hyun et al. LWT 162 (2022) 113474

Trends in Food Science & Technology, 71, 155–169. https://doi.org/10.1016/j. Park, S. J., Garcia, C. V., Shin, G. H., & Kim, J. T. (2017). Development of nanostructured
tifs.2017.11.009 lipid carriers for the encapsulation and controlled release of vitamin D3. Food
Araujo, V. H. S., da Silva, P. B., Szlachetka, I. O., da Silva, S. W., Fonseca-Santos, B., Chemistry, 225, 213–219. https://doi.org/10.1016/j.foodchem.2017.01.015
Chorilli, M., et al. (2020). The influence of NLC composition on curcumin loading Park, S. Y., Kang, Y. G., Bae, Y. H., Kim, S. R., Park, H. J., Kang, Y. S., et al. (2013). Role
under a physicochemical perspective and in vitro evaluation. Colloids and Surfaces A: of Sp in the regulation of Notch1 gene expression by curcumin. Korean Society for
Physicochemical and Engineering Aspects, 602(5), Article 125070. https://doi.org/ Biotechnology and Bioengineering Journal, 28(1), 1–6. https://doi.org/10.7841/
10.1016/j.colsurfa.2020.125070 ksbbj.2013.28.1.1
Balasubramanian, S., & Eckert, R. L. (2006). Curcumin suppresses AP1 transcription Puglia, C., Frasca, G., Musumeci, T., Rizza, L., Puglisi, G., Bonina, F., et al. (2012).
factor-dependent differentiation and activates apoptosis in human epidermal Curcumin loaded NLC induces histone hypoacetylation in the CNS after
keratinocytes. Journal of Biological Chemistry, 282(9), 6707–6715. https://doi.org/ intraperitoneal administration in mice. European Journal of Pharmaceutics and
10.1074/jbc.M606003200 Biopharmaceutics, 81, 288–293. https://doi.org/10.1016/j.ejpb.2012.03.015
Behbahani, E. S., Ghaedi, M., Abbaspour, M., Rostamizadeh, K., & Dashtian, K. (2019). Rapalli, V., Kaul, V., Waghule, T., Gorantla, S., Sharma, S., Roy, A., et al. (2020).
Curcumin loaded nanostructured lipid carriers: In vitro digestion and release studies. Curcumin loaded nanostructured lipid carriers for enhanced skin retained topical
Polyhedron, 164, 113–122. https://doi.org/10.1016/j.poly.2019.02.002 delivery: Optimization, scale-up, in-vitro characterization and assessment of ex-vivo
Dolatabadi, S., Karimi, M., Nasirizadeh, S., Hatamipour, M., Golmohammadzadeh, S., skin deposition. European Journal of Pharmaceutifcal Sciences, 152, Article 105438.
Jaafari, M. R., et al. (2021). Preparation, characterization and in vivo https://doi.org/10.1016/j.ejps.2020.105438
pharmacokinetic evaluation of curcuminoids-loaded solid lipid nanoparticles (SLNs) Rastall, R. (2007). Novel enzyme technology for food applications. In X. Xu,
and nanostructured lipid carriers (NLCs). Journal of Drug Delivery Science and J. B. Kristensen, & H. Zhang (Eds.), Production of structured lipids with functional health
Technology, 62, Article 102352. https://doi.org/10.1016/j.jddst.2021.102352 benefits (pp. 270–284). Woodhead Publishing.
Esatbeyoglu, T., Ulbrich, K., Rehberg, C., Rohn, S., & Rimbach, G. (2015). Thermal Riaz, A., Ahmed, N., Khan, M. I., Haq, I. ul, Rehman, A. ur, Khan, G. M., et al. (2019).
stability, antioxidant, and anti-inflammatory activity of curcumin and its Formulation of topical NLCs to target macrophages for cutaneous leishmaniasis.
degradation product 4-vinyl guaiacol. Food & Function, 6, 887–893. https://doi.org/ Journal of Drug Delivery Science and Technology, 54, Article 101232. https://doi.org/
10.1039/C4FO00790E 10.1016/j.jddst.2019.101232
Fujisawa, S., Atsumi, T., Ishihara, M., & Kadoma, Y. (2004). Cytotoxicity, ROS- Saarela, M. (2011a). Functional foods. In A. Turpeinen, & P. Merimaa (Eds.), Functional
generation activity and radical-scavenging activity of curcumin and related fats and spreads (2nd ed., pp. 383–400). Woodhead Publishing.
compounds. Anticancer Research, 24(2B), 563–570. Saarela, M. (2011b). Functional foods. In S. B. Myrie, & P. J. H. Jones (Eds.), Functional
Gu, L., Sun, R., Wang, W., & Xia, Q. (2022). Nanostructured lipid carriers for the foods and obesity (2nd ed., pp. 234–260). Woodhead Publishing.
encapsulation of phloretin: Preparation and in vitro characterization studies. Salvi, V. R., & Pawar, P. (2019). Nanostructured lipid carriers (NLC) system: A novel drug
Chemistry and Physics of Lipids, 242, Article 105150. https://doi.org/10.1016/j. targeting carrier. Journal of Drug Delivery Science and Technology, 51, 255–267.
chemphyslip.2021.105150 https://doi.org/10.1016/j.jddst.2019.02.017
Henry, C. J. K. (2007). Novel food ingredients for weight control. In I. Rudkowska, & Santadkha, T., Skolpap, W., & Thitapakorn, V. (2021). Diffusion modeling and in vitro
P. J. H. Jones (Eds.), Medium-chain and structured triglycerides: Their role in weight release kinetics studies of curcumin-loaded superparamagnetic nanomicelles in
control (pp. 305–325). Woodhead Publishing. cancer drug delivery system. Journal of Pharmaceutical Sciences. https://doi.org/
Hussain, Z., Thu, H. E., Amjad, M. W., Hussain, F., Ahmed, T. A., Khan, S., et al. (2017). 10.1016/j.xphs.2021.11.015
Exploring recent developments to improve antioxidant, anti-inflammatory and Selvam, C., Jachak, S. M., Thilagavathi, R., & Chakraborti, A. K. (2005). Design,
antimicrobial efficacy of curcumin: A review of new trends and future perspectives. synthesis, biological evaluation and molecular docking of curcumin analogues as
Materials Science And Engineering: C, 77, 1316–1326. https://doi.org/10.1016/j. antioxidant, cyclooxygenase inhibitory and anti-inflammatory agents. Bioorganic &
msec.2017.03.226 Medicinal Chemistry Letters, 15(7), 1793–1797. https://doi.org/10.1016/j.
Jo, Y. J., Kim, H. Y., & Kwon, Y. J. (2016). Influence of biopolymer emulsifier on the bmcl.2005.02.039
characteristics of curcumin nanoemulsions. Food Engineering Progress, 20(4), Sezer, A. D. (2012). Recent advances in novel drug carrier systems. In A. A. Attama,
321–327. https://doi.org/10.13050/foodengprog.2016.20.4.321 M. A. Momoh, & P. F. Builders (Eds.), Lipid nanoparticulate drug delivery systems: A
Jun, Y. K., Lim, Y. M., & Jin, B. S. (2015). Characteristics of α-Tocopherol-loaded revolution in dosage form design and development (pp. 107–140). Licensee InTech.
nanostructured lipid carriers and their stabilization effect. Applied Chemistry for Singleton, V., & Rossi, J. (1965). Colorimetry of total phenolics with phosphomolybdic-
Engineering, 26(6), 659–665. https://doi.org/10.14478/ace.2015.1082 phosphotungstic acid reagents. American Journal of Enology and Viticulture, 16,
Kharat, M., Du, Z., Zhang, G., & McClements, D. J. (2017). Physical and chemical 144–158.
stability of curcumin in aqueous solutions and emulsions: Impact of pH, Soltanzadeh, M., Peighambardoust, S. H., Ghanbarzadeh, B., Mohammadi, M., &
temperature, and molecular, environment. Journal of Agricultural and Food Chemistry, Lorenzo, J. M. (2021). Chitosan nanoparticles encapsulation lemongrass
65(8), 1525–1532. https://doi.org/10.1021/acs.jafc.6b04815 (Cymbopogon commutatus) essential oil: Physicochemical, structural, antimicrobial
Kim, C. H., Park, E. S., & Jeon, Y. H. (2008). Curcumin attenuates chronic constriction and in-vitro release properties. International Journal of Biological Macromolecules,
nerve injury-induced neuropathic pain in rats. Korean Journal of Medicinal Crop 192, 1084–1097. https://doi.org/10.1016/j.ijbiomac.2021.10.070
Science, 16(3), 183–187. Tamjidi, F., Shahedi, M., Varshosaz, J., & Nasirpour, A. (2013). Nanostructured lipid
Kim, E. J., Yi, S., Lee, H. S., Cho, H. J., & Yoon, J. H. (2004). Inhibitory effect of curcumin carriers (NLC): A potential delivery system for bioactive food molecules. Innovative
on the growth of MCF-7 human breast cancer cells. Journal of Korean Association of Food Science & Emerging Technologies, 19, 29–43. https://doi.org/10.1016/j.
Cancer Prevention, 9(4), 224–252. ifset.2013.03.002
Klucker, M. F., Dalencon, F., Probeck, P., & Haensler, J. (2012). AF03, an alternative United States Pharmacopeial Convention. (2009). United States Pharmacopeia 32 –
squalene emulsion-based vaccine adjuvant prepared by a phase inversion National formulary 27 (p. 726). Rockville, MD: United States Pharmacopeia
temperature method. Journal of Pharmaceutical Sciences, 101(12). https://doi.org/ Convention.
10.1002/jps.23311, 4490-4450. Valencia, M. S., Silva Junior, M. F., Xavier-Junior, F. H., Veras, B. O.,
Madane, R. G., & Mahajan, H. S. (2014). Curcumin-loaded nanostructured lipid carriers Albuquerque, P. B. S., Oliveira Borba, E. F., et al. (2021). Characterization of
(NLC) for nasal administration: Design, characterization, and in vivo study. Drug curcumin-loaded lecithin-chitosan bioactive nanoparticles. Carbohydrate Polymer
Delivery, 23(4), 1326–1334. https://doi.org/10.3109/10717544.2014.975382 Technologies and Applications, 2, Article 100119. https://doi.org/10.1016/j.
Mao, Y., & McClements, D. J. (2012). Influence of electrostatic heteroaggregation of lipid carpta.2021.100119
droplets on their stability and digestibility under simulated gastrointestinal Van Aken, G. A., Bomhof, E., Zoet, F. D., Verbeek, M., & Oosterveld, A. (2011).
conditions. Food & Function, 3(10), 1025–1034. https://doi.org/10.1039/ Differences in in vitro gastric behaviour between homogenized milk and emulsions
c2fo30108c stabilised by Tween 80, whey protein, or whey protein and caseinate. Food
Montoto, S. S., Sbaraglini, M. L., Talevi, A., Couyoupetrou, M., Ianni, M. D., Pesce, G. O., Hydrocolloids, 25(4), 781–788. https://doi.org/10.1016/j.foodhyd.2010.09.016
et al. (2018). Carbamazepine-loaded solid lipid nanoparticles and nanostructured Varshosaz, J., Eskandari, S., & Tabakhian, M. (2010). Production and optimization of
lipid carriers: Physicochemical characterization and in vitro/in vivo evaluation. valproic acid nanostructured lipid carriers by the Taguchi design. Pharmaceutical
Colloids and Surfaces B: Biointerfaces, 167, 73–81. https://doi.org/10.1016/j. Development and Technology, 15(1), 89–96. https://doi.org/10.3109/
colsurfb.2018.03.052 10837450903013568
Muller, R. H., Radtke, M., & Wissing, S. A. (2002). Nanostructured lipid matrices for Yuan, H., Chen, J., Du, Y. Z., Hu, F. Q., Zeng, S., Zhao, H. L., et al. (2007). Studies on oral
improved microencapsulation of drugs. International Journal of Pharmaceutics, 242 absorption of stearic acid SLN by a novel fluorometric method. Colloids and Surfaces
(1–2), 121–128. https://doi.org/10.1016/S0378-5173(02)00180-1 B: Biointerfaces, 58(2), 157–164. https://doi.org/10.1016/j.colsurfb.2007.03.002
Nanji, A. A., Jokelainen, K., Tipoe, G. L., Rahemtulla, A., Thomas, P., Dannenberg, A. J., Yu, Q., Hu, X., Ma, Y., Xie, Y., Lu, Y., Qi, J., et al. (2016). Lipids-based nanostructured
et al. (2003). Curcumin prevents alcohol-induced liver disease in rats by inhibiting lipid carriers (NLCs) for improved oral bioavailability of sirolimus. Drug Delivery, 23
the expression of NF-kappa B-dependent genes. American Journal of Physiology. (4). https://doi.org/10.3109/10717544.2016.1153744, 1496-1475.
Gastrointestinal and Liver Physiology, 284(2), G321–G327. https://doi.org/10.1152/
ajpgi.00230.2002

You might also like