You are on page 1of 9

Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

Contents lists available at ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

Loading of water-insoluble celastrol into niosome hydrogels for improved T


topical permeation and anti-psoriasis activity
Shikang Menga, Lin Sunb, Lun Wanga, Zibei Lina, Zeyu Liua, Long Xia, Zhenping Wangc,

Ying Zhenga,
a
State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
b
Zhuhai Campus of Zunyi Medical University, Department of Pharmaceutical Sciences, China
c
Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Psoriasis is a severe disfiguring skin disease affecting approximately 3% of people worldwide and negatively
Celastrol affecting their daily lives. The pathogenesis of psoriasis is complicated, and typical therapeutic strategies for
Niosomes psoriasis mainly focus on anti-inflammation. Considering the side effects, withdrawal rebound, high cost, and
Psoriasis many other disadvantages of existing treatments, we developed a new topical therapeutic formulation consisting
Topical permeation
of niosomes loaded with celastrol, a triterpenoid extracted from Tripterygium. Celastrol niosomes were prepared
by the thin film hydration method and probe sonication. The niosomes were composed of Span 20, Span 60, and
cholesterol at a weight ratio of 3:1:1. The particle size of the niosomes was approximately 147 nm, with yield of
up to 90%. Celastrol niosomes showed improved in vitro permeation ability compared to the raw drug. In our in
vivo study, celastrol niosomes effectively alleviated erythema and scaling on the dorsal skin of psoriasis mouse
models. Spleen weight and the levels of cytokines, including IL-22, IL-23, and IL-17, decreased after the treat-
ment, indicating the high therapeutic potential of this formulation for psoriasis. In conclusion, encapsulation of
celastrol by niosomes increased the water-solubility and permeation of celastrol into the skin, significantly
improving its anti-psoriasis activity in mice.

1. Background countries of different latitudes. Thus, it is now believed that a dry-cold


climate might be a major environmental factor inducing psoriasis [5].
1.1. Psoriasis Moreover, bacterial infections, injury, mental pressure, and heredity
are considered to be related to psoriasis development [6]. The patho-
Psoriasis, a chronic non-infectious disease, has been reported genesis of psoriasis is still not completely clear, but it has been found to
worldwide. Because registration of psoriasis cases is not compulsory, be closely related to the IL-17/IL-23 axis [7,8]. It was reported that only
there are few studies about the onset of psoriasis and these studies are approximately 40% of psoriasis patients are satisfied with the treatment
not very reliable [1]. It is roughly estimated that 3% of people world- prescribed by their dermatologist and that approximately over 70% of
wide have psoriasis [2,3]. Psoriasis could be divided into five types patients report low satisfaction regarding treatment [4]. Therefore, to
according to its pathological manifestation, and the most common type overcome these unmet clinical needs, it is important to develop new
being plaque psoriasis, which causes pain and itching in affected pa- effective anti-psoriasis drugs.
tients. In addition, it also causes large areas of erythema or scaling on
the skin, resulting in severe disfigurement [4]. All these symptoms 1.2. Anti-psoriasis treatment
might cause huge negative impacts on patients’ daily lives, such as
difficulties in choosing clothing, taking a shower, and performing out- For mild or moderate psoriasis, topical treatments are usually ad-
door activities. ministered. UVB treatment and combination medication are adminis-
The incidence of psoriasis was found not to be significantly asso- tered concomitantly to increase the therapeutic efficacy and improve
ciated with gender, age, or race; however, its incidence is different in patient satisfaction [9]. Severe psoriasis is usually treated with oral


Corresponding author.
E-mail addresses: mb65807@umac.mo (S. Meng), sakura528@126.com (L. Sun), kenwanglun668@gmail.com (L. Wang), mb75810@umac.mo (Z. Lin),
liuzeyujoey@163.com (Z. Liu), xl281213606@163.com (L. Xi), z7wang@ucsd.edu (Z. Wang), yzheng@umac.mo (Y. Zheng).

https://doi.org/10.1016/j.colsurfb.2019.110352
Received 13 March 2019; Received in revised form 15 June 2019; Accepted 6 July 2019
Available online 08 July 2019
0927-7765/ © 2019 Elsevier B.V. All rights reserved.
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

systemic drugs, such as methotrexate, acitretin, and cyclosporine. Most Table 1


of these chemical drugs have side effects, and their therapeutic effects Primer sequences of mouse genes used in qPCR analysis.
are not always satisfactory [10]. Recent biologics, which are mainly Primer Base sequence (5’ to 3’)
monoclonal antibodies targeting cytokines such as IL-17, IL-23, and
TNF-α, have higher therapeutic effect than the chemical drugs [11]. IL-1β (forward) CCCTGCAGCTGGAGAGTGTGGA
IL-1β (reverse) TGTGCTCTGCTTGTGAGGTGCTG
These biologics are suitable for all types of psoriasis, but owing to high
IL-6 (forward) CCTCTCTGCAAGAGACTTCCAT
price and drug withdrawal rebound, biologics are currently only used IL-6 (reverse) AGTCTCCTCTCCGGACTTGT
for treating moderate-to-severe psoriasis [9]. IL-17 F (forward) ACCCGTGAAACAGCCATGGTCAAG
Tripterygium wilfordii Hook f. has a long history as a traditional IL-17 F (reverse) CCCATGGGGAACTGGAGCGG
Chinese herbal medicine, and it is mainly used for treating rheumatoid IL-22 (forward) CAGCTCCTGTCACATCAGCGGT
IL-22 (reverse) AGGTCCAGTTCCCCAATCGCCT
arthritis and psoriasis [12,13]. Celastrol (Cel), a pentacyclic triterpene
IL-23 (forward) TCCTCCAGCCAGAGGATCACCC
isolated from Tripterygium wilfordii Hook f., had been widely reported IL-23 (reverse) AGAGTTGCTGCTCCGTGGGC
to have antioxidant, anti-inflammatory, and antitumor activity [14]. It IFN-γ (forward) TAACTCAAGTGGCATAGATGTGGAAG
is suspected that Cel exerts its anti-inflammatory activity by inhibiting IFN-γ (reverse) GACGCTTATGTTGTTGCTGATGG
STAT3 (forward) GAAGCCGACCCAGGTGC
the differentiation of Th17 cells through the blockage of pSTAT3 acti-
STAT3 (reverse) GTCACGTCTCTGCAGCTTCT
vation and the induction of microphage apoptosis by downregulating GAPDH (forward) GGGCTCTCTGCTCCTCCCTGT
the NF-kB pathway [15–18]. However, the solubility of Cel in water is GAPDH (reverse) CGGCCAAATCCGTTCACACCG
extremely poor [19]. As there were few literatures reporting topical
delivery systems for Cel, it is necessary to investigate a topical delivery
system which can enhance the solubility, skin permeation, and bioac- 2.2. Animals
tivity of Cel.
Female C57/BL6 mice (18˜22 g) aged 7–9 weeks were provided
1.3. Transdermal formulations from the FHS faculty of the University of Macau and then housed under
specific pathogen-free conditions. All experimental protocols using
Compared to other routes of administration, non-invasive adminis- animals were ratified by the Animal Ethics Committee of University of
tration through the skin allows drugs to avoid the extreme environment Macau, and all experiments shown below were performed according to
in the gastrointestinal tract and the first-pass metabolism of the liver, as the NIH Guidelines for the Care and Use of Laboratory Animals.
well as prevents systemic toxicity by the drugs. Dosing time and ad-
ministration site can also be accurately regulated [20]. The skin consists 2.3. Formula screening
of epidermis, dermis, and subcutaneous tissue. The stratum corneum, the
outermost layer of the epidermis consists of multilayers of dead kera- Cel Nio were prepared by the thin film hydration method with so-
tinocytes and intercellular lipids [21]. The stratum corneum protects the nication. Briefly, a mixture of a nonionic surfactant, cholesterol, and Cel
tissue from dehydration and resists external bacterial invasion, thereby at a specific weight was dissolved in a 2:1 chloroform:methanol solu-
functioning as a strong barrier protecting the internal environment tion (3 mL) in a flask. The organic solvent was removed using a rotary
[22]. Vesicles are a common lipid-based transdermal formulation, evaporator, leaving a layer of membrane material on the wall of the
which include liposomes and niosomes etc. Compared with other flask. The dried surfactant film was then hydrated with 5 mL of water at
transdermal agents, lipid-based nanoparticles may enhance the trans- 60 °C for 30 min with gentle spinning. After that, the suspension was
dermal permeation ability of drugs by changing the structure of the probe-sonicated (125 W, 3 mm probe, 0.5 s/0.5 s) for 4 min to obtain
stratum corneum lipids based on the Like Dissolves Like Theory. These Cel Nio. To increase the elasticity of Nio, Span 20 was used in combi-
nanoparticles may also function as a drug reservoir that can release a nation with Span 60 at a weight ratio of 1/3 to 3/1, and cholesterol was
drug at a steady rate, allowing prolonged release and activity of the used at a weight proportion of 1/2 to 1/5. The hydration period was
drug [23]. Niosomes (Nio), a type of vesicle whose bilayer membrane is screened from 10 to 60 min. The hydration volume was decided ac-
mainly made of nonionic surfactant incorporating cholesterol, have cording to the model of the sonication probe. The probe sonication
higher chemical stability than liposomes because of the surfactants period was screened from 1 to 10 min. All screenings were single-factor
used. Nio also contain a larger amount of surfactant than liposomes, screenings, and the screening parameters were size distribution, poly-
which may also offer a stronger penetration ability [24]. In the devel- dispersity index (PDI), and drug loading.
opment of new formulations, both hydrophilic and lipophilic drugs
could be loaded into Nio. 2.4. Characterization of Cel Nio

2. Materials and methods 2.4.1. Size distribution, zeta potential, and yield
The size distribution and zeta potential of Cel Nio was measured by
2.1. Materials using a Zetasizer Nano-Series (Malvern, United Kingdom). The samples
for particle size measurement were diluted approximately five fold with
Cel were obtained from Chengdu Pufei De Biotech Co., Ltd. water. Size distribution was expressed as percentage of intensity. The
(Chengdu, China). Span 20, Span 60, and cholesterol were all pur- PDI value was determined to evaluate the quality of size distribution.
chased from Aladdin Industrial Corporation (Shanghai, China). For yield measurement, samples were prepared by diluting Cel Nio
Carbopol 974 powder was obtained from Chineway (Shanghai, China). by 10 fold with absolute ethanol. Next, the Cel concentration in the
Imiquimod cream (IMQ; 5%) as Aldara and tacrolimus ointment (0.1%) preparation was measured by HPLC according to the method mentioned
as Protopic were purchased from Kangaiduo Drug Store (Guangzhou, above. Yield (Y%) was then calculated using the following formula:
China). Paraformaldehyde (4%) was purchased from Phygene (Fuzhou,
W
China). High-performance liquid chromatography (HPLC) and LC/MS/ Y%= × 100%
Wtotal
MS systems using acetonitrile and methanol were purchased from
Merck (Darmstadt, Germany). Absolute ethanol and chloroform were where, W and Wtotal are the amount of Cel measured with HPLC and the
purchased from Damao Chemical Reagent Factory (Tianjin, China). total amount of Cel put into the preparation, respectively. Settled Nio
Hair remover spray foam (Linco Care Ltd, Manchester, England) was were stored under 4 °C.
purchased from Sasa (Macau, China). Yield (Y%) of Cel Nio was measured with an Agilent 1200 Series

2
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

Fig. 1. Particle size distribution (A) and TEM image of Cel Nio (B, C). Bar, 100 nm.

HPLC-DAD system (Agilent Technologies, Santa Clara, USA). A C18 2.5.2. In vitro permeation studies
column (Agilent Zorbax; 4.6 mm × 250 mm, 5 μm) equipped with a In vitro permeation studies were performed using a vertical Franz
guard column was used for the separation at room temperature. The diffusion cell and a 1-cm2 permeation square (PermeGear, USA). Skin
mobile phase consisted of methanol-0.4% H3PO4 solution (85:15, v/v), tissues were prepared according to the method described above. The
and the elution rate was 1 mL/min. All samples were injected into the receiving medium consisted of 20% ethanol in phosphate buffered so-
column at a volume of 10 μL. The detection wavelength of Cel was lution (PBS) to maintain sink conditions [25]. The skin was paved and
430 nm. sandwiched between two glass containers with the stratum corneum
facing upwards. Next, 8 mL of ethanol solution was added to the re-
ceiving cells, which were then stirred at a steady rate at 32 °C in a water
2.4.2. Morphology bath. Subsequently, 0.4 g of Cel Nio gels (0.02%, w/w) or Cel gels
Transmission electron microscopy (TEM) was performed to in- (0.02%, w/w) were loaded into the donor cells and sealed. At each time
vestigate the morphology of Cel Nio. A few drops of the diluted Nio point of 6, 12, 14, 16, 18, 20, 22, and 24 h, 1.5 mL of receiving medium
were dried on a carbon-coated copper grid for approximately 1 min. was collected from the receiving cells and the same volume of fresh
The free Nio particles were absorbed by a piece of filter paper beneath receiving medium was added to the receiving cells.
the mesh. Phosphotungstic acid (2%) was used for negative staining. At the end of the permeation test, psoriatic skin was washed care-
The sample was observed with a Hitachi H-7650 electron microscope fully, submerged and chopped in a certain amount of dry ethanol, and
(Hitachi, Tokyo, Japan) at an accelerating voltage of 80 kV. then subjected to bath sonication to extract Cel from the skin. LCeMS/
MS was used to quantify Cel content in the skin and receiving medium.
Each group was tested thrice.
2.5. In vitro permeation studies
LC–MS/MS quantitative analysis was performed with an ABI 4000
Q-Trap™ hybrid triple quadrupole linear ion trap mass spectrometer
2.5.1. Preparation of psoriatic skin
(Applied Biosystems, Foster City, CA) used in the positive ion mode,
The dorsal hair of C57BL/6 mice was removed using a razor com-
accompanied by an Agilent 1100 HPLC system (Agilent Technologies,
bined with a depilatory cream. To prevent the depilatory cream from
Santa Clara, USA). The separation was performed using a C18 column
affecting the anti-psoriasis effect of Cel, the back of the mice was wa-
(Agilent Zorbax; 4.6 mm × 250 mm, 5 μm) with a guard column
shed with water after hair removal, and the treatment was administered
(Agilent Zorbax) at 30 °C. The mobile phase was run at 1.0 mL/min, and
three days after hair removal. Cream containing 3 mg of IMQ was ap-
it consisted of 90% acetonitrile in 5% water and 10% aqueous formic
plied topically every day in the morning. After five days of adminis-
acid (0.1%), both containing 2 mM ammonium acetate. The multiple
tration, all mice were sacrificed using CO2, and the entire back skin was
reaction monitoring mode was used for quantification. The iron tran-
isolated as psoriatic skin. All subcutaneous tissues were carefully re-
sition of Cel was 451.3 → 201.1 m/z, whereas that of hydrocortisone,
moved using a tweezer.

3
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

Fig. 2. Psoriatic symptoms in the dorsal skin of mice induced by topical application of IMQ. On the sixth day, photos of the dorsal skin were taken (A). H & E staining
(B, bar =200 μm) was performed to show changes in the inner skin after treatment with Blank Nio gel, Cel gel, Cel Nio gel, and tacrolimus ointment (positive
control). Enlarged figure of the normal (left) and IMQ model (right) groups are also presented for detail (C, Bar =100 μm).

an internal standard, was 363.1→ 121.2 m/z [26–28]. The analysis mouse (4 mg/kg) for Cel and 0.1 mg/day/mouse for tacrolimus [30].
conditions were as follows: DP, 50 V; CE, 30 V; ion-spray voltage,
4500 V; ion source temperature, 350 °C; detecting time, 200 ms. 2.6.3. Histological evaluation
Animal skin samples were collected, fixed with 4% paraformalde-
2.6. In vivo study of Cel Nio gels in psoriasis mouse models hyde, and embedded in paraffin. After deparaffinization and rehydra-
tion, the sections were stained with hematoxylin and eosin (H & E). A
2.6.1. Preparation of formulation microscope (BDS 200; Aote, China) was used to observe the sections.
To increase the retention time of the topical agents on the skin,
adequate viscosity is needed. Briefly, 0.8% (w/w) Carbopol 974 was 2.6.4. Psoriasis Area Severity Index (PASI) evaluation
dissolved in a certain amount of water with stirring overnight. A certain PASI value was evaluated before each IMQ administration to assess
amount of Cel Nio was added to the Carbopol 974 solution, and the the severity of inflammation of the skin [31]. The severity of erythema
mixture was stirred for 15 min until homogenized. The pH of the gels was scored from 0 to 4 (0, none; 4, severe). The overall score was the
was then adjusted to 7 by the dropwise addition of triethanolamine. To sum of two scores. PASI evaluation lasted for six days.
prepare drug suspension gels, Cel was ground and added to blank
Carbopol 974 gels using the same method of preparation. 2.6.5. Weight ratio of spleen to body (spleen/body wt%)
The spleen is the largest immune-related organ in the human body
2.6.2. Establishment of imiquimod-induced psoriatic mouse models [32]. Its main function is to participate in immune reactions, phago-
IMQ, a potent immune activator and an TLR7/8 ligand, was re- cytosis, and the elimination of aging red blood cells, bacteria, or foreign
ported to induce and exacerbate psoriasis [29]. Thus, it was used to as a antigens; produce lymphocytes and monocytes; and store blood. Spleen
model drug in this study. Briefly, female C57BL/6 mice were allocated weight is considered an indicator of immune stimulation, and an in-
at random into six groups of six mice each. IMQ (active compound crease in spleen/body weight ratio may indicate proliferation of im-
dosage: 3.125 mg per day) was applied to 6 cm2 of the dorsal skin. Four mune cells in the spleen, which probably reflects the activation of the
hours after IMQ administration, 0.4 g of Cel Nio gel, Cel gel, or 0.3 g of immune system [33]. Mouse spleens were isolated after sacrifice and
tacrolimus ointment (a positive control) was applied topically to the quickly weighed to avoid errors due to dehydration.
dorsal skin on the back of the mice of the respective treatment groups. A
group of mice administered only IMQ at the same dosage served as the 2.6.6. Measurement of Cel content in the skin and blood using LC–MS/MS
model group. Each group was administered its respective treatment After five days of administration, the mice were sacrificed using
once a day for five days. The theoretical dosage was 0.08 mg/day/ CO2, and total blood samples were collected via the ophthalmic artery,

4
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

Fig. 3. PASI scores of skin lesions in all groups including erythema and scaling were recorded for six days; each score ranged from 0 to 4, and the overall score ranged
from 0 to 8. *Significantly different compared with the IMQ model group (p < 0.05), (n = 6). Cel, celastrol; Nio, niosomes.

Table 2
Cel content in the skin and blood following application of Cel formulation
(0.08 mg/day/mouse) on IMQ-induced psoriatic mouse models.
Groups CCel in skin (ng/mg) CCel in blood (ng/mL)

0.02% Cel gel 0.86 ± 0.03 ND


0.02% Cel Nio gel 1.67 ± 0.05* ND

Data are shown as mean ± SD (n = 6). *Significantly different compared with


the Cel gel group (p < 0.05). ND, not detected; Cel, celastrol; Nio, niosomes;
CCel in skin, Cel concentration in the skin; CCel in blood, Cel concentration in blood.

and 4 °C for 5 min, and 200 μL of the supernatant was filtered using a
0.22-μm filter to obtain skin samples. Before the determination of Cel
content, internal standard solution (50 μg/mL hydrocortisone in me-
thanol) was mixed with blood samples at a ratio of 1:5 (v/v)”, Next,
300 μL of ethyl acetate was added to the mixture, which was then ex-
tracted by vortex mixing for 2 min [34]. After centrifuging the mixture
at 12,500 rpm for 15 min, 200 μL of the supernatant was collected and
evaporated. The remaining solids were dissolved in 100 μL of acetoni-
trile, and all samples were analyzed for Cel content with LC–MS/MS.

2.6.7. Cytokine level evaluation


Fig. 4. Spleen/body weight ratio was calculated after the mice were sacrificed. RIPA Lysis Buffer (Beyotime, China) containing 1 mM of phe-
*
Significantly different compared with the IMQ model group (p < 0.05), Cel, nylmethanesulfonyl fluoride was used to extract total protein from the
celastrol; Nio, niosomes. skin tissue homogenate. Total protein concentration was determined
using a BCA Protein Assay Kit (Tiangen Biotech, China). Total protein
and the dorsal skin was isolated. Plasma samples were obtained im- was diluted to approximately 500 μg/mL before the IL-23P40, IL-22, IL-
mediately by centrifugation at 2000 ×g for 10 min and then stored at 17A, and IFN-γ levels were analyzed using a Bio-Plex™ 200 system (Bio-
−20 °C until analysis. Skin samples were washed with PBS and cut into Rad, USA) according to the manufacturer’s instructions.
small pieces; each 100 mg of the skin was placed in 1 mL of dry ethanol.
Next, the skin samples were homogenized using a Tissuelyser II 2.6.8. Quantitative PCR (qPCR)
(Qiagen, CA, USA) combined with grinding beads at a frequency of 30/s Total RNA was extracted by homogenizing 100 mg of mice skin in
for 15 min. Subsequently, the samples were centrifuged at 10,000 rpm 1 mL of RNAiso Plus (TaKaRa, Korea Biomedical Inc.). The isolated total

5
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

Fig. 5. Inflammatory cytokine content in the skin of mice from all groups. A. IL-22; B. IL-23p40; C. IL-17; D. IFN-γ. *Significantly different compared with the IMQ
model group, p < 0.05. #Significantly different compared with the Cel gel group, p < 0.05. (n = 6). Values are shown as mean and SD. Cel, celastrol; Nio,
niosomes.

RNA was then reverse-transcribed into cDNA using a PrimeScript RT 3. Results and discussion
Reagent Kit (TaKaRa, Bio Inc.) on a C1000 touch thermal cycler system
(Bio-Rad). Afterwards, qPCR was conducted using SYBR Premix Ex Taq 3.1. Characterization of Cel Nio
II (TaKaRa, Bio Inc.) and a Mx3005 P qPCR System (Agilent
Technologies, USA). The primer sequences used are shown in Table 1. After preliminary screening of the formulation (data not shown), a
We used the comparative ΔΔCT method to quantify gene expression. Span 20, Span 60, and cholesterol ratio of 3:1:1 (w/w/w), hydration
Target gene expression levels in the test samples were normalized to time of 30 min, and sonication time of 2 min were selected. This opti-
endogenous GAPDH levels and expressed as fold differences relative to mized formulation had a smaller particle size and higher drug loading
GAPDH gene expression in the untreated baseline controls. All assays ability. Therefore, this formulation was selected to be used in the
were performed in triplicate, and the experiments were repeated at subsequent experiment. A graph of size distribution is shown in
least three times. Fig. 1(A). The particle size of Cel Nio was approximately 147.4 nm ±
Data are showed as mean ± SD. Significant differences between 5.6 nm with a PDI of 0.258 ± 0.02, indicating a homogenous popu-
different groups were determined by repeated measures or one-way lation for a colloidal system [35]. The zeta potential was -48.9 mV ±
ANOVA using GraphPad Prism 7, and p < 0.05 was considered sig- 1.1 mV, indicating a good colloidal stability [36]. The Y% was
nificant. 90.42% ± 3.38%. TEM results of Cel Nio are shown in Fig. 1(B, C). The
results showed that numerous Nio fused in the absence of water, and all
single Nio had spherical shape. Cel crystals were not observed in the
TEM results. These size analysis results correlated well with the results
of size distribution analysis.

6
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

Fig. 6. The mRNA levels of cytokines in the skin of mice after administration of the different formulations. A, IL-22; B, IL-23; C, IL-1β; D, IL-6; E, IFN-γ; F, IL-17; G,
STAT3. *Significantly different compared with the IMQ only group, p < 0.05. #Significantly different compared with the Cel gel group, p < 0.05, (n = 6). Values
are shown as mean and SD. Cel, celastrol; Nio, niosomes.

The permeation mechanism of formulations containing nonionic- 3.3. Histology evaluation


surfactant, such as Nio, was reported previously [37]. Briefly, surfac-
tant can penetrate into intercellular spaces and increase the fluidity of Histology evaluation results are shown in Fig. 2. Compared to the
the lipid. The penetrating surfactant can also bind with keratin fila- normal group, the IMQ model group showed obviously thickened skin
ments and disrupt the intercellular structure. In addition, nonionic covered with white desquamation. After treatment with different for-
surfactants are reported to emulsify sebum, thereby raising the ther- mulations, white scales and erythema were apparently reduced in the
modynamic coefficient of drugs and improving their permeation ability. Cel Nio gel-treated groups and tacrolimus-treated group. H & E-stained
skin sections showed that inflammatory cells, hyperkeratosis and
parakeratosis were reduced in the Cel Nio gel group compared with the
3.2. In vitro permeation study IMQ model group.

Drug content in the skin was measured by HPLC. For the Cel Nio gel 3.4. PASI evaluation
group, the Cel content was 465.3 ± 84.1 ng/cm2. For the Cel gel
group, only 35.5 ± 1.6 ng/cm2 of Cel was detected, indicating that the Skin erythema and scaling condition in all groups were scored, and
enhanced solubility of Cel in Nio may enhance its skin permeation. the results are shown in Fig. 3. The skin condition of mice in the Blank
However, Cel was not detected in the receiving medium even at the 24- Nio gel and Cel gel groups were slightly better than that in the IMQ
h time point. model group, suggesting that Blank Nio gel and Cel gel had a slight
positive effect against inflammation in psoriatic skin, possibly a
cleaning or moisturizing effect. However, the Cel Nio gel group showed

7
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

a much lower scaling and erythema score than the Blank Nio gel and Acknowledgement
Cel gel groups. After six days, the least and most severe pathological
skin conditions were observed in the positive control and IMQ model This study was financially supported by the Macao Science and
groups, respectively. Technology Development Fund (0013/2018/A1).

References
3.5. Spleen/body weight ratio evaluation

[1] W.H. Organization, Global Report on Psoriasis. 2016, WHO Library Cataloguing-in-
As shown in Fig. 4, after five-day administration of IMQ, the average Publication Data, 2016.
spleen/weight ratio in the IMQ model group was approximately three [2] J. Kim, J.G. Krueger, Psoriasis and Other Skin Inflammatory Diseases,
times that of the normal group, indicating a higher level of immune Inflammation: From Molecular and Cellular Mechanisms to the Clinic, (2018), pp.
1091–1104.
cells in the spleen. Compared to the normal group, the Cel Nio gel-
[3] J.M. Crow, Psoriasis uncovered: science is finally getting to grips with this enig-
treated and tacrolimus-treated groups both showed a significant de- matic autoimmune disease, Nature 492 (2012) S50–S50.
crease in spleen weight ratio, which was more obvious than that shown [4] L. Dubertret, U. Mrowietz, A. Ranki, P. Van De Kerkhof, S. Chimenti, T. Lotti,
by the group treated with Blank gel or Cel gel. G. Schäfer, European patient perspectives on the impact of psoriasis: the EUROPSO
patient membership survey, Br. J. Dermatol. 155 (2006) 729–736.
[5] C. Ferrándiz, S. Puig, R. Pujol, A. Smandía, Prevalence of psoriasis in Spain
(Epiderma Project: phase I), J. Eur. Acad. Dermatol. Venereol. 15 (2001) 20–23.
3.6. Measurement of Cel content in the skin and blood [6] N. Baumgarth, C.L. Bevins, Autoimmune disease: skin deep but complex, Nature
449 (2007) 551.
Cel content in mouse skin and blood was measured in the groups [7] P. Mudigonda, T. Mudigonda, A.N. Feneran, H.S. Alamdari, L. Sandoval,
S.R. Feldman, Interleukin-23 and interleukin-17: importance in pathogenesis and
treated with Cel gel and Cel Nio gel. As shown in Table 2, Cel Nio gel-
therapy of psoriasis, Dermatol. Online J. 18 (2012) 1.
treated mice showed higher drug content in the skin than the Cel gel [8] L. Grine, L. Dejager, C. Libert, R.E. Vandenbroucke, An inflammatory triangle in
groups, Furthermore, combined with the results of the in vitro per- psoriasis: TNF, type I IFNs and IL-17, Cytokine Growth Factor Rev. 26 (2015)
25–33.
meation test, it was indicated that incorporation of Cel into Nio im-
[9] S. Rizvi, K. Chaudhari, B.A. Syed, The Psoriasis Drugs Market, Nature Publishing
proved the permeation of Cel. While no blood Cel content was detected Group, 2015.
in the Cel gel and Cel Nio gel group. Therefore, more studies need to be [10] J.M. Crow, Silencing psoriasis, Nature 492 (2012) S58.
performed to explain how does Cel take effect in vivo. [11] D.B. Dubin, W. Tanner, R. Ellis, Biologics for Psoriasis, Nature Publishing Group,
2003.
[12] C. Wu, H.-Z. Jin, D. Shu, F. Li, C.-X. He, J. Qiao, X.-L. Yu, Y. Zhang, Y.-B. He, T.-
J. Liu, Efficacy and safety of Tripterygium wilfordii Hook F versus acitretin in
3.7. Cytokine level measurement moderate to severe psoriasis vulgaris: a randomized clinical trial, Chin. Med. J. 128
(2015) 443.
The levels of IL-22, IL-23p40, IL-17, and IFN-γ were measured, and [13] S. Meng, Z. Lin, Y. Wang, Z. Wang, P. Li, Y. Zheng, Psoriasis therapy by Chinese
medicine and modern agents, Chin. Med. 13 (2018) 16.
the results are shown in Fig. 5. Compared with those in the normal
[14] S.H. Venkatesha, K.D. Moudgil, Celastrol and Its Role in Controlling Chronic
group, IL-17, IL-23p40, IFN-γ, and IL-22 levels were significantly higher Diseases, Anti-Inflammatory Nutraceuticals and Chronic Diseases, Springer, 2016,
after administration of IMQ, indicating that topical administration of pp. 267–289.
IMQ induced the secretion of IL-23p40, IL-17, IFN-γ, and IL-22. The Cel [15] S.-R. Chen, Y. Dai, J. Zhao, L. Lin, Y. Wang, Y. Wang, A mechanistic overview of
triptolide and celastrol, natural products from Tripterygium wilfordii hook F, Front.
Nio gel and tacrolimus groups both showed obvious inhibition of IL- Pharmacol. 9 (2018) 104.
23p40, IL-17, IFN-γ, and IL-22 release. Moreover, the levels of these [16] B. Astry, S.H. Venkatesha, A. Laurence, A. Christensen-Quick, A. Garzino-demo,
cytokines in the Cel gel and Blank Nio gel groups were significantly M.B. Frieman, J.J. O’Shea, K.D. Moudgil, Celastrol, a Chinese herbal compound,
controls autoimmune inflammation by altering the balance of pathogenic and
higher than those in the Cel Nio gel group, indicating that Cel Nio gel regulatory T cells in the target organ, Clin. Immunol. 157 (2015) 228–238.
formulation had a good anti-inflammatory effect. [17] L. Hong, Establishment of a Mouse Model of Lupus-like Syndrome and Anti-in-
flammatory Immunosuppressive Effect of Tripterine, Shanghai: PhD thesis of Fudan
University (2006).
3.8. qPCR analysis [18] C. Mingxiang, F. Yujing, W. Dingyong, Y. Wen, Research progress of tripterine,
Chin. Patent Med. 32 (2010) 473–476.
[19] ChemSpider, Royal Society of Chemistry, 2015.
qPCR was used to quantify the mRNA levels of inflammatory cyto- [20] G. Verma, Transdermal drug delivery system, advance development and evaluation-
kine genes, which represent the severity of psoriasis. The results are a review, Int. J. Pharm. Sci. Res. 8 (2017) 385.
shown in Fig. 6. Compared with the normal group, the mRNA levels of [21] L. Norlén, Update of technologies for examining the stratum corneum at the mo-
lecular level, Br. J. Dermatol. 171 (2014) 13–18.
IL-22, IL-23, IL-1β, IL-6, IFN-γ, and IL-17 F significantly increased in the [22] M.B. Brown, G.P. Martin, S.A. Jones, F.K. Akomeah, Dermal and transdermal drug
IMQ model group. However, the mRNA levels of these cytokines de- delivery systems: current and future prospects, Drug Deliv. 13 (2006) 175–187.
creased in both Cel gel and Cel Nio gel groups. These results indicated [23] P.L. Honeywell-Nguyen, J.A. Bouwstra, Vesicles as a tool for transdermal and
dermal delivery, Drug Discov. Today Technol. 2 (2005) 67–74.
that Cel possessed a good anti-inflammatory effect and that this effect [24] R. Sharma, R. Mahatma, M. Bharkatiya, A. Goyal, Niosomes -as potential drug
was improved by the incorporation of Cel into Nio. Compared with delivery system, Int. J. Drug Res. Technol. 2 (2017) 8.
those in the IMQ model group, the levels of IL-1β, IL-6, etc. in the Cel [25] Y. Chen, L. Zhou, L. Yuan, Z.-h. Zhang, X. Liu, Q. Wu, Formulation, characteriza-
tion, and evaluation of in vitro skin permeation and in vivo pharmacodynamics of
gel group also decreased, probably because the Cel dose was much surface-charged tripterine-loaded nanostructured lipid carriers, Int. J.
higher than the minimum effective dose and the amount of drug par- Nanomedicine 7 (2012) 3023.
ticles that were in direct contact with the skin was sufficient to exert a [26] X.-k. OuYang, M.-q. Cai, X.-h. Chen, M.-c. Jin, Development and validation of a
liquid chromatography coupled with atmospheric-pressure chemical ionization ion
therapeutic effect on the immune system.
trap mass spectrometric method for the simultaneous determination of triptolide,
tripdiolide, and tripterine in human serum, J. Anal. Toxicol. 32 (2008) 737–743.
[27] Y. Huang, D. Zhou, T. Hang, Z. Wu, J. Liu, Q. Xu, X. Xie, J. Zuo, Z. Wang, Y. Zhou,
4. Conclusion Preparation, characterization, and assessment of the antiglioma effects of liposomal
celastrol, Anticancer Drugs 23 (2012) 515–524.
In this study, Nio hydrogels loaded with Cel were developed as an [28] S. Wu, W. Li, T. Mujamdar, T. Smith, M. Bryant, F.L. Tse, Supported liquid ex-
traction in combination with LC‐MS/MS for high‐throughput quantitative analysis
effective treatment of psoriasis-like mouse model induced by IMQ. Cel of hydrocortisone in mouse serum, Biomed. Chromatogr. 24 (2010) 632–638.
Nio had an average particle size of approximately147 nm with small [29] J.K. Wu, G. Siller, G. Strutton, Psoriasis induced by topical imiquimod, Australas. J.
PDI. Encapsulation of Cel into Nio increased the water-solubility and Dermatol. 45 (2004) 47–50.
[30] L. Sun, Z. Liu, L. Wang, D. Cun, H.H. Tong, R. Yan, X. Chen, R. Wang, Y. Zheng,
permeation of Cel in the skin, significantly improving its anti-psoriasis Enhanced topical penetration, system exposure and anti-psoriasis activity of two
activity in the mice model. Thus, the results indicated the potential particle-sized, curcumin-loaded PLGA nanoparticles in hydrogel, J. Control. Release
application of Cel Nio hydrogel in psoriasis therapy. 254 (2017) 44–54.

8
S. Meng, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110352

[31] L. van der Fits, S. Mourits, J.S. Voerman, M. Kant, L. Boon, J.D. Laman, [34] Q. Xu, M. Huang, M. Jin, Q. Ren, LC–APCI–MS–MS for the determination of ce-
F. Cornelissen, A.-M. Mus, E. Florencia, E.P. Prens, Imiquimod-induced psoriasis- lastrol in human whole blood, Chromatographia 66 (2007) 735–739.
like skin inflammation in mice is mediated via the IL-23/IL-17 axis, J. Immunol. [35] S. Singh, P. Parashar, J. Kanoujia, I. Singh, S. Saha, S.A. Saraf, Transdermal po-
182 (2009) 5836–5845. tential and anti-gout efficacy of Febuxostat from niosomal gel, J. Drug Deliv. Sci.
[32] A. De Porto, A. Lammers, R. Bennink, I. Ten Berge, P. Speelman, J. Hoekstra, Technol. 39 (2017) 348–361.
Assessment of splenic function, Eur. J. Clin. Microbiol. Infect. Dis. 29 (2010) [36] T.M. Riddick, Control of colloid stability through zeta potential, Blood 10 (1968).
1465–1473. [37] I. Som, K. Bhatia, M. Yasir, Status of surfactants as penetration enhancers in
[33] M.F. Cesta, Normal structure, function, and histology of the spleen, Toxicol. Pathol. transdermal drug delivery, J. Pharm. Bioallied Sci. 4 (2012) 2.
34 (2006) 455–465.

You might also like