Sunteți pe pagina 1din 10

Research Article

Cite This: ACS Appl. Mater. Interfaces 2018, 10, 9315−9324 www.acsami.org

Novel Solid Lipid Nanoparticle with Endosomal Escape Function for


Oral Delivery of Insulin
Yining Xu,† Yaxian Zheng,† Lei Wu, Xi Zhu, Zhirong Zhang, and Yuan Huang*
Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan
University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041 Sichuan, China
*
S Supporting Information

ABSTRACT: Although nanoparticles (NPs) have been demonstrated


as promising tools for improving oral absorption of biotherapeutics,
most of them still have very limited oral bioavailability. Lyso−endosomal
degradation in epithelial cells is one of the important but often-neglected
physiological barriers, limiting the transport of cargoes across the
intestinal epithelium. We herein reported a solid lipid nanoparticle
(SLN) platform with a unique feature of endosomal escape for oral
protein drug delivery. The SLNs consisted of a solid-lipid shell, which
contained an endosomal escape agent (GLFEAIEGFIENGWEGMIDG-
WYG, HA2), and an aqueous core that is loaded with insulin (INS HA2-
O-SLNs). SLNs without and with the HA2 peptide in the aqueous core
(INS SLNs and INS HA2-W-SLNs, respectively) were used as the
control groups. Our study showed that INS HA2-O-SLNs effectively
facilitated the escape of the loaded insulin from the acidic endosomes,
which preserved the biological activity of insulin to a greater extent during the intracellular transport. The spatial location of the
HA2 peptide was demonstrated to determine the endosomal escape efficiency. As demonstrated in the intracellular trafficking of
SLNs, INS HA2-O-SLNs displayed much less distribution in late endosomes and lysosomes. Meanwhile, insulin in INS HA2-O-
SLNs exhibited the highest transepithelial permeation efficiency, with 2.19 and 1.72 folds higher accumulated amount in the
basolateral side as compared to that in INS SLNs and INS HA2-W-SLNs. In addition, insulin from INS HA2-O-SLNs exhibited
the highest insulin permeation in various regions of small intestines. INS HA2-O-SLNs generated an excellent hypoglycemic
response following oral administration in diabetic rats. Thus, such functional SLNs demonstrated a great potency for oral delivery
of peptide/protein drugs.
KEYWORDS: solid lipid nanoparticles, insulin, endosomal escape, HA2 peptides, oral delivery

1. INTRODUCTION the terminal enzymatic degradative compartments.12 This


Therapeutic peptides and proteins have been extensively lysosomal degradation would drastically disrupt the protein
utilized in the treatment of numerous diseases for their integrity and thus impeded the transport of drugs into blood
specificity and high potency.1 However, these biotherapeutics circulation.2 Therefore, lyso−endosomal entrapment is a
are currently only limited to parenteral administration, which formidable but often-neglected barrier. An innovative and
result in unwanted pain, poor patient compliance, and several practical strategy utilizing endosomal escape might be desired
side effects. Although oral administration is much more for oral protein delivery.
preferred, bioavailability of these drugs is often limited by To date, extensive efforts for endosomal escape have been
several physiological barriers in the gastrointestinal tract made on cancer therapies and gene transfer during the past
(GIT).2,3 Since 1980s, nanotechnology has been reported to years.13−15 However, few research studies have been focused on
protect drugs from harsh conditions in the GIT.4−6 However, it the nanoplatforms combining the endosomal escaping strategy
is still a big challenge to improve the drug permeability across to improve the oral bioavailability of biotherapeutics.16 The
the intestinal epithelium.7−9 hemagglutinin-2 (GLFEAIEGFIENGWEGMIDGWYG, HA2)
Endocytic pathway is the major route for most nanoparticle peptide, derived from influenza virus coat, was introduced as a
(NP) uptake.10,11 However, the amount of NPs across the nontoxic fusogenic agent. The protonation of the HA2 peptide
intestinal epithelium was always much less than those engulfed could induce a conformational change to α-helixes in response
in cells. The dilemma of “easy endocytosis and hard to endosomal acidification and destabilize the membrane. The
transepithelial transport” is one of the major problems that
hinders the therapeutic effect of proteins in circulation. After Received: January 10, 2018
internalization, these NPs are commonly entrapped in Accepted: February 27, 2018
endosomes and eventually degraded in lysosomes, which are Published: February 27, 2018

© 2018 American Chemical Society 9315 DOI: 10.1021/acsami.8b00507


ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

contents were supposed to avoid lysosomal degradation, and (HPLC) (Agilent 1200 series and Alltech, respectively) method, and
then, the transepithelial transport of drugs would be further free FITC-INS was quantified by a multimode reader.10
improved.17 Therefore, we hypothesized that a well-designed 2.3. Colloidal Stability of SLNs. The colloidal stability of the
nanocarrier with the HA2 peptide might held out a SLNs was investigated by incubating SLNs with simulated gastric fluid
[SGF, with and without pepsin (0.32%, w/v)] and simulated intestinal
considerable promise for oral delivery of biomacromolecules. fluid [SIF, with and without trypsin (1%, w/v)] at 37 °C.23
Herein, a novel delivery system was constructed with the Furthermore, SGF was preneutralized by sodium bicarbonate solution
function of endosomal escape for orally administered protein (3%, w/v).24,25 At predetermined time intervals, the particle size of
drugs, aiming to protect these drugs from lysosomal SLNs was determined. TEM was also used to confirm the results.
degradation and enhance their oral bioavailability. The water 2.4. Drug and HA2 Peptide Release of SLNs. The in vitro
in oil in water (W/O/W) double emulsion was used to co- release of insulin and HA2 peptide from SLNs was investigated using
encapsulate hydrophilic insulin (INS, model protein drug) and the dialysis method.26−28 Briefly, 2 mL of SLNs was added in dialysis
the hydrophobic HA2 peptide (endosomal escape agent) in tubing (MWCO: 100 kDa) and then dialyzed against 1000 mL of
solid lipid nanoparticles (SLNs). INS HA2-W-SLNs (HA2 release medium (pH 2, 5.5, and 6.8) at 37 °C. At a predetermined
peptide loaded in the inner aqueous phase of SLNs) and INS time, the concentrations of insulin and HA2 peptide in dialysis tubing
were determined by RP-HPLC. The in vitro release in the pH 2
HA2-O-SLNs (HA2 peptide mainly loaded in the outer oil medium neutralized by sodium bicarbonate solution was also
phase of SLNs) were respectively prepared and supposed to performed.
have different release kinetics of the fusogenic agent. The effect 2.5. Cell Experiments. 2.5.1. Cell Culture and Cytotoxicity
of spatial location of the HA2 peptide in SLNs was thoroughly Assay. Human colon carcinoma Caco-2 cells were obtained from the
investigated via cellular uptake and intracellular trafficking. Institute of Biochemistry and Cell Biology (Shanghai, China) and
Furthermore, transepithelial transport efficiency of formulations cultured as the method described before.10,29 The in vitro cytotoxicity
was also conducted in vitro as well as ex vivo. Finally, in vivo of SLNs was evaluated. In brief, the Caco-2 cells were incubated with
studies were performed in diabetic rats to elucidate the SLNs for 3 h and were applied to methylthiazolyl tetrazolium (MTT)
assay.10 Hank’s balanced salt solution (HBSS)-treated group was used
pharmacological and pharmacokinetics of INS SLNs with the
as the control group.
HA2 peptide after oral administration. 2.5.2. In Vitro Cellular Uptake. 2.5.2.1. Flow Cytometry Method.
The cellular uptake of SLNs was quantified by flow cytometry.30 In
2. MATERIALS AND METHODS brief, Caco-2 cells were seeded in 12-well plates for 5 days. Then,
2.1. Materials. Porcine insulin was gained from Wanbang Caco-2 cells were incubated with fluorescence-labeled SLNs (FITC-
Biochemical Co., Ltd. GLFEAIEGFIENGWEGMIDGWYG (HA2) insulin, 100 μg/mL) at 37 °C for 3 h. Afterward, the cells were
peptide was gained from Chinese Peptide Co., Ltd. Soybean lecithin trypsinized and redispersed in cold phosphate-buffered saline (PBS)
was supplied by Taiwei Pharmaceutical Co., Ltd. Glycerol tripalmitate and then analyzed by flow cytometry analysis (CYTOMICS FC500,
was gained from Alfa Aesar Co., Ltd. Pluronic F68 was gained from Beckman Coulter, Miami, FL, USA).
Nanjing Weier Chemical Co., Ltd. Stearic acid was gained from 2.5.2.2. Endocytic Mechanism of SLNs. To investigate the
Huzhou Zhanwang Pharmaceutical Co., Ltd. Fluorescein isothiocya- endocytic mechanism of SLNs, the inhibitors of metabolism (5 mM
nate (FITC) and tetramethyl rhodamine isothiocyanate (TRITC) sodium azide), clathrin-mediated endocytosis (30 μM chlorproma-
were gained from Sigma-Aldrich. Rabbit anti-Rab5 and mouse anti- zine), caveolae-mediated endocytosis (500 nM filipin), and micro-
Rab7 antibodies were obtained from Abcam. LysoTracker Red was pinocytosis (12 μg/mL amiloride) were incubated with Caco-2 cells at
purchased from Invitrogen. Other chemicals were of analytic grade. 37 °C for 30 min.31,32 Afterward, the cells were incubated with SLNs
2.2. Preparation and Characterization of SLNs. Insulin-loaded and the same amount of inhibitors for 3 h. The internalization was
SLNs (INS SLNs) as well as the two kinds of HA2 peptide and insulin analyzed by flow cytometry.
dual-loaded SLNs were produced by the micelle-double emulsion 2.5.2.3. Enzyme-Linked Immunosorbent Assay (ELISA) Method.
method.18,19 As for the preparation of INS SLNs, 5 mg of insulin and The ELISA method was selected to test the internalization of insulin
25 mg of sodium cholate were dissolved in pH 2 hydrochloric acid with activity in cells. Caco-2 cells were incubated with various SLN
(HCl) solution. Then, 1 mL of dichloromethane containing 15 mg of suspensions (insulin, 100 μg/mL) at 37 °C for 3 h. Then, the cell
soybean lecithin, 15 mg of tripalmitin, and 1 mg of stearic acid was extracts containing active insulin were obtained by several freeze−thaw
added. The mixture was emulsified by probe sonication for each cycles. The concentration of active insulin in supernatants of cell
ultrasound 5 s, interval 5 s at 80 W within 2 min. Pluronic F68 solution extracts was measured by using the porcine insulin ELISA kit (R&D
(4 mL; 2%, w/v) was then added and sonicated in the same way. After System, Inc., MN, USA)33 and calibrated by the BCA assay kit
eliminating the organic solvent by evaporation for 30 min at 37 °C, (KeyGen Biotech Co., Ltd., Nanjing, China).
this emulsion was diluted with Pluronic F68 solution. INS HA2-W- 2.5.3. Intracellular Endo−Lysosomal Trafficking of SLNs.
SLNs or INS HA2-O-SLNs were prepared by using the aforemen- 2.5.3.1. Colocalization of SLNs in Endosomes and Lysosomes.
tioned method, besides that the HA2 peptide was added in the inner The localization of SLNs with different endosomes (early endosomes
aqueous phase (INS HA2-W-SLNs) or oil phase of mixed solvents and late endosomes) was analyzed using immunofluorescence
(dichloromethane/methanol 9:1) (INS HA2-O-SLNs). Fluorescent- staining.10 Briefly, the cells were then incubated with SLNs at 37 °C
labeled SLNs were prepared using FITC-labeled insulin (FITC-INS) for 1 h. After incubation time, the cells were rinsed by PBS, fixed with
in the method as mentioned above.7,20,21 Meanwhile, TRITC and 4% paraformaldehyde for 15 min, permeabilized by 1% Triton X-100
FITC were used to fluorescently label INS and HA2 peptide, for 10 min, and blocked with 5% normal goat serum for 1 h. Then,
respectively. The structures of the different SLNs were visualized primary rabbit anti-Rab5 and mouse anti-Rab7 antibodies were added
under confocal laser scanning microscopy (CLSM, Live 5 DUO, Carl separately at 4 °C overnight. The secondary antibodies 594-labeled
Zeiss, Jena, Germany). Various SLNs were characterized by Malvern goat anti-rabbit and anti-mouse IgG were incubated with the cells at 37
Zetasizer Nano ZS90. The size distribution was performed by °C for 2 h. Additionally, to observe the localization of SLNs with
NanoSight LM10. Transmission electron microscopy (TEM, Hitachi lysosomes, Caco-2 cells were incubated with 50 nM LysoTracker
H-600, Tokyo, Japan) was applied to observe the morphologies of probe at 37 °C for 30 min. DAPI was used to stain cell nuclei before
SLNs.22 To evaluate the entrapment efficiency (EE %) and drug CLSM observation.10
loading (DL %) of insulin and the HA2 peptide,19 SLNs were 2.5.3.2. Hemolysis Assay. To determine the pH-sensitive
dispersed in pH 2 HCl solutions and centrifuged at 13 000 rpm for 30 amphiphilic endosomal escape of various SLNs, hemolytic assay was
min. The amounts of insulin and HA2 peptide in supernatants were used as previous studies.34,35 Briefly, red blood cells (RBCs) obtained
quantified by a reverse-phase high performance liquid chromatography from Sprague-Dawley (SD) rats were diluted 1:50 in PBS at pH 7.4,

9316 DOI: 10.1021/acsami.8b00507


ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

Figure 1. (A) Schematic diagram of SLNs. (B) Size distribution of INS SLNs (a), INS HA2-W-SLNs (b), and INS HA2-O-SLNs (c) performed by
Malvern NanoSight LM10 system. (C) TEM images of INS SLNs (a), INS HA2-W-SLNs (b), and INS HA2-O-SLNs (c). Scale bar: 100 nm.

6.5, and 5.5. Then, INS SLNs, INS HA2-W-SLNs, INS HA2-O-SLNs, supplement of water. Before oral administration of SLNs, 0.5 mL of
HA2 (HA2, 30 μg/mL), and Triton X-100 (1%, w/v) were incubated sodium bicarbonate solution (3%, w/v) was orally administrated to
with the RBC solutions at 37 °C for 2 h. A multimode reader was neutralize the gastric acid. Then, free insulin solution (50 IU/kg), INS
applied to detect the absorbance of the supernatant at 540 nm. 1% SLNs (50 IU/kg), INS HA2-W-SLNs (50 IU/Kg ) and INS HA2-O-
Triton X-100 was used as the reference for 100% hemolytic activity. SLNs (50 IU/kg) were intragastrically administrated. Also, insulin
2.5.4. Transepithelial Transport Study. The transepithelial trans- solution (5 IU/kg) was subcutaneously administrated. At different
port study of the tested samples was investigated on the Caco-2 time intervals, blood glucose levels of rats were determined by a
monolayers seeded on Transwell inserts.36 Cell monolayers were first glucose meter. Meanwhile, plasma insulin levels were measured by the
equilibrated at 37 °C by HBSS. After equilibration, the cell monolayers ELISA kit. The pharmacological availability (PA %) and relative
were incubated with INS SLNs, INS HA2-W-SLNs, INS HA2-O- bioavailability (FR %) to subcutaneous injection were calculated using
SLNs, and free insulin (insulin, 100 μg/mL) for 6 h at 37 °C. The the following formulas:
amount of permeated insulin in the basolateral chambers was AACoral × doses.c.
measured by the ELISA kit. The transepithelial electrical resistant PA (%) = × 100
AACs.c. × doseoral
(TEER) values were measured before and after incubation to explore
the integrity of cell monolayers. AUCoral × doses.c.
2.6. Ex Vivo Ligated Intestinal Loop Assay. The permeation of FR (%) = × 100
insulin in the intestinal loop was evaluated by ex vivo ligated intestinal AUCs.c. × doseoral
loop assay.37 The animal study protocol was approved by the 2.8. Statistical Analyses. All experiments were performed in
Institutional Animal Care and Use Committee of Sichuan University. triplicate unless otherwise stated and presented as mean ± standard
Male SD rats (220−250 g) were sacrificed. Then, 2 cm sections of the deviation. Statistical analyses of the data were conducted with SPSS
duodenum, jejunum, and ileum were obtained, carefully washed, and program 16.0 and two-tail Student’s t-test. Statistical significance was
ligated at both ends. Then, the suspensions of SLNs (pH 5.5 or pH defined when p < 0.05.
6.8) were injected into the loops. In addition, the sections were
incubated with pH 7.4 medium at 37 °C for 2 h. The amount of 3. RESULTS AND DISCUSSION
permeated insulin in the medium was determined by the ELISA kit.
2.7. Pharmacological and Pharmacokinetics Studies. The 3.1. Preparation and Characterization of SLNs. SLNs
pharmacological and pharmacokinetics studies of various SLNs were were prepared by the W/O/W double emulsion strategy, which
performed on diabetic rats.38 The male SD rats (220−250 g) were has been commonly used to load hydrophobic and hydrophilic
injected by streptozotocin (70 mg/kg) solution to induce diabetes. macromolecules.18,39 The primary W/O emulsion was first
Rats with the fasting blood glucose above 16 mM were diabetic obtained, and the emulsion was then emulsified in a continuous
models. Prior to the experiments, rats were fasted overnight with the aqueous phase to form the double emulsion, which contained
9317 DOI: 10.1021/acsami.8b00507
ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

Table 1. Characterization of Various Insulin-Loaded SLNs and FITC-Insulin-Loaded SLNs Prepared by the Optimized
Formulationa
samples size (nm) PDI zeta (mV) EEinsulin (%) EEHA2 (%) DL (%)
INS SLN 171.2 ± 1.6 0.23 ± 0.02 −9.2 ± 0.2 97.93 ± 0.13 8.03 ± 0.19
INS HA2-W-SLN 148.8 ± 4.2 0.17 ± 0.02 −13.1 ± 1.3 98.90 ± 0.35 75.97 ± 0.65 7.23 ± 0.26
INS HA2-O-SLN 161.6 ± 3.7 0.25 ± 0.03 −16.1 ± 0.5 98.16 ± 0.80 92.67 ± 1.11 7.52 ± 0.31
FITC-INS SLN 172.4 ± 1.5 0.22 ± 0.03 −8.4 ± 1.1 96.60 ± 0.33 7.91 ± 0.15
FITC-INS HA2-W-SLN 157.8 ± 3.1 0.24 ± 0.02 −11.0 ± 0.3 97.64 ± 0.15 78.64 ± 0.91 7.42 ± 0.26
FITC-INS HA2-O-SLN 160.2 ± 2.0 0.26 ± 0.03 −14.6 ± 0.9 97.60 ± 0.26 90.83 ± 0.76 7.71 ± 0.24
a
Data shown as mean ± SD (n = 3).

Figure 2. Variation in the particle size of insulin-loaded SLNs following incubation in (A) SGF with 0.32% pepsin, (B) neutralized SGF with 0.32%
pepsin, (C) SIF with 1% trypsin at predetermined time intervals (mean ± SD, n = 3). *p < 0.05 vs INS SLNs at 0 h; #p < 0.05 vs INS HA2-W-SLNs
at 0 h; &p < 0.05 vs INS HA2-O-SLNs at 0 h. (D) TEM images of INS HA2-O-SLNs (a), INS HA2-O-SLNs in SGF with 0.32% pepsin (b), INS
HA2-O-SLNs in neutralized SGF with 0.32% pepsin (c), and INS HA2-O-SLNs in SIF with 1% trypsin (d). Scale bar: 100 nm.

insulin in the inner aqueous phase, lipid matrix in the oil phase, distribution. Also, the mean particle sizes of the formulated
and Pluronic F68 as the emulsifier in the outer aqueous phase. SLNs were in a range of 150 nm to 170 nm (Table 1, Figure
Finally, the evaporation of the solvent from the W/O/W 1B). Excitingly, the optimal SLNs exhibited high insulin
emulsion allowed the formation of SLNs.40 The combined use entrapment efficiency (over 97%) and drug loading efficiency
of soybean lecithin, tripalmitin, and stearic acid has been (over 7%), indicating that introducing appropriate noncovalent
previously proven to increase the crystallinity of NPs, which interactions (hydrophobic effect, hydrogen bonding, π-effects,
could prevent the leakage of the incorporated insulin (Figure etc.) and preparation methods could efficiently solve the
S1).18 Meanwhile, sodium cholate,41 an amphipathic bio- contradiction between the hydrophobic lipid matrix and
surfactant, was introduced to encapsulate hydrophilic insulin in hydrophilic proteins.42−44
the internal aqueous phase to further improve the entrapment Meanwhile, both INS HA2-W-SLNs and INS HA2-O-SLNs
efficiency and provide the sufficient protection for drugs via the showed high HA2 peptide entrapment efficiency (over 75%),
hydrophobic effect between the amphipathic biosurfactant and demonstrating the capacity of the vehicles encapsulating both
peptides.18,42 hydrophobic and hydrophilic agents. Insulin-loaded SLNs (INS
The formulation and preparation processes were optimized SLNs) were negatively surface charged (−9.2 mV), while
(Table S1). HA2 peptide-containing SLNs were prepared by insulin and HA2 peptide dual-loaded SLNs (INS HA2-O-SLNs
loading HA2 peptide in the internal aqueous phase or oil phase and INS HA2-W-SLNs) showed even more negative surface
(INS HA2-W-SLNs or INS HA2-O-SLNs) (Figure 1A). For charge (−16.1 and −13.1 mV) because of the existence of the
preparation of INS HA2-O-SLNs, a mixed organic solvent was anionic HA2 peptide.45 The different surface charges between
used to increase the solubility of HA2 peptide, achieving the INS HA2-O-SLNs and INS HA2-W-SLNs suggested that the
successful entrapment of HA2 peptide mainly in the outer lipid HA2 peptide was mainly installed in different layers of the
shell. The characterizations of the obtained SLNs were particles. In addition, the confocal images of TRITC-insulin
presented in Table 1. Small PDI index (under 0.3) indicated and FITC-HA2 peptide dual-loaded SLNs also confirmed the
the stability of various SLNs in terms of narrow size results (Figure S2). Moreover, the preparation technique
9318 DOI: 10.1021/acsami.8b00507
ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

Figure 3. Accumulation release profiles of insulin against PBS at (A) pH 5.5 and (B) pH 6.8 (mean ± SD, n = 3). *p < 0.05 vs INS HA2-W-SLNs of
INS HA2 SLNs; #p < 0.05 vs INS HA2-W-SLNs of INS HA2-O-SLNs. Accumulation release profiles of HA2 peptide against PBS at (C) pH 5.5 and
(D) pH 6.8 (mean ± SD, n = 3). ^p < 0.05 vs INS HA2-W-SLNs of INS HA2-O-SLNs.

avoided the high cost of grafting HA2 peptide either with HA2-O-SLNs and INS HA2-W-SLNs showed different trends
drugs46 or nanocarriers.47 The characterizations of the obtained of release for insulin and HA2 peptide. The release profile of
FITC-insulin-loaded SLNs are shown in Table 1, which were insulin from INS HA2-W-SLNs was more sustained compared
similar to those of insulin-loaded SLNs. The TEM images with that of INS HA2-O-SLNs and INS SLNs. After incubation
showed that the morphology of all SLNs was spherical in shape for 12 h, approximately 55 and 35% encapsulated insulin were
(Figure 1C). released from the INS HA2-W-SLNs in the pH 6.8 and 5.5
3.2. Colloidal Stability of SLNs. For orally delivered NPs, buffer, respectively, which were significantly lower compared
it is of importance to overcome the harsh environment of the with that from the INS SLNs and INS HA2-O-SLNs (∼70 and
GIT before NPs intrude the epithelia cells. Thus, the evaluation ∼50%, p < 0.05) (Figure 3A,B). Interestingly, the release at pH
of the colloidal stability of SLNs in simulated GI fluid (SGF and 5.5 was lower than that at pH 6.8 for all of the samples. As
SIF) is essential. The aggregation of SLNs was observed in insulin (isoelectric point = 5.3) in media at pH 5.5 was
SGF, whereas SLNs remained unchanged in neutralized SGF dominantly in nonionic form, the solubility at pH 5.5 was much
(Figures 2A,B and S3A,B). Various SLNs presented no lower than that at pH 6.8. Therefore, the decrease of the release
significant aggregation or disintegration in SIF with or without rate at pH 5.5 might be due to the low solubility of insulin.49,50
trypsin (1%, w/v) (Figures 2C and S3C). Additionally, regular Additionally, a much faster release rate of HA2 peptide
and smooth morphology of particles was observed when SLNs located in the external oil phase (INS HA2-O-SLNs) was also
were dispersed in enzyme-containing neutralized SGF and SIF observed compared with that of HA2 peptide located in the
(Figure 2D), suggesting that the colloidal stability of SLNs internal aqueous phase (INS HA2-W-SLNs) at pH 5.5 (Figure
would not be affected by the main proteases in the GIT. 3C), which was similar to the insulin release because of the
However, SLNs in SGF could be observed by aggregation and interaction of HA2 peptide and insulin in the aqueous phase. A
morphological changes (Figure 2D(b)). In the present study, similar phenomenon was also observed in pH 6.8 (Figure 3D).
before oral administration of SLNs, sodium bicarbonate This result demonstrated that the spatial location for HA2
solution was intragastrically administrated to neutralize the peptide could greatly influence the release rate. Therefore, with
gastric acid following previous reports.24,25 In clinical setting, a much faster release rate of HA2 peptide loaded in the external
enteric-coated gelatin capsules or tablets could also be utilized oil phase, instead of the internal aqueous phase, INS HA2-O-
to prevent the exposure of SLNs in acidic gastric fluids. SLNs were expected to acquire a desirable ability of rapid
3.3. Drug and HA2 Peptide Release of SLNs. The in endosomal escape.
vitro release profiles of insulin and HA2 peptide in pH 2 buffer 3.4. Cell Experiments. 3.4.1. Caco-2 Cell Viability Study.
are shown in Figure S4. Approximately 40% insulin and 65% Caco-2 cells were used as the cell model for the cell
HA2 peptide released from INS HA2-O-SLNs within 2 h. experiments because of the advantage of perfectly mimicking
While in the neutralized medium, both HA2 peptide and the enterocytes of the intestinal gut. The biocompatibility of
insulin showed sustained release from SLNs. Moreover, the SLNs was evaluated by MTT assay. As the viability of both
release rate of HA2 peptide from INS HA2-W-SLNs was insulin- and FITC-labeled insulin encapsulated in SLNs was
significantly lower than that from INS HA2-O-SLNs. ranged from 80 to 120%, all SLNs at test concentrations
Then, we studied insulin and HA2 peptide release in pH 5.5 exhibited no cytotoxicity (Figure S5).
and 6.8 media, which mimicked the pH environments in the 3.4.2. Cellular Uptake. To investigate whether the addition
acidic endosomes48 and small intestine,18 respectively. INS of HA2 peptide could affect the cellular uptake of SLNs, flow
9319 DOI: 10.1021/acsami.8b00507
ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

Figure 4. (A) Flow cytometry quantification of internalization of FITC-labeled INS SLNs, INS HA2-W-SLNs, and INS HA2-O-SLNs (mean ± SD,
n = 3). (B) Endocytic mechanism of SLNs in terms of energy dependence, clathrin-mediated endocytosis, caveolae-mediated endocytosis, and
micropinocytosis. *p < 0.05 vs control group of INS SLNs; &p < 0.05 vs control group of INS HA2-W-SLNs; #p < 0.05 vs control group of INS
HA2-O-SLNs. (C) ELISA quantification of INS SLNs, INS HA2-W-SLNs, and INS HA2-O-SLNs on Caco-2 cells at tested insulin concentration
(mean ± SD, n = 3). **p < 0.01 vs INS HA2-O-SLNs. (D) CLSM images of SLNs (green) and Caco-2 cells stained with early endosomes (red) and
late endosomes (red). (E) CLSM images of SLNs (green) and Caco-2 cells stained with lysosomes (red). Yellow color denotes the overlay of
colocalization of red and green fluorescence. DAPI staining of nuclei (blue).

cytometry was utilized to quantify the amount of fluorescence- 3.4.3. Endosomal Escape of SLNs. SLNs have been
labeled SLNs internalized into cells. Interestingly, as shown in confirmed to involve in clathrin-mediated endocytosis. After
Figure 4A, the detected fluorescent intensity showed no the sequential transport through early and late endosomes,
significant difference among all samples, indicating that the SLNs transported through an intracellular endolysosomal
endocytosis was not influenced by the addition of HA2 peptide. pathway will enable the biotherapeutics delivered to unique
Furthermore, the endocytic mechanism of various SLNs was enzymatic degradative compartments named lysosomes.53 Rab
evaluated. All tested formulations exhibited obvious reduction GTPases play a vital role on vesicular transport in the
of cellular uptake in the presence of sodium azide, endolysosomal pathway.54 Rab5 and Rab7 are expressed in
chlorpromazine, and amiloride (Figure 4B), suggesting that early endosomes and late endosomes, respectively.55,56 As
clathrin-mediated endocytosis was the endocytic mechanism of expected, the colocalization of FITC-labeled INS SLNs, INS
SLNs involved. HA2-W-SLNs, and INS HA2-O-SLNs with early endosome
Meanwhile, the retained active insulin after cellular internal- compartments (Rab5) was observed (Figure 4D), suggesting
ization was determined by ELISA assay. The activity of proteins that all formulations were involved in early endosomes. It was
with fragile structures was easily disrupted under harsh reported that after endocytosis, most NPs would first be
conditions.51,52 During the transport processes, the internalized transported to early endosomes, and then, they could be
insulin might lose biological activity because of the complex trafficked to other organelles.57,58 Interestingly, INS HA2-O-
environment within cells.33 The ELISA method used in our SLNs, showing faster release of HA2 peptide, displayed less
study proved that insulin contained in all samples could retain colocalization with late endosomes and lysosomes. While INS
partly biological activity after cellular internalization. More SLNs and INS HA2-W-SLNs significantly overlaid with the late
excitingly, INS HA2-O-SLNs displayed the greatest insulin endosomes and lysosomes. Thus, the results indicated the
activity (2.64 folds, p < 0.05) compared with INS SLNs and crucial role of HA2 peptide on the endosomal escape of cargoes
INS HA2-W-SLNs, which exhibited similar amounts of active by destabilizing the membrane of endosomes. For INS HA2-W-
insulin in cells (Figure 4C). These results indicated that HA2 SLNs, however, HA2 peptide and insulin would be exposed in
peptide with the appropriate application played a crucial role in late endosomes or lysosomes simultaneously after degradation
protecting the biological activity of insulin though it did not of the lipid materials,19,59 resulting in the entrapment and
elevate the uptake of SLNs. degradation of drugs in lysosomes. In accordance with the in
9320 DOI: 10.1021/acsami.8b00507
ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

Figure 5. (A) Relative hemolysis efficiency of 1% triton X-100, various insulin loaded SLNs and HA2 peptide at pH 7.4, 6.5, and 5.5, and PBS as the
control group. *p < 0.05 vs pH 7.4; #p < 0.05 vs pH 6.5. (B) Transepithelial transport of insulin and insulin-loaded SLNs. *p < 0.05 vs INS; #p < 0.05
vs INS SLNs; &p < 0.05 vs INS HA2-W-SLNs. (C) TEER value of Caco-2 cell monolayers before and after incubation of insulin or SLNs. (D)
Permeated amount of transported insulin in ligated intestinal loop assay. *p < 0.05 vs INS SLNs; #p < 0.05 vs INS HA2-W-SLNs; &p < 0.05 vs INS
HA2-O-SLNs in duodenum at pH 6.8.

vitro cellular uptake assays, INS HA2-O-SLNs demonstrated insulin of INS HA2-W-SLNs merely presented a slight increase
the highest efficiency of endosomal escape by the interactions of transepithelial transport efficiency than that of free insulin
between HA2 and the membrane of late endosomes, protecting and INS SLNs (p < 0.05). Thus, INS HA2-O-SLNs were
insulin from the transport to lysosomes. confirmed to promote the transepithelial transport, which
Meanwhile, the mechanism of endosomal escape of SLNs might be due to the protection of insulin during the
was further investigated using hemolytic assay.34,35 It was intracellular trafficking. Meanwhile, the TEER value remained
demonstrated that the relative hemolytic activity of INS HA2- unchanged after incubation of various SLNs (Figure 5C),
O-SLNs increased with the acidification of maturing endo- indicating that the monolayer remained intact during the
somes (Figure 5A). The hemolysis of INS HA2-O-SLNs is transcellular transport of insulin.
15.53 and 52.61% at pH 6.5 and 5.5, respectively, suggesting 3.5. Ex Vivo Ligated Intestinal Loop Assay. The oral
the excellent ability of INS HA2-O-SLNs to interact with late intestinal absorption of INS HA2-O-SLNs was further
endosomes; while, INS HA2-O-SLNs exhibited low hemolytic investigated by the ex vivo ligated intestinal loop assay (Figure
activity (2.85%) at pH 7.4, indicating the low toxicity on cells. 5D).37 Consistent with the in vitro behavior of SLNs, INS
In contrast, INS HA2-W-SLNs presented relative low pH- HA2-O-SLNs showed 1.48 fold higher intestinal transport than
sensitive amphiphilic endosomal escape. The hemolytic activity INS SLNs and INS HA2-W-SLNs in jejunum. Interestingly, the
of INS SLNs did not increase with the acidification of absorption of SLNs in jejunum was much higher than that in
endosomes. This result was consistent with the intracellular duodenum and ileum, which might be due to the more dense
trafficking of various SLNs observed under CLSM, and INS jejunum villi distribution in the small intestine.60 Moreover,
HA2-O-SLNs were confirmed to display the pH-sensitive INS HA2-O-SLNs dispersed in pH 5.5 enhanced insulin
amphiphilic endosomal escape. transport through the duodenum, while no significant increase
3.4.4. Transepithelial Transport Study. INS HA2-O-SLNs was observed in INS SLNs or INS HA2-W-SLNs in the same
have been confirmed to protect insulin from lysosomal condition. The result suggested that INS HA2-O-SLNs could
degradation by successful endosomal escape. The permeation potentially enhance the oral absorption of insulin in duodenum.
of insulin through Caco-2 cell monolayers was further However, SLNs might not have enough retention time in
investigated. As shown in Figure 5B, after loaded into SLNs, duodenum on animal models. The enhanced duodenal
insulin showed the enhanced transepithelial transport efficiency absorption of INS HA2-O-SLNs may be weakened by intestinal
(p < 0.05). Of note, insulin of INS HA2-O-SLNs exhibited the flow, rapid dilution, as well as spreading.61
highest accumulated amount in the basolateral side, which was 3.6. Pharmacological and Pharmacokinetics Studies.
respectively 4.30-fold (p < 0.05) and 2.19-fold (p < 0.05) The in vivo pharmacological and pharmacokinetics effect of
higher than that of free insulin and INS SLNs. By contrast, various SLNs were determined on diabetic rats. As shown in
9321 DOI: 10.1021/acsami.8b00507
ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

Figure 6. (A) Blood glucose level (% of initial) and (B) serum insulin level of rats following administration of various formulations (mean ± SD, n =
5). *p < 0.05 vs insulin solution. #p < 0.05 vs INS SLNs. &p < 0.05 vs INS HA2-W-SLNs.

Figure 6A, subcutaneously injected insulin (5 IU/kg) lipid shell containing endosomal escape agent (HA2 peptides)
represented remarkable hypoglycemia effect and the blood and an aqueous core containing insulin. HA2 peptide in the
glucose level decreased to the minimum at 2 h, whereas oral external oil phase (INS HA2-O-SLNs) was demonstrated to
administration of insulin (50 IU/kg) failed to reduce the blood possess a much faster release compared with that of HA2 in the
glucose level. Oral administration of INS SLNs, INS HA2-W- external aqueous phase (INS HA2-W-SLNs). When exposed to
SLNs, and INS HA2-O-SLNs generated significant hypoglyce- acidic endosomes, the released HA2 peptide helped cargoes
mic response (p < 0.05), indicating that bioactivity of insulin escape from degradation in the lysosomal pathway by rapidly
encapsulated in SLNs was protected against various pH disrupting the late endosomal membranes. In vitro cell
environments and enzymatic digestion. Excitingly, INS HA2- experiments confirmed that INS HA2-O-SLNs could retain
O-SLNs presented the most excellent hypoglycemic effect with the highest biological activity of internalized insulin. As
37% blood glucose decrease at 3 h. The pharmacological demonstrated in the intracellular trafficking of SLNs, INS
availability (PA %) of different samples related to subcutaneous HA2-O-SLNs displayed much less distribution in late endo-
injection is shown in Table 2. INS HA2-O-SLNs demonstrated somes and lysosomes. Meanwhile, INS HA2-O-SLNs exhibited
PA % of 7.32%, which was 4 and 1.7 folds higher than free the highest transepithelial transport efficiency, with 2.19 folds
insulin solution and INS HA2-W-SLNs. (p < 0.05) and 1.72 folds (p < 0.05) higher intestinal transport
as compared to INS SLNs and INS HA2-W-SLNs.
Table 2. Pharmacological and Pharmacokinetic Parameters Furthermore, the ex vivo ligated intestinal loop assay
of Insulin Formulations in a Diabetic Rat Modela demonstrated that insulin from INS HA2-O-SLNs exhibited
the highest permeation in various regions of intestines. Finally,
samples dose (IU/kg) AUC (mIU·h/L) PA (%) F (%) the developed SLNs were confirmed to increase the serum
SC insulin 5 278.1 ± 69.2 100 insulin concentration and generate an excellent hypoglycemic
oral insulin 50 47.3 ± 6.1 1.83 1.70 response, suggesting the importance of endosomal escape for
INS SLN 50 96.5 ± 10.5 3.40 3.47 orally delivered proteins and peptides. The excellent in vitro−in
INS HA2-W-SLN 50 96.6 ± 23.2 4.33 3.47 vivo correlation confirmed the great potency for SLNs with
INS HA2-O-SLN 50 152.1 ± 12.0 7.32 5.47 endosomal escape function, and the strategy could be further
a
AUC: area under the plasma concentration−time curve; PA %: applied in the oral delivery of other biomacromolecules.


pharmacological bioavailability; F %: relative bioavailability.
ASSOCIATED CONTENT
The serum insulin concentration is shown in Figure 6B. *
S Supporting Information
Compared with subcutaneously administrated free insulin, SLN The Supporting Information is available free of charge on the
administration by gavage showed a relatively slower increase in ACS Publications website at DOI: 10.1021/acsami.8b00507.
the serum insulin concentration. At the dose of 50 IU/kg, INS The chemical structure of glycerol tripalmitate and
HA2-O-SLNs showed a significant higher relative bioavailability stearic acid; Physicochemical characterization of SLNs;
of 5.47%, which was 3.2-fold higher than free insulin.
CLSM images of HA2-O-SLNs and HA2-W-SLNs;
Therefore, the in vivo−in vitro correlation was confirmed.
accumulation release profiles of insulin and HA2 in pH
Taken previous results together, HA2 peptide in INS HA2-O-
SLNs could be appropriately released and destabilize the 2; and Caco-2 cell viability treated with various SLNs by
membrane of endosomes to achieve the endosomal escape of MTT assays (PDF)
insulin. The designed delivery system showed the great potency
as a promising strategy to overcome degradation of insulin in
lysosomes and further enhanced the oral bioavailability of
■ AUTHOR INFORMATION
Corresponding Author
insulin.
*E-mail: huangyuan0@163.com. Phone/Fax: +86-28-
4. CONCLUSIONS 85501617.
In summary, we rationally developed the functional nano- ORCID
vehicles for the efficient oral absorption of biomacromolecules. Zhirong Zhang: 0000-0001-9118-5394
The optimized SLNs (INS HA2-O-SLNs) were comprised of a Yuan Huang: 0000-0003-3410-8602
9322 DOI: 10.1021/acsami.8b00507
ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

Author Contributions (16) Malhaire, H.; Gimel, J.-C.; Roger, E.; Benoît, J.-P.; Lagarce, F.

Y.X. and Y.Z. contributed equally to this study and shared first How to Design the Surface of Peptide-Loaded Nanoparticles for
authorship. Efficient Oral Bioavailability? Adv. Drug Delivery Rev. 2016, 106, 320−
336.
Notes (17) Lear, J. D.; DeGrado, W. F. Membrane Binding and
The authors declare no competing financial interest. Conformational Properties of Peptides Representing the Nh2


Terminus of Influenza Ha-2. J. Biol. Chem. 1987, 262, 6500−6505.
(18) Chen, C.; Fan, T.; Jin, Y.; Zhou, Z.; Yang, Y.; Zhu, X.; Zhang,
ACKNOWLEDGMENTS
Z.-R.; Zhang, Q.; Huang, Y. Orally Delivered Salmon Calcitonin-
The authors gratefully acknowledge financial support from the Loaded Solid Lipid Nanoparticles Prepared by Micelle-Double
National Science Foundation for Distinguished Young Scholars Emulsion Method Via the Combined Use of Different Solid Lipids.
(81625023) and the Major Research Plan of National Natural Nanomedicine 2013, 8, 1085−1100.
Science Foundation of China (81690261). (19) Chen, C.; Zhu, X.; Dou, Y.; Xu, J.; Zhang, J.; Fan, T.; Du, J.; Liu,


K.; Deng, Y.; Zhao, L.; Huang, Y. Exendin-4 Loaded Nanoparticles
with a Lipid Shell and Aqueous Core Containing Micelles for
REFERENCES Enhanced Intestinal Absorption. J. Biomed. Nanotechnol. 2015, 11,
(1) Antosova, Z.; Mackova, M.; Kral, V.; Macek, T. Therapeutic 865−876.
Application of Peptides and Proteins: Parenteral Forever? Trends (20) Hentz, N. G.; Richardson, J. M.; Sportsman, J. R.; Daijo, J.;
Biotechnol. 2009, 27, 628−635. Sittampalam, G. S. Synthesis and Characterization of Insulin-
(2) Malhaire, H.; Gimel, J.-C.; Roger, E.; Benoît, J.-P.; Lagarce, F. Fluorescein Derivatives for Bioanalytical Applications. Anal. Chem.
How to Design the Surface of Peptide-Loaded Nanoparticles for 1997, 69, 4994−5000.
Efficient Oral Bioavailability? Adv. Drug Delivery Rev. 2016, 106, 320− (21) Zhang, P.; Xu, Y.; Zhu, X.; Huang, Y. Goblet Cell Targeting
336. Nanoparticle Containing Drug-Loaded Micelle Cores for Oral
(3) Lakkireddy, H. R.; Urmann, M.; Besenius, M.; Werner, U.; Delivery of Insulin. Int. J. Pharm. 2015, 496, 993−1005.
Haack, T.; Brun, P.; Alié, J.; Illel, B.; Hortala, L.; Vogel, R.; Bazile, D. (22) Cui, F.; Qian, F.; Yin, C. Preparation and Characterization of
Oral Delivery of Diabetes Peptides - Comparing Standard For- Mucoadhesive Polymer-Coated Nanoparticles. Int. J. Pharm. 2006,
mulations Incorporating Functional Excipients and Nanotechnologies 316, 154−161.
in the Translational Context. Adv. Drug Delivery Rev. 2016, 106, 196− (23) Cui, M.; Wu, W.; Hovgaard, L.; Lu, Y.; Chen, D.; Qi, J.
222. Liposomes Containing Cholesterol Analogues of Botanical Origin as
(4) Bakhru, S. H.; Furtado, S.; Morello, A. P.; Mathiowitz, E. Oral Drug Delivery Systems to Enhance the Oral Absorption of Insulin. Int.
Delivery of Proteins by Biodegradable Nanoparticles. Adv. Drug J. Pharm. 2015, 489, 277−284.
Delivery Rev. 2013, 65, 811−821. (24) Yoo, H. S.; Park, T. G. Biodegradable Nanoparticles Containing
(5) Aguirre, T. A. S.; Teijeiro-Osorio, D.; Rosa, M.; Coulter, I. S.; Protein-Fatty Acid Complexes for Oral Delivery of Salmon Calcitonin.
Alonso, M. J.; Brayden, D. J. Current Status of Selected Oral Peptide J. Pharm. Sci. 2004, 93, 488−495.
Technologies in Advanced Preclinical Development and in Clinical (25) Li, X.; Wang, C.; Liang, R.; Sun, F.; Shi, Y.; Wang, A.; Liu, W.;
Trials. Adv. Drug Delivery Rev. 2016, 106, 223−241. Sun, K.; Li, Y. The Glucose-Lowering Potential of Exenatide Delivered
(6) Morishita, M.; Peppas, N. A. Is the Oral Route Possible for Orally Via Goblet Cell-Targeting Nanoparticles. Pharm. Res. 2015, 32,
Peptide and Protein Drug Delivery? Drug Discovery Today 2006, 11, 1017−1027.
905−910. (26) Wang, X.-Q.; Zhang, Q. Ph-Sensitive Polymeric Nanoparticles
(7) Jin, Y.; Song, Y.; Zhu, X.; Zhou, D.; Chen, C.; Zhang, Z.; Huang, to Improve Oral Bioavailability of Peptide/Protein Drugs and Poorly
Y. Goblet Cell-Targeting Nanoparticles for Oral Insulin Delivery and Water-Soluble Drugs. Eur. J. Pharm. Biopharm. 2012, 82, 219−229.
the Influence of Mucus on Insulin Transport. Biomaterials 2012, 33, (27) Zhu, X.; Wu, J.; Shan, W.; Tao, W.; Zhao, L.; Lim, J.-M.;
1573−1582. D’Ortenzio, M.; Karnik, R.; Huang, Y.; Shi, J.; Farokhzad, O. C.
(8) Pan, Y.; Li, Y.-J.; Zhao, H.-Y.; Zheng, J.-M.; Xu, H.; Wei, G.; Hao,
Polymeric Nanoparticles Amenable to Simultaneous Installation of
J.-S.; Cui, F.-D. Bioadhesive Polysaccharide in Protein Delivery
Exterior Targeting and Interior Therapeutic Proteins. Angew. Chem.,
System: Chitosan Nanoparticles Improve the Intestinal Absorption
Int. Ed. 2016, 55, 3309−3312.
of Insulin in Vivo. Int. J. Pharm. 2002, 249, 139−147.
(28) Liu, C.; Shan, W.; Liu, M.; Zhu, X.; Xu, J.; Xu, Y.; Huang, Y. A
(9) Yun, Y.; Cho, Y. W.; Park, K. Nanoparticles for Oral Delivery:
Targeted Nanoparticles with Peptidic Ligands for Oral Protein Novel Ligand Conjugated Nanoparticles for Oral Insulin Delivery.
Delivery. Adv. Drug Delivery Rev. 2013, 65, 822−832. Drug Delivery 2016, 23, 2015−2025.
(10) Xu, Y.; Xu, J.; Shan, W.; Liu, M.; Cui, Y.; Li, L.; Liu, C.; Huang, (29) Sadeghi, A. M. M.; Dorkoosh, F. A.; Avadi, M. R.; Weinhold,
Y. The Transport Mechanism of Integrin Alphavbeta3 Receptor M.; Bayat, A.; Delie, F.; Gurny, R.; Larijani, B.; Rafiee-Tehrani, M.;
Targeting Nanoparticles in Caco-2 Cells. Int. J. Pharm. 2016, 500, 42− Junginger, H. E. Permeation Enhancer Effect of Chitosan and
53. Chitosan Derivatives: Comparison of Formulations as Soluble
(11) Bareford, L. M.; Swaan, P. W. Endocytic Mechanisms for Polymers and Nanoparticulate Systems on Insulin Absorption in
Targeted Drug Delivery. Adv. Drug Delivery Rev. 2007, 59, 748−758. Caco-2 Cells. Eur. J. Pharm. Biopharm. 2008, 70, 270−278.
(12) Luzio, J. P.; Gray, S. R.; Bright, N. A. Endosome-Lysosome (30) Perumal, O. P.; Inapagolla, R.; Kannan, S.; Kannan, R. M. The
Fusion. Biochem. Soc. Trans. 2010, 38, 1413−1416. Effect of Surface Functionality on Cellular Trafficking of Dendrimers.
(13) Huang, H. W.; Chen, F.-Y.; Lee, M.-T. Molecular Mechanism of Biomaterials 2008, 29, 3469−3476.
Peptide-Induced Pores in Membranes. Phys. Rev. Lett. 2004, 92, (31) Chen, C.; Zhu, X.; Dou, Y.; Xu, J.; Zhang, J.; Fan, T.; Du, J.; Liu,
198304. K.; Deng, Y.; Zhao, L. Exendin-4 Loaded Nanoparticles with a Lipid
(14) Lin, C.; Engbersen, J. F. J. Effect of Chemical Functionalities in Shell and Aqueous Core Containing Micelles for Enhanced Intestinal
Poly(Amido Amine)S for Non-Viral Gene Transfection. J. Controlled Absorption. J. Biomed. Nanotechnol. 2015, 11, 865−876.
Release 2008, 132, 267−272. (32) Shrestha, N.; Araújo, F.; Shahbazi, M.-A.; Mäkilä, E.; Gomes, M.
(15) Berg, K.; Selbo, P. K.; Prasmickaite, L.; Tjelle, T. E.; Sandvig, K.; J.; Herranz-Blanco, B.; Lindgren, R.; Granroth, S.; Kukk, E.; Salonen,
Moan, J.; Gaudernack, G.; Fodstad, O.; Kjolsrud, S.; Anholt, H.; Rodal, J.; Hirvonen, J.; Sarmento, B.; Santos, H. A. Thiolation and Cell-
G. H.; Rodal, S. K.; Hogset, A. Photochemical Internalization: A Novel Penetrating Peptide Surface Functionalization of Porous Silicon
Technology for Delivery of Macromolecules into Cytosol. Cancer Res. Nanoparticles for Oral Delivery of Insulin. Adv. Funct. Mater. 2016,
1999, 59, 1180−1183. 26, 3405−3416.

9323 DOI: 10.1021/acsami.8b00507


ACS Appl. Mater. Interfaces 2018, 10, 9315−9324
ACS Applied Materials & Interfaces Research Article

(33) Sajeesh, S.; Sharma, C. P. Cyclodextrin-Insulin Complex (52) Muheem, A.; Shakeel, F.; Jahangir, M. A.; Anwar, M.; Mallick,
Encapsulated Polymethacrylic Acid Based Nanoparticles for Oral N.; Jain, G. K.; Warsi, M. H.; Ahmad, F. J. A Review on the Strategies
Insulin Delivery. Int. J. Pharm. 2006, 325, 147−154. for Oral Delivery of Proteins and Peptides and Their Clinical
(34) Gujrati, M.; Vaidya, A.; Lu, Z.-R. Multifunctional Ph-Sensitive Perspectives. Saudi Pharm. J. 2016, 24, 413−428.
Amino Lipids for Sirna Delivery. Bioconjugate Chem. 2015, 27, 19−35. (53) Luzio, J. P.; Pryor, P. R.; Bright, N. A. Lysosomes: Fusion and
(35) Gujrati, M.; Malamas, A.; Shin, T.; Jin, E.; Sun, Y.; Lu, Z.-R. Function. Nat. Rev. Mol. Cell Biol. 2007, 8, 622−632.
Multifunctional Cationic Lipid-Based Nanoparticles Facilitate Endo- (54) Grosshans, B. L.; Ortiz, D.; Novick, P. Rabs and Their Effectors:
somal Escape and Reduction-Triggered Cytosolic Sirna Release. Mol. Achieving Specificity in Membrane Traffic. Proc. Natl. Acad. Sci. U.S.A.
Pharmaceutics 2014, 11, 2734−2744. 2006, 103, 11821−11827.
(36) Gamboa, J. M.; Leong, K. W. In Vitro and in Vivo Models for (55) Bucci, C.; Parton, R. G.; Mather, I. H.; Stunnenberg, H.;
the Study of Oral Delivery of Nanoparticles. Adv. Drug Delivery Rev. Simons, K.; Hoflack, B.; Zerial, M. The Small Gtpase Rab5 Functions
2013, 65, 800−810. as a Regulatory Factor in the Early Endocytic Pathway. Cell 1992, 70,
(37) Primard, C.; Rochereau, N.; Luciani, E.; Genin, C.; Delair, T.; 715−728.
Paul, S.; Verrier, B. Traffic of Poly (Lactic Acid) Nanoparticulate (56) Poteryaev, D.; Datta, S.; Ackema, K.; Zerial, M.; Spang, A.
Vaccine Vehicle from Intestinal Mucus to Sub-Epithelial Immune Identification of the Switch in Early-to-Late Endosome Transition. Cell
Competent Cells. Biomaterials 2010, 31, 6060−6068. 2010, 141, 497−508.
(38) Sarmento, B.; Martins, S.; Ferreira, D.; Souto, E. B. Oral Insulin (57) Aniento, F.; Emans, N.; Griffiths, G.; Gruenberg, J. Cytoplasmic
Delivery by Means of Solid Lipid Nanoparticles. Int. J. Nanomed. 2007, Dynein-Dependent Vesicular Transport from Early to Late Endo-
2, 743−749. somes. J. Cell Biol. 1993, 123, 1373−1387.
(39) Aditya, N. P.; Aditya, S.; Yang, H.; Kim, H. W.; Park, S. O.; Ko, (58) Luzio, J. P.; Mullock, B. M.; Pryor, P. R.; Lindsay, M. R.; James,
S. Co-Delivery of Hydrophobic Curcumin and Hydrophilic Catechin D. E.; Piper, R. C. Relationship between Endosomes and Lysosomes.
by a Water-in-Oil-in-Water Double Emulsion. Food Chem. 2015, 173, Biochem. Soc. Trans. 2001, 29, 476−480.
7−13. (59) Mantle, M.; Allen, A. A Colorimetric Assay for Glycoproteins
(40) Gallarate, M.; Trotta, M.; Battaglia, L.; Chirio, D. Preparation of Based on the Periodic Acid/Schiff Stain [Proceedings]. Biochem. Soc.
Solid Lipid Nanoparticles from W/O/W Emulsions: Preliminary Trans. 1978, 6, 607−609.
Studies on Insulin Encapsulation. J. Microencapsulation 2009, 26, 394− (60) Yusrizal; Chen, T. C. Effect of Adding Chicory Fructans in Feed
402. on Broiler Growth Performance, Serum Cholesterol and Intestinal
(41) Ninomiya, R.; Matsuoka, K.; Moroi, Y. Micelle Formation of Length. Int. J. Poult. Sci. 2003, 2, 214.
Sodium Chenodeoxycholate and Solubilization into the Micelles: (61) Watts, P. J.; Barrow, L.; Steed, K. P.; Wilson, C. G.; Spiller, R.
Comparison with Other Unconjugated Bile Salts. Biochim. Biophys. C.; Melia, C. D.; Davies, M. C. The Transit Rate of Different-Sized
Acta, Mol. Cell Biol. Lipids 2003, 1634, 116−125. Model Dosage Forms through the Human Colon and the Effects of a
(42) Abbas, M.; Zou, Q.; Li, S.; Yan, X. Self-Assembled Peptide- and Lactulose-Induced Catharsis. Int. J. Pharm. 1992, 87, 215−221.
Protein-Based Nanomaterials for Antitumor Photodynamic and
Photothermal Therapy. Adv. Mater. 2017, 29, 1605021.
(43) Zou, Q.; Abbas, M.; Zhao, L.; Li, S.; Shen, G.; Yan, X. Biological
Photothermal Nanodots Based on Self-Assembly of Peptide−
Porphyrin Conjugates for Antitumor Therapy. J. Am. Chem. Soc.
2017, 139, 1921−1927.
(44) Zhou, L.; Lv, F.; Liu, L.; Shen, G.; Yan, X.; Bazan, G. C.; Wang,
S. Cross-Linking of Thiolated Paclitaxel−Oligo(P-Phenylene Vinyl-
ene) Conjugates Aggregates inside Tumor Cells Leads to “Chemical
Locks” That Increase Drug Efficacy. Adv. Mater. 2018, 30, 1704888.
(45) Niikura, K.; Horisawa, K.; Doi, N. A Fusogenic Peptide from a
Sea Urchin Fertilization Protein Promotes Intracellular Delivery of
Biomacromolecules by Facilitating Endosomal Escape. J. Controlled
Release 2015, 212, 85−93.
(46) Wadia, J. S.; Stan, R. V.; Dowdy, S. F. Transducible Tat-Ha
Fusogenic Peptide Enhances Escape of Tat-Fusion Proteins after Lipid
Raft Macropinocytosis. Nat. Med. 2004, 10, 310−315.
(47) Zhou, Z.; Liu, Y.; Wu, L.; Li, L.; Huang, Y. Enhanced Nuclear
Delivery of Anti-Cancer Drugs Using Micelles Containing Releasable
Membrane Fusion Peptide and Nuclear-Targeting Retinoic Acid. J.
Mater. Chem. B 2017, 5, 7175−7185.
(48) Gruenberg, J.; van der Goot, F. G. Mechanisms of Pathogen
Entry through the Endosomal Compartments. Nat. Rev. Mol. Cell Biol.
2006, 7, 495−504.
(49) Pan, Y.; Li, Y.-j.; Zhao, H.-y.; Zheng, J.-m.; Xu, H.; Wei, G.; Hao,
J.-s. Bioadhesive Polysaccharide in Protein Delivery System: Chitosan
Nanoparticles Improve the Intestinal Absorption of Insulin in Vivo.
Int. J. Pharm. 2002, 249, 139−147.
(50) Tokumitsu, H.; Ichikawa, H.; Fukumori, Y. Chitosan-
Gadopentetic Acid Complex Nanoparticles for Gadolinium Neutron-
Capture Therapy of Cancer: Preparation by Novel Emulsion-Droplet
Coalescence Technique and Characterization. Pharm. Res. 1999, 16,
1830−1835.
(51) Erlkamp, M.; Marion, J.; Martinez, N.; Czeslik, C.; Peters, J.;
Winter, R. Influence of Pressure and Crowding on the Sub-
Nanosecond Dynamics of Globular Proteins. J. Phys. Chem. B 2015,
119, 4842−4848.

9324 DOI: 10.1021/acsami.8b00507


ACS Appl. Mater. Interfaces 2018, 10, 9315−9324

S-ar putea să vă placă și