Subscriber access provided by UNIV OF NEWCASTLE
Article
Biofunctional polymer-lipid hybrid high density lipoproteinmimicking nanoparticles loading anti-miR155 for combined antiatherogenic effects on macrophages Jing Lu, Yi Zhao, Xiaoju Zhou, Jian Hua He, Yun Yang, Cuiping Jiang, Zitong Qi, Wenli Zhang, and Jianping Liu Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.7b00436 • Publication Date (Web): 24 Jul 2017 Downloaded from http://pubs.acs.org on July 25, 2017
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
Biomacromolecules is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
4
Biofunctional polymer-lipid hybrid high density lipoprotein-mimicking nanoparticles loading anti-miR155 for combined antiatherogenic effects on macrophages
5
Jing Lu, Yi Zhao, Xiaoju Zhou, Jian Hua He, Yun Yang, Cuiping Jiang, Zitong Qi,
6
Wenli Zhang and Jianping Liu*
7
Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China
8
KEYWORDS
9
Biofunction, polymer-lipid hybrid HNP, acid-labile PEI, anti-miR155, combination
1 2 3
10
therapy, atherosclerosis.
11
ABSTRACT
12
A biofunctional polymer-lipid hybrid high density lipoprotein-mimicking
13
nanoparticle
(HNP)
loading
anti-miR155
was
constructed
14
antiatherogenic effects on macrophages. The HNP consisted of an anti-miR155 core
15
condensed by acid-labile polyethylenimine (acid-labile PEI) polymers and a lipid
16
bilayer coat that was decorated with apolipoprotein A-1, termed acid-labile PEI/HNP.
17
The acid-labile PEI was synthesized with low molecular weight PEI and
18
glutaraldehyde to reduce the cytotoxicity and facilitate nucleic acids escaping from
19
acidic endolysosomes. The increased silencing efficiency of acid-labile PEI/HNP was
20
ascribed to the clathrin-mediated endocytosis and successful endolysosomal escape.
21
Decreased intracellular reactive oxygen species production and DiI-oxLDL uptake
1
ACS Paragon Plus Environment
for
combined
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
22
revealed the antioxidant activities of both anti-miR155 and HNP. Cholesterol efflux
23
assay indicated the potential of HNP in reverse cholesterol transport. Collectively, the
24
acid-labile PEI/HNP not only realized the efficacy of anti-miR155 in macrophages,
25
but also exerted the anti-atherosclerotic biofunction of HNP.
26
INTRODUCTION
27
Atherosclerosis is a chronic inflammatory lesion with high morbidity and
28
mortality worldwide, which is caused by excess lipids deposition in the arterial blood
29
vessels.1 Macrophages situated within the intima play a pivotal role in atherogenic
30
programming, which internalize modified lipoproteins resulting in foam cells
31
formation and further promote the retention of lipoproteins. Excess lipids
32
accumulation and defective phagocytic clearance contribute to the formation of a
33
necrotic core, triggering myocardial infarction or stroke.2,3 The macrophage is
34
therefore pursued as a therapeutic target to prevent atherosclerosis events.
35
MicroRNA (miRNA)-based therapy has been increasingly recognized as a
36
promising therapeutic approach for atherosclerosis.4,5 MicroRNA-155 (miR155), a
37
class of endogenous noncoding RNA with about 23 nucleotides, has been reported to
38
be expressed specifically in macrophages of atherosclerotic plaques.6 As a multi-target
39
molecule, miR155 induces atherosclerosis by base paring with different target
40
mRNAs, causing series of impaired inflammatory responses and lipid metabolisms.
41
Tian et al. proved that miR155 promoted lipid uptake and reactive oxygen species
42
(ROS) production of macrophages to induce foam cells formation, and miR155
43
deficiency in apolipoprotein E-deficient (apo E−/−) mice mitigated atherogenesis via
2
ACS Paragon Plus Environment
Page 2 of 54
Page 3 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
44
reducing inflammatory responses of macrophages and enhancing macrophage
45
cholesterol efflux.6-8 Therefore, anti-miR155 with complementary sequences against
46
miR155 function in macrophages may be a promising strategy to halt atherogenesis.
47
However, free anti-miRNA is unstable in the plasma and may suffer nucleases
48
degradation and renal clearance.9 It is urgent to find an effective carrier for the direct
49
anti-miR155 delivery into macrophages.
50
Endogenous high density lipoproteins (HDLs) are dynamic natural nanoparticles,
51
existing mainly in two distinct structures, discoidal HDL and spherical HDL.10 The
52
discoidal HDL is comprised of a phospholipid bilayer circumscribed with two
53
apolipoprotein A-1 (apo A1) molecules, and the spherical HDL has a hydrophobic
54
lipids core surrounded by a phospholipid monolayer with apo A1 embedded. Apo A1,
55
the most important protein constituent of HDL, is proved to be with high affinity to
56
scavenger receptor type B-1 (SR-B1) expressed abundantly in macrophages and
57
cancer cells.10 Based on the long circulation half-life, excellent biocompatibility and
58
SR-B1-specific targeting, considerable interests have been focused on utilizing
59
recombinant HDL nanoparticles (rHDL) for the delivery of nucleic acids. Biomimetic
60
rHDL nanocarriers with cholesterylated nucleic acids adsorbed onto the surface for
61
the direct cytosolic delivery of cargos achieved improved gene silencing.11-14
62
McMahon constructed HDL inspired-hybrid vehicles through assembling templated
63
lipoprotein particles with single-stranded RNA complements of short-interfering RNA
64
(siRNA) after formulation with a cationic lipid whereby RNA was also localized to
65
the particle surface.15 Shahzad et al. incorporated siRNA into rHDL, facilitating
3
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
66
highly efficient systemic delivery mediated by the over-expressed SR-B1 in cancer
67
cells.16 These contents make the rHDL nanocarriers appealing for anti-miR155
68
delivery.
69
In addition to the superiority as nanocarriers, rHDL has been explored as a
70
potential anti-atherosclerosis therapeutics.17,18 Intravenous infusion of rHDL has been
71
shown to have an anti-atherosclerotic effect in animals19-21 and human models22,23.
72
The benefits of rHDL therapy appear to result from the reverse cholesterol transport
73
(RCT) pathway in much the same way as endogenous HDL.24,25 HDL removes
74
cholesterol from the lipid-loaded foam cells via efflux through their surface receptors,
75
ATP-binding cassette subfamily members A1 (ABCA1), G1(ABCG1) and SR-B1, and
76
then the cholesterol-loaded HDL is transported to the liver for elimination.10
77
Moreover, HDL may have benefits beyond RCT in its atheroprotective role, including
78
the antioxidant functions26,27, anti-inflammatory effects28,29, and endothelial cells
79
protective actions30,31, which have also been manifested in the progress of rHDL
80
regressing atherosclerotic plaques.19 We attempted to combine the unique
81
anti-atherosclerotic biological function of rHDL with the anti-miR155 efficacy for
82
effective atheroprotection. Notably, rHDL has been tested as a biofunctional
83
therapeutic agent for other diseases. For instance, Yang32 prepared gold core HDL
84
nanoparticles as therapeutic agents for lymphoma by binding to SR-B1 and interfering
85
with cellular cholesterol flux and Numata33 employed nanodiscs as therapeutic
86
delivery agents to inhibit respiratory syncytial virus infection in the lung. The intrinsic
87
therapeutic effect of HDL nanostructures was defined as “theralivery” by Mutharasan
4
ACS Paragon Plus Environment
Page 4 of 54
Page 5 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
88
recently.34
89
In this study, we constructed a HDL‐mimicking nanoparticle (HNP) containing a
90
condensed anti-miR155 core and a lipid bilayer envelope structure modified with apo
91
A1 (Scheme 1), providing greater protection for nucleic acids from the degradation
92
and loss in systemic circulation. Due to lacking of the ability of lipids core to
93
condense nucleic acids, a templated anti-miR155 nanoparticle was used as an
94
alternative core inside the HNP. Furthermore, the alternative core would not interfere
95
with the biological functions of HNP which has been demonstrated that HDL
96
biomimics with a PLGA core or a gold nanoparticle core were still capable of
97
mediating cholesterol efflux.35,36 The stable core-shell structure allowed a sufficient
98
lipidic bilayer space for cholesterol storage.35 Branched polyethylenimine (PEI, 25
99
kDa), a cationic polymer, is often employed to condense nucleic acids with high
100
transfection efficiency based on the ‘proton sponge’ hypothesis.37 However, the high
101
cytotoxicity of PEI limits the application owing to the interaction of free PEI with
102
cellular components which disturbs the normal cellular process after the
103
internalization and release of nucleic acids.38 To reduce the cytotoxicity after entering
104
the cell, a high molecular weight PEI with acid-labile linkers was synthesized
105
between low molecular weight PEI (1.8 kDa) and glutaraldehyde. Then the acid-labile
106
PEI was bound to anti-miR155 and inserted into the HNP, termed acid-labile PEI/HNP.
107
The polymer of acid-labile PEI was characterized by the
108
fourier-transform infrared spectrometer (FT-IR), gel permeation chromatography
109
(GPC) and the buffering capacity. The physicochemical properties of acid-labile
5
ACS Paragon Plus Environment
13
C NMR spectroscopy,
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 6 of 54
110
PEI/HNP, such as the content determination, particle size, zeta potential, morphology
111
and stability test were evaluated. Furthermore, in vitro cell viability, cellular uptake,
112
internalization mechanisms, in vitro gene silencing were assessed in detail. The
113
anti-atherosclerotic efficacies involving the ROS production, DiI-oxidized low
114
density lipoproteins (DiI-oxLDL) uptake, and cholesterol efflux were also validated.
115
Scheme 1. Schematics of the acid-labile PEI/HNP.
116 117
2. MATERIALS AND EXPERIMENTAL SECTION
118
2.1
119
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenylterazolium bromide (MTT) and block lipid
120
transport 1 (BLT-1) were purchased from Sigma-Aldrich (USA). Branched PEI (1.8
121
kDa), heparin sodium, chlorpromazine hydrochloride, genistein, and amiloride were
122
purchased from Aladdin (Shanghai, China). Glutaraldehyde was bought from
123
Sinopharm Chemical Reagent Co., Ltd (Shanghai, China). Egg phospholipid (Lipoid
124
S-100) was purchased from Lipoid GmbH (Germany). Quant-iT™ RiboGreen® RNA
125
Reagent was purchased from Thermo Fisher Scientific (USA). Cy5.5 mono
Materials.
Branched
PEI
(25
kDa),
cholesterol,
6
ACS Paragon Plus Environment
sodium
cholate,
Page 7 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
126
N-hydroxysuccinamide ester (Cy5.5 NHS ester) was from APExBIO (Houston, TX,
127
USA). DiI-oxLDL was obtained from Yiyuan Biotech. Co., Ltd (Guangzhou, China).
128
RNase A, GelRed, Triton X-100, Hoechst 33342 and ROS Assay Kit were bought
129
from Keygen Biotech Co., Ltd (Jiangsu, China). Lyso-Tracker Red and Radio
130
immunoprecipitation assay (RIPA lysis buffer) were purchased from Beyotime
131
Institute of Biotechnology (Shanghai, China). Raw 264.7 cells were kindly gifted
132
from Atherosclerosis Research Centre, Nanjing Medical University (Nanjing, China).
133
The apo A1 (97% purity) was isolated from the albumin waste as described
134
previously.35 25-[N-[(7-nitro-2-1,3-benzoxadiazol-4-yl)methyl]amino]-27-norcholest-
135
erol(NBD-cholesterol) was obtained from Cayman Chemical (Michigan, USA).
136
Anti-miR155
(sequence:
137
anti-miRNA
negative
138
5’-ACAACUUAUUACGCACCUA ACUA-3’) and FAM-labled anti-miR155 were
139
synthesized by Genepharm Co., Ltd (Shanghai, China).
140
2.2. Synthesis and Characterization of acid-labile PEI
5’-ACCCCUAUCAC control
(anti-miRNA
AAUUAGCAUUAA-3’), N.C.,
sequence:
141
First, 0.6 g PEI (1.8 kDa) dissolved in dichloromethane (0.05 M) was added to
142
the round bottom flask. Then glutaraldehyde (25% aqueous solution, 364 µL)
143
dissolved in dichloromethane (0.005 M) was added dropwise into the clear PEI
144
solution over 2 h under vigorous stirring at room temperature through dropping funnel.
145
After another 20 h stirring, the solvent was removed by evaporation. Subsequently,
146
the collected viscous residue was dissolved in deionized water again and further
147
purified by dialysis in deionized water (MWCO 8000) for 24 h. The dialysate was
7
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 8 of 54
148
lyophilized for 2 days to achieve yellowish products which was stored at -20 °C until
149
further use.
150
The characteristics of the acid-labile PEI were analyzed by
13
C NMR spectrum
151
(AVANCE AV-300, Bruker, Germany) and FT-IR (Tensor 27, Bruker, Germany). GPC
152
(Shimadzu, Japan) was used to determine the average molecular weight of acid-labile
153
PEI. The mobile phase was ddH2O at the flow rate of 1 mL/min and the column
154
temperature was maintained at 40 °C. Samples of PEG with average molecular
155
weights ranging from 3070-21600 Da were used as standards for calibration.
156
2.3. Degradation and Buffering Capacity of acid-labile PEI
157
GPC was employed to measure the degradation rates of acid-labile PEI under
158
different pH conditions. Five mg/mL acid-labile PEI solution dissolved in deionized
159
water was titrated with HCl to reach the initial pH of 4.5, 5.4 and 7.4, respectively.
160
The average molecular weight of polymer was determined at various time points at
161
37 °C. The half-life of the polymer degradation was defined as the time required for
162
the 50% average molecular weight.
163
The buffering capability of acid-labile PEI was measured by acid-base titration.
164
Briefly, 1 mg/mL polymer dispersed in 0.1 M NaCl was prepared and the initial pH of
165
the polymer solution was adjusted to 10.0 with 1 M NaOH. Then 0.1 M HCl was
166
added dropwise to titrate the solution. The pH values were recorded with a pH Meter
167
(Mettler-Toledo, Switzerland). NaCl solution was titrated in the same manner as a
168
blank control.
169
2.4. Synthesis of Cy5.5 Labeled apoA 1
8
ACS Paragon Plus Environment
Page 9 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
170
To label apo A1, 25 mg apo A1 was dissolved in 9 mL of sodium bicarbonate
171
solution (0.1 M, pH 8.3-8.5). Five mg Cy5.5 NHS ester in 1 mL of DMSO was added
172
to the apo A1 solution, and then the reaction was stirred for 12 h in the dark at room
173
temperature. The resulting product was dialyzed (MWCO 8000-14000 Da) against
174
PBS (pH 7.4) and lyophilized for further use.
175
2.5. Preparation of acid-labile PEI/HNP
176
2.5.1. Preparation of acid-labile PEI core
177
Polymer was dissolved in RNase-free water with the final concentration of 1
178
µg/µL. To form the acid-labile PEI core, anti-miR155 solution in RNase-free water
179
(0.1 nmol/µL) was added to the polycationic solution slowly at a desired N/P ratio.
180
After vortexing for 30 s, the condensed anti-miR155 suspension was incubated at
181
room temperature for 30 min.
182
2.5.2. Preparation of acid-labile PEI/HNP
183
Lipid film hydration-extrusion method was used to prepare the hybrid
184
nanoparticle by enveloping the acid-labile PEI core into liposome, called acid-labile
185
PEI/lipo. Briefly, the lipid compositions composed of phospholipid and cholesterol
186
(10:1, w/w) dissolved in 1 mL of chloroform were mixed into an eggplant flask,
187
evaporating to dryness under vacuum at 40 °C. Then 1 mL condensed anti-miR155
188
complexation was added to the lipid film for electrostatic binding (lipids:cationic
189
complexation = 6:1, w/w), followed by agitation for 5 min in an ice-bath
190
ultrahomogenizer JY92II (Ningbo, China) to hydrate the lipids. Finally, the acid-labile
191
PEI/lipo was obtained by extrusion (Lipex, ATS, Canada) through a 200 nm
9
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
192
polycarbonate membrane 3 times to further package the polycationic core into the
193
lipid bilayers. Sodium cholate dialysis method was carried out to construct the HNP39.
194
250 µL sodium cholate (7 mg/mL in 20 mM Tris-Hcl 8.0 ) and 500 µL apo A1 (14
195
mg/mL in 20 mM Tris-Hcl 8.0) were added to the acid-labile PEI/lipo successively
196
and the final volume was adjusted to 2 mL. The final mixture was then incubated for
197
12 h by stirring gently at 4 °C, followed by dialysis against pH 8.0 Tris-HCl for 48 h
198
to remove excess sodium cholate.
199
2.6. Quantification of anti-miR155 and apo A1
200
The final acid-labile/HNPs were spun down at 15000 rpm for 30 min (4 °C) and
201
then the concentrated nanoparticles were re-dispersed in the Tris-HCl solution (20
202
mM, pH 8.0). Anti-miR155 was quantified by releasing the free anti-miR155 from
203
acid-labile/HNP via Triton X-100 and heparin. Then the releasing anti-miR155 was
204
stained with Ribogreen dye and measured by a microplate reader (POLARstar Omega,
205
BMG Labtech, Germany) at an excitation and emission wavelength of 480 nm and
206
520 nm. For quantification of apo A1, the acid-labile/HNP was prepared with
207
Cy5.5-apo A1 and analyzed at an excitation and emission wavelength of 675 nm and
208
693 nm. Anti-miR155 and apo A1 were quantified against a standard curve, which
209
was established from each of the fluorescently labeled molecules.
210
2.7. Particle Size, Zeta Potential and Morphology Measurements
211
The average particle size and zeta potential of acid-labile PEI core, acid-labile
212
PEI/lipo and acid-labile PEI/HNP were determined by ZetaPlus particle size and zeta
213
potential analyzer (Brookhaven Instruments, USA). Particles were dispersed in
10
ACS Paragon Plus Environment
Page 10 of 54
Page 11 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
214
deionized water and measured in triplicate.
215
The morphological observation was performed by transmission electron
216
microscopy (TEM) (H-7650, Hitachi, Japan). Briefly, a drop of the prepared sample
217
was negatively stained with 2% w/v phosphotungstic acid aqueous solution for 10 min
218
on a perforated carbon film-coated copper grid for analysis.
219
2.8. Serum Stability and RNase Protection Assay
220
A serum stability test was performed to investigate the ability of HNP to protect
221
anti-miRNA from serum nuclease degradation. Briefly, acid-labile PEI/HNP and
222
naked anti-miR155 at an anti-miR155 concentration of 0.1 µg/µL were exposed to
223
10% and 50% fetal bovine serum (FBS) and then incubated at 37 °C for various time
224
periods.
225
RNase protection assay was performed to evaluate the integrity of anti-miR155
226
released from HNP. RNase A (50 µg/mL, 2 µL) was incubated with samples (0.1
227
µg/µL anti-miR155, 10 µL) at 37 °C for 30 min, 1 h and 2 h. At predetermined time
228
point, RNase A was inactivated with EDTA (3 µL, 25 mM) for 20 min at 37 °C.
229
For both serum stability and RNase protection test, free anti-miR-155 needed to
230
be liberated from nanoparticles. 10% Triton X-100 (v/v) causing destruction of lipid
231
bilayer and 0.1% heparin (w/v) dissociating free anti-miR155 were added to aboved
232
samples and incubated at room temperature for 10 min. Afterwards, aliquots of each
233
sample adding with loading buffer were loaded onto a 1.0% (w/v) agarose gel
234
containing GelRed and then electrophoresed for 15 min at a voltage of 90 V in 1 ×
235
TBE buffer solution.
11
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
236
Page 12 of 54
2.9. Cell culture and Cell Viability Assay
237
Mouse macrophage cell line Raw 264.7 cells were cultured in DMEM
238
supplemented with 10% FBS, 100 U/mL penicillin G sodium and 100 µg/mL
239
streptomycin sulfate at 37 °C and 5% CO2.
240
The cytotoxicity of various complexes was evaluated by MTT assay. Raw 264.7
241
cells were seeded in 96-well plates at a density of 1 × 104 cells per well and incubated
242
overnight. After the removal of media, cells were treated with different complexes for
243
24 h. Twenty µL MTT agent (5 mg/mL in PBS) was then added to each well and
244
incubated for another 4 h. Subsequently, the supernatant was replaced by 150 µL
245
dimethyl sulfoxide (DMSO). The optical density values in each well were measured
246
by a microplate reader (Biotek ELx800, USA) at the absorbance of 570 nm. Untreated
247
cells were used as control and cell viability was defined as a percentage of the
248
untreated cells.
249
2.10. Cellular Uptake Study and Active Targeting of acid-labile PEI/HNP to
250
SR-B1
251
To
assess
the
cellular
uptake
of
different
complexes,
fluorescent
252
FAM-anti-miR155 was delivered into Raw 264.7 cells. Raw 264.7 cells (2 × 105
253
cells/well) were plated in 12-well plates and incubated until the 80% confluence.
254
Then the media were renewed by fresh serum-free DMEM containing naked
255
FAM-anti-miR155 alone, 1.8 kDa PEI core, 25 kDa PEI core, acid-labile PEI core,
256
acid-labile PEI/lipo and acid-labile PEI/HNP at a FAM-anti-miR155 concentration of
257
200 nM and the incubation lasted for 6 h. In order to study the active targeting of
12
ACS Paragon Plus Environment
Page 13 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
258
acid-labile PEI/HNP, the SR-B1 receptor was blocked with BLT-1 (dissolved in
259
DMSO), pre-incubated with Raw 264.7 cells at 100 µM for 2 h at 37°C prior to
260
acid-labile PEI core, acid-labile PEI/lipo and acid-labile PEI/HNP treatment. After the
261
treatment for 6 h, the solution was withdrawn and the cells were washed, collected
262
and resuspended with PBS 3 times. The cell fluorescence was analyzed by flow
263
cytometry (MACSQuant, Miltenyi Biotec, Gladbach, Germany) at 493/518 nm and a
264
total of 10,000 events were measured for each sample.
265
2.11. Cellular Trafficking Pathway
266
Raw 264.7 cells (2 × 105 cells/well) were seeded in 12-well plates and incubated
267
for 24 h. Energy-dependent cellular uptake experiments were performed by
268
pre-incubating the cells at 4 °C or with 1 mg/mL NaN3 at 37 °C for 30 min. After this
269
pre-incubation, acid-labile PEI/HNP at a FAM-anti-miR155 concentration of 200 nM
270
was added and incubated for another 6 h at the previous temperature. To investigate
271
the influence of uptake inhibitors on cellular uptake pathway, the cells were
272
pre-incubated for 30 min at 37 °C with different endocytosis inhibitors at the
273
following concentrations: chlorpromazine hydrochloride 10 µg/mL, genistein 54.04
274
µg/mL and amiloride 13.3 µg/mL. Acid-labile PEI/HNP (200 nM) was then added and
275
incubated for 6 h. Subsequently, the cells were washed, harvested, and resuspended 3
276
times with PBS. The mean fluorescence intensity was measured using a flow
277
cytometry. Cells with just the presence of acid-labile PEI/HNP were used as controls.
278
2.12. Intracellular Distribution
279
Raw 264.7 cells (2 × 105) were plated on 15-mm glass-bottom dishes growing
13
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
280
overnight and then exposed to 200 nM acid-labile PEI/HNP. Lyso-Tracker Red (1.5
281
µM) was added and incubated for 0.5 h at a predetermined time, followed by a wash
282
step with PBS for three times. Then cells were fixed with 4% formaldehyde for 15
283
min and stained with Hoechst 33342 (10 µg/mL) for 10 min. Finally, the cells were
284
washed three times with PBS and imaged using an laser scanning confocal
285
microscope (LSM700, Carl Zeiss, Germany).
286
2.13. Assessment of Gene Silencing
287
For quantitative real-time PCR (qRT-PCR) analysis, Raw 264.7 cells (5×105
288
cells/well) were seeded in 6-well plates for 24 h before experiments. The cells were
289
transfected with naked anti-miR155 alone, 1.8 kDa PEI core, 1.8 kDa PEI/lipo, 1.8
290
kDa PEI/HNP, 25 kDa PEI core, 25 kDa PEI/lipo, 25 kDa PEI/HNP, acid-labile PEI
291
core, acid-labile PEI/lipo and acid-labile PEI/HNP (anti-miR155 concentration of 200
292
nM), respectively. After 6 h of cell incubation, the media were exchanged for fresh
293
media, and the cells were continued to incubate for 48 h.
294
The total mRNA was isolated using TriZol reagent (Invitrogen, Grand Island, NY)
295
according to the manufacturer's protocol. Then the miR155 cDNA was synthesized by
296
MicroRNA reverse transcription Kit (GenePharm, Shanghai, China), and the
297
qRT-PCR reactions (Applied Biosystems, USA) were performed using SYBR Master
298
Mix (GenePharm, Shanghai, China). The relative expression of miR155 was
299
calculated using ∆∆Ct method and normalized with U6 snRNA.
300
2.14. DiI-oxLDL Uptake
301
Raw 264.7 cells were seeded in 12-well plates and transfected with naked
14
ACS Paragon Plus Environment
Page 14 of 54
Page 15 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
302
anti-miR155 alone, acid-labile PEI core, acid-labile PEI/lipo and acid-labile PEI/HNP
303
for 48 h. The negative controls were set as acid-labile PEI core N.C, acid-labile
304
PEI/lipo N.C, acid-labile PEI/HNP N.C.. Then the cells were loaded with 40 µg/mL
305
DiI-oxLDL for another 6 h. Blank cells treated with DiI-oxLDL were set as positive
306
controls. At the end of incubation period, the cells were washed, fixed with 4%
307
formaldehyde for 15 min and stained with Hoechst 33342 (10 µg/mL) for 10 min. The
308
images were captured by fluorescence microscope (IX 71, Olympus, Japan).
309
2.15. Intracellular ROS Measurement
310
Intracellular ROS levels were monitored by 2', 7'-Dichlorodihy-drofluorescin
311
diacetate assay (DCFH-DA) probe. Raw 264.7 cells were transfected with naked
312
anti-miR155 alone, acid-labile PEI core, acid-labile PEI/lipo, acid-labile PEI/HNP,
313
acid-labile PEI core N.C., acid-labile PEI/lipo N.C. and acid-labile PEI/HNP N.C.
314
(200 nM) for 48 h and then stimulated with 40 µg/mL oxLDL for 6 h. The cells were
315
subsequently incubated with 10 µM DCFH-DA for 20 minutes at 37 °C, followed by
316
washing with PBS three times. Blank cells treated only with oxLDL or DCFH-DA
317
were used as positive controls and negative controls respectively. The mean
318
fluorescence intensity was analyzed using a flow cytometry operated at an excitation
319
wavelength of 488 nm and an emission wavelength of 525 nm.
320
2.16. Cholesterol Efflux Assay
321
Raw 264.7 cells plated in 24-well plates were stimulated with 40 µg/mL oxLDL
322
and labled with 2 µg/mL NBD-cholesterol for 24 h. The cells were washed and
323
incubated with 200 nM nanoparticles in red-free medium for an additional 48 h. Then
15
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 16 of 54
324
supernatant was collected and the cells were lysed in RIPA lysis buffer. The amounts
325
of cholesterol in media and cells were assayed by a microplate reader (POLARstar
326
Omega, BMG Labtech, Germany) at an excitation and emission wavelength of 485
327
nm and 535 nm. The percentage of cholesterol efflux was calculated as (cholesterol in
328
medium)/(cholesterol in medium + cholesterol in cells) × 100 %.
329
2.17. Statistical Analysis
330
Results were reported as mean ± standard deviation (SD), and a minimum of
331
triplicates were performed in each experiment. Statistical analysis was tested by
332
Student's t-test for two groups and ANOVA for multiple groups. Statistically
333
significant difference was considered at p < 0.05, highly significance was p < 0.01.
334
All statistical analyses were performed by SPSS 19.0 (IBM Corporation, USA).
335
3. RESULTS AND DISCUSSION
336
3.1. Synthesis and Characteristics of acid-labile PEI
337
To reduce the cytotoxicity, degradable PEIs with acid-labile imine linkers were
338
synthesized by Schiff's base reaction between low molecular weight PEI (1.8 kDa)
339
and glutaraldehyde (Scheme 2).38,40 Formation of the polymers was confirmed by 13C
340
NMR spectrum, FT-IR and GPC. As shown in Figure 1A, the appearance of a carbon
341
signal at 164.0582 ppm corroborated the formation of imine linkers in
342
spectrum and the result was also confirmed by the absorbance at ~1658.4 cm−1 in the
343
FT-IR spectrum (Figure 1B). Additionally, the average molecular weight of the
344
acid-labile PEI was determined to be 28.5 kDa by GPC.
345
13
C NMR
The accumulation of non-biodegradable polymers may be a serious problem in
16
ACS Paragon Plus Environment
Page 17 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
346
vivo. Simulating the pH of extracellular environment, endosomes and lysosomes, the
347
degradability of acid-labile PEI was examined in Figure 1C. The half-life of the
348
polymer degradation was 0.9 h, 3.6 h, and 129 h, respectively, at the corresponding
349
pH condition of 4.5, 5.4 and 7.4. The buffering capacity of polymeric gene delivery
350
system is closely related to the transfection efficiency. The high buffering capacity of
351
PEI ascribes to the high content of amino groups, thus facilitating endolysosomal
352
escape of nucleic acids. As seen in Figure 1D, acid-labile PEI exhibited high buffering
353
capacity, even similar with 25 kDa PEI. It is generally accepted that the cytotoxicity
354
and transfection efficiency of PEI are positively correlated with the molecular weight.
355
Higher molecular weight PEI (i.e. 25 kDa) has significant cytotoxicity and high
356
transfection efficiency, while lower molecular weight PEI (i.e. 1.8 kDa) shows low
357
transfection efficiency but negligible cytotoxicity.38 Therefore, crosslinking low
358
molecular weight PEI into high molecular weight PEI with degradable linkers may be
359
an alternative strategy to overcome the high cytotoxicity and mediate efficient gene
360
transfection.
361 362
Scheme 2. The synthesis of acid-labile PEI.
363
17
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
364 365
Figure 1. Characterization of the acid-labile PEI in (A) 13C NMR and (B) FT-IR. (C)
366
Degradation of the acid-labile PEI in different pH conditions. (D) pH buffering
367
capacity of the acid-labile PEI, 25k PEI and 1.8k PEI.
368
3.2. Preparation and Characterization of acid-labile PEI/HNP
369
Before encapsulating anti-miR155 inside the core of HNP, acid-labile PEI was
370
first used to condense the anti-miR155. Agarose gel electrophoresis was performed to
371
investigate whether anti-miR155 molecules were retarded completely by acid-labile
372
PEIs. When the N/P ratio reached 5/1, almost all RNA molecules were complexed and
373
there was no free anti-miR155 band observable in Figure 3A. Dynamic light
374
scattering measurement revealed the changes of particle size at various N/P ratios in
375
Figure 2B, N/P ratio of 5/1 with the minimum size (179.2 ± 1.6 nm) was chosen as the
376
positively charged core (31.75 ± 1.69 mV) for the next step. The core was then
377
packaged in a lipid bilayer by thin-film hydration method, followed by extrusion to
378
apply a mechanical energy. After build-up of the lipid shell on the core (191.2 ± 0.8
379
nm), the zeta potential shifted from positive values to negative values (-24.87 ± 1.37
380
mV), indicating that the lipid bilayer was coated. Afterwards, apo A1 was assembled
18
ACS Paragon Plus Environment
Page 18 of 54
Page 19 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
381
to form the final HNP by sodium cholate dialysis method with the diameter of 180.6 ±
382
0.4 nm and the zeta potential of -37.44 ± 1.63 mV. Fluorescent quantitative analysis of
383
acid-labile PEI/HNP revealed that the amount of anti-miR155 and apo A1 were
384
respectively 24000 nM and 103.5 nM.
385
The morphological characteristics of different nanoparticles were directly
386
visualized by TEM as illustrated in Figure 2A. The image of acid-labile PEI core
387
demonstrated the spherical shape formation. Acid-labile PEI/HNP appeared as a black
388
core surrounded by a rim, confirming the expected core-shell type HNP.
389 390
Figure 2. (A) TEM images of the acid-labile PEI core and acid-labile PEI/HNP. (B)
391
Changes of particle size and zeta potential of acid-labile PEI/anti-miRNA155
392
complexes at different N/P ratios. (C) Particle size and zeta potential of acid-labile
393
PEI core, acid-labile PEI/lipo and acid-labile PEI/HNP.
394
3.3. Serum Stability and RNase Protection Assay
395
When gene delivery vectors enter into the blood circulation, the nonspecific
396
interactions of vectors with serum components will influence the transfection
397
efficiency. Gel retardation assay was carried out to confirm the serum stability of
398
acid-labile PEI/HNP. Figure 3B is the electrophoregram incubated in 10% and 50%
19
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
399
FBS, respectively. It can be seen that the bands of naked anti-miR155 gradually
400
blurred with time in 10% FBS and disappeared completely after 24 h in 50% FBS.
401
However, when anti-miR155 was incorporated into the HNP, there was almost no
402
degradation in both 10% and 50% FBS during 48 h, indicating that HNP protected
403
nucleic acids from the degradation and loss in circulatory system.
404
Another important factor affecting the efficient gene delivery is the enzymatic
405
degradation in vitro and in vivo. The RNase protection ability of vectors was
406
examined in Figure 3C. Naked anti-miR155 was quickly degraded when exposed to
407
the RNase A. In comparison with naked anti-miR155, the released anti-miR bands in
408
HNP were clear and intact. The result implied that protecting anti-miRNAs against
409
nucleases by desirable delivery vectors is necessary.
410 411
Figure 3. (A) Gel retardation assay of acid-labile PEI/anti-miR155 complexes at
412
various N/P ratios. (B) Serum stability test of (a) naked anti-miR155 and (b)
413
acid-labile PEI/HNP in 10% and 50% FBS. (C) Gel electrophoresis of naked
414
anti-miR155 and acid-labile PEI/HNP incubated for different periods with RNase A.
20
ACS Paragon Plus Environment
Page 20 of 54
Page 21 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
415
3.4. Cell Viability Assay
416
The safety of materials is one of an important indicator for ideal gene vectors. In
417
order to compare the cytotoxicity of acid-labile PEI, 1.8 kDa PEI and 25 kDa PEI,
418
three polymers were complexed with anti-miR155 at similar particle size and zeta
419
potential and MTT assay was performed at different polymers concentration. The
420
results in Figure 4A showed that the cell viability of acid-labile PEI and 1.8 kDa PEI
421
remained unchanged as the polymer concentration increasing. However, the cells
422
treated with 25 kDa PEI had significantly higher cytotoxicity compared to those
423
treated with acid-labile PEI and 1.8 kDa PEI, and the viability dropped dramatically
424
to 6% when the polymer concentration reached 50 µg/mL. Liu et al.41 showed that
425
mixing PEI with negatively charged nucleic acids could shield or neutralize the
426
positive charges of the PEI molecules, thus lowering part of the cytotoxicity.
427
Nevertheless, when nucleic acids are released in cytoplasm after internalization, free
428
PEIs will interact with cellular components and disturb the normal cellular
429
process,38,42 especially for high molecular weight PEI. A major benefit of this
430
acid-labile PEI is that it can be degraded into low molecular weight PEI, which is
431
almost non-toxic to cells. Therefore, we expected that the acid-labile PEI could
432
substitute the 25 kDa PEI with the characteristics of high transfection while
433
maintaining the low cytotoxicity. Besides, after loading the cationic core inside the
434
HNP, the cell viabilities of various nanoparticles at tested concentrations were also
435
evaluated in Figure 4B, and no obvious cytotoxicity was observed.
21
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
436 437
Figure 4. (A) Cell viability of three PEI core at various polymer concentrations (1-50
438
µg/mL). (B) Cell viability of different nanoparticles at an anti-miR155 concentration
439
of 200 nM.
440
3.5. Cellular Uptake Study and Active Targeting of acid-labile PEI/HNP to SR-B1
441
The uptake of different complexes was determined by using FAM labled
442
anti-miR155. As illustrated in Figure 5A and B, naked anti-miR155 was hardly
443
uptaked, suggesting that only with the help of vectors could the uptake be improved.
444
Acid-labile PEI core and 25 kDa PEI core had similar fluorescence signals which
445
were stronger than those of 1.8 kDa PEI core. Compared to acid-labile PEI core,
446
acid-labile PEI/lipo showed increased uptake which might be attributed to the polar
447
lipid bilayer facilitating the fusion with the cell membrane. Moreover, after modifying
448
with apo A1, the uptake of acid-labile PEI/HNP was about 2.1-fold higher than that of
449
non-targeted acid-labile PEI/lipo and about 3.2-fold higher than that of cationic
450
acid-labile PEI core in Raw 264.7 cells. To further confirm the role of SR-B1 in
451
mediating the uptake of acid-labile PEI/HNP, we also treated cells with the BLT-1, a
452
selective inhibitor of SR-B1. As exhibited in Figure 6, acid-labile PEI/HNP uptake
453
was decreased by 68.8% after the knock down of SR-B1 in Raw 264.7 cells.
454
Conversely, the uptake of acid-labile PEI core and acid-labile PEI/lipo in cells showed
455
no significant difference with BLT-1 treatment. The results demonstrated the active 22
ACS Paragon Plus Environment
Page 22 of 54
Page 23 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
456
targeting of the acid-labile PEI/HNP to SR-B1 in macrophages.
457 458
Figure 5. (A) and (B) Quantitative analysis the cellular uptake behaviors of different
459
nanoparticles. (C) Influence of different inhibitors on the cellular uptake pathway of
460
acid-labile PEI/HNP.
461 462
Figure 6. (A) and (B) Inhibitory effect of BLT-1 on uptake of acid-labile PEI core (a),
463
acid-labile PEI/lipo (b) and acid-labile PEI/HNP (c) by Raw 264.7 cells.
464
3.6. Mechanism of Cellular Uptake
465
3.6.1. Cellular Trafficking Pathway
466
The endocytosis of nanoparticle is an active and energy-dependent process, which
467
can be inhibited by low temperature as well as ATP enzyme inhibitors.43 Therefore,
468
the two factors are usually the most direct way to distinguish the endocytic pathway
469
and the non-endocytic pathway. Sodium azide (NaN3) which can inhibit the
23
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
470
production of energy by blocking the respiratory chain of mitochondria is widely used
471
as the inhibitor of energy dependence.43 As seen in Figure 5C, a decreasing cellular
472
uptake was observed in cells incubated at 4 °C or incubated with NaN3, clarifying the
473
energy-dependent endocytosis.
474
Generally, there are four types of endocytic pathway, including clathrin-mediated
475
endocytosis, caveolae-mediated endocytosis, macropinocytosis and phagocytosis.44 To
476
elucidate the cellular trafficking pathway of acid-labile PEI/HNP, different inhibitors
477
blocking specific pathway were used. Chlorpromazine hydrochloride prevents
478
clathrin-coated pit formation by promoting clathrin agglutination in late endosomes.45
479
In cells treated with chlorpromazine hydrochloride, there was a significant decrease
480
(68% reduction) in cellular uptake relative to the control. Genistein, a known inhibitor
481
of caveolae-mediated endocytosis, showed a considerably smaller impact on the
482
internalization in Raw 264.7 cell (17.6% reduction). Next, inhibitor of sodium-proton
483
exchange “amiloride” for the macropinocytosis inhibition44 exhibited a 38% reduction
484
in Raw 264.7 cell. It could be concluded that clathrin-mediated endocytosis was the
485
main uptake pathway for acid-labile PEI/HNP, and macropinocytosis was also an
486
effective uptake pathway in Raw 264.7 cells.
487
3.6.2. Intracellular Distribution
488
To further illuminate the trafficking mechanism of acid-labile PEI/HNP, the
489
intracellular behavior was investigated by confocal microscopy. FAM labeled
490
anti-miR155 was used to trace the nanoparticles, and the nucleus and acidic
491
endolysosomes were stained by Hoechst 33342 and Lyso-Tracker Red, respectively.
24
ACS Paragon Plus Environment
Page 24 of 54
Page 25 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
492
In Raw 264.7 cells, acid-labile/HNPs were observed bound to the cell membrane at 2
493
h (Figure 7). By 6 h post-treatment, intracellular green fluorescence began to appear
494
in larger and brighter compartments, suggestive of vesicles containing numerous
495
nanoparticles. Futhermore, the yellow fluorescence appeared in the cells marginally
496
which was the co-localization of FAM-anti-miR155 and Lyso-Tracker, declaring that
497
some nanoparticles had located in endolysosomes. After 16 h incubation, an increased
498
yellow fluorescence was visible in cells, indicating that most of the nanoparticles had
499
been entrapped within endolysosomes. This result was consistent with the conclusion
500
above that clathrin-mediated endocytosis was the primary endocytosis pathway. At the
501
time of 34 h, the yellow fluorescence of the image was weakened accompanied by the
502
appearance of large green fluorescence. It meant that the anti-miR155 had escaped
503
from the endolysosomes into cytoplasm successfully, mainly attributing to the high
504
buffering capacity of acid-labile PEI.
505
It is noteworthy that the intracellular trafficking and uptake mechanism of HDL
506
are dependent on different cell lines. For instance, the interaction of HDL with
507
hepatocytes involves the selective lipid uptake mediated by SR-B146 and the
508
holo-particle uptake mediated by SR-B1, CD36 and the mitochondrial β-chain of
509
ATP synthase and P2Y13.47,48 Yang and other researchers utilized the selective lipid
510
uptake pathway for HNP delivering nucleic acids directly into the cytoplasm thereby
511
bypassing the endosomal trapping.12-14 Researchers also took advantage of the
512
holo-particle uptake pathway of HNP for the target-specific delivery of siRNA.49
513
However, there has been no report about the selective lipid uptake pathway in
25
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
514
macrophages, which might be associated with the physiological functions of different
515
cells. In hepatocytes, mature HDL can unload free and esterified cholesterol by open a
516
non-aqueous channel in SR-B1, namely selective cholesteryl ester uptake pathway for
517
depletion.48,50 In contrast, HDL is responsible for mediating cholesterol efflux in
518
macrophages and foam cells. And in our experiments, FAM-anti-miR155 and
519
Cy5.5-apo A1 were used to label the acid-labile/HNP for characterizing the uptake
520
pathway. As shown in Figure S1, green and red fluorescences were located within the
521
cytosolic compartments and observed in the same positions at 6 h, demonstrating a
522
holo-particle uptake pathway of HNP in macrophages.
523 524
Figure 7. Confocal microscopy shows intracellular distribution of acid-labile
525
PEI/HNP. Green fluorescence represents the FAM-anti-miR155. Red fluorescence is
526
from Lyso-Tracker Red endolysosomes staining. Blue fluorescence is from the
527
nucleus stained with Hoechst 33342. Yellow fluorescence is from the co-localization
528
of green and red.
529
3.7. In vitro Gene Silencing Efficiency
530
Figure 8 shows the miR155 expression levels in different formulations containing 26
ACS Paragon Plus Environment
Page 26 of 54
Page 27 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
531
anti-miR155 relative to the untreated group examined by qRT-PCR. Naked
532
anti-miR155 could hardly penetrate the cell membrane, resulting in low transfection
533
efficiency. Therefore, the acid-labile PEI as a vector with low cytotoxicity and high
534
transfection efficiency was synthesized. Packaging anti-miR155 into 1.8 kDa PEI and
535
25 kDa PEI formulations were used as the negative control and positive control
536
groups, respectively. As expected, acid-labile PEI and 25 kDa PEI treated groups
537
exhibited a similar and significantly reduced expression of miR155 levels compared
538
to the untreated group. There was almost no silencing efficiency in 1.8 kDa PEI
539
treated group. The high transfection efficiency of acid-labile PEI and 25 kDa PEI
540
formulations was attributed to the high charge density and the high buffering capacity,
541
thus triggering anti-miR155 escape from endolysosomes. Subsequently, the
542
transfection efficiency in each polymer treated group was compared. Treatment with
543
the acid-labile PEI or 25 kDa PEI/lipo caused a slight reduction in the expression of
544
miR155 levels to those of acid-labile PEI or 25 kDa PEI core. The positive surface
545
charges of acid-labile PEI or 25 kDa PEI core and the polar lipid bilayer of acid-labile
546
PEI or 25 kDa PEI/lipo were favorable for the cellular uptake, which contributed to
547
the increased gene silencing efficiency. In acid-labile PEI or 25 kDa PEI/HNP treated
548
group, the miR155 expression was strongly inhibited compared to that in acid-labile
549
PEI or 25 kDa PEI/lipo treated group, and the reduction was 22% and 23.5%
550
respectively, exhibiting the advantage of specific targeting of HNP.
551
27
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
552 553
Figure 8. In vitro gene silencing efficiency after the transfection with different
554
nanoparticles at an anti-miR155 concentration of 200 nM.
555
3.8. DiI-oxLDL Uptake
556
Lipid uptake plays a central role in macrophage-derived foam cell formation.51
557
Tian proved that miR155 promoted lipid uptake and ROS production to enchance
558
macrophages-derived foam cell formation in vitro via direct suppression of HMG
559
box-transcription protein1 (HBP1). Conversely, inhibition of miR155 reduced lipid
560
uptake at a dose dependent manner.7 In order to explore the efficacy of various
561
formulations containing anti-miR155, DiI-oxLDL uptake assay was performed to
562
evaluate the intracellular lipid contents (Figure 9). Compared to the positive control
563
group, there was no obvious inhibition on lipid uptake in the treatment of naked
564
anti-miR155 alone. When the anti-miR155 was packaged in different formulations,
565
the lipid uptake gradually decreased in the order of acid-labile PEI core, acid-labile
566
PEI/lipo and acid-labile PEI/HNP group. Excluding the effect of anti-miR155, the
567
blank HNP also exhibited the function of inhibiting lipid uptake in acid-labile
568
PEI/HNP loaded with negative control anti-miRNA relative to that in acid-labile PEI
569
core N.C. and acid-labile PEI/lipo N.C. groups. Cho et al. also demonstrated that 28
ACS Paragon Plus Environment
Page 28 of 54
Page 29 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
570
rHDL treatment had the antioxidant activity by inhibiting cellular uptake of oxLDL.19
571
Furthermore, to exclude a potential competition between ox-LDL and HNP,
572
incubating acid-labile PEI/HNP N.C. with Raw 254.7 cells, with or without washing
573
before stimulated with oxLDL was performed to make a comparison. There was no
574
significant difference in the uptake of DiI-oxLDL whether there was a washing after
575
the treatment of acid-labile PEI/HNP N.C. (Figure S2), meaning that the reduction of
576
DiI-oxLDL uptake was not caused by the competition of the HNP. Above all, these
577
findings suggested that both anti-miR155 and blank HNP inhibited the
578
oxLDL-induced foam cell formation.
579
Recent studies have revealed the potential anti-atherosclerotic effect of miR155
580
inhibition. MiR155 deficiency in apo E-deficient mice resulted in decreased
581
macrophage inflammations and lipid dispositions and attenuated atherogenesis by
582
interfering with different pathway.6-8 However, others reported that LDL receptor
583
deficiency (LDLR−/−) mice with miR155−/− bone marrow cells showed a contradictory
584
outcome with enchanced atherosclerosis.52 The inconsistent results are speculated to
585
be dependent on different animal models of the disease. The detailed atheroprotection
586
of anti-miR155 in our experiments needs to be further verified in atherosclerotic
587
animal models.
588 589
Figure 9. DiI-oxLDL uptake images captured by fluorescence microscope.
29
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
590
3.9. ROS Production
591
ROS production is another important factor effects foam cell formation, which
592
has been proved that transfection of miR155 in Raw 264.7 cells enhanced ROS
593
production, while inhibition of miR155 presented the opposite effects.7 As shown in
594
Figure 10A, exposure of cells to formulations containing anti-miR155 led to a marked
595
decrease in ROS production compared to the blank cells only irritated with oxLDL.
596
The production of ROS in various formulations decreased in the following order:
597
acid-labile PEI core group, acid-labile PEI/lipo group and acid-labile PEI/HNP group,
598
respectively. We next examined whether blank HNP affects ROS generation.
599
Interestingly, acid-labile PEI/HNP N.C. showed a significantly suppression of ROS
600
generation relative to the positive control. Tölle reported that HDL had the capacity to
601
inhibit NAD(P)H oxidase-dependent ROS generation in vascular smooth muscle
602
cells.53 In Figure S3, the production of ROS also displayed no obvious difference with
603
or without washing after the treatment of acid-labile PEI/HNP N.C., eliminating the
604
potential competition between oxLDL and HNP. It is hoped that this prepared
605
acid-labile PEI/HNP can not only play the role of the anti-miR155, but also exert the
606
biological function of the HNP itself.
607 608
Figure 10. (A) ROS production and (B) Cholesterol efflux assay in Raw 264.7 cells
609
after the incubation with different nanoparticles. 30
ACS Paragon Plus Environment
Page 30 of 54
Page 31 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
610
3.10. Cholesterol Efflux Assay
611
It has been well known that cholesterol efflux from macrophages to HDL
612
represents an important mechanism for maintaining cholesterol homeostasis. To
613
examine whether miR155 and HNP have the capacity of mediating cholesterol efflux,
614
NBD-cholesterol and ox-LDL were pre-incubated with Raw 264.7 cells. As seen in
615
Figure 10B, there was a small increase of cholesterol efflux in the presence of
616
acid-labile PEI/lipo and acid-labile PEI core compared to the control and naked
617
anti-miR155 group, whereas acid-labile PEI/HNP group displayed significantly
618
increased cholesterol efflux. Moreover, we found that cholesterol efflux to blank HNP
619
was significantly increased compared to the control, but efflux to acid-labile PEI/lipo
620
N.C. and acid-labile PEI core N.C. was not changed, manifesting the potential RCT
621
function of HNP. It was reported that SR-B1-mediated HDL endocytosis leads to the
622
resecretion of HDL.48,54 During the retro-endocytosis, HDL interacted with lipid
623
droplets in foam cells for the exchange of cholesterol to HDL, thus facilitating
624
cholesterol efflux. Besides, we speculated that the lipids bilayer structure also
625
provided a space for sequestering more cholesterol as demonstrated by Luthi.35
626
CONCLUSIONS
627
In summary, a biomimetic polymer-lipid hybrid HNP loading anti-miR155 was
628
established successfully for combined antiatherogenic effects on macrophages with
629
nanocarrier and nucleic acid. The developed HNP realized the macrophages-specific
630
targeting and escaped from the endolysosomes via clathrin-mediated endocytosis with
631
high transfection efficiency. Besides, in vitro cells confirmed that the HNP closely
31
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
632
mimicking endogenous HDL in the unique anti-atherosclerotic biological function had
633
the ability of antioxidation and mediating cholesterol efflux. Further in vivo
634
pharmacodynamics of both anti-miR155 and nanocarrier are still required to be
635
carried out in atherosclerotic animal models.
636
ASSOCIATED CONTENT
637
Supporting Information
638
Confocal images of acid-labile PEI/HNP, DiI-oxLDL uptake images of acid-labile
639
PEI/HNP N.C. and the ROS production of acid-labile PEI/HNP N.C..
640
AUTHOR INFORMATION
641
Corresponding Authors
642
*E-mail:
[email protected]. Fax:+86-25-83271293
643
Notes
644
The authors declare no competing financial interests.
645
ACKNOWLEDGMENTS
646
The work was financially supported by National Natural Science Foundation of China
647
Project (No. 81273466) and Priority Academic Program Development of Jiangsu
648
Higher Education Institutions. We also thank you Minhui Sun, Yingjian Hou, and
649
Xiaonan Ma, for technical support from Cellular and Molecular Biology Center of
650
China Pharmaceutical University.
651
REFERENCES
652
(1) Weber, C.; Noels H. Atherosclerosis: current pathogenesis and therapeutic
653
options. Nat. Med. 2011, 17, 1410-1422.
32
ACS Paragon Plus Environment
Page 32 of 54
Page 33 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
654
(2) Moore, K. J.; Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell
655
2011, 145, 341-355.
656
(3) Li, A. C.; Glass, C. K. The macrophage foam cell as a target for therapeutic
657
intervention. Nat. Med. 2002, 8, 1235-1243.
658
(4) Madrigal-Matute, J.; Rotllan, N.; Aranda, J. F.; Fern á ndez-Hernando, C.
659
MicroRNAs and atherosclerosis. Curr. Atheroscler. Rep. 2013, 15, 1-8.
660
(5) Condorelli, G.; Latronico, M. V. G.; Cavarretta, E. MicroRNAs in cardiovascular
661
diseases: current knowledge and the road ahead. J. Am. Coll. Cardiol. 2014, 63,
662
2177-2187.
663
(6) Nazari-Jahantigh, M.; Wei, Y.; Noels, H.; Akhtar, S.; Zhou, Z.; R.Koenen, R.; Heyll,
664
K.; Gremse, F.; Kiessling, F.; Grommes, J.; Weber, C.; Schober, A. MicroRNA-155
665
promotes atherosclerosis by repressing Bcl6 in macrophages. J. Clin. Invest. 2012,
666
122, 4190-4202.
667
(7) Tian, F. J.; An, L. N.; Wang, G. K.; Zhu, J. Q.; Zhang, Y. Y.; Zeng, A.; Zou, J.;
668
Zhu, R. F.; Han, X. S.; Shen, N.; Yang, H. T.; Zhao, X. X.; Huang, S.; Qin, Y. W.;
669
Jing, Q. Elevated microRNA-155 promotes foam cell formation by targeting HBP1 in
670
atherogenesis. Cardiovasc. Res. 2014, 103, 100-110.
671
(8) Du, F.; Yu, F.; Wang, Y.; Hui, Y.; Carnevale, K.; Fu, M.; Lu, H.; Fan, D.
672
MicroRNA-155 deficiency results in deceased macrophage inflammation and
673
attenuated atherogenesis in apolipoprotein E-deficient Mice. Arterioscl. Throm. Vas.
674
2014, ATVBAHA. 113.302701.
33
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
675
(9) Singh, A.; Talekar, M.; Raikar, A.; Aimiji, M. Macrophage-targeted delivery
676
systems for nucleic acid therapy of inflammatory diseases. J. Controlled Release
677
2014, 190, 515-530.
678
(10) Damiano, M. G.; Mutharasan, R. K.; Tripathy, S.; McMahon, K. M.; Thaxton, C.
679
S. Templated high density lipoprotein nanoparticles as potential therapies and for
680
molecular delivery. Adv. Drug Delivery Rev. 2013, 65, 649-662.
681
(11) McMahon, K. M.; Mutharasan, R. K.; Tripathy, S.; Veliceasa, D.; Bobeica, M.;
682
Shumaker, D.K.; Luthi, A. J.; Helfand,B. T.; Ardehail, H.; Mirkin, C. A.; Volpert, D.;
683
Thaxton, C. S. Biomimetic high density lipoprotein nanoparticles for nucleic acid
684
delivery. Nano. Lett. 2011, 11, 1208-1214.
685
(12) Yang, M.; Jin, H.;Chen, J.; Ding, L.; Ng, K. K.; Lin, Q. Y.; Lovell, J. F.; Zhang,
686
Z. H.; Zheng, G. Efficient cytosolic delivery of siRNA using HDL‐mimicking
687
nanoparticles. Small 2011, 7, 568-573.
688
(13) Lin, Q.; Chen, J.; Jin, H.; Ng, K. K.; Yang, M.; Cao, W. G.; Ding, L. L.; Zhang,
689
Z. H.; Zheng, G. Efficient systemic delivery of siRNA by using high-density
690
lipoprotein-mimicking peptide lipid nanoparticles. Nanomedicine 2012, 7, 1813-1825.
691
(14) Zhang, Z.; Cao, W.; Jin, H.; Lovell, J. F.; Yang, M.; Ding, L. L.; Chen, J.;
692
Corbin, L.; Luo, Q. M.; Zheng, G. Biomimetic nanocarrier for direct cytosolic drug
693
delivery. Angew. Chem. Int. Edit. 2009, 48, 9171-9175.
694
(15) McMahon, K. M.; Plebanek, M. P.; Thaxton, C. S. Properties of Native High‐
695
Density Lipoproteins Inspire Synthesis of Actively Targeted In Vivo siRNA Delivery
696
Vehicles. Adv. Funct. Mater. 2016, 26, 7824-7835.
34
ACS Paragon Plus Environment
Page 34 of 54
Page 35 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
697
(16) Shahzad, M. M. K.; Mangala, L. S.; Han, H. D.; Lu, C. H.; Bottsford-Miller, J.;
698
Nishimura, M.; Mora, E. M.; Lee, J. W.; Stone, R. L. Targeted delivery of small
699
interfering RNA using reconstituted high-density lipoprotein nanoparticles. Neoplasia
700
2011, 13, 309IN3-319IN8.
701
(17) Krause, B. R.; Remaley, A. T. Reconstituted HDL for the acute treatment of
702
acute coronary syndrome. Curr. Opin. Lipidol. 2013, 24, 480-486.
703
(18) Newton, R. S.; Krause, B. R. HDL therapy for the acute treatment of
704
atherosclerosis. Atherosclerosis Sup. 2002, 3, 31-38.
705
(19) Cho, K. H.; Kim, J. R. A reconstituted HDL containing V156K or R173C apoA-I
706
exhibited anti-inflammatory activity in apo-E deficient mice and showed resistance to
707
myeloperoxidase-mediated oxidation. Exp. Mol. Med. 2009, 41, 417-428.
708
(20) Badimon, J. J.; Badimon, L.; Galvez, A.; Dische, R.; Fuster, V. High density
709
lipoprotein plasma fractions inhibit aortic fatty streaks in cholesterol-fed rabbits.
710
Scand. J. Clin. Lab. Inv. 1989, 60, 455-461.
711
(21) Badimon, J. J.; Badimon, L,; Fuster, V. Regression of atherosclerotic lesions by
712
high density lipoprotein plasma fraction in the cholesterol-fed rabbit. J. Cin. Invest.
713
1990, 85, 1234.
714
(22) Tardif, J. C.; Grégoire, J.; L’Allier, P. L.; Ibrahim, R.; Lespérance, J.; Heinonen,
715
T. M.; Kouz, S.; Berry, C.; Basser, R.; Lavoie, M. A.; Guertin, M. C.; Rodes-Cabau,
716
J. Effects of reconstituted high-density lipoprotein infusions on coronary
717
atherosclerosis: a randomized controlled trial. Jama 2007, 297, 1675-1682.
35
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
718
(23) Shaw, J. A.; Bobik, A.; Murphy, A.; Kanellakis, P.; Blombery, P.;
719
Mukhamedova, N.; Woollard, K.; Lyon, S.; Sviridov, D.; Dart, A. M. Infusion of
720
reconstituted high-density lipoprotein leads to acute changes in human atherosclerotic
721
plaque. Circ. Res. 2008, 103, 1084-1091.
722
(24) Orekhov, A. N.; Misharin, A. Y.; Tertov, V. V.; Khashimov, K.; Pokrovsky, S.
723
N.; Repin, V. S.; Smirnov, V. N. Artificial HDL as an anti-atherosclerotic drug.
724
Lancet 1984, 324, 1149-1150.
725
(25) Koizumi, J.; Kano, M.; Okabayashi, K.; Jadhav, A.; Thompson, G. R. Behavior
726
of human apolipoprotein AI: phospholipid and apoHDL: phospholipid complexes in
727
vitro and after injection into rabbits. J. Lipid. Res. 1988, 29, 1405-1415.
728
(26) Kontush, A.; Chapman, M. J. Antiatherogenic function of HDL particle
729
subpopulations: focus on antioxidative activities. Curr. Opin. Lipidol. 2010, 21,
730
312-318.
731
(27) Kontush, A.; Chapman, M. J. Antiatherogenic small, dense HDL-guardian angel
732
of the arterial wall. Nat. Clin. Pract. Card. 2006, 3, 144-153.
733
(28) Cockerill, G. W.; Rye, K. A.; Gamble, J. R.; Vadas, M. A.; Barter, P. J.
734
High-density lipoproteins inhibit cytokine-induced expression of endothelial cell
735
adhesion molecules. Arterioscl. Throm. Vas. 1995, 15, 1987-1994.
736
(29) Ansell, B. J.; Navab, M.; Hama, S.; Kamranpour, N.; Fonarow, G.; Hough, G.;
737
Rahmani, S.; Mottahedeh, R.; Dave, R.; Reddy, S. T.; Fogelman, A. M.
738
Inflammatory/antiinflammatory properties of high-density lipoprotein distinguish
739
patients from control subjects better than high-density lipoprotein cholesterol levels
36
ACS Paragon Plus Environment
Page 36 of 54
Page 37 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
740
and are favorably affected by simvastatin treatment. Circulation 2003, 108,
741
2751-2756.
742
(30) Sugano, M.; Tsuchida, K.; Makino, N. High-density lipoproteins protect
743
endothelial cells from tumor necrosis factor-α-induced apoptosis. Biochem. Bioph.
744
Res. Co. 2000, 272, 872-876.
745
(31) Nofer, J. R.; Van Der Giet, M.; Tölle, M.; Wolinska, I.; Lipinski, K. W.; Baba, H.
746
A.; Tietge, U. J.; Godecke, A.; Ishii, I.; Kleuser, B.; Schafers, M.; Fobker, M.; Zidek,
747
W.; Assmann, G.; Chun, J.; Levkau, B. HDL induces NO-dependent vasorelaxation
748
via the lysophospholipid receptor S1P 3. J. Clin. Invest. 2004, 113, 569-581.
749
(32) Yang, S; Damiano, M. G.; Zhang, H.; Tripathy, S.; Luthi, A. J.; Rink, J. S.;
750
Ugolkov, A. V.; Singh, A. T. k.; Dave, S. S.; Gordon, L. I.; Thaxton, C. S.
751
Biomimetic, synthetic HDL nanostructures for lymphoma. P. Natl. Acad. Sci. 2013,
752
110, 2511-2516.
753
(33) Numata, M.; Grinkova, Y. V.; Mitchell, J. R.; Chu, H. W.; Sligar, S. G.; Voelker,
754
D. R. Nanodiscs as a therapeutic delivery agent: inhibition of respiratory syncytial
755
virus infection in the lung. Int. J. Nanomed. 2013, 8, 1417-1427.
756
(34) Mutharasan, R. K.; Foit, L.; Thaxton, C. S. High-density lipoproteins for
757
therapeutic delivery systems. J. Mater. Chem. B 2016, 4, 188-197.
758
(35) Luthi, A. J.; Zhang, H.; Kim, D.; Giljohann, D. A.; Mirkin, C. A.; Thaxton, C. S.
759
Tailoring of biomimetic high-density lipoprotein nanostructures changes cholesterol
760
binding and efflux. ACS Nano 2011, 6, 276-285.
37
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 38 of 54
761
(36) Zhang, M. Y,; He, J. H.; Jiang, C.P.; Zhang, W. L.; Yang, Y.; Wang, Z. Y.; Liu, J.
762
P. Plaque-hyaluronidase-responsive high-density-lipoprotein-mimetic nanoparticles
763
for
764
anti-atherosclerotic therapy. Int. J. Nanomed. 2017, 12, 533.
765
(37) Behr, J. P. The proton sponge: a trick to enter cells the viruses did not exploit.
766
CHIMIA International Journal for Chemistry. 1997, 51, 34-36.
767
(38) Kim, Y. H.; Park, J. H.; Lee, M.; Kim, Y. H.; Park, T. G.; Kim, S. W. Polyethylene
768
with acid-labile linkages as a biodegradable gene carrier. J. Controlled Release 2005,
769
103, 209-219.
770
(39) He, H.; Liu, L.; Bai, H.; Wang, J.; Zhang, Y.; Zhang, W. L.; Zhang, M. Y.; Wu,
771
Z.; Liu, J. P. Arachidonic acid-modified lovastatin discoidal reconstituted high density
772
lipoprotein markedly decreases the drug leakage during the remodeling behaviors
773
induced by lecithin cholesterol acyltransferase. Pharm. Res. 2014, 31, 1689-1709.
774
(40) Chen, S.; Jin, T. Poly-cross-linked PEI through aromatically conjugated imine
775
linkages as a new class of pH-responsive nucleic acids packing cationic polymers.
776
Front. Pharmacol. 2016, 7.
777
(41) Liu, Z.; Niu, D.; Zhang, J.; Zhang, W.; Yao, Y.; Li, P.; Gong, J. Amphiphilic
778
core-shell nanoparticles containing dense polyethyleneimine shells for efficient
779
delivery of microRNA to Kupffer cells. Int. J. Nanomed. 2016, 11, 2785.
780
(42) Godbey, W. T.; Wu, K. K.; Mikos, A. G. Poly (ethylenimine)-mediated gene
781
delivery affects endothelial cell function and viability. Biomaterials 2001, 22,
782
471-480.
multistage
intimal-macrophage-targeted
drug
delivery
38
ACS Paragon Plus Environment
and
enhanced
Page 39 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
783
(43) Liu, P.; Sun, Y.; Wang, Q.; Sun, Y.; Li, H.; Duan, Y. Intracellular trafficking and
784
cellular uptake mechanism of mPEG-PLGA-PLL and mPEG-PLGA-PLL-Gal
785
nanoparticles for targeted delivery to hepatomas. Biomaterials 2014, 35, 760-770.
786
(44) Xiang, S.; Tong, H.; Shi, Q.; Fernandes, J. C.; Jin, T.; Dai, K.; Zhang, X. Uptake
787
mechanisms of non-viral gene delivery. J. Controlled Release 2012, 158, 371-378.
788
(45) Wang, L. H.; Rothberg, K. G.; Anderson, R. G. W. Mis-assembly of clathrin
789
lattices on endosomes reveals a regulatory switch for coated pit formation. J. Cell Biol.
790
1993, 123, 1107-1107.
791
(46) Rodrigueza, W. V.; Thuahnai, S. T.; Temel, R. E.; Lund-Katz, S.; Phillips, M. C.;
792
Williams, D. L. Mechanism of scavenger receptor class B type I-mediated selective
793
uptake of cholesteryl esters from high density lipoprotein to adrenal cells. J. Biol.
794
Chem. 1999, 274, 20344-20350.
795
(47) Röhrl, C.; Pagler, T. A.; Strobl, W.; Ellinger, A.; Neumuller, J.; Pasvelka, M.;
796
Stangl, H.; Meisslitzer-Ruppitsch, C. Characterization of endocytic compartments
797
after holo-high density lipoprotein particle uptake in HepG2 cells. Histochem. Cell
798
Biol. 2010, 133, 261-272.
799
(48) Röhrl, C.; Stangl, H. HDL endocytosis and resecretion. Bba-mol. Cell Biol. L.
800
2013, 1831, 1626-1633.
801
(49) Rui, M.; Tang, H.; Li, Y.; Wei, X.; Xu, Y. Recombinant high density lipoprotein
802
nanoparticles for target-specific delivery of siRNA. Pharm. Res. 2013, 30, 1203-1214.
803
(50) Rodrigueza, W. V.; Thuahnai, S. T.; Temel, R. E.; Lund-Katz, S.; Phillips, M. C.;
804
Williams, D. L. Mechanism of scavenger receptor class B type I-mediated selective
39
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
805
uptake of cholesteryl esters from high density lipoprotein to adrenal cells. J. Biol.
806
Chem. 1999, 274, 20344-20350.
807
(51) Li, A. C.; Glass, C. K. The macrophage foam cell as a target for therapeutic
808
intervention. Nat. Med. 2002, 8, 1235-1243.
809
(52) Donners, M. M. P. C.; Wolfs, I. M. J.; Stöger, L. J.; Vorst, E.; Pottgens, C.;
810
Heymans, S.; Schroen, B.; Gijbels, M.; Winher, M. Hematopoietic miR155 deficiency
811
enhances atherosclerosis and decreases plaque stability in hyperlipidemic mice. Plos
812
One 2012, 7, e35877.
813
(53) Tölle, M.; Pawlak, A.; Schuchardt, M.; Kawamura, A.; Tietge, U. J.; Lorkowski,
814
S.; Keul, P.; Assmann, G.; Chun, J.; Levkau, B.; Giet, M.; Nofer, J. R. HDL-associated
815
lysosphingolipids inhibit NAD (P) H oxidase-dependent monocyte chemoattractant
816
protein-1 production. Arterioscl. Throm. Vas. 2008, 28, 1542-1548.
817
(54) Pagler, T. A.; Rhode, S.; Neuhofer, A.; Laggner, H.; Strobl, W.; Hinterndorfer, C.;
818
Volf, I.; Pavelka, M.; Eckhardt, E. R. M.; Westhuyzen, D. R. Schutz, G. J.; Stangl, H.
819
SR-BI-mediated high density lipoprotein (HDL) endocytosis leads to HDL resecretion
820
facilitating cholesterol efflux. J. Biol. Chem. 2006, 281, 11193-11204.
821 822 823 824 825 826
40
ACS Paragon Plus Environment
Page 40 of 54
Page 41 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
827
For Table of Contents Use Only
831
Biofunctional polymer-lipid hybrid high density lipoprotein-mimicking nanoparticles loading anti-miR155 for combined antiatherogenic effects on macrophages
832
Jing Lu, Yi Zhao, Xiaoju Zhou, Jian Hua He, Yun Yang, Cuiping Jiang, Zitong Qi,
833
Wenli Zhang and Jianping Liu*
834
Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China
828 829 830
835 836
Schematic illustration of the acid-labile PEI/HNP trafficking in Raw 264.7 cell.
41
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Schematic illustration of the acid-labile PEI/HNP trafficking in Raw 264.7 cell. 35x14mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 42 of 54
Page 43 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 1. Characterization of the acid-labile PEI in (A) 13C NMR and (B) FT-IR. (C) Degradation of the acidlabile PEI in different pH conditions. (D) pH buffering capacity of the acid-labile PEI, 25k PEI and 1.8k PEI. 64x50mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 2. (A) TEM images of the acid-labile PEI core and acid-labile PEI/HNP. (B) Changes of particle size and zeta potential of acid-labile PEI/anti-miRNA155 complexes at different N/P ratios. (C) Particle size and zeta potential of acid-labile PEI core, acid-labile PEI/lipo and acid-labile PEI/HNP. 53x34mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 44 of 54
Page 45 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 3. (A) Gel retardation assay of acid-labile PEI/anti-miR155 complexes at various N/P ratios. (B) Serum stability test of (a) naked anti-miR155 and (b) acid-labile PEI/HNP in 10% and 50% FBS. (C) Gel electrophoresis of naked anti-miR155 and acid-labile PEI/HNP incubated for different periods with RNase A. 74x73mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 4. (A) Cell viability of three PEI core at various polymer concentrations (1-50 µg/mL). (B) Cell viability of different nanoparticles at an anti-miR155 concentration of 200 nM. 33x13mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 46 of 54
Page 47 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 5. (A) and (B) Quantitative analysis the cellular uptake behaviors of different nanoparticles. (C) Influence of different inhibitors on the cellular uptake pathway of acid-labile PEI/HNP. 42x11mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 6. (A) and (B) Inhibitory effect of BLT-1 on uptake of acid-labile PEI core (a), acid-labile PEI/lipo (b) and acid-labile PEI/HNP (c) by Raw 264.7 cells. 61x45mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 48 of 54
Page 49 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 7. Confocal microscopy shows intracellular distribution of acid-labile PEI/HNP. Green fluorescence represents the FAM-anti-miR155. Red fluorescence is from Lyso-Tracker Red endolysosomes staining. Blue fluorescence is from the nucleus stained with Hoechst 33342. Yellow fluorescence is from the co-localization of green and red. 66x53mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 8. In vitro gene silencing efficiency after the transfection with different nanoparticles at an antimiR155 concentration of 200 nM. 57x40mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 50 of 54
Page 51 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Figure 9. DiI-oxLDL uptake images captured by fluorescence microscope. 31x12mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Figure 10. (A) ROS production and (B) Cholesterol efflux assay in Raw 264.7 cells after the incubation with different nanoparticles. 34x14mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 52 of 54
Page 53 of 54
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
Scheme 1. Schematics of the acid-labile PEI/HNP. 77x72mm (300 x 300 DPI)
ACS Paragon Plus Environment
Biomacromolecules
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Scheme 2. The synthesis of acid-labile PEI. 177x53mm (300 x 300 DPI)
ACS Paragon Plus Environment
Page 54 of 54