Accepted Manuscript: 10.1016/j.jconrel.2018.04.006
Accepted Manuscript: 10.1016/j.jconrel.2018.04.006
Accepted Manuscript: 10.1016/j.jconrel.2018.04.006
Jun Gi Rho, Hwa Seung Han, Ji Hye Han, Hansang Lee, Van Quy
Nguyen, Wang Hee Lee, Seunglee Kwon, Sungeun Heo, Juhwan
Yoon, Han Ho Shin, Eun-young Lee, Hoin Kang, Siyoung Yang,
Eun Kyung Lee, Jae Hyung Park, Wook Kim
PII: S0168-3659(18)30180-9
DOI: doi:10.1016/j.jconrel.2018.04.006
Reference: COREL 9233
To appear in: Journal of Controlled Release
Received date: 22 February 2018
Revised date: 28 March 2018
Accepted date: 5 April 2018
Please cite this article as: Jun Gi Rho, Hwa Seung Han, Ji Hye Han, Hansang Lee, Van Quy
Nguyen, Wang Hee Lee, Seunglee Kwon, Sungeun Heo, Juhwan Yoon, Han Ho Shin, Eun-
young Lee, Hoin Kang, Siyoung Yang, Eun Kyung Lee, Jae Hyung Park, Wook Kim , Self-
assembled hyaluronic acid nanoparticles: Implications as a nanomedicine for treatment of
type 2 diabetes. The address for the corresponding author was captured as affiliation for
all authors. Please check if appropriate. Corel(2018), doi:10.1016/j.jconrel.2018.04.006
This is a PDF file of an unedited manuscript that has been accepted for publication. As
a service to our customers we are providing this early version of the manuscript. The
manuscript will undergo copyediting, typesetting, and review of the resulting proof before
it is published in its final form. Please note that during the production process errors may
be discovered which could affect the content, and all legal disclaimers that apply to the
journal pertain.
ACCEPTED MANUSCRIPT
Jun Gi Rhoa,1 , Hwa Seung Hanb,1 , Ji Hye Hana, Hansang Leeb, Van Quy Nguyenb, Wang Hee
Leea, Seunglee Kwonb, Sungeun Heoa, Juhwan Yoona, Han Ho Shina, Eun-young Leea, Hoin
Kangc, Siyoung Yangd, Eun Kyung Leec, Jae Hyung Parkb,e,2 , and Wook Kima,2
a
Department of Molecular Science & Technology, Ajou University, Suwon 16499, Republic
of Korea.
PT
b
School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of
Korea.
c
Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul
RI
06591, Republic of Korea.
d
Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Republic
SC
of Korea.
e
Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon
16419, Republic of Korea.
NU
1
These authors contributed equally to this work.
2
To whom correspondence may be addreseed. Email: [email protected] or
[email protected].
MA
T ED
C EP
AC
ACCEPTED MANUSCRIPT
Abstract
PT
tissues (eWATs), but these were rarely observed in the eWATs of CD44-/- mice. In addition,
CD44 expression and HA-NP accumulation in the eWATs were increased in mice with diet-
induced obesity (DIO) compared to lean mice. Interestingly, treatment with HA-NPs in DIO
RI
mice suppressed adipose tissue inflammation as indicated by reduced macrophage content, the
production of proinflammatory cytokines and NLRP3 inflammasome activity in eWATs,
SC
leading to improved insulin sensitivity and normalized blood glucose levels. Collectively,
these results suggest that an empty HA-NP itself can be a therapeutic agent for the treatment
of type 2 diabetes.
NU
Keywords: self-assembled nanoparticles, hyaluronic acid, nanomedicine, inflammation,
insulin resistance, type 2 diabetes
MA
T ED
C EP
AC
ACCEPTED MANUSCRIPT
1. Introduction
Hyaluronic acid (HA) has excellent biological characteristics such as biocompatible,
biodegradable, non-toxic, and receptor-binding properties [1-4]. Accordingly, HA and its
derivatives have been investigated extensively for biomedical and pharmaceutical applications,
such as tissue engineering, arthritis treatment, ocular surgery, drug delivery, and molecular
imaging [1-5]. In particular, HA and its derivatives have emerged as the constituent of the
drug carrier for the target-specific and long-acting delivery of various chemicals and
biopharmaceuticals including proteins, peptides, and nucleotides. Also, their functional
groups at the backbone allow facile chemical modification, and this has enabled HA to be
used in various formulations to prepare the nanomedicines in the form of conjugates,
nanotubes, dendrimers, liposomes and self-assembled nanoparticles [5-9].
PT
Of the nanomedicines, the amphiphilic HA-based conjugates can be self-assembled
into nano-sized carriers for the delivery of a wide range of hydrophobic drugs as well as
imaging agents. Especially, self-assembled HA nanoparticles (HA-NPs), comprising
RI
hydrophobic cores surrounded by a hydrophilic HA shell, have been used widely as cancer-
targeting nanocarriers for therapeutic or diagnostic agents, because HA-NPs specifically bind
SC
to the CD44, the major cell-surface HA receptor overexpressed on cancer cells [6, 10-14].
Moreover, self-assembled HA-NPs have exhibited the potential as the carrier to selectively
deliver the drug to the inflamed joint of rheumatoid arthritis and to the atherosclerotic lesion
after systematic administration, due to the active targeting mechanism via CD44 highly
NU
expressed on activated macrophages [15, 16]. In addition to drug delivery, HA-NPs also have
utilized as targeted magnetic resonance (MR), positron emission tomography (PET) and
ultrasound imaging agents for biomedical applications [17-19].
MA
Interestingly, it has been recently demonstrated that CD44 on pro- inflammatory cells
in obese adipose tissue is involved in the development of adipose tissue inflammation and
insulin resistance in rodent models and human patients with type 2 diabetes (T2D) [20-23].
Therefore, it can be hypothesized that the CD44-targeting biomolecules play a role in insulin
ED
resistance and glycemic control. Here, we report the potential of self-assembled HA-NPs as a
therapeutic agent, not as a drug carrier, for the treatment of T2D. For the in vitro study, the
competitive inhibitory behavior of HA-NPs for free low molecular weight (LMW) HA, which
cause a pro-inflammatory response as an endogenous danger signal, was investigated by
T
evaluating the cellular uptake and pro-inflammatory gene expression by free LMW HA-
EP
preferentially accumulate in adipose tissue of DIO mice due to increased CD44 expression,
block LMW HA binding to CD44, and suppress LMW HA- induced pro-inflammatory
AC
2. Experimental Section
2.1 Materials
Sodium hyaluronate (MW = 235 kDa and 12 kDa) was purchased from Lifecore
Biomedical (Chaska, MN, USA). 5β-Cholanic acid (CA), Propargylamine (98%), sodium
cyanoborohydride (95%), ε-caprolactone, tin(II) 2-ethylhexanoate, sodium azide (NaN3), N-
hydroxysuccinimide (NHS), p-toluenesulfonyl chloride (p-TsCl), 1-ethyl-3(3-
(dimethylamino)propyl)-carbodiimide·hydrochloride (EDC·HCl), ethylenediamine, and
copper(II) sulfate pentahydrate (CuSO 4 •5H2 O) were obtained from Sigma-Aldrich (St. Louis,
MO, USA). Water, used for synthesis and characterization in this experiment, was purified
using an AquaMax-Ultra water purification system (Younglin Co., Anyang, South Korea). All
other chemicals were analytical grade and were used without further purification.
ACCEPTED MANUSCRIPT
PT
using a HR-TEM (JEOL-2100F, Tokyo, Japan) operated at an accelerating voltage of 200 kV.
For TEM images, samples were dispersed in deionized water (1 mg/ml) and dropped on a
200-mesh copper grid. The nanoparticles were then treated with 1 % uranyl acetate for
RI
negative staining. The size and zeta potential of the nanoparticles were determined at 25 °C
using a DLS (Nano ZS90, Malvern Instruments, Worcestershire, UK). The particle sizes were
SC
determined using the nanoparticles in PBS (pH 7.4), whereas zeta potentials were obtained
those in distilled water. 12k-HA-CA and HA-PCL conjugates were synthesized as previously
reported [10, 25]. To analyze the chemical structures using 1H-NMR, 12k-HA-CA and HA-
PCL NPs were dissolved in CD3OD/D2O (1v/1v) and DMSO-d6/D2O (1v/1v), respectively.
NU
2.3 In vivo biodistribution of HA-CA NPs.
All experiments with live animals were performed in compliance with relevant laws
MA
NPs (1mg/ml) were injected into the tail vein of male C57BL/6 mice (n=3) fed a STD or HFD
for 12 weeks. The organs and tissues were dissected from mice at pre-determined time points,
and the time-dependent biodistribution of the NPs were monitored using the eXplore Optix
system (ART Advanced Research Technologies Inc., Montreal, Canada). All data were
T
calculated using the region of interest function of the Analysis Workstation software (ART
EP
Advanced Research Technologies, Inc., Montreal, Canada), and values are presented as the
means with standard deviations for three animals. For histological analysis, the dissected
eWATs were stained as previously reported [26]. In brief, the tissues were fixed in 4%
C
formalin solution, dehydrated with a graded ethanol series, and embedded in paraffin. The
paraffin tissues were sliced into 6 µm in thickness and incubated overnight with anti-CD44
AC
antibody (BD Bioscience, CA, USA) diluted at 1:200 in a solution containing 3% BSA. After
three washes in PBS, immunofluorescence was detected following incubation with Ale xa
Fluor 488 anti- mouse antibody diluted at 1:200. The fluorescence images were obtained using
an LSM 510 META NLO confocal laser scanning microscope (Carl Zeiss Microimaging
GmbH, Jena, Germany).
2.4 Mice.
CD44-/- (C57BL/6J background) mice were obtained from The Jackson Laboratory
(Bar Harbor, ME, USA) and DBA/2 mice were obtained from Orient bio (Seongnam, South
Korea). The mice were housed individually and maintained under a 12-h light-dark cycle and
fed ad libitum (ad- lib) with a STD (NIH-31 rodent diet; 4% of calories from fat). Four-week-
old male C57BL/6J or DBA/2 mice were provided with ad- lib access to water and either STD
or a HFD (D12492, Research Diets, New Brunswick, NJ, USA; 60% of calories from fat) for
12 or 20 weeks, and were randomly allocated to each experimental group. The only criterion
ACCEPTED MANUSCRIPT
was that the mean weight was matched among the groups. Mice fed the HFD received
phosphate buffered saline (PBS) as vehicle, HA-CA, or HA-PCL NPs by daily i.v. injection,
according to two different protocols. In the first one, mice fed with a HFD for 12 weeks were
injected with 10 or 20 mg per kg body weight per day HA-CA or PBS for 4 weeks, with age-
matched lean control mice being fed with a STD. In the second protocol, mice fed with the
HFD for 20 weeks received PBS, HA-CA, or HA-PCL NPs daily for 4 weeks by i.v. injection
of 20 mg/kg, with age-matched lean control mice being fed with the STD. Body weight and
food intake were monitored daily. At the end of the treatment period, mice were subjected to
intraperitoneal (i.p.) glucose tolerance and insulin tolerance tests, as described below. Mice
were then euthanized using CO2 gas, and their eWAT, liver, and blood were collected to
determine endocrine and biochemical parameters, macrophage contents, and pro-
PT
inflammatory gene expression.
RI
Blood glucose levels were determined using an AGM 4000 glucometer (Allmedicus,
Anyang, South Korea). Plasma insulin was measured using a Mouse Insulin ELISA Kit
SC
(ALPCO, Salem, NH, USA). HOMA-IR in the mice was calculated using the following
equation: (fasting glucose concentration × fasting insulin concentration)/405. Intrahepatic
triglyceride (TG) content was determined on liver extracts using a serum TG determination kit
(Sigma-Aldrich), according to the manufacturer’s instructions.
NU
2.6 Intraperitoneal glucose tolerance test (IPGTT) and insulin tolerance tests (ITT).
Mice were fasted overnight and then injected with glucose (2 g/kg, i.p.), followed by
MA
tail blood collection at 0, 15, 30, 60, 90, and 120 minutes. Blood glucose levels were
determined using the AGM-4000 glucometer (Allmedicus). For the insulin tolerance tests,
mice were fasted for 4 h and then i.p. injected with 0.75 U/kg insulin (Sigma-Aldrich); blood
glucose levels were determined at 0, 15, 30, 60, 90, and 120 minutes.
ED
measured using a Mouse IL-1/IL-1F2 Immunoassay kit (R&D systems). NLRP3, PYCARD,
EP
caspase-1, IL-1β, and IL-18 mRNA levels in the eWAT were measured by quantitative real-
time PCR analysis, as described below.
C
embedded in paraffin, and sectioned onto glass slides. Tissue slides were stained with
hematoxylin-eosin (H&E) to evaluate the tissue morphology, following standardized
protocols. To evaluate lipid droplets in the liver, frozen liver sections were stained with Oil
Red O and counterstained with hematoxylin. Macrophage infiltration into the eWAT was
evaluated by immunohistochemistry (IHC) using primary antibodies recognizing CD68. The
NLRP3 inflammasome in the eWAT was evaluated by IHC using primary antibodies
recognizing NLRP3 and ASC. Information on the antibodies used for immunostaining is
listed in Table S3. Each bound antibody was revealed using the appropriate Elite ABC
horseradish peroxidase (HRP)-diaminobenzediene (DAB) system (Vector Labs, Burlingame,
CA, USA). Slides were viewed using a Leica DMi8 microscope (Leica, Buffalo Grove, IL,
USA).
Total RNAs were isolated from frozen mouse tissues or cell lines using the TRIzol
reagent (Thermo Fisher Scientific, Waltham, MA, USA), according to the manufacturer’s
protocol. All extractions were followed by DNase I treatment (Invitrogen). After reverse
transcription (RT) using random hexamers and reverse transcriptase (Toyobo, Osaka, Japan),
the mRNA abundance was assessed using a CFX ConnectTM Real- Time PCR Detection
System (Bio-rad, Hercules, CA, USA) and the SYBR green PCR master mix (Kapa
Biosystems, Wilmington, MA, USA). The 18S or GAPDH gene was used for normalization
to quantify the relative mRNA expression levels. The gene-specific primer sets are listed in
Table S4.
PT
BMDMs were obtained from male C57BL/6 CD44+/+ and CD44-/- mice by flushing
the tibia with PBS. The cells (2 x 106 cells/well) were cultured in 6-well plate with a RPMI
1640 medium (Gibco, Grand Island, NY) containing 10 % heat- inactivated fetal bovine serum
RI
(FBS), 1 % penicillin/streptomycin and 10 μg/ml M-CSF at 37 ˚C in a humidified 5% CO 2
atmosphere. The cells were then incubated for 7 days with medium change every 3 days. For
competitive inhibition studies with free HA (235 kDa) and HA-CA NPs, BMDMs (1 x 105
SC
cells/well) were seeded in 6-well plates and incubated for 24 h. The cells were pre-treated
with 2 ml of HA-CA NPs or HA-PCL NPs (0.5, 1, or 5 mg/ml) for 60 min. Subsequently,
without the removal of the NPs, Cy5.5-labeled free HA (100 μg/ml) was immediately treated
NU
in the cells, followed by incubation for an additional 30 min. The cells were then washed
twice with PBS (pH 7.4) and fixed with a 4% paraformaldehyde solution. The intracellular
localization of Cy5.5- labeled free HA was observed using confocal microscopy. To evaluate
MA
the effects of free HA and HA-CA NPs on pro- inflammatory gene expression, RAW264.7
macrophages (1 x 105 cells/well) and BMDMs (1 x 106 cells/well) were seeded in 6-well
plates for 24 h and washed twice with PBS (p H 7.4). The cells were then treated for 3 h with
free HA (50, 100, 200 and 500 μg/ml) or HA-CA NPs (50, 100, 200 and 500 μg/ml), or with
free HA (100 or 500 μg/ml) in the absence or presence of HA-CA NPs (100 μg/ml) in the
ED
media (1 ml/well) without FBS. Cells were washed twice in ice-cold sterile PBS before
harvesting. Pro- inflammatory gene expression was determined using quantitative real-time
PCR analysis, as described above.
T
EP
using GraphPad Prism (GraphPad Software). A P value of < 0.05 was considered statistically
significant.
AC
PT
RI
SC
NU
Fig 1. Characteristics of self-assembled HA-CA NPs. (A) Schematic illustration of HA-CA
MA
NPs for treatment of type 2 diabetes. (B) TEM images and size distribution of HA-CA NPs.
Scale bar, 500 nm (Inset’ 50 nm). (C). Fluorescence images of major organs and adipose
tissues after intravenous administration of Cy5.5-HA-CA NPs at predetermined time intervals.
Li: Liver, Lu: Lung, Sp: Spleen, Ki: Kidney, He: Heart, Pa: Pancreas, eWAT: Epididymal
ED
adipose tissue, sWAT: Subcutaneous adipose tissue; BAT: Brown adipose tissue. (D) Mean
fluorescence intensities of Cy5.5-HA-CA NPs. Data represent mean ± SD (n = 3 mice per
group). (E) Confocal microscopic images of CD44 and Cy5.5-HA-CA NP in the eWAT of
T
the in vivo biodistribution was non- invasively monitored for 24 h using a real-time near-
infrared fluorescence (NIRF) imaging technique (Fig. 1C and Fig. S3A). For DIO mice fed a
AC
HFD, strong NIRF signals were found in the liver and kidney, which might be due to uptake
by the reticuloendothelial system and the renal excretion, respectively [27-29]. Interestingly,
considerable signal was observed for 24 h in all adipose tiss ue, in which epididymal white
adipose tissue (eWAT) exhibited stronger signal, compared to other tissues or organs such as
subcutaneous WAT (sWAT), brown adipose tissue (BAT), lung, spleen, heart, and pancreas
(Fig. 1C and D). This result was further supported by the histological analysis in DIO mice, as
indicated by strong fluorescent signals in the eWAT due to the presence of Cy5.5-HA-CA
NPs (Fig. 1E). The fluorescent signals in the eWAT were increased by HFD feeding and,
conversely, it was largely reduced in CD44- nullt (CD44-/-) mice (Fig. S3A). The
immunohistochemical analysis further confirmed that the fluorescent signals of Cy5.5-HA-
CA NPs and CD44 in the eWAT of HFD- fed wild-type (CD44+/+) DIO mice were much
stronger than those in the eWAT of STD-fed CD44+/+ mice, but both signals were rarely
detected in the eWAT of STD- fed CD44-/- mice (Fig. S3B). Accordingly, the co- localization
of HA-CA NPs with CD44 was obviously detected in HFD- fed CD44+/+ DIO mice and to a
ACCEPTED MANUSCRIPT
somewhat lesser degree in STD- fed CD44+/+ mice, but not in STD-fed CD44-/- mice. Given
that CD44 expression was largely induced by the HFD feeding [20, 21, 23], these results
suggest that the interaction between HA-CA NPs and CD44 could be responsible for the
effective targeting of the HA-CA NPs in the eWAT.
PT
Cy5.5-labeled free HA in the absence of HA-CA NPs. However, the cellular uptake of Cy5.5-
labeled free HA decreased in a dose-dependent manner when the cells were pretreated with an
excess amount of HA-CA NPs. In addition, very weak fluorescence intensities of Cy5.5 were
RI
detected when CD44-/- BMDMs were treated with free HA and HA-CA NPs. These results
imply that HA-CA NP and free HA compete for binding to CD44. As LMW free HA-
SC
dependent CD44 signaling induces the expression of pro- inflammatory cytokines and CD44
[21, 32], we compared the effect of LMW free HA on the expression of pro-inflammatory
genes and CD44 in the absence or presence of HA-CA NPs using CD44+/+ and CD44-/-
BMDMs. We found that the expression levels of CD44, TNF-α, NLRP3, and IL-1β were
NU
increased by LMW free HA, but not by HA-CA NP, in CD44+/+ BMDMs, whereas these
LMW free HA- induced expressions were inhibited by pre-treatment with HA-CA NP (Fig.
2B). Moreover, LMW free HA-induced expression of the pro- inflammatory genes was not
MA
observed in CD44-/- BMDMs. Taken together, it is likely that HA-CA NP inhibits pro-
inflammatory function of LMW free HA by altering the LMW free HA-binding capacity of
CD44 as a competitive inhibitor.
T ED
C EP
AC
Fig. 2. In vitro competitive inhibition assay using LMW free HA in the absence or presence
of HA-CA NPs. (A) Confocal images of CD44+/+ and CD44-/- BMDMs incubated with
Cy5.5-labeled free HA in the absence or presence of HA-CA NPs. BMDMs were treated with
HA-CAs (0.5, 1, or 5 mg/ml) for 60 min, followed by incubation with Cy5.5-labeled free HA
(100 μg/ml) for 30 min. Scale bars, 50 μm. (B) Pro- inflammatory gene expression in
ACCEPTED MANUSCRIPT
CD44+/+ and CD44-/- BMDMs treated with free HA and/or HA-CA NP. BMDMs were
treated for 3 h with free HA (500 μg/ml) in the absence or presence of HA-CA NPs (100
μg/ml). Data represent mean ± SEM from at least three indep endent experiments. * P < 0.05,
**
P < 0.01 versus PBS by two-tailed Student’s t test.
3.4 Self-Assembled HA-CA NPs Reduce Body Weight and Improve the Impaired
Glucose Tolerance and Insulin Sensitivity.
It has been recently demonstrated that LMW HA and CD44 are involved in the
pathogenesis of T2D [20-22, 33, 34]. In particular, both LMW HA and CD44 levels in DIO
mice are positively correlated with insulin resistance and glycemic control [20-23, 33, 34].
Thus, these studies have proposed that interrupting LMW HA-CD44 interaction could have
PT
therapeutic benefits for the treatment of T2D. Indeed, blocking LMW HA-CD44 interaction
in DIO mice by recombinant hyaluronidase PH20, CD44 deficiency or anti-CD44 antibody
treatment reversed HFD- induced insulin resistance and glycemic control [20, 21, 23, 33, 34].
RI
As HA-CA NPs alter the LMW free HA-binding capacity of CD44 as a competitive inhibitor,
we tested whether HA-CA NPs cause metabolic effects in DIO mice. We first performed daily
SC
intravenous (i.v.) injections of HA-CA NPs at 10 or 20 mg/kg/day for 4 weeks in DIO mice
fed a HFD for 12 weeks. At the end of the treatment period, we observed that injection of 20
mg/kg HA-CA NP caused a reduction in body weight, eWAT mass, fasting blood glucose,
and the homeostatic model assessment of insulin resistance (HOMA-IR) index (Fig. S4A-D).
NU
HA-CA NP at a dose of 20 mg/kg/day also normalized the impaired glucose tolerance and
improved insulin sensitivity (Fig. S4E and F). These effects were not observed at a dose of 10
mg/kg (Fig. S4A-F). To more achieve the full spectrum of T2D symptoms, especially
MA
inflammation and insulin resistance, and further confirm the metabolic effects of HA-CA NPs,
we performed daily i.v. injections of HA-CA NPs at 20 mg/kg for 4 weeks in DIO mice fed
HFD for 20 weeks and evaluated the metabolic parameters at the end of the treatment period.
Consistent with the results in Fig. S4, HA-CA NPs treatment caused statistically significant
ED
reductions in body weight and food intake (Fig. 3A-C). HA-CA NPs were also effective in
normalizing fasting blood glucose levels, and attenuating glucose intolerance and insulin
resistance (Fig. 3D-F). These results suggest that HA-CA NPs could be used to interrupt
LMW HA-CD44 interaction, leading to amelioration of insulin sensitivity and glycemic
T
control.
C EP
AC
ACCEPTED MANUSCRIPT
PT
RI
SC
Fig. 3. Effects of self-assembled HA-CA NP treatment on body weight, glucose tolerance,
and insulin resistance in DIO mice. (A–C) Effects of HA-CA NP treatment (20 mg/kg) in DIO
NU
mice on body weight (A), weight change (B), and food intake (C) compared with PBS-treated
controls. Data represent the mean ± SEM. n = 3 mice per group for the standard chow diet
(STD); n = 5 mice per group for HFD. *P < 0.05, **P < 0.01 versus HFD-PBS group by two-
tailed Student’s t test for bar graphs and two-way ANOVA for comparison of multiple time
MA
points in line graphs. (D–F) Effects of HA-CA NP treatment (20 mg/kg) in the same DIO
mice on fasting blood glucose (D) along with IPGTT (E) and ITT (F). Data represent the
mean ± SEM. n = 3 mice per group for the STD; n = 4–7 mice per group for HFD. †P < 0.05,
††
P < 0.01 versus STD group; *P < 0.05, **P < 0.01 versus HFD-PBS group by two-tailed
ED
Student’s t test.
(epididymal) adipose tissue inflammation and insulin resistance in rodents and humans. CD44
AC
is expressed on inflammatory cells of obese adipose tissue, and CD44 levels are positively
correlated with adipose tissue inflammation, insulin resistance, and glycemic control in both
humans and rodents [20-23]. Accordingly, CD44-/- mice exhibited a significantly improved
adipose tissue inflammation and insulin sensitivity in DIO mice, and anti-CD44 antibody
suppressed adipose tissue inflammation and reduced insulin resistance and hyperglycemia [20,
21, 23]. Given that HA-CA NPs suppress pro- inflammatory function of LMW free HAs by
altering the LMW free HA-binding capacity of CD44 (Fig. 2), we investigated their effects on
eWAT inflammation. In DIO mice, the accumulation of CD68 + macrophages forming crown-
like structures (CLSs) surrounding adipocytes in eWAT was observed frequently, as also
indicated by higher CD68 expression compared with that in STD lean littermates (Fig. 4A).
However, HA-CA NP treatment reduced macrophage infiltration and the e xpressions of CD68.
Consistent with previous reports [20-23], an HFD feeding led to accumulation of CD44 +
inflammatory cells and increased expression of CD44 in the eWAT compared with the STD
lean littermates, suggesting that most infiltrating cells in the CLS are macrophages and are
ACCEPTED MANUSCRIPT
positive for CD44 (Fig. 4A). This increase was also reduced by treatment with HA-CA NPs.
We also found that the mRNA expression levels of NF-κB, MCP-1 and TNF-α were
normalized by HA-CA NP treatment (Fig. 4B-D).
PT
RI
SC
NU
MA
ED
treated DIO mice. Scale bars, 250 μm. Boxed areas were magnified, shown at the bottom, for
EP
better visualization. CD68 and CD44 mRNA levels measured in eWATs are shown at the
bottom. (B) Expression of NF-κB mRNA in the same eWATs as in (A). (C and D) Pro-
inflammatory gene expression (MCP-1 and TNF-α) in the same eWATs as in (A). Data
represent the mean ± SEM. n = 3 mice for STD; n = 3–7 per group for HFD. †P < 0.05, ††P <
C
0.01 versus STD group; * P < 0.05, ** P < 0.01 versus HFD-PBS group by two-tailed Student’s
AC
t test.
higher caspase-1 activity in the eWAT, in conjunction with higher IL-1β levels in both the
eWAT (Fig. 5D and E) and blood (Fig. S5). In contrast, the same parameters in HA-CA NP-
treated DIO mice were similar to or only slightly different from their levels in the STD lean
littermates (Fig. 5A-E and Fig. S5). These results indicate that HA-CA NPs have therapeutic
effects on adipose tissue inflammation and insulin resistance by suppressing the activation of
NLRP3 inflammasome.
PT
RI
SC
NU
MA
ED
NLRP3 (A) and ASC (B) in eWATs from lean and HA-CA NP-treated DIO mice. Scale bars,
250 μm. NLRP3 and PYCARD mRNA levels measured in eWATs are shown on the right.
EP
(C–E) Caspase-1, IL-1β, and IL-18 mRNA levels (C) as well as caspase-1 activity (D) and IL-
1β protein (E) in the same eWATs as in (A). n = 3 mice for STD; n = 3–7 mice per group for
HFD. Data represent the mean ± SEM. †P < 0.05, ††P < 0.01 versus STD group; * P < 0.05, ** P
C
3.7 Other Self-Assembled HA-NP, HA-PCL NP, Has Similar Effects with HA-CA NP on
Adipose Tissue Inflammation and Insulin Resistance.
Hydrophobic CA used in HA-CA NP synthesis is a bile acid derivative [45]. Bile acids
and their derivatives play critical roles in the regulation of glucose and energy metabolism,
and inflammation [46]. In addition, pro-inflammatory response can be affected by the MW of
HA in the conjugate [32, 47]. To confirm the effect of the hydrophobic moiety and MW of
HA, we have prepared two more HA-NPs, consisting of HA (MW = 12 kDa) as the
hydrophilic shell and CA or polycaprolactone (PCL) as the hydrophobic core (Fig. S1, S6A,
and S7A). The chemical structures of 12k-HA-CA and HA-PCL NPs were analyzed using 1 H-
NMR (Fig. S2A and B), and TEM images indicated that they are spherical in shape (Fig. S6B
and S7B). Compare to HA-CA NP, 12k-HA-CA and HA-PCL NPs had similar particle sizes
with slightly lower zeta potential values (Tables S1 and S2). In addition, 12k-HA-CA and
HA-PCL NPs showed similar effects with HA-CA NP all in vitro experiments. Briefly, in
vitro competitive inhibition assay showed that both the NPs and LMW free HA compete for
ACCEPTED MANUSCRIPT
binding to CD44 (Fig. S6C and S7C). Similar to HA-CA NP, 12k-HA-CA NP suppressed
LMW free HA- induced expressions of CD44, TNF-α, and NLRP3 in RAW264.7
macrophages (Fig. S6D). For in vivo experiment using DIO mice, HA-PCL NP significantly
reduced body weight and food intake, normalized fasting blood glucose, and attenuated
glucose intolerance and insulin resistance (Fig. S8A-F). HA-PCL NPs also reversed
macrophage infiltration into eWATs and suppressed pro-inflammatory gene expression and
NLRP3 inflammasome activation in DIO mice (Fig. S9 and S10). These results imply that,
irrespective of hydrophobic constituents (CA and PCL) and the molecular weights of free HA
(12 kDa and 235 kDa), the therapeutic effects of HA-CA NPs on adipose tissue inflammation,
glycemic control and insulin resistance are attributed to the presence of self- assembled
hydrophilic HA shell with multiple binding sites to CD44.
PT
In normal tissue, the most widely distributed native form of HA is a high- molecular-
weight (HMW, MW > 1000 kDa) polymer present in all tissues and body fluid [1, 48]. Under
pathological conditions, including inflammation and tissue injury, HMW HA undergoes
RI
dynamic degradation, resulting in accumulation of LMW species (MW < 500 kDa), which
trigger and promote a pro- inflammatory response as an endogenous danger signal [1, 48-51].
SC
In contrast, HMW HAs exert protective anti- inflammatory effects through reducing gene
expression and synthesis of pro- inflammatory cytokines [1, 48-51]. These differential
biological activities of HMW and LMW HAs are likely due to the ability of HA to induce
CD44 clustering on cell surface. Recent studies reported that CD44 has two different
NU
conformational forms and HA size-dependent conformational switching between the low- and
high-affinity binding states induce clustering of CD44 [52-55]. In contrast to a LMW HA, a
HMW HA binds simultaneously to many CD44 receptors through multivalent or repeated
MA
assembled HA-NP exerts anti- inflammatory effects through the same mechanism with a
HMW HA (Fig. 1A).
4. Conclusions
T
In summary, we report a novel therapeutic role of empty HA-NPs without any drug in
EP
adipose tissue inflammation, insulin resistance and glycemic control. The in vitro studies
demonstrated that HA-NPs effectively suppressed the LMW free HA- induced expressions of
pro-inflammatory mediators by acting as a competitive inhibitor of HA-CD44 interactions in
C
macrophages. These results suggest that HA-NPs can exert anti- inflammatory effects by
interrupting the LMW HA interaction with CD44. Indeed, treatment with HA-NPs in DIO
AC
mice inhibited macrophage infiltration into adipose tissue and production of pro-inflammatory
cytokines, leading to an attenuated adipose tissue inflammation, and normalized fasting blood
glucose levels and insulin sensitivity. Therefore, our results identify novel function of a self-
assembled HA-NP, which could be a therapeutic agent for the treatment of T2D by breaking
the links between adipose tissue inflammation and insulin resistance. Ultimately, we can
expect that self-assembled HA-NP loaded with specific drugs might further improve the
therapeutic outcome, exert simultaneous multiple effects or reduce side effects.
Acknowledgements
This work was supported by the Basic Science Research Program through the National
Research Foundation of Korea (NRF), funded by the Ministry of Science, ICT & Future
(2016R1E1A1A01941213, 2015R1A2A2A05001390, and 20100027955).
ACCEPTED MANUSCRIPT
References
[1] D. Jiang, J. Liang, P.W. Noble, Hyaluronan as an Immune Regulator in Human Diseases,
Physiol. Rev. 91 (2011) 221-264.
[2] R. Duncan, The dawning era of polymer therapeutics, Nat. Rev. Drug Discov. 2 (2003)
347-360.
[3] V.P. Torchilin, Nanocarriers, Pharm. Res. 24 (2007) 2333-2334.
[4] Y. Zhu, C. Crewe, P.E. Scherer, Hyaluronan in adipose tissue: Beyond dermal filler and
therapeutic carrier, Sci. Transl. Med. 8 (2016) 323ps324-323ps324.
[5] E.J. Oh, K. Park, K.S. Kim, J. Kim, J.-A. Yang, et al., Target specific and long-acting
delivery of protein, peptide, and nucleotide therapeutics using hyaluronic acid derivatives, J.
Control. Release 141 (2010) 2-12.
PT
[6] N.V. Rao, H.Y. Yoon, H.S. Han, H. Ko, S. Son, et al., Recent developments in hyaluronic
acid-based nanomedicine for targeted cancer treatment, Expert Opin. Drug Deliv. 13 (2016)
239-252.
RI
[7] H.-J. Cho, I.-S. Yoon, H.Y. Yoon, H. Koo, Y.-J. Jin, et al., Polyethylene glycol-conjugated
hyaluronic acid-ceramide self-assembled nanoparticles for targeted delivery of doxorubicin,
SC
Biomaterials 33 (2012) 1190-1200.
[8] R.R. Wakaskar, Promising effects of nanomedicine in cancer drug delivery, J. Drug Target.
26 (2018) 319-324.
[9] R.R. Wakaskar, General overview of lipid–polymer hybrid nanoparticles, dendrimers,
NU
micelles, liposomes, spongosomes and cubosomes, J. Drug Target. 26 (2018) 311-318.
[10] K.Y. Choi, H. Chung, K.H. Min, H.Y. Yoon, K. Kim, et al., Self-assembled hyaluronic
acid nanoparticles for active tumor targeting, Biomaterials 31 (2010) 106-114.
MA
[11] G. Liu, K.Y. Choi, A. Bhirde, M. Swierczewska, J. Yin, et al., Sticky Nanoparticles: A
Platform for siRNA Delivery by a Bis(zinc(II) dipicolylamine)-Functionalized, Self-
Assembled Nanoconjugate, Angew. Chem.-Int. Edit. 51 (2012) 445-449.
[12] M. Swierczewska, K.Y. Choi, E.L. Mertz, X. Huang, F. Zhang, et al., A Facile, One-Step
ED
[14] H.S. Han, K.Y. Choi, H. Ko, J. Jeon, G. Saravanakumar, et al., Bioreducible core-
EP
crosslinked hyaluronic acid micelle for targeted cancer therapy, J. Control. Release 200
(2015) 158-166.
[15] R. Heo, J.-S. Park, H.J. Jang, S.-H. Kim, J.M. Shin, et al., Hyaluronan nanoparticles
C
[16] G.Y. Lee, J.-H. Kim, K.Y. Choi, H.Y. Yoon, K. Kim, et al., Hyaluronic acid nanoparticles
for active targeting atherosclerosis, Biomaterials 53 (2015) 341-348.
[17] H.S. Min, S. Son, T.W. Lee, H. Koo, H.Y. Yoon, et al., Liver‐ Specific and Echogenic
Hyaluronic Acid Nanoparticles Facilitating Liver Cancer Discrimination, Adv. Funct. Mater.
23 (2013) 5518-5529.
[18] L. Zhang, S. Gao, F. Zhang, K. Yang, Q. Ma, et al., Activatable Hyaluronic Acid
Nanoparticle as a Theranostic Agent for Optical/Photoacoustic Image-Guided Photothermal
Therapy, ACS Nano 8 (2014) 12250-12258.
[19] M. Kamat, K. El-Boubbou, D.C. Zhu, T. Lansdell, X. Lu, et al., Hyaluronic Acid
Immobilized Magnetic Nanoparticles for Active Targeting and Imaging of Macrophages,
Bioconjugate Chem. 21 (2010) 2128-2135.
[20] K. Kodama, M. Horikoshi, K. Toda, S. Yamada, K. Hara, et al., Expression-based
genome-wide association study links the receptor CD44 in adipose tissue with type 2 diabetes,
Proc. Natl. Acad. Sci. U.S.A. 109 (2012) 7049-7054.
ACCEPTED MANUSCRIPT
PT
vivo biodistribution, J. Mater. Chem. 19 (2009) 4102-4107.
[25] H.S. Han, T. Thambi, K.Y. Choi, S. Son, H. Ko, et al., Bioreducible Shell-Cross- Linked
Hyaluronic Acid Nanoparticles for Tumor-Targeted Drug Delivery, Biomacromolecules 16
RI
(2015) 447-456.
[26] Isabelle K. Vila, P.-M. Badin, M.-A. Marques, L. Monbrun, C. Lefort, et al., Immune
SC
Cell Toll- like Receptor 4 Mediates the Development of Obesity- and Endotoxemia-Associated
Adipose Tissue Fibrosis, Cell Reports 7 (2014) 1116-1129,
https://doi.org/10.1016/j.celrep.2014.03.062.
[27] V.M. Platt, F.C. Szoka, Anticancer Therapeutics: Targeting Macromolecules and
NU
Nanocarriers to Hyaluronan or CD44, a Hyaluronan Receptor, Mol. Pharm. 5 (2008) 474-486.
[28] B. Zhou, J.A. Weigel, L. Fauss, P.H. Weigel, Identification of the Hyaluronan Receptor
for Endocytosis (HARE), J. Biol. Chem. 275 (2000) 37733-37741.
MA
[29] R. Masereeuw, F.G.M. Russel, Mechanisms and clinical implications of renal drug
excretion*, Drug Metab. Rev. 33 (2001) 299-351.
[30] Y. Zhong, J. Zhang, R. Cheng, C. Deng, F. Meng, et al., Reversibly crosslinked
hyaluronic acid nanoparticles for active targeting and intelligent delivery of doxorubicin to
ED
drug resistant CD44+ human breast tumor xenografts, J. Control. Release 205 (2015) 144-154.
[31] D.-S. Liang, W.-J. Zhang, A.-T. Wang, H.-T. Su, H.-J. Zhong, et al., Treating metastatic
triple negative breast cancer with CD44/neuropilin dual molecular targets of multifunctional
nanoparticles, Biomaterials 137 (2017) 23-36.
T
[32] A.R. Jordan, R.R. Racine, M.J.P. Hennig, V.B. Lokeshwar, The Role of CD44 in Disease
EP
[34] L. Kang, L. Lantier, A. Kennedy, J.S. Bonner, W.H. Mayes, et al., Hyaluronan
Accumulates With High-Fat Feeding and Contributes to Insulin Resistance, Diabetes 62
AC
(2013) 1888-1896.
[35] G.S. Hotamisligil, Inflammation and metabolic disorders, Nature 444 (2006) 860-867.
[36] M.Y. Donath, S.E. Shoelson, Type 2 diabetes as an inflammatory disease, Nat. Rev.
Immunol. 11 (2011) 98-107.
[37] K. Schroder, J. Tschopp, The Inflammasomes, Cell 140 821-832.
[38] J.J. Shin, E.K. Lee, T.J. Park, W. Kim, Damage-associated molecular patterns and their
pathological relevance in diabetes mellitus, Ageing Res. Rev. 24 (2015) 66-76.
[39] L. Agostini, F. Martinon, K. Burns, M.F. McDermott, P.N. Hawkins, et al., NALP3
Forms an IL-1β-Processing Inflammasome with Increased Activity in Muckle-Wells
Autoinflammatory Disorder, Immunity 20 (2004) 319-325.
[40] K. Schroder, R. Zhou, J. Tschopp, The NLRP3 Inflammasome: A Sensor for Metabolic
Danger?, Science 327 (2010) 296-300.
[41] R. Stienstra, L.A.B. Joosten, T. Koenen, B. van Tits, J.A. van Diepen, et al., The
Inflammasome-Mediated Caspase-1 Activation Controls Adipocyte Differentiation and
ACCEPTED MANUSCRIPT
PT
Expression, J. Pharmacol. Sci. 107 (2008) 285-294.
[46] T. Li, J.Y.L. Chiang, Bile Acid Signaling in Metabolic Disease a nd Drug Therapy,
Pharmacol. Rev. 66 (2014) 948-983.
RI
[47] S. Misra, V.C. Hascall, R.R. Markwald, S. Ghatak, Interactions between Hyaluronan and
Its Receptors (CD44, RHAMM) Regulate the Activities of Inflammation and Cancer, Front.
SC
Immunol. 6 (2015).
[48] T.C. Laurent, J.R. Fraser, Hyaluronan, The FASEB J. 6 (1992) 2397-2404.
[49] P. Teder, R.W. Vandivier, D. Jiang, J. Liang, L. Cohn, et al., Resolution of Lung
Inflammation by CD44, Science 296 (2002) 155-158.
NU
[50] C.M. McKee, M.B. Penno, M. Cowman, M.D. Burdick, R.M. Strieter, et al., Hyaluronan
(HA) fragments induce chemokine gene expression in alveolar macrophages. The role of HA
size and CD44, J. Clin. Invest. 98 (1996) 2403-2413, http://doi.org/10.1172/JCI119054.
MA
[51] D. Jiang, J. Liang, J. Fan, S. Yu, S. Chen, et al., Regulation of lung injury and repair by
Toll-like receptors and hyaluronan, Nat. Med. 11 (2005) 1173-1179.
[52] R. Stern, A.A. Asari, K.N. Sugahara, Hyaluronan fragments: An information-rich system,
Eur. J. Cell Biol. 85 (2006) 699-715.
ED
[53] S. Banerji, A.J. Wright, M. Noble, D.J. Mahoney, I.D. Campbell, et al., Structures of the
Cd44-hyaluronan complex provide insight into a fundamental carbohydrate-protein
interaction, Nat. Struct. Mol. Biol. 14 (2007) 234-239.
[54] C. Yang, M. Cao, H. Liu, Y. He, J. Xu, et al., The High and Low Molecular Weight
T
Forms of Hyaluronan Have Distinct Effects on CD44 Clustering, J. Biol. Chem. 287 (2012)
EP
43094-43107.
[55] A.C. Petrey, C.A. de la Motte, Hyaluronan, a Crucial Regulator of Inflammation, Front.
Immunol. 5 (2014) 101.
C
[56] P.M. Wolny, S. Banerji, C. Gounou, A.R. Brisson, A.J. Day, et al., Analysis of CD44-
Hyaluronan Interactions in an Artificial Membrane System: INSIGHTS INTO THE
AC
Figure caption
PT
Fig. 2. In vitro competitive inhibition assay using LMW free HA in the absence or presence
of HA-CA NPs. (A) Confocal images of CD44+/+ and CD44-/- BMDMs incubated with
RI
Cy5.5-labeled free HA in the absence or presence of HA-CA NPs. BMDMs were treated with
HA-CAs (0.5, 1, or 5 mg/ml) for 60 min, followed by incubation with Cy5.5-labeled free HA
(100 μg/ml) for 30 min. Scale bars, 50 μm. (B) Pro-inflammatory gene expression in
SC
CD44+/+ and CD44-/- BMDMs treated with free HA and/or HA-CA NP. BMDMs were
treated for 3 h with free HA (500 μg/ml) in the absence or presence of HA-CA NPs (100
μg/ml). Data represent mean ± SEM from at least three independent experiments. * P < 0.05,
NU
**
P < 0.01 versus PBS by two-tailed Student’s t test.
and insulin resistance in DIO mice. (A–C) Effects of HA-CA NP treatment (20 mg/kg) in DIO
mice on body weight (A), weight change (B), and food intake (C) compared with PBS-treated
controls. Data represent the mean ± SEM. n = 3 mice per group for the standard chow diet
(STD); n = 5 mice per group for HFD. *P < 0.05, **P < 0.01 versus HFD-PBS group by two-
tailed Student’s t test for bar graphs and two-way ANOVA for comparison of multiple time
ED
points in line graphs. (D–F) Effects of HA-CA NP treatment (20 mg/kg) in the same DIO
mice on fasting blood glucose (D) along with IPGTT (E) and ITT (F). Data represent the
mean ± SEM. n = 3 mice per group for the STD; n = 4–7 mice per group for HFD. †P < 0.05,
T
††
P < 0.01 versus STD group; *P < 0.05, **P < 0.01 versus HFD-PBS group by two-tailed
EP
Student’s t test.
DIO mice. (A) Representative eWAT H&E staining and macrophage content determined by
immunohistochemical staining for CD68 and CD44 in eWATs from lean and HA-CA NP-
AC
treated DIO mice. Scale bars, 250 μm. Boxed areas were magnified, shown at the bottom, for
better visualization. CD68 and CD44 mRNA levels measured in eWATs are shown at the
bottom. (B) Expression of NF-κB mRNA in the same eWATs as in (A). (C and D) Pro-
inflammatory gene expression (MCP-1 and TNF-α) in the same eWATs as in (A). Data
represent the mean ± SEM. n = 3 mice for STD; n = 3–7 per group for HFD. †P < 0.05, ††P <
0.01 versus STD group; * P < 0.05, ** P < 0.01 versus HFD-PBS group by two-tailed Student’s
t test.
HFD. Data represent the mean ± SEM. †P < 0.05, ††P < 0.01 versus STD group; * P < 0.05, ** P
< 0.01 versus HFD-PBS group by two-tailed Student’s t test.
PT
RI
SC
NU
MA
T ED
C EP
AC
ACCEPTED MANUSCRIPT
Graphical abstract
PT
RI
SC
NU
MA
ED
T
C EP
AC