You are on page 1of 16

Biomaterials 264 (2021) 120390

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Targeted silver nanoparticles for rheumatoid arthritis therapy via


macrophage apoptosis and Re-polarization
Yihua Yang a, b, 1, Lina Guo a, 1, Zhe Wang a, Peng Liu a, Xuanjun Liu a, Jinsong Ding a,
Wenhu Zhou a, *
a
Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
b
Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmaceutical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China

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

Keywords: Infiltration of inflammatory cells, especially the M1 macrophages that secrete various types of inflammation
Silver nanoparticles cytokines, play crucial roles in the pathogenesis of rheumatoid arthritis (RA). To relief synovial inflammation,
ROS scavenge M1 macrophages must be eliminated or switched to anti-inflammatory M2 phenotype. We herein developed folic
Macrophage polarization
acid modified silver nanoparticles (FA-AgNPs) that can actively deliver into M1 macrophages to synergistically
Rheumatoid arthritis
induce M1 macrophages reduction and M2 macrophages polarization for effective RA treatment. The AgNPs was
Targeted therapy
Nanomedicine facilely prepared, PEGylated and modified with FA to realize M1 macrophages targeting delivery via folate re­
ceptor overexpressed on M1 macrophages surface. After entering cells, FA-AgNPs dissolved and released Ag+ in
response to intracellular glutathione (GSH), which is the key element to exert a series of anti-inflammatory
functions, such as M1 macrophages apoptosis and reactive oxygen species (ROS) scavenging to facilitate M2
macrophages polarization, both of which contributed to RA treatment. This nano-system could passively accu­
mulate into inflamed joints, permit potent anti-inflammatory activity, and impose strong therapeutic efficacy in
mice RA models with high biosafety. After treatment, FA-AgNPs could be gradually cleared from the body mainly
via feces without tissue accumulation, and did not show any appreciable long-term toxicity. This work declares
the first example of using bio-active nanoparticles for RA treatment without loading any drugs, and highlights
the potential of FA-AgNPs for targeted RA therapy via simultaneous M1 macrophage apoptosis and M1-to-M2
macrophages re-polarization.

1. Introduction expected [7], accompanied by serious risks such as tuberculosis reac­


tivation and serious infections [8]. Therefore, novel therapeutic strate­
Rheumatoid arthritis (RA) is a type of chronic auto-immune disorder gies for RA treatment are still urgently warranted.
that affects ~1% of the adult population [1]. The disease is character­ Recent years have witnessed a surge in the development of nano­
ized by progressive inflammation and persistent synovitis, leading to medicine for combating RA, with the purpose of bypassing the defects of
bone and cartilage destruction, functional incapability, and ultimately the current treatments. Due to the abnormal vessels and inflammatory
disability [2,3]. Currently, RA is still an incurable disease, and there are cell infiltration, the vascular permeability is remarkably improved in RA
three classes of medications commonly used in clinic, i.e., non-steroidal site, which allows for the passive accumulation of nanocarriers through
anti-inflammatory drugs (NSAIDs), glucocorticoids (GCs), and so-called “ELVIS” effect (extravasation through leaky vasculature and
disease-modifying anti-rheumatic drugs (DMARDs). While they can subsequent inflammatory cell-mediated sequestration) [9], similar to
improve the symptoms and slow arthritis progression, high dosage and the EPR effect observed in solid tumors. Because of this, a number of
frequent administration are often required to obtain satisfactory effi­ nano-drug delivery systems have been demonstrated to effectively
cacy, which inevitably cause untoward side-effects [4,5]. The biological deliver various kinds of drugs into RA site [10–13], resulting in
agents are also developed to prevent joint destruction by suppressing the enhanced drug accumulation and reduced side-effects [14,15]. More­
cytokines’ level [6], but the therapeutic benefits are not as good as over, since the inflammation-associated cells pathologically overexpress

* Corresponding author.
E-mail address: zhouwenhuyaoji@163.com (W. Zhou).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biomaterials.2020.120390
Received 22 June 2020; Received in revised form 1 September 2020; Accepted 16 September 2020
Available online 19 September 2020
0142-9612/© 2020 Elsevier Ltd. All rights reserved.
Y. Yang et al. Biomaterials 264 (2021) 120390

certain receptors on cell membrane, actively targeted drug delivery Lipopolysaccharide (LPS) was from Biosharp Co., Ltd (Beijing, China).
systems were also designed via surface modifications of specific recog­ Bovine Type II collagen was obtained from Chondrex (Washington DC,
nition ligands [10,16]. While significant efforts have been made, this USA). Silver nitrate, sodium borohydride (NaBH4), 3-(4,5-dimethylth­
field is still in its infancy and there is still deficiency of nanomedicines iazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), complete Freund’s
for effective RA management. adjuvant and incomplete Freund’s adjuvant were from Sigma-Aldrich
In RA, the synovial tissue is infiltrated by various inflammatory cells, Co., Ltd (St. Louis, MO, USA). Chlorpromazine hydrochloride, nystatin
especially macrophages, which play crucial roles in the pathophysio­ and colchicine were purchased from Aladdin (Shanghai, China). L-
logical responses [17]. The activated macrophages, termed M1 macro­ cysteine was from Shanghai Boao biotechnology Co., Ltd (Shanghai,
phages, produce a series of inflammation cytokines, such as TNF-α, China). Annexin V-FITC Apoptosis Detection Kit was provided by BD
IL-1β, and IL-6, to sustain and aggravate joint inflammation [18]. biosciences Co., Ltd (San Jose, CA, USA). TRIzol reagent, First Strand
Therefore, the M1 macrophages have been considered as an vital ther­ cDNA Synthesis Kit and qPCR Detection Kit were bought from Thermo
apeutic target to relive symptoms of RA [19]. For example, different Fisher Scientific Co., Ltd (MA, USA). The primers of IL-1β, IL-6, TNF-α,
nano-vehicles have been developed for targeted delivery of metho­ Arg-1, IL-10 and housekeeping gene of GAPDH were designed by Qingke
trexate (MTX) into M1 macrophages [10,20]. Beside direct damaging Biotech (Changsha, China). The antibodies of folate receptor beta, CD68,
M1 macrophages using MTX, alternative strategies have also been iNOS and CD206 were from GeneTex (USA), Abzoom (USA), Boster
attempted to switch M1 type macrophages into anti-inflammatory M2 (Wuhan, China) and Protein tech (Wuhan, China), respectively. TUNEL
type by regulating microenvironment in inflamed joints [11,21]. M2 Apoptosis Assay Kit was purchased from Boster (Wuhan, China). Dul­
macrophages can secrete anti-inflammatory cytokines to relief inflam­ becco’s modified Eagle’s medium (DMEM), penicillin-streptomycin so­
mation, while in arthritic joints the macrophages are predominantly lution, phosphate buffered saline (PBS), and fetal bovine serum (FBS)
polarized to M1 subtype due to the specific inflammatory environment, were purchased from Gibco Life Technologies, Inc. (Grand Island, NY,
such as hypoxia-inducible factor (HIF-1α) expression and the high USA). Methanol (HPLC grade) was purchased from Tedia Co., Ltd (USA).
reactive oxygen species (ROS) level [22]. As such, several studies have
employed ROS scavenging nanoenzymes to drive M2 polarization for 2.1.2. Cells
combating inflammatory diseases [23,24]. Collectively, targeting M1 Murine macrophage cells (RAW264.7) were obtained from Xiangya
macrophages by direct triggering cell death and inducing M1-to-M2 cell center (Changsha, China). The cells were cultured in DMEM medium
phenotypic transition could be a promising strategy for RA treatment, supplemented with 10% FBS, 1% penicillin (50 U/mL) and streptomycin
while successful strategies to simultaneously achieve both goals are still (50 U/mL) in a 5% CO2 atmosphere at 37 ◦ C.
lacking.
AgNPs are a type of highly commercialized nanomaterial for 2.1.3. Animals
biomedical applications owing to ease of synthesis, readily modification, Male DBA/1 J mice (7–8 weeks old, 20 ± 2 g) were purchased from
and broad biological activities [25–30]. While it is still under debate, the Cavens animal Co., Ltd. (Changzhou, China), and maintained in a sterile
mechanisms of AgNPs for bio-functions are considered to be ROS environment and allowed free access to food and water. All animal ex­
regulation, inflammasome formation, and the release of silver ions to periments were approved by the Experimental Animal Ethics Committee
cause cell apoptosis [31–34]. Recently, AgNPs have been used as vehi­ of Central South University and were carried out in accordance with the
cles to deliver anti-inflammatory drugs for RA treatment [35,36], and requirements the National Act on the Use of Experimental Animals
interestingly, it was indicated that AgNPs on its own showed (People’s Republic of China).
anti-inflammatory activity and could promote M2 polarization [37,38].
We hypothesize that AgNPs with ROS regulation, cell apoptosis, and 2.2. Synthesis of LA-PEG-FA
anti-inflammatory activities might be a potential nanomedicine for RA
therapy, but we are unaware of any published attempts on this front. Under magnetic stirring, FA (500 μmol, 220.5 mg), DCC (550 μmol,
In current work, we studied the therapeutic potential of AgNPs for 113.5 mg) and NHS (550 μmol, 63.5 mg) were mixed in anhydrous
RA and demonstrated that AgNPs were highly effective for alleviating DMSO (20 mL). After 6 h reaction, the solution was added to a DMSO
inflammation via synergistic inducing M1 macrophage apoptosis and solution (10 mL) containing LA-PEG3400-NH2 (100 μmol, 360 mg). After
M2 macrophage polarization (Scheme 1). The AgNPs were PEGylated to 72 h reaction, 15 mL deionized water was added, and the insoluble
improve the colloidal stability, and decorated with folic acid (FA) to precipitate was removed by filtration. The product was sequentially
achieve active targeting towards the M1 macrophages. After physico­ dialyzed against PBS (for 72 h) and water (for 48 h) with MWCO of 3500
chemical characterizations, the modified AgNPs (termed FA-AgNPs) Da. The dialysis medium was replaced every 12 h. Then the product was
were investigated for in vitro/vivo targetability as well as therapeutic lyophilized for 24 h and stored at − 20 ◦ C for further characterizations.
effect. To have fundamental understanding, the mechanisms of AgNPs
for anti-rheumatic activity were also explored. Finally, the therapeutic 2.3. Preparation and characterizations of FA-AgNPs
effects and safety of AgNPs were assessed after systemic administration
using an in vivo RA mice model. This work demonstrated for the first AgNPs was synthesized according to the previous reported procedure
time that AgNPs can potentially be used for therapy of RA. [39]. Briefly, 1.2 mL trisodium citrate (100 mM) and 4 mL freshly pre­
pared NaBH4 (100 mM) were mixed with 390.8 mL water via stirring in
2. Materials and methods ice bath. Then, 4 mL AgNO3 (10 mM) was added, followed by 30 min
reaction. AgNPs was collected by centrifugation and resuspended in
2.1. Materials, cells and animals water. Then, the FA-AgNPs was prepared by conjugating LA-PEG-FA
with AgNPs through Ag–S bond. Briefly, 36 mL of AgNPs (0.42 nM)
2.1.1. Materials were mixed with 20 mg of LA-PEG-FA under magnetic stirring in the
Folic acid (FA) was from Sinopharm Chemical Reagent Co., Ltd dark. After 24 h reaction, the resultant nanoparticles were collected by
(Shanghai, China), and α-lipoyl-ω-amino poly (ethylene glycol) (LA- centrifugation at 20,000 rpm for 30 min at 4 ◦ C, and the pellets were
PEG-NH2) was purchased from Hunan huateng pharmaceutical Co., Ltd. washed and resuspended in water, and stored at 4 ◦ C for further use. The
(Changsha, China). Methylene blue (MB), Glutathione (GSH) and 2′ ,7′ - particle size, polydispersity index (PDI) and zeta potential were deter­
dichlorodihydrofluorescein diacetate (DCFH-DA) probe were obtained mined by using Malvern Zeta Sizer Nano series (Nano ZS, Malvern In­
from Macklin Co., Ltd (Shanghai, China). LysoTracker Red was pur­ struments, UK) at 25 ◦ C. The morphologies were observed by using
chased from Solarbio Biotech Co., Ltd (Beijing, China). transmission electron microscopy (TEM) (Titan G2-F20, FEI, USA). The

2
Y. Yang et al.
3

Biomaterials 264 (2021) 120390


Scheme 1. Schematic illustration of the therapeutic mechanism of FA-AgNPs against RA. FA-AgNPs dissolve and release Ag+ in response to intracellular GSH, which synergistically induces M1 macrophages apoptosis
and scavenge ROS to cause the polarization of M1 macrophages to M2 phenotype in inflamed synovial joints.
Y. Yang et al. Biomaterials 264 (2021) 120390

stability studies were conducted by adding various concentrations of 2.7. In vitro cytotoxicity study
NaCl, and the UV–vis spectra of the nanoparticles were measured by UV
Spectrophotometer. To prepare FITC-labelled NPs, the LA-PEG-FITC RAW264.7 cells were seeded into 96-well plates at density of 5 × 103
instead of LA-PEG-FA was used and performed as described above. per well and activated by LPS (10 μg/mL) for 48 h. After removing the
medium and washing twice with PBS, the cells were incubated with
2.4. Ag+ release study different formulations (diluted by cell culture medium) for 48 h. Then,
20 μL of MTT solution (5 mg/mL) was added. After 4 h, 100 μL of DMSO
The Ag+ release was assessed by membrane-free dissolution method. was added, and the absorbance was detected at 490 nm using microplate
The release media was 10 mM phosphate buffer solutions (pH 7.4, 5.5) reader (Infinite M200 PRO, TECAN, Austria).
without or with 10 mM GSH. Typically, FA-AgNPs were dissolved by
different media under mechanical shaking (100 rpm) at 37 ◦ C. At pre­ 2.8. Cellular apoptosis assay
determined intervals, sample was withdrawn and centrifuged, the su­
pernatant of Ag+ content was detected by an inductively coupled plasma RAW264.7 cells were seeded onto 6-well plates at density of 2 × 105
mass spectrometer (ICP-MS). cells per well and activated by LPS (10 μg/mL) for 48 h. Subsequently,
0.9 nM of FA-AgNPs were added for 4, 12, 48 h incubation, respectively.
2.5. ROS scavenging activity assay The cells were collected and suspended with 500 μL PBS. Then, 5 μL
FITC-Annexin and 5 μL PI were added. The apoptosis rate was deter­
2.5.1. Catalase-like activity mined by flow cytometry (FACSVerse, BD, USA).
The catalase-like activity of AgNPs and AgNO3 was studied by
measuring the oxygen generation using portable dissolved oxygen meter
2.9. Imaging of reactive oxygen species
(JPBJ-609L, INESA Scientific Instrument Co., Ltd., China). Briefly,
AgNPs (42 nM) or AgNO3 (10 mM) was added to an equal volume of
The RAW264.7 cells were seeded onto 24-well plates at density of 4
H2O2 (100 mM), and the oxygen generation was monitored every 10 s
× 104 cells per well, and stimulated by LPS (10 μg/mL). After 48 h, the
for 160 s.
cells were treated with 0.8 nM of AgNPs, FA-AgNPs, or 10.3 μM AgNO3
for 24 h. Then, 10 μM of DCFH-DA was added and incubated for 30 min.
2.5.2. Hydroxyl radical scavenging activity
The cells were then imaged by fluorescent microscopy. To quantify the
The hydroxyl radical (⋅OH) was generated via Mn2+-mediated
ROS level, the cells were seeded onto 6-well plates at density of 2 × 105
fenton-like reaction according to our previous reported procedure [40].
cells per well, and the same treatments were performed as described
Meanwhile, AgNPs (8.4 nM) or AgNO3 (2 mM) and MB (10 μg/mL) was
above. The cells were harvested and the fluorescence intensity was
added. After incubating at 37 ◦ C for 30 min, the UV–vis spectra within
measured by flow cytometry (FACSVerse, BD, USA).
400–800 nm was measured using microplate reader (Infinite M200 PRO,
TECAN, Austria).
2.10. Macrophage phenotype transition study
2.6. Cellular uptake study
For immunofluorescence staining, RAW 264.7 cells were seeded in a
The RAW264.7 cells were seeded onto 24-well plates at density of 4 24-well plate with coverslips and activated with LPS (10 μg/mL) for 48
× 104 cells per well, and cultured for 48 h in the absence or presence of h. The cells were then incubated with FA-AgNPs (0.8 nM) for 48 h,
lipopolysaccharide (LPS, 10 μg/mL). Then, the cells were treated with followed by fixing with 4% paraformaldehyde. Then, the cells were
FITC-labelled NPs for 2 h, followed by washing three times with PBS (pH incubated with primary antibodies against CD68 (1:100), iNOS (1:50) or
7.4). The cells were fixed with 4% paraformaldehyde for 20 min, and CD206 (1:50) at 4 ◦ C overnight and then incubated with respective
stained with DAPI for 10 min. Internalization was observed by using fluorescent-labelled secondary antibodies for 30 min. The nuclei were
fluorescent microscopy. To quantify the internalization, the cells were stained with DAPI and the cells were mounted. The samples were
seeded onto 6-well plates at density of 2 × 105 cells per well, and the observed with a fluorescent microscope. For RT-PCR quantification,
same treatments were performed as described above. After washing in RAW264.7 cells were seeded onto 6-well plates at density of 2 × 105
triplet with PBS (pH 7.4), the cells were collected and the fluorescence cells per well and activated by LPS (10 μg/mL) for 48 h. Then, 0.8 nM of
intensity was assessed by flow cytometry (FACSVerse, BD, USA). To FA-AgNPs were added. After 48 h, the cells were harvested and total
examine the uptake mechanism, the cells were pretreated with 1 mM RNA was extracted using TRIzol reagent. The levels of mRNAs
free folic acid (FA) for 1 h before incubating with FITC-labelled NPs. (including IL-1β, IL-6, TNF-α, Arg-1 and IL-10) were determined by RT-
To study the uptake mechanism, the activated macrophages were PCR (CFX-Connect, BIO-RAD, USA). Primer sequences were designed by
treated with chlorpromazine (10 μg/mL), nystatin (15 μg/mL) or soft of primer 5.0 (Table 1).
colchicine (5 μg/mL) for 1 h under cell culture conditions. After this
pretreatment, the medium was replaced by serum-free medium con­ 2.11. Mice model of collagen-induced arthritis
taining FITC-labelled FA-AgNPs and incubated for 4 h followed by
washing three times with PBS. Subsequently, the cells were harvested, The collagen-induced arthritis (CIA) mice model was established
washed with PBS for several times and resuspended in PBS. The fluo­ according to a previous report [41]. First, two emulsions were prepared
rescence intensity was analyzed by flow cytometry (FACSVerse, BD, by mixing 2 mg/mL bovine type II collagen with an equal volume of
USA) with 10,000 events collected.
To examine the subcellular localization, the cells were seeded in 35 Table 1
mm confocal dishes at density of 2 × 105 cells per dish, incubated Primer sequences used for real-time PCR.
overnight and activated with LPS (10 μg/mL) for 48 h. Then, the me­ Gene Forward Primer (5′ to 3′ ) Reverse Primer (5′ to 3′ )
dium was removed and FITC-labelled FA-AgNPs were added. After in­
IL-1β ATGAAGGGCTGCTTCCAAAC TCTCCACAGCCACAATGAGT
cubation for 2 h, 4 h or 12 h, the cells were washed three times with PBS IL-6 GGAGCCCACCAAGAACGATA ACCAGCATCAGTCCCAAGAA
and incubated with pre-warmed Lysotracker Red (1:20,000) for 1 h at TNF-α CTCATGCACCACCATCAAGG ACCTGACCACTCTCCCTTTG
37 ◦ C. Then the cells were fixed with 4% paraformaldehyde for 15 min Arg-1 TGGCTTGCGAGACGTAGAC GCTCAGGTGAATCGGCCTTT
and the nuclei were stained with DAPI (1 μg/mL), followed by observing IL-10 CTGGACAACATACTGCTAACCG GGGCATCACTTCTACCAGGTAA
GAPDH GGGTCCCAGCTTAGGTTCAT CCAATACGGCCAAATCCGTT
using confocal laser scanning microscope (CLSM).

4
Y. Yang et al. Biomaterials 264 (2021) 120390

Complete Freund’s Adjuvant (for emulsion A) or Incomplete Freund’s microscope.


Adjuvant (for emulsion B) for overnight stirring in ice bath. On day 0,
the mice were administrated with 0.1 mL emulsion A via tail intravenous 2.17. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP Nick
injection. On day 21, a booster injection was given with emulsion B. End Labeling (TUNEL) staining
Mice were monitored daily for arthritis progression.
After deparaffinization, rehydration and permeation with proteinase
2.12. In vivo biodistribution of the nanoparticles K and 0.1% Triton X100, the sections of ankle joints were blocked by 3%
H2O2-methanol solution. After that, TUNEL staining was conducted at
The CIA or healthy mice were injected intravenously with FITC 37 ◦ C for 60 min. Then, the sections were stained with DAPI. The images
labelled FA-AgNPs, and the hind paws of mice were isolated and visu­ were recorded with a fluorescent microscope.
alized by in vivo imaging system (Caliper, Hopkington, MA, USA) 10 h
after administration. To quantify the Ag content, the hind paws were 2.18. Safety evaluation in vivo
grinded with liquid nitrogen, and placed in a mixed acid matrix of aqua
regia with heating overnight at 90 ◦ C, followed by an additional heating To evaluate the safety of each formulation, the serum of mice was
at 140 ◦ C for 6 h to completely digest any organic compounds, and the collected after treatment, and the levels of aspartate aminotransferase
solution was analyzed by ICP-MS (Agilent 7500C). (AST), alanine aminotransferase (ALT), blood urea nitrogen (BUN) and
creatinine (Cre) were measured by using the standard kits according to
2.13. Pharmacokinetic and excretion studies the manufacturer’s instructions.

The healthy mice were injected intravenously with FA-AgNPs, and 2.19. Long-term toxicity test
the venous blood, urine, and feces samples were collected at pre-
designated timepoints. The samples were mineralized in a high- The healthy mice were injected with FA-AgNPs (0.652 nmol/kg)
pressure microwave oven, followed by digestion using aqua regia as every other day for overall 10 times. After the final injection, the body
described above. The Ag content was quantified by ICP-MS (Agilent weights were monitored, and the mice were sacrificed at day 28 to
7500C). measure the hematological and clinical biochemistry indexes. The main
organs were collected for histological analysis. To study the tissue
2.14. Therapeutic efficacy in vivo accumulation, the healthy mice were treated with FA-AgNPs as
described above. The mice were sacrificed at day 1, 7, or 28 after the
The CIA mice were divided into 4 groups (n = 6) for administration final injection, and the main organs were collected to measure the Ag
of saline, MTX (5 mg/kg BW), MTX-AgNPs (0.652 nmol/kg BW) or FA- content.
AgNPs (0.652 nmol/kg BW). In parallel, the normal healthy mice
without any treatment were used as control. The treatment was started 2.20. Statistical analysis
at day 30 after the first immunization, and the mice were injected
intravenously with different formulations every other day for overall 10 All quantitative data were expressed as mean ± SD from at least
times. The hind paw thickness and arthritis score were recorded every triplicate measurements. Differences between two comparative groups
other day. The arthritis scores of four paws were evaluated from 0 to 4 were assessed using the Student’s t-test, and the significance among
according to the following scale: 0 = no evidence of erythema or multiple groups was examined by the one-way analysis of variance
swelling, 1 = erythema and mild swelling, 2 = erythema and mild (ANOVA). Significance was measured at the following thresholds: *P <
swelling extending from the ankle to the tarsals, 3 = erythema and 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
moderate swelling extending from the ankle to metatarsal joints, and 4
= erythema and severe swelling encompassing the ankle, foot, and digits 3. Results and discussions
or ankylosis of the limb [41]. The maximum arthritis score of every
mouse was 16. On day 50 after first immunization, joint tissues were 3.1. Preparation and characterizations of FA-AgNPs
sampled, and the total RNA was extracted by TRIzol reagent. The mRNA
levels of the pro-inflammatory cytokines were quantitated using The heterofunctional α-lipoyl-ω-folic poly (ethylene glycol) (abbre­
RT-PCR. viated as LA-PEG-FA) was synthesized by conjugating FA with α-lipoyl-
ω-amino poly (ethylene glycol) (LA-PEG-NH2, MW = 3600) through
2.15. Histological analysis DCC/NHS coupling chemistry, and the conjugation was confirmed by
UV–vis, FT-IR and 1H- NMR characterizations (Figure S1). The conju­
After treatments, the mice were sacrificed, and ankle joints, spleen gation efficiency was quantified by HPLC measurement, and ~45% of
and thymus were collected and fixed in 4% paraformaldehyde over­ LA-PEG was modified with FA (Figure S2), indicating high conjugation
night. Ankle joints were decalcified with 10% EDTA solution for 40 days. ratio. Citrate capped silver nanoparticles (AgNPs) were synthesized
Samples were embedded in paraffin and sliced into 4 μm-thick sections. using NaBH4 as reductant according to the procedure reported previ­
The tissue sections were stained with H&E. The images of sections were ously [39]. From transmission electron microscopy (TEM), the particles
obtained with a light microscope (Model IX71 Olympus, Tokyo, Japan). were roughly spherical with uniformed size (Fig. 1A). The LA-PEG-FA
The histological changes with synovial inflammation and cartilage was anchored on the surface of AgNPs via Ag-sulfide bond (Scheme
erosion of ankle joints were evaluated by HSS (histopathological scores 1), resulting in surface FA-modified AgNPs (termed as FA-AgNPs), which
of synovium) [42,43]. were purified by centrifugation to remove unreacted LA-PEG-FA. The
FA-AgNPs can be easily dispersed in aqueous solution, and the spherical
2.16. Immunofluorescence staining morphology of FA-AgNPs was retained (Fig. 1B). After surface
LA-PEG-FA modification, the zeta potential of AgNPs changed from
After deparaffinization, rehydration and antigen retrieval, the sec­ − 16.6 mV to − 4.2 mV, due to the surface displacement of negatively
tions of ankle joints were incubated with primary antibodies against charged citrate by LA-PEG-FA.
iNOS (1:50) and CD206 (1:100) at 4 ◦ C overnight, and then incubated Both AgNPs and FA-AgNPs showed strong UV absorption peak at
with fluorescent-labelled secondary antibodies for 30 min. After being ~400 nm, indicating no aggregation during the LA-PEG-FA conjugation
counterstained with DAPI, the sections were imaged using a fluorescent (Fig. 1C) [39]. We then studied the colloidal stability of the

5
Y. Yang et al. Biomaterials 264 (2021) 120390

Fig. 1. TEM micrographs of (A) AgNPs and (B) FA-AgNPs. (C) UV–vis spectra of AgNPs and FA-AgNPs. Colloidal stability of (D) AgNPs and (E) FA-AgNPs as a
function of NaCl concentration indicated by UV–vis spectra. Inset: The photographs of these samples. Storage stability of (F) AgNPs and (G) FA-AgNPs at 4 ◦ C and
25 ◦ C. Size was measured at predetermined time within 60 d. Data were presented as mean ± SD (n = 3). (H) In vitro Ag+ release from FA-AgNPs under various
conditions. The pHs were buffered by phosphate buffer solution, and the GSH concentration was 10 mM.

nanoparticles. With increasing NaCl concentration, the brown color of After 2 h incubation, the activated macrophages displayed strong
AgNPs gradually faded, concomitant with the decrease of UV absorption green fluorescence throughout the cells, indicating substantial cellular
(Fig. 1D). Therefore, AgNPs alone are prone to aggregation at high salt uptake (Fig. 2A). For the normal macrophages (without LPS stimula­
concentration, probably due to the charge screen effect of the salt to tion), in contrast, only weak green fluorescence was seen in part of the
attenuate the negative repulsion between each AgNPs. After LA-PEG-FA cells. We reason that this difference can be ascribed to the over­
modification, in contrast, the stability of FA-AgNPs was markedly expression of folate receptor beta (FR-β) on the membrane of the acti­
enhanced to tolerate 200 mM NaCl without obvious color and UV ab­ vated macrophages, which facilitates the cellular uptake of FA-AgNPs
sorption changes (Fig. 1E). In addition, the LA-PEG-FA conjugation through the specific interaction between FA and FR [48,49]. To confirm
could stabilize AgNPs for at least 60 d (Fig. 1F and G). This improved this, we assessed the FR-β on cell surface by immunofluorescence
stability can be attributable to the steric hindrance effect provided by staining, which confirmed the significant up-expression of FR-β after LPS
PEG. activation (Figure S4). To further declare the contribution of FR-β for
AgNPs are known to undergo gradual dissolution to release Ag+ in FA-AgNPs internalization, we pre-treated the cells with free FA to
aqueous solution, leading to non-specific toxic effects on various or­ abolish the FR-β pathway, and in this case, we can hardly observe green
ganisms during in vivo circulation [44,45]. To predict the biological fate fluorescence inside cells. For comparison, we also prepared PEGylated
of the FA-AgNPs, the release of Ag+ was studied at different conditions. AgNPs with methotrexate (MTX) modification (MTX-AgNPs). MTX is a
We first dissolved the nanoparticles into phosphate buffer solution (pH FA analogue, while its binding affinity toward folate receptor is >
7.4) to mimic physiological environment. After 48 h, no Ag+ was 100-fold lower than that of FA [50]. In addition, after conjugation with
detected by ICP-MS (Fig. 1H, black trace). The pH was then decreased to PEG polymer, the bioactivity of MTX was strongly passivated
5.5, but again Ag+ barely released (Fig. 1H, red trace). Finally, we (Figure S5). Therefore, MTX-AgNPs is an excellent control to study the
simulated the cytoplasm condition by adding 10 mM GSH, and in this targetability of FA-AgNPs. As expected, MTX-AgNPs also showed mini­
case a time-dependent Ag+ release was observed, with a cumulative mal internalization. All these results confirmed that the FR-mediated
release ~80% after 48 h (Fig. 1H, blue trace). This GSH-responsive Ag+ uptake pathway plays a major role for intracellular delivery of
release would be ascribed to the thiophilic nature of Ag+ to strongly bind FA-AgNPs into the activated macrophages.
GSH, thus facilitating the dissolution of Ag+ from AgNPs surface. Note To have a quantitative characterization, the flow cytometry experi­
that the GSH level is ~2–10 mM in living cells while only 2–20 μM in ments were performed (Fig. 2B), and the fluorescent intensities were
extracellular fluids [46]. Therefore, the FA-AgNPs could remain stable quantified (Fig. 2C). For FA-AgNPs, the activated macrophages dis­
under physiological conditions, but rapidly release Ag+ to exert bio­ played ~5-fold higher fluorescence than normal macrophages, demon­
logical functions after delivering into the target cells, thus minimizing strating the targeting ability. However, the intensity of MTX-AgNPs was
the undesirable side-effects. rather low in both activated macrophages and normal macrophages.
Free FA pre-treatment, on the other hand, strongly decreased the in­
3.2. In vitro cellular uptake tensity of FA-AgNPs in the activated macrophages. Overall, the flow
cytometry results were highly in accordance with the above fluorescent
We next explored cellular uptake of the FA-AgNPs by using RAW microscopy studies. It should be noted that the normal macrophages
264.7 cells (a macrophage cell line) as model. The cells were activated showed only minimal uptake for both MTX-AgNPs and FA-AgNPs, which
by lipopolysaccharide (LPS) to stimulate the inflammation statement for can be attributable to the hydrophilic layer of PEG on particle surface
M1 polarization according to a previous report [47], and the successful [51]. Therefore, the surface PEG modification would protect the nano­
activation was validated by significant increases of cytokines including particles from non-specific mononuclear phagocyte system (MPS)
TNF-α, IL-1β and IL-6 (Figure S3). To track the FA-AgNPs inside cells, the clearance, which is important for prolonged circulation half-life and
particles were labelled with FITC fluorophore, and the location of the better targeting delivery toward RA site through “ELVIS” effect [14,52].
cells were visualized by staining the cell nuclei with DAPI. In general, the nanoparticles are internalized by the cytomembrane

6
Y. Yang et al. Biomaterials 264 (2021) 120390

Fig. 2. (A) Fluorescent images of RAW 264.7 cells and activated RAW 264.7 cells (by LPS) treating with FITC-labelled FA-AgNPs or MTX-AgNPs for 2 h. The FR-
mediated uptake of FA-AgNPs were explored by pretreating the cells with 1 mM free FA. Scale bar = 100 μm. (B) Flow cytometry images of RAW 264.7 cells and
activated RAW 264.7 cells after incubating with FITC-labelled FA-AgNPs (in absence or presence of 1 mM free FA) or MTX-AgNPs for 2 h. (C) Quantified fluorescence
intensity from (B). For B and C, the control means the cells without any treatment. (D) Relative cellular internalization of FA-AgNPs without or with pretreatment of
various endocytosis inhibitors. (E) CLSM images of activated RAW264.7 cells incubated with FITC-labelled FA-AgNPs for various time periods. Scale bar = 25 μm.

to form endosomes through different endocytosis pathways. To study internalized by clathrin-mediated endocytosis, which is in agreement
the detailed uptake mechanism, the cells were pre-treated with several with other FA-modified nanoparticles reported previously [55]. To
inhibitors, including nystatin, chlorpromazine (CPZ) and colchicine, further track the intracellular fate of FA-AgNPs, time-dependent cellular
which prevents caveolin-, clathrin- and macropinocytosis-mediated uptake was studied. After 2 h incubation, the fluorescence of FA-AgNPs
endocytosis, respectively [53,54]. All types of inhibitors were able to showed good co-localization with endo- and lysosomes stained red by
decrease FA-AgNPs uptake to some extent, and among them CPZ was the Lysotracker, confirming the endocytotic internalization (Fig. 2E). The
most effective one (Fig. 2D). Therefore, FA-AgNPs was predominately green fluorescence was intensified with longer incubation time because

7
Y. Yang et al. Biomaterials 264 (2021) 120390

of more FA-AgNPs assimilation, and the fluorescence was mostly local­ AgNPs, likely due to the higher cellular uptake induced by FA decora­
ized in the cytoplasm with minimal distribution in cell nuclei. tion. For normal macrophages, on the contrary, the trend was reversed
that AgNPs was much more toxic than FA-AgNPs (~3-fold higher). This
can be explained by surface PEG modification that impedes FA-AgNPs
3.3. In vitro cytotoxicity
uptake by normal macrophages. These results suggest a selective
toxicity of FA-AgNPs towards the inflammatory cells against the normal
Having demonstrated targeting capability of FA-AgNPs, we then
macrophages (the IC50 value was 2-fold lower). Therefore, the FA-AgNPs
surveyed cellular cytotoxicity by using MTT test, and the unmodified
could specifically suppress inflammation cells with much less influence
AgNPs were used as control. After 48 h treatment, the viability of both
on the essential functions of immunity system.
normal macrophages (Fig. 3A) and activated macrophages (Fig. 3B)
The cytotoxicity of AgNPs has been reported in several cancer cells,
decreased in a dose-dependent manner. Notably, the activities of these
which proposed that the release of Ag+ in the cytosol plays a critical role
two nanoparticles were variant dependent on the cell lines. To quantify
[32]. From the above experiments, we have demonstrated that
these differences, the IC50 values were measured (Fig. 3C). For activated
FA-AgNPs could release Ag+ with the aid of GSH. To explore effect of
macrophages, the FA-AgNPs showed ~3-fold higher activity than

Fig. 3. In vitro viability test of (A) RAW 264.7 cells and (B) activated RAW 264.7 cells in presence of different concentrations of AgNPs and FA-AgNPs after 48 h
incubation. (C) IC50 values calculated from (A) and (B). (D) Viability of the activated RAW 264.7 cells in presence of 0.5 mM Cys as a function of FA-AgNPs
concentration after 48 h incubation. (E) The apoptosis of the activated RAW 264.7 cells after treatment of 0.9 nM FA-AgNPs with variant time periods.

8
Y. Yang et al. Biomaterials 264 (2021) 120390

Ag+ release, we first incubated the activated macrophages with different 3.4. ROS scavenging capability
concentrations of AgNO3, and the IC50 value was calculated to be 11.9
μM (Figure S6), corresponding to 0.05 nM FA-AgNPs with the same mass Oxidative stress refers to the production of reactive oxygen species
amount of Ag atom. Therefore, free Ag+ was much more toxic than (ROS) to cause intracellular anti-oxidant imbalance, leading to a series
FA-AgNPs (the IC50 value of FA-AgNPs was 0.92 nM). We then incubated of cellular dysfunctions. Previous studies have shown that ROS are
the cells with FA-AgNPs in presence of 0.5 mM cysteine (Cys), a strong implicated in the pathophysiology of RA, and strategies for regulating
Ag+ chelator that can abolish the biological functions of Ag+ inside cells. ROS have been employed for the treatment of RA [24,56]. There are
Cys itself was non-toxic to cells (Figure S7), but it can strongly alleviate different types of ROS in the activated macrophages, including
the toxic effect of FA-AgNPs (Fig. 3D). Therefore, the released Ag+ from non-radical species (H2O2, singlet oxygen 1O2, etc.) and radical species
FA-AgNPs should play a vital role for cell cytotoxicity. (superoxide anion O⋅-2 , hydroxyl radical ⋅OH, etc.). To study the ROS
To further study the cellular inhibitory effect of FA-AgNPs, the scavenging activity of FA-AgNPs in activated macrophages, the
Annexin V/PI assay was carried out to evaluate the mode and extent of 2′ -7′ -dichlorodihydrofluorescein diacetate (DCFH-DA) was used as a
cell death. With 0.9 nM FA-AgNPs treatment, the percentage of probe to fluorescently monitor the total intracellular ROS level. After
apoptotic cells gradually increased over time, reaching 50% after 48 h diffusion into cells, the DCFH-DA probe is deacetylated to DCFH by
incubation (Fig. 3E). This result was highly consistent with the IC50 cellular esterase, and further oxidized by ROS to yield green fluorescent
value measured by MTT assay. Therefore, the FA-AgNPs damage cells DCF for ROS monitoring. Before LPS stimulation, no apparent fluores­
mainly through apoptotic cell death. cence was detected inside cells, indicating low ROS level (Fig. 4A). The
LPS treatment significantly improved the ROS level as evidenced by the

Fig. 4. (A) Fluorescent images and (B) flow cytometry of the cells with different treatments and then stained with ROS fluorescent probe DCFH-DA. Scale bar = 100
μm. (C) Quantified fluorescence intensity from (B). (D) The kinetics of O2 generation monitored by portable dissolved oxygen meter. (E) Hydroxyl radical scavenging
activity of AgNPs and Ag+ probed by MB.

9
Y. Yang et al. Biomaterials 264 (2021) 120390

bright fluorescence emission, leading to severe oxidative stress. We then 3.5. In vitro M1 to M2 phenotypic transition of macrophages
treated the activated macrophages with AgNPs or FA-AgNPs for 24 h,
and as expected, the ROS level markedly decreased (Fig. 4A), confirming Macrophage can exist in two different phenotypes, i.e. pro-
strong ROS scavenging activity. inflammatory (M1) and anti-inflammatory (M2) phenotypes. In RA
It is interesting to note that several studies have shown the ROS joints, the macrophages are primarily of the M1 phenotype that pro­
regulation capability of AgNPs [31,57], and AgNPs is known to kill motes RA progression by secreting various inflammatory cytokines [59].
cancer cells by producing ROS to damage intracellular constituents and Therefore, switching macrophage from M1 to M2 phenotype is a po­
cause cell apoptosis [31]. In our case, however, AgNPs act as ROS tential method for the treatment of RA [24]. To examine capability of
scavenger to down-regulate the ROS level. These discrepancy results FA-AgNPs for reprogramming macrophage, the phenotypes of the cells
may be due to the different intracellular environment, as the activated were analyzed by immunofluorescence staining of CD68 (the
macrophage is pathologically featured with high oxidative pressure. pan-macrophage marker), iNOS (M1 marker), and CD206 (M2 marker).
Such environment-dependent properties of nano-materials have been After FA-AgNPs treatment, the iNOS signal decreased while the immu­
noticed previously. For example, iron oxide nanoparticles could switch nity of CD206 becomes more pronounced (Fig. 5A), suggesting effective
peroxidase-like activity for free radical production to catalase-like ac­ repolarization of macrophages from M1 to M2 type. To confirm this
tivity for ROS scavenging with pH increase [58]. We reason that the ROS result, the mRNA expression of macrophage markers, including IL-1β,
regulation activity of AgNPs is partially controlled by intracellular ROS IL-6, TNF-α, Arg-1 and IL-10, were measured by using RT-PCR.
level. Compared to the non-treated cells, FA-AgNPs could significantly
Since FA-AgNPs could rapidly release Ag+ in response to intracel­ decrease the expression of M1 markers (Fig. 5B), while increase the M2
lular GSH, we next test whether such released Ag+ has any effect on ROS markers (Fig. 5C). Therefore, FA-AgNPs can exert anti-inflammatory
scavenging. To do this, the cells were treated with free Ag+, and inter­ activity by modulating macrophage polarization, and subsequent regu­
estingly, the ROS level was significantly decreased. The relative fluo­ lating inflammation cytokines expression.
rescence intensity of each group was measured by flow cytometry
(Fig. 4B), which allowed for quantitative comparison (Fig. 4C). The LPS
stimulation increased the fluorescence by ~40-fold, indicating the 3.6. In vivo pharmacokinetics, excretion, and targetability
activation of the macrophage. The fluorescence intensity for AgNPs, FA-
AgNPs and Ag+-treated groups decreased by 14, 21, and 36-fold Having confirmed the therapeutic activity of FA-AgNPs in cell level,
respectively, compared with the non-treated control after 24 h incuba­ we next studied the in vivo performances of the FA-AgNPs. The phar­
tion. The ROS scavenging activity was in order of Ag+ > FA-AgNPs > macokinetics and excretion of FA-AgNPs were first investigated in
AgNPs. Collectively, these findings revealed that FA-AgNPs could healthy mice after a single intravenous injection by measuring the Ag
effectively decrease intracellular ROS level to alleviate inflammation, content using ICP-MS. The FA-AgNPs displayed a typical long-
which is partly achieved by releasing Ag+. To further explore the circulation profile, with half-life of ~104 h (Fig. 6A), likely attribut­
mechanism, we also studied the ROS scavenging activity in test tube by able to its surface PEG modification. Notably, over 80% of the Ag con­
using hydrogen peroxide (H2O2) and hydroxyl radical (⋅OH) as example tent was cleared after 16 d, indicating biodegradability of the
(Fig. 4D and E), since they are two major types of ROS in RA site. nanoparticles. From Fig. 6B, it was clear seen that Ag was predominantly
Interestingly, the AgNPs could catalyze H2O2 decomposition while Ag+ excreted via feces, which is consistent with previous report [60]. The
could effectively scavenge ⋅OH, confirming their ROS scavenging cumulative excretion reached ~50% at day 8, in line with the above t1/2
activity. data, and ~80% of total body burden was excreted at day 16, indicating
minimal body accumulation of Ag.
We then studied the in vivo behavior in collagen-induced arthritis

Fig. 5. (A) Immunofluorescence staining of CD68 (green), iNOS and CD206 (red), and nuclei (blue) on macrophages without or with treatment of FA-AgNPs. Scale
bar = 100 μm. The mRNA levels of (B) M1 macrophage markers (IL-1β, IL-6 and TNF-α), and (C) M2 macrophage markers (Arg-1 and IL-10) in activated macrophages
without and with treatment of FA-AgNPs. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

10
Y. Yang et al. Biomaterials 264 (2021) 120390

Fig. 6. (A) Kinetics of blood Ag concentration in mice following a single intravenous injection of FA-AgNPs. (B) Accumulated Ag content excreted in feces and urine
(inset). (C) Fluorescent image of FITC-labelled FA-AgNPs in joints of CIA mouse and normal mouse 10 h after injection of FA-AgNPs. (D) Quantified intensity from
(C). (E) The amount of Ag accumulated in the paws of CIA mouse and normal mouse 10 h after intravenous injection.

(CIA) model mice. The mice were injected with collagen to build the CIA reduce the cytokines almost to normal level, demonstrating a good anti-
model according to a previous reported protocol [41], and the arthritis inflammation activity. The cytokines levels also significantly decreased
symptoms were fully developed 30 days after immunization, charac­ after MTX-AgNPs and MTX treatment, while the efficacy was much
terized by severe swelling around the ankle, foot and digits. To evaluate lower. Furthermore, the phenotypic alteration of synovial macrophages
the in vivo targeting efficiency, the FITC-labelled FA-AgNPs were intra­ in RA joints were evaluated by immunofluorescent staining of M1 (iNOS,
venously administrated, and the fluorescence was detected using an red) and M2 (CD206, green) markers (Fig. 7G). Compared with that of
optical imaging system. Strong emission was observed in the inflamed normal mice, level of M1 macrophage-specific biomarker in inflamed
joints of CIA mouse, while the signal in the noninflamed paws of normal joints significantly increased, whereas the injection of each formulation
mouse was much weaker (Fig. 6C), demonstrating the in vivo targeting could reduce its level to different extent, accompanied by the increase of
ability of the nanoparticles to selectively accumulate into the inflamed M2 macrophage-specific biomarker. As expected, the FA-AgNPs dis­
joint. Such passively accumulation of nanoparticles into inflamed joints played the most obvious for M1-to-M2 phenotypic transition, which
has been demonstrated previously, which can be ascribed to the “ELVIS” contributed to the best anti-inflammatory efficacy.
effect [11,61,62]. We then quantified the fluorescent intensity, and We then evaluate the final outcomes by histological examination
~3-fold stronger intensity was obtained in inflamed paw (Fig. 6D). To (Fig. 7H). The mice with saline treatment showed significant inflam­
accurately identify the accumulation of FA-AgNPs in the inflamed joints, mation, involving synovial hyperplasia, pannus formation and bone
we measured the mass of Ag in the paws using ICP-MS, and the result destruction in the joints. The MTX-AgNPs and MTX-treated group
was quite consistent with the fluorescent observation (Fig. 6E). showed an obvious improvement of the symptoms, with less synovial
cell lining and milder bone destruction. Among them, the mice treated
with FA-AgNPs displayed the mildest inflammation with smooth artic­
3.7. In vivo therapeutic efficacy on CIA mice ulation of the cartilage surface. We quantified the histological changes
with synovial inflammation and cartilage erosion of ankle joint by HSS,
The therapeutic efficacy was measured by treating the CIA mice with and the scores were in order of FA-AgNPs < MTX-AgNPs < MTX < Saline
FA-AgNPs every other day for 20 days, and MTX was chosen as clinical (Fig. 7I). In addition, based on TUNEL staining, the macrophage
standard treatment. To confirm the targeted therapy of FA-AgNPs in apoptosis was observed after various treatment, and the overall trend
vivo, MTX-AgNPs were also tested for comparison. Compared with the was consistent with the above therapeutic efficacy (Fig. 7J). Collec­
saline-treated mice, the mice subjected with each formulation showed tively, all these in vivo results suggest good therapeutic effect of FA-
obvious improvement in the degree of swelling as well as much less AgNPs, which is significantly better than that of the commercial drug
redness (Fig. 7A). While MTX and MTX-AgNPs displayed comparable of MTX.
therapeutic effect (Fig. 7B and C), the FA-AgNPs achieved significantly
decreased clinical score and better end outcome, demonstrating the
superiority of targeted FA-AgNPs. Swelling ratios were quantified by the 3.8. Safety evaluation of FA-AgNPs in vivo
examination of joint thickness, the results in each group agreed with the
clinical scores (Fig. 7D and E). We then measured the levels of pro- For the safety concerns, the hepatotoxicity was evaluated by
inflammatory cytokines as another index to evaluate the therapeutic measuring the serum levels of liver enzymes alanine transaminase (ALT)
efficacy. For the saline-treated mice, the cytokines including TNF-α, IL- and aspartate aminotransferase (AST) after treatment (Fig. 8A), and the
1β and IL-6 remained at high levels (Fig. 7F), while FA-AgNPs could nephrotoxicity was tested by measuring blood urea nitrogen (BUN) and

11
Y. Yang et al. Biomaterials 264 (2021) 120390

(caption on next page)

12
Y. Yang et al. Biomaterials 264 (2021) 120390

Fig. 7. (A) Photographs of inflamed joints before and after different treatments. (B) The clinical scores calculated for the inflamed joints with different treatments as
a function of time. (C) The clinical score after 20 days of different treatments. (D) The paw thickness of the inflamed joints for different treatments as a function of
time. (E) The paw thickness after 20 days of different treatments. (F) The mRNA levels of TNF-α, IL-1β and IL-6 in joints of CIA mice after different treatments.
Healthy, un-treated animals served as control (normal). (G) Immunofluorescence analysis of M1 (iNOS, red) and M2 (CD206) macrophage markers in the joint after
treatment. Scale bars = 50 μm. (H) Hematoxylin-eosin (H&E) and (J) TUNEL staining of ankle joints from mice treated with different formulations. Arrows indicate
finger-like pannus formation and the asterisk stands for bone destruction. Scale bar = 100 μm. (I) Histopathology evaluations of joint synovium by HSS determined
from (H). (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

Fig. 8. Evaluation of the (A) hepatotoxicity and (B) nephrotoxicity of each treatment by measuring the serum levels of ALT, AST, BUN and Cre. (C) Histopathological
analysis of spleen and thymus after treatments by staining with hematoxylin and eosin (H&E). Scale bar = 100 μm.

creatinine (Cre) (Fig. 8B). All these parameters were not changed after most of the nanoparticles were removed from each organ after 28 days,
FA-AgNPs and MTX-AgNPs treatment compared to normal animals, which is consistent with the above excretion results.
suggesting minimal acute toxicity to liver and kidney. For MTX-treated After 28 days treatment, the serum biochemical and hematological
group, by contrast, considerable increase of AST and Cre levels were parameters were measured to quantitatively evaluate the potential
noticed, which may be a sign of liver and renal damage after long-term toxicity of FA-AgNPs. Overall, while some parameters showed relatively
MTX administration. Indeed, the severe side-effects of MTX are major large standard deviations due to inter-animal variation, none of them
limitations for its clinical RA therapy applications. Therefore, the FA- showed any significant changes compared to those of the mice without
AgNPs seems to be a better choice due to the higher efficacy and bio- any treatment. For routine blood examination, all parameters, such as
safety. red blood cells, white blood cells, and platelets, were at a normal level
As FA-AgNPs function as an immunomodulator for RA therapy, we (Fig. 9C), indicating no significant risk associated with FA-AgNPs
then explored whether the nanoparticles had any effect on immune or­ treatment. In serum biochemical analysis, various parameters were
gans by histopathological analysis (Fig. 8C). No obvious change was measured with particular attention paid to liver (e.g., ALP, ALT, AST)
observed from the hematoxylin and eosin (H&E) stained organs of and kidney (e.g., Cre, BUN) (Fig. 9D). Likewise, all indicators were
spleen and thymus. Collectively, all these results implied that FA-AgNPs within normal ranges, suggesting no obvious injury to these organs.
present a highly safe and effective platform for RA therapy. Based on these results, the tissue damage was further evaluated by H&E
staining for the major organs (Fig. 9E), and no apparent histopatho­
3.9. Long-term toxicology evaluation of FA-AgNPs in vivo logical abnormalities or lesions were observed in each organ. Collec­
tively, all these data indicate no significant long-term toxicity of FA-
As FA-AgNPs could be retained in mice body for a long period of time AgNPs up to 28 days.
after multiple injection, a systematic study was performed to evaluate
the in vivo long-term toxicity. Body weight usually reflects the health 4. Conclusions
condition of the treated mice. In general, the average body weights
gradually increased during the observation period of 28 days after FA- In summary, we developed FA-AgNPs that can alleviate inflamma­
AgNPs treatment (Fig. 9A), suggesting a well tolerance of the mice to­ tion by synergistic M1 macrophages reduction and M2 macrophages
ward FA-AgNPs at the therapeutically efficacious dose. We also induction for effective RA treatment. Mechanistic studies indicated that
measured the tissue Ag content in main organs at day 1, 7, and 28 after the intracellular released Ag+ plays critical roles for this synergistic ef­
the final injection. The FA-AgNPs were mainly distributed in spleen, fect, which caused M1 macrophages apoptosis and ROS clearance to
liver and lungs (Fig. 9B), while low level of Ag accumulation in kidney allow M1-to-M2 re-polarization. FA-AgNPs were PEGylated to improve
was observed, further suggesting that the FA-AgNPs was not primarily colloidal stability and reduce non-specific opsonization, which enabled
excreted by urine. The Ag content gradually decreased overtime, and passive accumulation into RA site through “ELVIS” effect. Surface FA

13
Y. Yang et al. Biomaterials 264 (2021) 120390

Fig. 9. (A) Body weight of the mice treated with multiple injections of FA-AgNPs for 28 days. (B) Distribution of Ag in main organs following multiple injection of
FA-AgNPs at different timepoints. (C) Hematological and (D) Serum biochemical parameters following multiple intravenous exposure to FA-AgNPs at day 28. (E)
Hematoxylin and eosin stained images of major organs from mice with multiple injection of FA-AgNPs at 28 days post-injection.

modification further mediated active delivery of the nano-system into CrediT author statement
M1 macrophages to attenuate the joints inflammation in RA mice, which
showed much better efficacy than the current standard treatment of Yihua Yang: Methodology, Conceptualization, performed the ex­
MTX with higher biosafety. Besides, the FA-AgNPs could be excreted periments, Writing - original draft, read and approved the final manu­
from mice body and did not cause any obvious long-term toxicity in vivo. script. Lina Guo: Methodology, Conceptualization, performed the
Given the high commercialization of AgNPs for medical applications, we experiments, Writing - original draft, read and approved the final
can envisage that FA-AgNPs would be a promising clinical therapeutic manuscript. Zhe Wang: performed the experiments, read and approved
modality for RA therapy. the final manuscript. Peng Liu: performed the experiments, read and
approved the final manuscript. Xuanjun Liu: performed the experi­
Data availability statement ments, read and approved the final manuscript. Jinsong Ding: Formal
analysis, Validation, read and approved the final manuscript. Wenhu
The raw data and processed data required to reproduce these find­ Zhou: Methodology, Conceptualization, Writing - original draft, read
ings are available from the corresponding author upon request. and approved the final manuscript.

14
Y. Yang et al. Biomaterials 264 (2021) 120390

Declaration of competing interest [24] J. Kim, H.Y. Kim, S.Y. Song, S.H. Go, H.S. Sohn, S. Baik, et al., Synergistic oxygen
generation and reactive oxygen species scavenging by manganese ferrite/ceria Co-
decorated nanoparticles for rheumatoid arthritis treatment, ACS Nano 13 (2019)
The authors declare that they have no known competing financial 3206–3217.
interests or personal relationships that could have appeared to influence [25] E.I. Alarcon, K. Udekwu, M. Skog, N.L. Pacioni, K.G. Stamplecoskie, M. Gonzalez-
the work reported in this paper. Bejar, et al., The biocompatibility and antibacterial properties of collagen-
stabilized, photochemically prepared silver nanoparticles, Biomaterials 33 (2012)
4947–4956.
Acknowledgement [26] Z.M. Xiu, Q.B. Zhang, H.L. Puppala, V.L. Colvin, P.J.J. Alvarez, Negligible particle-
specific antibacterial activity of silver nanoparticles, Nano Lett. 12 (2012)
4271–4275.
This work was supported by Innovation-Driven Project of Central [27] R.W.-Y. Sun, R. Chen, N.P.Y. Chung, C.-M. Ho, C.-L.S. Lin, C.-M. Che, Silver
South University (No. 20170030010004), National Natural Science nanoparticles fabricated in Hepes buffer exhibit cytoprotective activities toward
Foundation of China (No. 21804144, 81573374), Hunan Engineering HIV-1 infected cells, Chem. Commun. (2005) 5059–5061.
[28] R. Bhattacharya, P. Mukherjee, Biological properties of "naked" metal
Research Center for Optimization of Drug Formulation and Early Clin­ nanoparticles, Adv. Drug Deliv. Rev. 60 (2008) 1289–1306.
ical Evaluation (No. 2015TP2005). [29] J. Liu, Y. Zhao, Q. Guo, Z. Wang, H. Wang, Y. Yang, et al., TAT-modified nanosilver
for combating multidrug-resistant cancer, Biomaterials 33 (2012) 6155–6161.
[30] A. Panacek, M. Kolar, R. Vecerova, R. Prucek, J. Soukupova, V. Krystof, et al.,
Appendix A. Supplementary data Antifungal activity of silver nanoparticles against Candida spp, Biomaterials 30
(2009) 6333–6340.
Supplementary data to this article can be found online at https://doi. [31] D.W. Guo, L.Y. Zhu, Z.H. Huang, H.X. Zhou, Y. Ge, W.J. Ma, et al., Anti-leukemia
activity of PVP-coated silver nanoparticles via generation of reactive oxygen
org/10.1016/j.biomaterials.2020.120390.
species and release of silver ions, Biomaterials 34 (2013) 7884–7894.
[32] V. De Matteis, M.A. Malvindi, A. Galeone, V. Brunetti, E. De Luca, S. Kote, et al.,
References Negligible particle-specific toxicity mechanism of silver nanoparticles: the role of
Ag+ ion release in the cytosol, Nanomed. Nanotechnol. Biol. Med. 11 (2015)
[1] J.M.W. Hazes, J.J. Luime, The epidemiology of early inflammatory arthritis, Nat. 731–739.
Rev. Rheumatol. 7 (2011) 381–390. [33] E.T. Hwang, J.H. Lee, Y.J. Chae, Y.S. Kim, B.C. Kim, B.I. Sang, et al., Analysis of the
[2] G. Schett, E. Gravallese, Bone erosion in rheumatoid arthritis: mechanisms, toxic mode of action of silver nanoparticles using stress-specific bioluminescent
diagnosis and treatment, Nat. Rev. Rheumatol. 8 (2012) 656–664. bacteria, Small 4 (2008) 746–750.
[3] I.B. McInnes, G. Schett, The pathogenesis of rheumatoid arthritis, N. Engl. J. Med. [34] E.-J. Yang, S. Kim, J.S. Kim, I.-H. Choi, Inflammasome formation and IL-1β release
365 (2011) 2205–2219. by human blood monocytes in response to silver nanoparticles, Biomaterials 33
[4] Z.A. Khan, R. Tripathi, B. Mishra, Methotrexate: a detailed review on drug delivery (2012) 6858–6867.
and clinical aspects, Expet Opin. Drug Deliv. 9 (2012) 151–169. [35] A. Mani, C. Vasanthi, V. Gopal, D. Chellathai, Role of phyto-stabilised silver
[5] J.N. Hoes, J.W.G. Jacobs, F. Buttgereit, J.W.J. Bijlsma, Current view of nanoparticles in suppressing adjuvant induced arthritis in rats, Int. Immunopharm.
glucocorticoid co-therapy with DMARDs in rheumatoid arthritis, Nat. Rev. 41 (2016) 17–23.
Rheumatol. 6 (2010) 693–702. [36] K. Rao, T. Roome, S. Aziz, A. Razzak, G. Abbas, M. Imran, et al., Bergenin loaded
[6] H. Lee, S.H. Bhang, J.H. Lee, H. Kim, S.K. Hahn, Tocilizumab-alendronate gum xanthan stabilized silver nanoparticles suppress synovial inflammation
conjugate for treatment of rheumatoid arthritis, Bioconjugate Chem. 28 (2017) through modulation of the immune response and oxidative stress in adjuvant
1084–1092. induced arthritic rats, J. Mater. Chem. B 6 (2018) 4486–4501.
[7] D.L. Scott, F. Wolfe, T.W.J. Huizinga, Rheumatoid arthritis, Lancet 376 (2010) [37] A.N. Yilma, S.R. Singh, S. Dixit, V.A. Dennis, Anti-inflammatory effects of silver-
1094–1108. polyvinyl pyrrolidone (Ag-PVP) nanoparticles in mouse macrophages infected with
[8] J.A. Singh, G.A. Wells, R. Christensen, E.T. Ghogomu, L. Maxwell, J.K. MacDonald, live Chlamydia trachomatis, Int. J. Nanomed. 8 (2013) 2421–2432.
et al., Adverse effects of biologics: a network meta-analysis and Cochrane [38] Y.M.F. Chen, M. Guan, R.Y. Ren, C.H. Gao, H. Cheng, Y. Li, et al., Improved
overview, Cochrane Database Syst. Rev. (2011) 60. immunoregulation of ultra-low-dose silver nanoparticle-loaded TiO2 nanotubes via
[9] D. Wang, S.R. Goldring, The bone, the joints and the balm of gilead, Mol. Pharm. 8 M2 macrophage polarization by regulating GLUTI and autophagy, Int. J. Nanomed.
(2011) 991–993. 15 (2020) 2011–2026.
[10] J.M. Shin, S.H. Kim, T. Thambi, D.G. You, J. Jeon, J.O. Lee, et al., A hyaluronic [39] X. Zhang, M.R. Servos, J. Liu, Fast pH-assisted functionalization of silver
acid-methotrexate conjugate for targeted therapy of rheumatoid arthritis, Chem. nanoparticles with monothiolated DNA, Chem. Commun. 48 (2012) 10114–10116.
Commun. 50 (2014) 7632–7635. [40] Y. Nie, D. Li, Y. Peng, S. Wang, S. Hu, M. Liu, et al., Metal organic framework
[11] Q. Wang, H. Jiang, Y. Li, W.F. Chen, H.M. Li, K. Peng, et al., Targeting NF-kB coated MnO2 nanosheets delivering doxorubicin and self-activated DNAzyme for
signaling with polymeric hybrid micelles that co-deliver siRNA and dexamethasone chemo-gene combinatorial treatment of cancer, Int. J. Pharm. 585 (2020) 119513.
for arthritis therapy, Biomaterials 122 (2017) 10–22. [41] D.D. Brand, K.A. Latham, E.F. Rosloniec, Collagen-induced arthritis, Nat. Protoc. 2
[12] S.M. Lee, H.J. Kim, Y.J. Ha, Y.N. Park, S.K. Lee, Y.B. Park, et al., Targeted chemo- (2007) 1269–1275.
photothermal treatments of rheumatoid arthritis using gold half-shell [42] H. Liu, J.X. Ding, J.C. Wang, Y.A. Wang, M.D. Yang, Y.B. Zhang, et al., Remission of
multifunctional nanoparticles, ACS Nano 7 (2013) 50–57. collagen-induced arthritis through combination therapy of microfracture and
[13] R. Li, Y. He, Y. Zhu, L. Jiang, S. Zhang, J. Qin, et al., Route to rheumatoid arthritis transplantation of thermogel-encapsulated bone marrow mesenchymal stem cells,
by macrophage-derived microvesicle-coated nanoparticles, Nano Lett. 19 (2019) PloS One 10 (2015) 18.
124–134. [43] H. Liu, J.X. Ding, C.Y. Wang, J.C. Wang, Y.N. Wang, M.D. Yang, et al., Intra-
[14] Q. Wang, X. Sun, Recent advances in nanomedicines for the treatment of articular transplantation of allogeneic BMMSCs rehabilitates cartilage injury of
rheumatoid arthritis, Biomater Sci 5 (2017) 1407–1420. antigen-induced arthritis, Tissue Eng. 21 (2015) 2733–2743.
[15] M.D. Yang, X.R. Feng, J.X. Ding, F. Chang, X.S. Chen, Nanotherapeutics relieve [44] T.S. Peretyazhko, Q.B. Zhang, V.L. Colvin, Size-controlled dissolution of silver
rheumatoid arthritis, J. Contr. Release 252 (2017) 108–124. nanoparticles at neutral and acidic pH conditions: kinetics and size changes,
[16] T.P. Thomas, S.N. Goonewardena, I.J. Majoros, A. Kotlyar, Z.Y. Cao, P.R. Leroueil, Environ. Sci. Technol. 48 (2014) 11954–11961.
et al., Folate-targeted nanoparticles show efficacy in the treatment of inflammatory [45] S. Kittler, C. Greulich, J. Diendorf, M. Koller, M. Epple, Toxicity of silver
arthritis, Arthritis Rheum. 63 (2011) 2671–2680. nanoparticles increases during storage because of slow dissolution under release of
[17] I.A. Udalova, A. Mantovani, M. Feldmann, Macrophage heterogeneity in the silver ions, Chem. Mater. 22 (2010) 4548–4554.
context of rheumatoid arthritis, Nat. Rev. Rheumatol. 12 (2016) 472–485. [46] R. Cheng, F. Feng, F.H. Meng, C. Deng, J. Feijen, Z.Y. Zhong, Glutathione-
[18] J.S. Smolen, D. Aletaha, M. Koeller, M.H. Weisman, P. Emery, New therapies for responsive nano-vehicles as a promising platform for targeted intracellular drug
treatment of rheumatoid arthritis, Lancet 370 (2007) 1861–1874. and gene delivery, J. Contr. Release 152 (2011) 2–12.
[19] I.B. McInnes, G. Schett, Cytokines in the pathogenesis of rheumatoid arthritis, Nat. [47] M.D. Yang, J.X. Ding, X.R. Feng, F. Chang, Y.N. Wang, Z.L. Gao, et al., Scavenger
Rev. Immunol. 7 (2007) 429–442. receptor-mediated targeted treatment of collagen-induced arthritis by dextran
[20] R. Heo, D.G. You, W. Um, K.Y. Choi, S. Jeon, J.S. Park, et al., Dextran sulfate sulfate-methotrexate prodrug, Theranostics 7 (2017) 97–105.
nanoparticles as a theranostic nanomedicine for rheumatoid arthritis, Biomaterials [48] A. Rollett, T. Reiter, P. Nogueira, M. Cardinale, A. Loureiro, A. Gomes, et al., Folic
131 (2017) 15–26. acid-functionalized human serum albumin nanocapsules for targeted drug delivery
[21] S. Jain, T.H. Tran, M. Amiji, Macrophage repolarization with targeted alginate to chronically activated macrophages, Int. J. Pharm. 427 (2012) 460–466.
nanoparticles containing IL-10 plasmid DNA for the treatment of experimental [49] C. Yang, S. Gao, J. Kjems, Folic acid conjugated chitosan for targeted delivery of
arthritis, Biomaterials 61 (2015) 162–177. siRNA to activated macrophages in vitro and in vivo, J. Mater. Chem. B 2 (2014)
[22] U. Fearon, M. Canavan, M. Biniecka, D.J. Veale, Hypoxia, mitochondrial 8608–8615.
dysfunction and synovial invasiveness in rheumatoid arthritis, Nat. Rev. [50] P.T. Wong, S.K. Choi, Mechanisms and implications of dual-acting methotrexate in
Rheumatol. 12 (2016) 385–397. folate-targeted nanotherapeutic delivery, Int. J. Mol. Sci. 16 (2015) 1772–1790.
[23] F. Zeng, Y. Wu, X. Li, X. Ge, Q. Guo, X. Lou, et al., Custom-made ceria nanoparticles [51] C.D. Walkey, J.B. Olsen, H.B. Guo, A. Emili, W.C.W. Chan, Nanoparticle size and
show a neuroprotective effect by modulating phenotypic polarization of the surface chemistry determine serum Protein adsorption and macrophage uptake,
microglia, Angew. Chem. Int. Ed. 57 (2018) 5808–5812. J. Am. Chem. Soc. 134 (2012) 2139–2147.

15
Y. Yang et al. Biomaterials 264 (2021) 120390

[52] H.C. Fischer, L.C. Liu, K.S. Pang, W.C.W. Chan, Pharmacokinetics of nanoscale [57] C. Beer, R. Foldbjerg, Y. Hayashi, D.S. Sutherland, H. Autrup, Toxicity of silver
quantum dots: in vivo distribution, sequestration, and clearance in the rat, Adv. nanoparticles—nanoparticle or silver ion? Toxicol. Lett. 208 (2012) 286–292.
Funct. Mater. 16 (2006) 1299–1305. [58] Z. Chen, J.-J. Yin, Y.-T. Zhou, Y. Zhang, L. Song, M. Song, et al., Dual enzyme-like
[53] M. Liu, Y. Peng, Y.B. Nie, P. Liu, S. Hu, J.S. Ding, et al., Co-delivery of doxorubicin activities of iron oxide nanoparticles and their implication for diminishing
and DNAzyme using ZnO@polydopamine core-shell nanocomposites for chemo/ cytotoxicity, ACS Nano 6 (2012) 4001–4012.
gene/photothermal therapy, Acta Biomater. 110 (2020) 242–253. [59] S. Bhattacharya, A. Aggarwal, M2 macrophages and their role in rheumatic
[54] Y. Chen, C.J. Yang, J. Mao, H.G. Li, J.S. Ding, W.H. Zhou, Spermine modified diseases, Rheumatol. Int. 39 (2019) 769–780.
polymeric micelles with pH-sensitive drug release for targeted and enhanced [60] Y. Lee, P. Kim, J. Yoon, B. Lee, K. Choi, K.H. Kil, et al., Serum kinetics, distribution
antitumor therapy, RSC Adv. 9 (2019) 11026–11037. and excretion of silver in rabbits following 28 days after a single intravenous
[55] H. Yang, C.C. Lou, M.M. Xu, C.H. Wu, H. Miyoshi, Y.Y. Liu, Investigation of folate- injection of silver nanoparticles, Nanotoxicology 7 (2013) 1120–1130.
conjugated fluorescent silica nanoparticles for targeting delivery to folate receptor- [61] C.H. Li, H.M. Li, Q. Wang, M.L. Zhou, M. Li, T. Gong, et al., pH-sensitive polymeric
positive tumors and their internalization mechanism, Int. J. Nanomed. 6 (2011) micelles for targeted delivery to inflamed joints, J. Contr. Release 246 (2017)
2023–2032. 133–141.
[56] Q. Zhang, S.Y. Lin, S.R. Shi, T. Zhang, Q.Q. Ma, T.R. Tian, et al., Anti-inflammatory [62] Y.J. Kim, S.Y. Chae, C.H. Jin, M. Sivasubramanian, S. Son, K.Y. Choi, et al., Ionic
and antioxidative effects of tetrahedral DNA nanostructures via the modulation of complex systems based on hyaluronic acid and PEGylated TNF-related apoptosis-
macrophage responses, ACS Appl. Mater. Interfaces 10 (2018) 3421–3430. inducing ligand for treatment of rheumatoid arthritis, Biomaterials 31 (2010)
9057–9064.

16

You might also like