You are on page 1of 11

Materials Science & Engineering C 115 (2020) 111096

Contents lists available at ScienceDirect

Materials Science & Engineering C


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

Injectable anti-inflammatory hyaluronic acid hydrogel for osteoarthritic T


cartilage repair
Yinji Jina,1, Rachel H. Koha,1, Su-Hwan Kimb,c, Kyung Min Kimb, G. Kate Parkc,

Nathaniel S. Hwanga,b,c,
a
School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
b
Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
c
Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea

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

Keywords: Osteoarthritis (OA) is one of the most common cartilage disorder that results from breakdown of joint cartilage
Osteoarthritis and underlying bone tissues. Once OA is induced in the joint, subsequent immune reaction leads to chronic
Cartilage regeneration inflammation that results in the progression of OA and deconstruction of the cartilage. In this study, an injectable
Epigallocatechin-3-gallate hydrogel with epigallocatechin-3-gallate (EGCG) is introduced to control inflammation and enhance cartilage
Anti-inflammation
regeneration. EGCG has intrinsic properties that can modulate inflammation and scavenge radical species.
Injectable hydrogel
Hyaluronic acid (HA), as a major component of the cartilage ECM, is commonly used for cartilage tissue en-
gineering. In this study, EGCG was combined with tyramine-conjugated HA and gelatin to create a composite
hydrogel at an optimized concentration of 50 μM EGCG and 5% w/v HA. The composite hydrogel provided
protection to chondrocytes against the pro-inflammatory factor, IL-1β. Additionally, the composite hydrogel led
to chondrogenic regeneration in vitro. Histological analysis in vivo showed that EGCG-HA/Gelatin hybrid hy-
drogel minimized cartilage loss in surgically induced OA model. This study demonstrates that inflammation-
modulating HA-based hydrogel may provide a therapeutic option for OA treatment.

1. Introduction to have therapeutic effects such as anti-cancer, anti-oxidant, and anti-


inflammation for many diseases [7–10]. Found in green tea, epigallo-
Osteoarthritis (OA) is known as the most common chronic disease of catechin-3-gallate (EGCG) is one of the polyphenols that can scavenge
the cartilage. Risk factors include age, obesity and traumatic injury, radical oxygen species and prevent inflammation-induced oxidative
which lead to excessive or altered joint loading. As the cartilage breaks damage. Furthermore, EGCG has been shown to directly suppress in-
down due to wear-and-tear and/or mechanical stress, it causes swelling, flammation in various cell types, including vascular endothelial cells,
pain and inflammation, leading to extracellular matrix (ECM) loss [1]. fibroblasts, and chondrocytes [11,12]. EGCG has been shown to directly
The imbalance between anabolic and catabolic processes destroys inhibit upstream targets of pro-inflammatory signaling cascades by in-
normal cartilage ECM turnover and homeostasis [2,3]. The main cata- hibiting phosphorylation of p38 and JNK, and limiting nuclear trans-
bolic cytokine involved in OA pathogenesis is interleukin-1 beta (IL- location of NF-kB [8,13,14]. Over the last decade, many studies have
1β). OA patients have elevated levels of IL-1β in the synovium and demonstrated inflammation-regulating and chondroprotective effects of
cartilage [4]. IL-1β significantly affects the metabolism of the chon- EGCG on monolayer-cultured chondrocytes [12,15,16]. EGCG has been
drocytes and its ECM [5]. A previous study by Stove et al. demonstrated shown to inhibit metalloproteinases (MMP13) cyclooxygenase-2 (COX-
that IL-1β blocks the synthesis of ECM components such as type II 2), and nuclear factor-kappa B (NF-kB) in IL-1-stimulated chondrocytes
collagen (COL2) and aggrecan (ACAN) [6]. Thus, anti-inflammatory [17–19].
medications deserve attention in therapeutic strategies to delay OA Current treatments are limited to mitigation of OA symptoms by
progression. taking some analgesics and nonsteroidal anti-inflammatory drugs
Nowadays, many researchers have focused their attention on poly- (NSAIDs), or intra-articular injection of steroid drugs [20]. These
phenol that is a pharmaceutical-grade nutrient. Polyphenols are known treatments, when carried out long-term, have detrimental side effects


Corresponding author at: School of Chemical and Biological Engineering, Seoul National University, Republic of Korea.
E-mail address: nshwang@snu.ac.kr (N.S. Hwang).
1
These two authors contributed equally to this paper.

https://doi.org/10.1016/j.msec.2020.111096
Received 8 January 2020; Received in revised form 23 April 2020; Accepted 12 May 2020
Available online 15 May 2020
0928-4931/ © 2020 Elsevier B.V. All rights reserved.
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

on gastrointestinal and cardiovascular systems. Therefore, OA therapy 2.3. Hydrogel preparation


still needs development. The on-site delivery of drugs via controlled
releasing systems is a new breakthrough. For the delivery system, Synthesized HA_t was dissolved in PBS at 4% or 10% (w/v). In ad-
hyaluronic acid (HA)-based hydrogel has been established as a useful dition, gelatin was dissolved in PBS at 10% (w/v). Then HA_t and ge-
vehicle for drug delivery [21]. HA is a substance that occurs naturally in latin solutions were sterilized by 0.22 μm syringe filter and mixed at 1:1
the joints and is known as a major component of GAG. HA is widely ratio to make 2% HTG (2% HA_t and 5% gelatin) and 5% HTG (5% HA_t
used in many tissue engineering approaches because of its biocompat- and 5% gelatin) hydrogel precursors. 0% HTG hydrogel precursor was
ibility, biodegradability and low immunogenicity. Since the elimination composed of 5% (w/v) gelatin solution only (without HA_t). Upon
of injected HA occurs rapidly, HA is usually crosslinked to last longer precursor solution preparation, 6 μM of tyrosinase (SA_Ty) was added
[22,23]. So, HA-based hydrogels are widely studied for OA treatment. into the precursor solution to induce enzymatic cross-linking as pre-
However, HA-based hydrogels have limitation cell adhesion [24]. Ge- viously described [28]. HTG hydrogel containing EGCG (HTG-E) were
latin, the production of collagen hydrolysis, contains many Arg-Gly-Asp fabricated by physical mixing of EGCG (0–100 μM) into HTG hydrogel
(RGD) residues that can provide a good cell adhesive and proliferative precursor solution prior to addition of tyrosinase.
environment [25,26]. In this study, we fabricated hybrid hydrogels that
consist of HA and gelatin to mimic the cartilage ECM. For HA and ge- 2.4. Rheological test
latin hydrogel formation, HA was conjugated with tyramine (HA_t) to
crosslink via oxidative reaction of tyramine residues [27]. Recently, we For rheological measurements, hydrogels were fabricated in 8 mm
have reported enzyme-based hydrogel crosslinking systems [27,28]. diameter and 2 mm thickness cylindrical PDMS mold. The test was
Enzyme-mediated HA_t hydrogel has advantages, including rapid carried out in Anton Paar Korea's demo lab using a rheometer (MCR
crosslinking, non-cytotoxicity, and injectability. 302, Anton-Paar). Amplitude sweep was performed with increasing
In this study, we investigated EGCG-loaded HA/gelatin (HTG-E) shear strain (%) from 0.1 to 10%. Frequency sweep was carried out at a
hybrid hydrogel for OA therapy. We first studied EGCG and HA/gelatin constant strain of 1% from 0.1 to 10 Hz. To determine the gelation time,
(HTG) hybrid hydrogel, independently. The evaluation of the anti-in- time-dependent storage (G′) and loss (G″) moduli at 1% shear strain and
flammatory effect of EGCG was performed by OA-like chondrocyte 10 Hz measured upon adding SA_Ty. The time at which G′ crosses over
laden hydrogel. The chondrogenic potential of HTG hydrogel was in- G″ indicates sol-to-gel transition, which corresponds to gelation.
vestigated by cartilage formation for 3 weeks in vitro. Finally, we
combined the composite hydrogel with EGCG to provide a suitable 3- 2.5. FT-IR analysis and swelling ratio analysis
dimensional (3D) environment for the growth of inflammation-induced
chondrocyte in vitro. The in vivo evaluation of EGCG-loaded composite For FT-IR analysis, prepared HTG hydrogels and HTG-E hydrogels
hydrogel demonstrated a synergetic anti-inflammatory and chon- were lyophilized and analyzed via FT-IR Spectrometer (PerkinElmer).
droprotective effects on OA therapy. We have established a novel For measuring the swelling ratio (Q), hydrogels (n = 3) were immersed
method for OA therapy with inflammation-modulating HA and gelatin- in PBS for 24 h. The swollen (Wwet) and dry (Wdry) weights of the hy-
based hybrid hydrogels. drogels were measured. The swelling ratio was calculated by the fol-
lowing Eq. (1).
2. Materials and methods Wwet
Q=
Wdry (1)
2.1. Porcine articular chondrocyte isolation

Articular cartilage was harvested from femoral condyle of domestic 2.6. Enzymatic degradation test
pigs, which were purchased from a local slaughterhouse. Cartilage
tissue was minced into 1mm3 pieces and digested with 0.2% (w/v) type Hydrogels were prepared at the same volume (50 μL/construct;
II collagenase for 18 h on an orbital shaker at 37 °C. Subsequently, the n = 3/time point). 10 units of hyaluronidase (Worthington) and col-
digested cell suspension was strained by 70 μm cell strainer and cen- lagenase II (Worthington) were used independently for enzymatic de-
trifuged. The cell pellet was washed 2–3 times with PBS and cultured in gradation test. Enzyme solution was changed daily. Each time point
high glucose Dulbecco's modified Eagles' medium (Hyclone), supple- samples were collected and lyophilized, and the dry weight was mea-
mented with 10% (v/v) fetal bovine serum (Corning), 1% (v/v) peni- sured. The weight loss of the dry weight of hydrogel was calculated.
cillin/streptomycin (Gibco), 10 mM HEPES (Gibco), 0.1 mM NEAA
(Gibco), 8 μg mL−1 proline (Sigma-Aldrich), and 50 μg mL−1 ascorbic 2.7. Cell encapsulation
acid (Sigma-Aldrich).
Articular chondrocytes (Passage 1–2) were prepared for 3D cell
2.2. Tyramine-conjugated HA synthesis encapsulation. Cells were initially suspended with HTG hydrogel pre-
cursor solution containing EGCG (HTG-E). Then, SA_Ty was added for
HA_tyramine (HA_t) synthesis was performed by EDC/NHS coupling hydrogel formation and cell encapsulation. The final cell concentration
protocol which was demonstrated by previous work [27]. Briefly, so- in the hydrogel was 2 × 107 cells mL−1. After gelation, cell-laden
dium hyaluronate (Lifecore Biomedical) was dissolved into 0.2 M 2-(N- hydrogels were transferred to 24-well plate and supplied with DMEM,
morpholino) ethanesulfonic acid (MES) buffered saline (Thermo Fisher supplemented with 1% penicillin/streptomycin, 1% ITS + Premix (BD
Scientific) to make 1% (w/v) HA solution. While stirring, N-hydro- Biosciences), 100 nM dexamethasone (Sigma-Aldrich), 50 mg mL−1
xysulfosuccinimide (sulfo-NHS; Sigma-Aldrich) and N-(3-dimethylami- ascorbic acid-2-phosphate (Sigma-Aldrich), and 10 ng mL−1 TGF-β3
nopropyl)-N-ethylcarbodiimide (EDC; Sigma-Aldrich) were added to (Life technologies). The media was changed every 2 days. Encapsulated
the HA solution. Tyramine hydrochloride (Sigma-Aldrich) was added at chondrocytes were treated with 10 ng mL−1 IL-1β to induce OA-like in
1:1 mole ratio to HA. The reaction was performed overnight, and the vitro condition.
product was dialyzed against distilled water for 3 days. Synthesized
HA_t was lyophilized for later use. 2.8. Cytotoxicity evaluation
HA_t was characterized via 1H NMR. Peaks at 7.3 and 7.5 ppm are
due to aromatic protons of tyramine. The tyramine content in HA_t was After cell encapsulation, cell cytotoxicity was measured by live/
determined to be 13.38%. dead assay kit (Invitrogen), according to the manufacturer's instruction.

2
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

Table 1 2.12. Histological analysis


Primer list.
Primer sequences (5′-3′) The in vitro hydrogel samples were collected after 3 weeks of cul-
ture, and mice with intra-articular injection were sacrificed 4 weeks
GAPDH Forward TCA GCT GCT GGG GAG TCA CA post-injection. All samples were fixed with 4% PFA at 4 °C for 24 h.
Reverse CCT AAG CCC CTC CCC TTC TT
Mice joint samples were decalcified in 0.5 M
IL-1β Forward TCT CTG GAC CCA AAG GAG GG
Reverse ACG GCT GCT TTC ACG GGT GA
Ethylenediaminetetraacetic acid (Sigma) for a week. Fixed cell-laden
TNF-a Forward AGC TGG TGG TGC CGA CAG AT hydrogels and mice joint samples were then embedded in paraffin
Reverse TGC GAT GCG GCT GAT GGT GT blocks and sectioned into 3-μm thickness slides. For histological
MMP13 Forward CCT GTG CTC CTG CCA TTT GG staining analysis, the sections were deparaffinized and rehydrated. For
Reverse GAA TGG GCA GCT CCA TGG CT
H&E staining, sections were stained with hematoxylin (Ricca Chemical
ADAMTS5 Forward CGC TCC CTG GCT GTC TTT GA
Reverse TCA AAG TGT AGC GCG CGT GC Company) for 5 min and Eosin-Y (Richard-Allan Scientific) for 1 min,
COL2 Forward TAG AGA GGT TTC CTG GGC CG followed by dehydration in graded series of ethanol. For Safranin-O
Reverse AGG AGG ACG CTG GAA CAG AG staining, the sections were dipped in 0.1% (w/v) Safranin-O solution
SOX9 Forward CAG TCC CAG CGA ACG CAC AT
(ScholAR Chemistry) for 5 min, followed by dehydration in ethanol.
Reverse TGC TGC TGC TGC TCG CTG TA
ACAN Forward CTG CCC CGA AAC ATC ACC GA
Types I, II and X collagen immunofluorescence staining was per-
Reverse GTA AAG GGC TCC TCA GGC TC formed on sectioned slices of in vivo samples. Deparaffinized sections
were permeabilized in 0.1% (v/v) Triton-X (Sigma), and blocked with
10% (v/v) normal goat serum for 45 min. Sections were incubated with
Cell-laden hydrogels were stained for 20 min and washed with PBS. anti-collagen II (ab34712, Abcam; 1:100 dilution), anti-collagen X
Live/dead images were captured via LSM 720 confocal microscope (ab58632, Abcam; 1:100 dilution) or anti-collagen I (ab3471, Abcam;
(Zeiss) or EVOS FL Imaging System AMF4300 (Life Technologies). Cell 1:100 dilution) at 4 °C for overnight. After washing thrice with 0.1%
viability was quantified via ImageJ software. BSA/PBS, sections were incubated with secondary antibodies (Goat
anti-rabbit Alexa-Fluor 546 or 488; Life Technologies). The nuclei were
stained with DAPI for 5 min at room temperature. Imaging was carried
2.9. Real-time PCR analysis out using EVOS FL Imaging System AMF4300 (Life Technologies).
ImageJ was used to quantify the average cartilage thickness from
Cell-laden hydrogels (n = 3) were collected on week 3 for gene Safranin-O staining and the mean fluorescence intensity of immuno-
expression analysis. Samples were ground within TRIzol reagent (Life fluorescence stainings.
Technologies) and total mRNA was extracted. The RNA concentration
was measured using Infinite 200 PRO NanoQuant Microplate Readers 2.13. Statistical analysis
(TECAN). 1 μg of RNA was used for cDNA synthesis by M-MLV cDNA
Synthesis Kit (Enzynomics), according to the manufacturer's instruc- All data are presented as mean ± standard deviation (SD).
tion. Real-time PCR was performed using SYBR Green PCR Master Mix Statistical significance between groups was determined by one-way
(Enzynomics) and ABI StepOnePlus™ Real-Time RCR system (Applied ANOVA with *p < 0.05, **p < 0.01, ***p < 0.005.
Biosystems). Primer sequence of glyceraldehyde 3-phosphate dehy-
drogenase (GAPDH, housekeeping gene), type II collagen (COL2), Sex 3. Results
determining region Y box 9 (SOX 9), aggrecan (ACAN), Tumor necrosis
factor-alpha (TNF-α), interleukin 1 beta (IL-1β), Matrix metalloproteinase 3.1. Effect of EGCG on 3D-encapsulated chondrocytes
13 (MMP13) and A disintegrin and metalloproteinase with thrombospondin
motifs 5 (ADAMTS5) are listed in Table 1. The relative gene expression EGCG is a polyphenolic bioactive compound isolated from green tea
was calculated by −2ΔΔCt method. extracts [30]. Many studies have demonstrated the therapeutic effects
of EGCG on inflammatory diseases [31,32]. However, previous studies
that claim the therapeutic potential of EGCG were limited to 2D cell
2.10. Biochemical assay culture. In this study, we aimed to fabricate immune-modulating hy-
drogel composed of EGCG and apply this hydrogel to OA disease model
For biochemical analysis, cell-laden hydrogels (n = 3) were di- for functional recovery. EGCG-incorporated hydrogels were fabricated
gested using papain solution (Worthington) for 16 h at 60 °C. DNA by introducing various amounts of EGCG into the mixture of HA_t and
content was quantified by Quant-iT™ Pico-Green®dsDNA assay kit gelatin. We have previously reported the enzyme kinetics of SA_Ty for
(Invitrogen) according to the manufacturer's instruction. sGAG accu- facilitated enzyme-based crosslinking of phenolic compounds [27].
mulation was measured by dimethylmethylene blue (DMMB) spectro- Therefore, a combination of HA_t, gelatin and EGCG can be crosslinked
photometric assay at A525. For total collagen content, the hydro- to form a hydrogel in a rapid manner by SA_Ty. EGCG-loaded HA_t/
xyproline amount was measured by chloramine-T colorimetric assay. gelatin (HTG-E) hydrogels were examined for their in vitro anti-in-
flammatory responses and subsequently, applied to murine OA model
to evaluate therapeutic efficacy (Fig. 1).
2.11. Intra-articular injection In order to optimize EGCG concentration, we examined cytotoxicity
and anti-inflammatory effects of EGCG on OA chondrocyte in a 3D
Twenty male Balb/c mice (10-week-old) were used for the in vivo environment. Chondrocytes (2 × 107 cells/construct, 50 μL/construct)
experiment following the protocols approved by the Institutional were encapsulated in 5% (w/v) gelatin hydrogel crosslinked with
Animal Care and Use Committee (IACUC) of Seoul National University. SA_Ty, containing various concentrations of EGCG (0–100 μM).
The mice were randomly divided into 5 groups (n = 4/group): normal, Encapsulated chondrocytes were then treated with 10 ng mL−1 IL-1β
PBS, EGCG 50 μM, 5% HTG and 5% HTG-E. OA induction surgery was throughout the entire culture period to re-create an OA-like condition in
performed by anterior cruciate ligament transection (ACLT) [29]. A vitro. After 1 day of encapsulation, live/dead staining showed that
week after surgery, 5 μL of PBS, 50 μM EGCG, 5% HTG or 5% HTG-E EGCG did not cause significant cytotoxicity in 3D culture systems
were injected bilaterally into the knee. After 4 weeks post-injection, the (Fig. 2A, B). OA chondrocytes maintained above 90% viability even at
knee joint samples were collected for histological analysis. 100 μM EGCG concentration. Interestingly, in the monolayer culture

3
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

Fig. 1. Schematic illustration of evaluating inflammation-modulatory function of HA-based hydrogel.

system, EGCG concentration above 20 μM showed cytotoxicity, which inflammatory cytokine in synovial fluid that causes inflammatory
may be attributed to its low stability in free-standing form (Supple- symptoms in OA. To mimic OA inflammation, chondrocytes were en-
mentary Fig. 3A). The polyphenol can undergo fast degradation in capsulated in the gelatin-based hydrogel and maintained with culture
aqueous solution via auto-oxidation or epimerization, and the de- media containing IL-1β (10 ng mL−1). After 2 days, cell-laden con-
gradation products may have damaging effects on cells [33]. These structs were collected for real-time PCR analysis (Fig. 2C). The in-
results suggest that SA_Ty successfully incorporated EGCG into the flammatory and catabolic genes were all increased after IL-1β stimu-
hydrogel as crosslinkers and allowed limited free-standing EGCG in the lation. However, EGCG-dependent downregulation of inflammatory
medium. We observed that EGCG did not cause any cytotoxicity to response genes was observed. In particular, gelatin hydrogel with >
chondrocyte when incorporated into the hydrogel. Therefore, hydrogels 30 μM EGCG concentration resulted in significant downregulation of
incorporated with EGCG can support cell survival for long-term culti- IL-1β, TNF-α, MMP13, and ADAMTS5. Gene expression of IL-1β and
vation. TNF-α showed similar levels as that of the control group with the 30 μM
The anti-inflammatory effect of EGCG on monolayer-expanded EGCG incorporation. In addition, MMP13 and ADAMTS5 gene expres-
chondrocytes was reported in several studies [12,16,34]. These studies sion levels were downregulated to that of the control group with 50 μM
demonstrated that EGCG suppressed the expressions of pro-in- EGCG incorporation. It demonstrated that EGCG has the ability to
flammatory genes such as COX2, MMP1, MMP13, and ADMTS5 [35,36]. regulate inflammation-induced by IL-1β in the hydrogels. Based on real-
We evaluated whether incorporation of EGCG into the hydrogel would time PCR analysis, we selected 50 μM EGCG for further in vitro and in
exhibit similar effects on OA chondrocytes. IL-1β is the main pro- vivo applications.

Fig. 2. EGCG effect on 3D encapsulated OA chondrocytes. (A) Live/dead assay for dose-dependent EGCG treatment; Live cell was stained with by Calcein AM (green)
and dead cell was stained by Ethidium homodimer-1 (red). Scale bar = 100 μm. (B) Cell viability assay was measured by AlamarBlue™ reagent. (C) Real-time PCR
analysis of IL-1β, TNF-α, MMP13 and ADAMTS5 genes (n = 3), RFI = relative fold induction. *p < 0.05, **p < 0.01, ***p < 0.005. (For interpretation of the
references to color in this figure legend, the reader is referred to the web version of this article.)

4
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

Fig. 3. Characterization of HTG hydrogel. (A) Gross images of 0%, 2% and 5% HTG hydrogels. The final concentration of each hydrogel was composed of 0%, 2%, 5%
(w/v) HA_t and 5% (w/v) gelatin. (B) SEM images of internal pore structure at 100× magnification. (C) Rheological analysis of HTG hydrogels to determine gelation
time. G′ = storage modulus and G″ = loss modulus. (D) Rheological analysis of HTG to determine viscoelastic properties of pre-formed hydrogels. (E) Swelling ratio
(n = 3). (F) Enzymatic degradation of HTG hydrogels using 10 U/mL of each enzyme (n = 3). *p < 0.05, **p < 0.01, ***p < 0.005. (For interpretation of the
references to color in this figure, the reader is referred to the web version of this article.)

3.2. Characterization of HTG hydrogel and its chondrogenic effects sweep exhibited HA_t concentration-dependent manner. 5% HTG hy-
drogel showed the highest storage modulus (G′). The swelling ratio
It is well known that HA promotes chondrogenesis because HA is a analysis showed reciprocal trends as the storage modulus G′ (Fig. 3E).
major component of the cartilage ECM [37]. As a natural polymer, non- 5% HTG hydrogel showed the lowest degree of water uptake. Both
crosslinked, unmodified HA has limitations in use for cartilage tissue rheology and swelling test results confirmed the highest crosslinking
engineering, such as rapid clearance and low mechanical properties. degree and strength of 5% HTG hydrogel.
Several approaches to crosslink HA have been studied [38]. In a pre- We also measured enzymatic degradation of hydrogels (Fig. 3F).
vious study, our lab reported the fabrication of HA-based injectable The hydrogels were exposed to hyaluronidase or collagenase
hydrogel [27]. Briefly, tyramine hydrochloride was conjugated onto HA (10 U mL−1). After 2 weeks of hyaluronidase degradation, all groups
backbone via EDC/NHS chemistry. Then, tyramine-conjugated were degraded completely. However, in the collagenase degradation
HA(HA_t) was mixed with gelatin and crosslinked via tyrosinase- test, all groups except 0% HTG hydrogel remained after 2 weeks. This
mediated oxidative reaction. This enzyme-based crosslinking is rela- demonstrated that gelatin-related crosslinking undergoes rapid enzy-
tively rapid and can be applied as an injectable system. 2% or 5% (w/v) matic degradation and HA_t-related crosslinking is strong so that the
HA_t was mixed with 5% (w/v) gelatin to produce 2% or 5% HTG hy- degradation is not easy.
drogels. We used gelatin without HA_t as a control hydrogel (0% HTG).
Since gelatin itself has tyrosinase-reactive functional groups (i.e. amine 3.3. Evaluation of chondrogenic effects of HTG hydrogel
and tyrosine), HA/Gelatin hydrogel without HA_t showed brown color
(0% HTG), as a result of SA_Ty crosslinking. Additionally, HA/Gelatin To evaluate whether HTG hydrogel can be applied to cartilage tissue
(HTG) hydrogels showed gradually darker brown color as HA_t con- regeneration, porcine articular chondrocytes (2 × 107 cells/construct)
centration was increased (Fig. 3A). The rheological properties of HTG were encapsulated in HTG hydrogels and evaluated the tissue con-
hydrogels were evaluated by the rheometer (Fig. 3C & D). Gelation structs after 3 weeks of culture. After 3 weeks of culture, constructs
time, which can be determined from the time point at which G′ exceeds were harvested and biochemical analysis was performed. We quantified
G″, was 420 s, 108 s and 132 s for 0%, 2% and 5% HTG, respectively. As sulfated GAG amount by DMMB assay and collagen content by hydro-
expected, gelation time was much faster for HA_t-containing groups xyproline content analysis. All the measurements were normalized to
than 0% HTG hydrogel. However, 5% HTG hydrogel exhibited slightly the respective DNA amount. 0% HTG hydrogel on day 1 served as
slower gelation time than 2% HTG hydrogel, presumably due to high control. As HA_t content increased, overall collagen and GAG contents
viscosity of 5% HTG precursor solution. Both amplitude and frequency within the hydrogel also increased (Fig. 4A). Additionally, 5% HTG

5
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

Fig. 4. Comparison of the chondrogenic properties of 2% and 5% HTG hydrogel through in vitro cell encapsulation. (A) Biochemical analysis of sGAG, collagen and
DNA content (n = 3). 0% HTG hydrogel collected at day 1 was served as control. (B) Histological analysis of H&E, Safranin-O and type II collagen im-
munohistochemistry. Scale bar = 200 μm (C) Real-time PCR results of COL2, SOX9, and ACAN genes (n = 3). 0% HTG hydrogel collected on day 1 served as control.
RFI = Relative fold induction. *p < 0.05.

6
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

exhibited the highest level of collagen compared to 0% and 2% HTG Furthermore, in 5% HTG-E, cartilage surface and thickness (Fig. 6B)
groups. Histological analysis confirmed with biochemical analysis were completely intact, showing no sign of wear and tear.
(Fig. 4B). H&E staining showed reduced cellularity in 5% HTG com- Types I and X collagen, which are absent in healthy articular car-
pared to the 2% HTG, confirming chondrocytes within 5% HTG pro- tilage, have been additionally examined to observe chondrocyte ded-
duced more ECM. Additionally, Safranin-O staining showed intense red- ifferentiation and hypertrophy in OA mouse knee joints. Elevated ex-
stained area in 5% HTG hydrogel compared to 2% HTG hydrogel as a pression levels of collagen I and X were observed in PBS-injected OA
result of an increased in GAG accumulation within the hydrogel. Fi- joints, while OA joints that received any form of treatment indicated
nally, type II collagen immunostaining showed that the cells in 5% HTG lower expression levels of these markers. In particular, the mean
hydrogel retained circular morphology and type II collagen was re- fluorescence intensity of 5% HTG-E group had no significant difference
tained within the cellular periphery. Cells in 2% HTG hydrogels were with that of normal group (Fig. 6C). These results indicate that the
spindle-shaped and elongated. Furthermore, counterstaining with DAPI combination of EGCG and HTG hydrogel retarded the progression of
confirmed increased cellularity within 2% HTG hydrogel. Interestingly, OA, while each of the components alone also had mild beneficial ef-
0% HTG (HA/Gelatin hydrogel without HA_t), resulted in complete fects.
degradation within 5 days of culture. This may be due to the reduced
crosslinking of the hydrogel as a result of 0% HA_t along with active 4. Discussion
cellular reorganization and cell-mediated degradation.
We also measured cartilage-specific gene expression levels of the OA is a type of cartilage degenerative disease that results from the
cell-laden HTG hydrogels. Real-time PCR was used to compare the level breakdown of joint cartilage and underlying bone [39]. With OA pro-
of chondrogenic genes such as COL2, SOX9 and ACAN at week 3. The gression, ECM components in cartilage degrade gradually as pro-in-
data is shown in Fig. 4C. All gene expressions were normalized to 0% flammatory environment promotes ECM deconstruction. IL-1β is a main
HTG hydrogel collected on day 1. Real-time PCR data showed similar pro-inflammatory cytokines in osteoarthritic joint synovium. Therefore,
trends as biochemical analysis. Chondrogenic gene levels of 5% HTG the sequestration of pro-inflammatory cytokine like IL-1β may have
hydrogel group were higher than those of other groups. We confirmed significant therapeutic benefits in preventing OA progression and re-
that HTG composite hydrogel can promote chondrogenesis, with higher storing tissue homeostasis.
chondrogenic property as HA content increases. In recent years, several biomaterials or cell secretome-based ap-
proaches have attempted to modulate the immune response in a way
3.4. Synergetic effect of HTG hydrogel combined with EGCG (HTG-E) that can more effectively promote cartilage regeneration at the site of
injury [26,40–43]. These include the modulation in biomaterial surface
To confirm the synergetic effect of EGCG and HTG hydrogel, we chemistry to influence protein adsorption. The selectivity of adsorbed
encapsulated chondrocytes in 5% HTG hydrogels or 5% HTG-E hydro- proteins on biomaterials may mediate the interactions with immune
gels (5% HTG hydrogel with 50 μM EGCG) and treated with cells, which may ultimately control immune cell activation. In addition,
10 ng mL−1 of IL-1β for 3 weeks to mimic inflammatory OA environ- a strategy of chemical crosslinking of natural ECM has been employed
ment. EGCG was mixed with hydrogel precursors to evaluate its anti- to increase the stability of naturally derived biomaterials. Interestingly,
inflammatory effect. Fig. 5A showed histological analysis 5% HTG and recent evidence supports the notion that types of crosslinking strategies
5% HTG-E cultured for 3 weeks. Safranin-O staining indicated more may impact the immune response. Recently, Brown et al. showed that
GAG accumulation in 5% HTG-E group than 5% HTG group. Type II acellular ECM without chemical modification often results in M2
collagen immunostaining results showed collagen production in the macrophage response while carbodiimide (CDI) cross-linked scaffolds
pericellular area in both groups. However, the expression of type II induce M1 response [44]. In our study, we employed tyrosinase-in-
collagen protein was more prominent in 5% HTG-E hydrogel compared duced crosslinking of tyramine-conjugated HA and gelatin. Tyrosinase
to the EGCG-absent group. The presence of IL-1β leads to cartilage ECM activity is involved in our body as a natural crosslinking process that
degradation in OA knee joint. This may lead to reduced syntheses of induces the accumulation of melanin. Our previous report has sug-
ECM like GAG and type II collagen by chondrocyte in 5% HTG, which gested that SA_Ty based crosslinking of natural ECM (such as gelatin)
were preserved in EGCG-containing hydrogel. EGCG not only has an allowed a minimal immune response. Furthermore, SA_Ty-based
anti-inflammatory effect as shown in Fig. 2C but also a chon- crosslinking allowed a very stable hydrogel system that can withstand
droprotective effect in long-term inflammation. collagenase or hyaluronidase-induced degradation (Fig. 3F). In addi-
The chondrogenic and inflammatory gene expressions were eval- tion, SA_Ty-based crosslinking system not only allowed in situ hydrogel
uated by real-time PCR (Fig. 5B and C). As anticipated, inflammatory formation but also induced stable adhesion of the HTG hydrogel onto
gene expressions were down-regulated in the EGCG-containing group, the cartilage surface. In a previous study, Liu et al. fabricated a free
confirming EGCG's inflammatory effects in the HTG hydrogel in vitro. radical scavenging HA-EGCG conjugate hydrogel that is crosslinked via
The chondrogenic genes were upregulated in 5% HTG-E group, which is horseradish peroxide and hydrogen peroxide, which is injectable and
consistent with the histological analysis. anti-inflammatory like our HTG-E hydrogel [45]. However, under our
hydrogel system, tyramine residues conjugated on HA backbone and
3.5. In vivo OA treatment with HTG-E hydrogel in mouse OA model amine functional groups of gelatin enable a high degree of adhesion
onto cartilage surface, a huge advantage for intra-articular injection.
EGCG with HTG hydrogel was intra-articularly administered to OA- We and others have demonstrated injectability of a tyrosinase-based
induced mice to determine the effect on OA progression. A week after crosslinking system and adhesiveness of gelatin-based hydrogels on
OA induction surgery, PBS, 50 μM EGCG, 5% HTG, and 5% HTG-E were various tissue surface [27,28,46]. As the hydrogel can imbibe a large
each injected into the knee. The knee joints were collected for histo- amount of water, our HTG hydrogel may have contributed to the se-
logical analysis after 4 weeks (Fig. 6A). Safranin-O staining and type II questration of inflammatory cytokines upon injection into the OA an-
collagen immunostaining demonstrated that the PBS injection group imal model, creating a local anti-inflammatory microenvironment that
had very severely eroded and thinned cartilage and notable GAG and influenced the regeneration.
type II collagen loss. Although less severe than the PBS group, 50 μM In this study, we also incorporated EGCG and hyaluronic acid into
EGCG and 5% HTG hydrogel groups showed some degree of cartilage our hydrogel platform to the added benefit of controlling the OA in-
ECM loss and surface erosion compared to normal cartilage. When flammatory microenvironment. EGCG is a polyphenolic compound that
EGCG and the hydrogel were injected together as in 5% HTG-E group, has been shown to possess anti-inflammatory activity with a potential
GAG and collagen content were up to par with that of native cartilage. for anti-arthritic effects [15]. Due to its phenolic functional groups, it

7
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

Fig. 5. In vitro evaluation of 5% HTG and 5% HTG-E hydrogel in OA mimetic environment. (A) Histological analysis of H&E, Safranin-O and type II collagen
immunohistochemistry. Scale bar = 200 μm (B) Real-time PCR results of IL-1β, MMP13, and ADAMTS5 (n = 3). (C) Real-time PCR results of COL2, SOX9, and ACAN
(n = 3). RFI = Relative fold induction. *p < 0.05, **p < 0.01.

can scavenge radical oxygen species (ROS), which are overproduced in have confirmed that chondrocytes in pathophysiological conditions
inflammatory osteoarthritic joints. Elevated ROS levels put oxidative exhibited significant downregulation of inflammatory gene expression
stress on chondrocytes, leading to cell senescence, apoptosis as well as levels when treated with EGCG (Fig. 2C). However, we noticed that
lower levels of ECM synthesis and elevated levels of ECM degradation. EGCG over 20 μM resulted in cellular cytotoxicity in monolayer (Sup-
By scavenging ROS, EGCG can rescue chondrocytes from inflammation- plementary Fig. 3A). However, when we observed cell viability in the
induced cell damage. Moreover, as mentioned previously, EGCG can HTG-E hydrogel, we were able to load up to 100 μM of EGCG onto the
suppress pro-inflammatory signaling cascades by inhibiting phosphor- HTG-E hydrogel with minimal cytotoxicity. This could be the result of
ylation of p38, JNK and nuclear translocation of NF-kB. Indeed, we covalent conjugation of EGCG (with its phenolic functional groups)

8
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

Fig. 6. Intra-articular injection of 5% HTG-E hydrogel in OA mouse joint. (A) Histological analysis by Safranin-O and immunofluorescence staining of types I, II, X
collagen. Scale bar = 200 μm. (B) Quantification of cartilage thickness. (C) Quantification of mean fluorescence intensity (MFI) of types I, II and X collagen
immunofluorescence staining. *p < 0.05, **p < 0.01, ***p < 0.005.

onto HA_t or gelatin, resulting in covalent incorporation of EGCG onto hydrogels promoted the GAG and collagen II accumulation [49,50]. In
the hydrogel. Indeed, physical and mechanical properties comparing accordance with the previous study, we also demonstrated that higher
5% HTG vs 5% HTG-E showed slightly decreased swelling ratio along the HA content more ECM was accumulated within the hydrogel [51].
with increased mechanical properties as a result of EGCG incorporation Histological analysis and immunostaining confirmed the increased ECM
(Supplementary Fig. 2). As a significant amount of added EGCG was accumulation in 5% HTG when compared to the 2% HTG constructs.
covalently incorporated onto the hydrogel, we hypothesize that EGCG- Interestingly, cellular phenotype and organization of type II collagen
induced cytotoxicity was minimized in the 3D hydrogel environment were drastically different in 5% HTG versus 2% HTG (Fig. 4B). In 5%
compared to the 2D culture platform. In addition, our study demon- HTG hydrogel, chondrocytes maintained circular phenotype and type II
strates the added benefit of incorporating EGCG onto the tyrosinase- collagen were typically located around the cells. However, within 2%
induced crosslinking hydrogel for effective anti-inflammatory control HTG hydrogel, cells appeared to be stretched and collagen orientation
via EGCG. was distinct. Relatively lower HA content in 2% HTG hydrogel may
HA is one of the most abundant glycosaminoglycans (GAGs) in the have allowed increased cell adhesion to gelatin within the hydrogel.
cartilage ECM. In addition, HA in synovial fluid provides lubrication, Although cellular adhesion to biomaterials is beneficial for cell survival
along with lubricin [47]. Even though the therapeutic effect of intra- and proliferation, cytoskeletal tension within chondrocytes is re-
articular injection of HA to OA joint has been reported the HA injection sponsible for dedifferentiation and phenotypical change to fibroblast-
provide only a transient treatment option for OA patient as it is not like morphology. This is one of the primary reasons why chondrocytes
directly involved in the cartilage regeneration and has relatively rapid cultured in 2D platform are not able to synthesize GAGs and down-
clearance [48]. As a way to provide a cell-friendly environment for the regulate chondrocyte-specific genes such as COL2, SOX9, and ACAN
cartilage and provide a lubricating component to the injected hydrogel, [52]. In contrast, 3D cultivation of chondrocytes can support the re-
we introduced tyramine-conjugated HA. differentiation by keeping the cell in spherical shape [53]. In addition
When chondrocytes were encapsulated in HTG hydrogel of various to cytoskeletal tension due to cell adhesion, different physical proper-
HA_t concentration, we observed a HA_t concentration-dependent ECM ties of the HTG hydrogels may have influenced chondrocyte phenotype
accumulation. Previously, crosslinked HA-based hydrogel has been within the hydrogel. Compared to 5% HTG hydrogel, 2% HTG hydrogel
extensively studied for cartilage tissue engineering for past decades. has a higher swelling ratio and larger internal pores as seen in SEM
Levett et al. and Moulisova et al. have reported crosslinked HA-based images (Fig. 3B and E). This implies that a larger living space is

9
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

available in 2% HTG hydrogel for encapsulated chondrocytes, leading CRediT authorship contribution statement
to a higher degree of cell spreading and dedifferentiation. Toh et al. and
many others have reported that chondrocytic phenotype is more well- Yin Ji Jin: Writing - original draft, Methodology, Formal analysis,
maintained on lower stiffness hydrogels [54,55]. Although 2% HTG has Investigation. Rachel H. Koh: Writing - review & editing,
lower stiffness than 5% HTG hydrogel, the relative availability of cell Methodology. Su-Hwan Kim: Conceptualization. Kyung Min Kim:
adhesion sites and size of cell living space seemingly have more influ- Resources. G. Kate Park: Resources. Nathaniel S. Hwang:
ence on chondrocyte phenotype and genotype. Conceptualization, Funding acquisition, Supervision.
Based on EGCG-induced anti-inflammation effects and optimized
HA_t concentration to induce optimal ECM production, we incorporated Declaration of competing interest
EGCG into 5% HTG hydrogel for the treatment of OA. Prior to appli-
cation in OA animal model, cell-laden hydrogels with or without EGCG The authors declare that they have no known competing financial
were cultured in IL-1β-containing media for 3 weeks to investigate interests or personal relationships that could have appeared to influ-
whether the HTG-E hydrogel could resist to OA environment. Our ence the work reported in this paper.
findings suggested that 5% HTG-E hydrogel provides a protective effect
from the OA-like inflammatory environment. The accumulation of ECM Acknowledgments
was detected in the 5% HTG-E group rather than in the 5% HTG group,
and the inflammatory gene was down-regulated through the 3-week The Institute of Engineering Research at Seoul National University
culture. On the other hand, the cartilage forming gene was upregulated provided research facilities for this work. This research was supported
in the 5% HTG-E group. In vivo evaluation of intra-articular injection of by National Research Foundation of Korea Grant funded by the Korean
5% HTG-E showed progressive results in OA treatment. OA was in- Government (NRF-2016R1E1A1A01943393). This research was a part
itiated by destabilization of the medial meniscus, and 5 μL of hydrogels of the project titled ‘Biochemical modification of marine based bio-
(5% HTG with or without EGCG) were injected. All the tissues were materials for therapeutic ischemic cardiovascular disease treatment’,
evaluated after 4 weeks. In the PBS injection control group, the carti- funded by the Ministry of Oceans and Fisheries, Korea.
lage was completely damaged. In 50 μM EGCG and 5% HTG hydrogel
injection groups, the cartilage was thinned out, but not completely Appendix A. Supplementary data
eroded. These results indicated that injection of 50 μM EGCG or 5%
HTG hydrogel alone provided therapeutic benefit to OA. Our study is in Supplementary data to this article can be found online at https://
accordance with previous studies that reported the beneficial effects of doi.org/10.1016/j.msec.2020.111096.
EGCG on the OA model. Leong et al., have reported that the daily in-
traperitoneal injection of EGCG to OA induced mice exhibited relief of References
OA-related pain and reduction of proinflammatory cytokine levels [56].
In another study by Natarajan et al., intra-articular injections of poly- [1] R. Altman, E. Asch, D. Bloch, G. Bole, D. Borenstein, K. Brandt, W. Christy,
phenols such as EGCG and tannic acid showed resistant to degradation T.D. Cooke, R. Greenwald, M. Hochberg, et al., Development of criteria for the
despite ongoing inflammation [57]. This study was performed with classification and reporting of osteoarthritis. Classification of osteoarthritis of the
knee, Diagn. Ther. Crit. Commit. Am. Rheum. Assoc. Arthritis Rheum. 29 (8) (1986)
EGCG and Tannic acid injection every 3 days for the significant out- 1039–1049.
come. Although intra-articular injection of drugs provides one option [2] M.B. Goldring, The role of the chondrocyte in osteoarthritis, Arthritis Rheum. 43 (9)
for drug delivery in a joint, the rapid clearance of the injected drug (2000) 1916–1926.
[3] M. Daheshia, J.Q. Yao, The interleukin 1beta pathway in the pathogenesis of os-
limits the therapeutic efficacy of the intra-articular injection. teoarthritis, J. Rheumatol. 35 (12) (2008) 2306–2312.
In our study, we have allowed crosslinking of EGCG into our hy- [4] P. Wojdasiewicz, L.A. Poniatowski, D. Szukiewicz, The role of inflammatory and
drogel. Our HTG-E hydrogel can be retained in the joint for a long time. anti-inflammatory cytokines in the pathogenesis of osteoarthritis, Mediat. Inflamm.
(2014) (2014) 561459.
Furthermore, due to the adhesive property of HTG-E hydrogel, anti- [5] K.B. Marcu, M. Otero, E. Olivotto, R.M. Borzi, M.B. Goldring, NF-kappaB signaling:
inflammatory EGCG components can be retained within the surface of multiple angles to target OA, Curr. Drug Targets 11 (5) (2010) 599–613.
articular cartilage. A recent study by Emoto et al. demonstrated the in [6] J. Stove, K. Huch, K.P. Gunther, H.P. Scharf, Interleukin-1beta induces different
gene expression of stromelysin, aggrecan and tumor-necrosis-factor-stimulated gene
situ cross-linkable HA hydrogels. This HA-based hydrogel was able to
6 in human osteoarthritic chondrocytes in vitro, Pathobiology 68 (3) (2000)
sustain the release of anti-cancer drugs within several days [58]. For the 144–149.
combination treatment of EGCG and HA-based hydrogel, 5% HTG hy- [7] J.T. Hwang, J. Ha, I.J. Park, S.K. Lee, H.W. Baik, Y.M. Kim, O.J. Park, Apoptotic
drogel injection group showed hyaline cartilage similar to that of the effect of EGCG in HT-29 colon cancer cells via AMPK signal pathway, Cancer Lett.
247 (1) (2007) 115–121.
normal control group. We did not detect any wear and ECM loss. The [8] J. Yang, Y. Han, C. Chen, H. Sun, D. He, J. Guo, B. Jiang, L. Zhou, C. Zeng, EGCG
injected HTG-E hydrogel may have formed a thin layer on the cartilage attenuates high glucose-induced endothelial cell inflammation by suppression of
surface to protect cartilage from physical abrasion. Furthermore, the PKC and NF-kappaB signaling in human umbilical vein endothelial cells, Life Sci. 92
(10) (2013) 589–597.
EGCG component within HTG-E hydrogel may provide an anti-in- [9] S. Valcic, A. Muders, N.E. Jacobsen, D.C. Liebler, B.N. Timmermann, Antioxidant
flammatory microenvironment that allowed the maintenance of ar- chemistry of green tea catechins. Identification of products of the reaction of
ticular cartilage. Based on our initial finding, we believe that this in- (−)-epigallocatechin gallate with peroxyl radicals, Chem. Res. Toxicol. 12 (4)
(1999) 382–386.
flammatory modulating hydrogel can be applied in various tissue [10] M. Nakamuta, N. Higashi, M. Kohjima, M. Fukushima, S. Ohta, K. Kotoh,
augment applications. N. Kobayashi, M. Enjoji, Epigallocatechin-3-gallate, a polyphenol component of
green tea, suppresses both collagen production and collagenase activity in hepatic
stellate cells, Int. J. Mol. Med. 16 (4) (2005) 677–681.
5. Conclusion
[11] S.H. Kim, K. Kim, B.S. Kim, Y.H. An, U.J. Lee, S.H. Lee, S.L. Kim, B.G. Kim,
N.S. Hwang, Fabrication of polyphenol-incorporated anti-inflammatory hydrogel
In summary, we demonstrated that HTG hydrogel promotes ECM via high-affinity enzymatic crosslinking for wet tissue adhesion, Biomaterials 242
(2020) 119905.
accumulation and EGCG-loaded composite hydrogel has anti-in-
[12] N. Akhtar, T.M. Haqqi, Epigallocatechin-3-gallate suppresses the global interleukin-
flammatory and chondroprotective capacities in IL-1β-induced OA-like 1beta-induced inflammatory response in human chondrocytes, Arthritis Res. Ther.
environment. Injection of the EGCG-loaded composite hydrogel to 13 (3) (2011) R93.
surgically induced OA knee joint can protect the cartilage from in- [13] Y. Zhong, Y.S. Chiou, M.H. Pan, F. Shahidi, Anti-inflammatory activity of lipophilic
epigallocatechin gallate (EGCG) derivatives in LPS-stimulated murine macro-
flammation and “wear-and-tear.” Therefore, we believe that this EGCG- phages, Food Chem. 134 (2) (2012) 742–748.
loaded injectable HTG hydrogel is a promising tissue engineering [14] I.H. Jung, D.E. Lee, J.H. Yun, A.R. Cho, C.S. Kim, Y.J. You, S.J. Kim, S.H. Choi, Anti-
strategy and has great potential for clinical recovery of OA. inflammatory effect of (−)-epigallocatechin-3-gallate on Porphyromonas gingivalis

10
Y. Jin, et al. Materials Science & Engineering C 115 (2020) 111096

lipopolysaccharide-stimulated fibroblasts and stem cells derived from human per- [36] M.N. Vankemmelbeke, G.C. Jones, C. Fowles, M.Z. Ilic, C.J. Handley, A.J. Day,
iodontal ligament, J. Periodontal Implant Sci. 42 (6) (2012) 185–195. C.G. Knight, J.S. Mort, D.J. Buttle, Selective inhibition of ADAMTS-1, -4 and -5 by
[15] R. Singh, N. Akhtar, T.M. Haqqi, Green tea polyphenol epigallocatechin-3-gallate: catechin gallate esters, Eur. J. Biochem. 270 (11) (2003) 2394–2403.
inflammation and arthritis. [corrected], Life Sci. 86 (25–26) (2010) 907–918. [37] J.R. Fraser, T.C. Laurent, U.B. Laurent, Hyaluronan: its nature, distribution, func-
[16] S. Ahmed, Green tea polyphenol epigallocatechin-3-gallate (EGCG) differentially tions and turnover, J. Intern. Med. 242 (1) (1997) 27–33.
inhibits interleukin-1-induced expression of matrix metalloproteinase-1 and -13 in [38] J.A. Burdick, G.D. Prestwich, Hyaluronic acid hydrogels for biomedical applica-
human chondrocytes, J. Pharmacol. Exp. Ther. 308 (2) (2003) 767–773. tions, Adv. Mater. 23 (12) (2011) H41–H56.
[17] Z. Rasheed, A.N. Anbazhagan, N. Akhtar, S. Ramamurthy, F.R. Voss, T.M. Haqqi, [39] T. Hayami, M. Pickarski, Y. Zhuo, G.A. Wesolowski, G.A. Rodan, L.T. Duong,
Green tea polyphenol epigallocatechin-3-gallate inhibits advanced glycation end Characterization of articular cartilage and subchondral bone changes in the rat
product-induced expression of tumor necrosis factor-alpha and matrix metallopro- anterior cruciate ligament transection and meniscectomized models of osteoar-
teinase-13 in human chondrocytes, Arthritis Res. Ther. 11 (3) (2009) R71. thritis, Bone 38 (2) (2006) 234–243.
[18] S. Ahmed, A. Rahman, A. Hasnain, M. Lalonde, V.M. Goldberg, T.M. Haqqi, Green [40] Y.G. Kim, U. Park, B.J. Park, K. Kim, Exosome-mediated bidirectional signaling
tea polyphenol epigallocatechin-3-gallate inhibits the IL-1 beta-induced activity and between mesenchymal stem cells and chondrocytes for enhanced chondrogenesis,
expression of cyclooxygenase-2 and nitric oxide synthase-2 in human chondrocytes, Biotechnol. Bioproc. E 24 (5) (2019) 734–744.
Free Radic. Biol. Med. 33 (8) (2002) 1097–1105. [41] M.E. Han, S.H. Kim, H.D. Kim, H.G. Yim, S.A. Bencherif, T.I. Kim, N.S. Hwang,
[19] R. Singh, S. Ahmed, N. Islam, V.M. Goldberg, T.M. Haqqi, Epigallocatechin-3-gal- Extracellular matrix-based cryogels for cartilage tissue engineering, Int. J. Biol.
late inhibits interleukin-1beta-induced expression of nitric oxide synthase and Macromol. 93 (Pt B) (2016) 1410–1419.
production of nitric oxide in human chondrocytes: suppression of nuclear factor [42] N.S. Hwang, S. Varghese, J. Elisseeff, Cartilage tissue engineering: directed differ-
kappaB activation by degradation of the inhibitor of nuclear factor kappaB, entiation of embryonic stem cells in three-dimensional hydrogel culture, Methods
Arthritis Rheum. 46 (8) (2002) 2079–2086. Mol. Biol. 407 (2007) 351–373.
[20] T.E. McAlindon, R.R. Bannuru, M.C. Sullivan, N.K. Arden, F. Berenbaum, [43] H.D. Kim, Y. Lee, Y. Kim, Y. Hwang, N.S. Hwang, Biomimetically reinforced poly-
S.M. Bierma-Zeinstra, G.A. Hawker, Y. Henrotin, D.J. Hunter, H. Kawaguchi, vinyl alcohol-based hybrid scaffolds for cartilage tissue engineering, Polymers
K. Kwoh, S. Lohmander, F. Rannou, E.M. Roos, M. Underwood, OARSI guidelines (Basel) 9 (12) (2017).
for the non-surgical management of knee osteoarthritis, Osteoarthr. Cartil. 22 (3) [44] B.N. Brown, J.E. Valentin, A.M. Stewart-Akers, G.P. McCabe, S.F. Badylak,
(2014) 363–388. Macrophage phenotype and remodeling outcomes in response to biologic scaffolds
[21] A. Rey-Rico, H. Madry, M. Cucchiarini, Hydrogel-based controlled delivery systems with and without a cellular component, Biomaterials 30 (8) (2009) 1482–1491.
for articular cartilage repair, Biomed. Res. Int. 2016 (2016) 1215263. [45] C. Liu, K.H. Bae, A. Yamashita, J.E. Chung, M. Kurisawa, Thiol-mediated synthesis
[22] T.J. Brown, U.B. Laurent, J.R. Fraser, Turnover of hyaluronan in synovial joints: of hyaluronic acid-epigallocatechin-3-O-gallate conjugates for the formation of in-
elimination of labelled hyaluronan from the knee joint of the rabbit, Exp. Physiol. jectable hydrogels with free radical scavenging property and degradation re-
76 (1) (1991) 125–134. sistance, Biomacromolecules 18 (10) (2017) 3143–3155.
[23] N.E. Larsen, H.D. Dursema, C.T. Pollak, E.M. Skrabut, Clearance kinetics of a hylan- [46] E.H. Kim, S. Lim, T.E. Kim, I.O. Jeon, Y.S. Choi, Preparation of in situ injectable
based viscosupplement after intra-articular and intravenous administration in an- chitosan/gelatin hydrogel using an acid-tolerant tyrosinase, Biotechnol. Bioproc. E
imal models, J Biomed Mater Res B Appl Biomater 100 (2) (2012) 457–462. 23 (5) (2018) 500–506.
[24] G. Camci-Unal, J.W. Nichol, H. Bae, H. Tekin, J. Bischoff, A. Khademhosseini, [47] Y. Lee, J. Choi, N.S. Hwang, Regulation of lubricin for functional cartilage tissue
Hydrogel surfaces to promote attachment and spreading of endothelial progenitor regeneration: a review, Biomater. Res. 22 (2018) 9.
cells, J. Tissue Eng. Regen. Med. 7 (5) (2013) 337–347. [48] K.N. Antonas, J.R. Fraser, K.D. Muirden, Distribution of biologically labelled
[25] P. Taddei, V. Chiono, A. Anghileri, G. Vozzi, G. Freddi, G. Ciardelli, Silk fibroin/ radioactive hyaluronic acid injected into joints, Ann. Rheum. Dis. 32 (2) (1973)
gelatin blend films crosslinked with enzymes for biomedical applications, 103–111.
Macromol. Biosci. 13 (11) (2013) 1492–1510. [49] P.A. Levett, D.W. Hutmacher, J. Malda, T.J. Klein, Hyaluronic acid enhances the
[26] H.D. Kim, J. Heo, Y. Hwang, S.Y. Kwak, O.K. Park, H. Kim, S. Varghese, mechanical properties of tissue-engineered cartilage constructs, PLoS One 9 (12)
N.S. Hwang, Extracellular-matrix-based and Arg-Gly-Asp-modified photo- (2014) e113216.
polymerizing hydrogels for cartilage tissue engineering, Tissue Eng. Part A 21 (3–4) [50] V. Moulisova, S. Poveda-Reyes, E. Sanmartin-Masia, L. Quintanilla-Sierra,
(2015) 757–766. M. Salmeron-Sanchez, G. Gallego Ferrer, Hybrid protein-glycosaminoglycan hy-
[27] S.H. Kim, Y.H. An, H.D. Kim, K. Kim, S.H. Lee, H.G. Yim, B.G. Kim, N.S. Hwang, drogels promote chondrogenic stem cell differentiation, ACS Omega 2 (11) (2017)
Enzyme-mediated tissue adhesive hydrogels for meniscus repair, Int. J. Biol. 7609–7620.
Macromol. 110 (2018) 479–487. [51] C. Chung, J.A. Burdick, Influence of three-dimensional hyaluronic acid micro-
[28] S.H. Kim, S.H. Lee, J.E. Lee, S.J. Park, K. Kim, I.S. Kim, Y.S. Lee, N.S. Hwang, environments on mesenchymal stem cell chondrogenesis, Tissue Eng. Part A 15 (2)
B.G. Kim, Tissue adhesive, rapid forming, and sprayable ECM hydrogel via re- (2009) 243–254.
combinant tyrosinase crosslinking, Biomaterials 178 (2018) 401–412. [52] M.M. Caron, P.J. Emans, M.M. Coolsen, L. Voss, D.A. Surtel, A. Cremers, L.W. van
[29] S. Kamekura, K. Hoshi, T. Shimoaka, U. Chung, H. Chikuda, T. Yamada, M. Uchida, Rhijn, T.J. Welting, Redifferentiation of dedifferentiated human articular chon-
N. Ogata, A. Seichi, K. Nakamura, H. Kawaguchi, Osteoarthritis development in drocytes: comparison of 2D and 3D cultures, Osteoarthr. Cartil. 20 (10) (2012)
novel experimental mouse models induced by knee joint instability, Osteoarthr. 1170–1178.
Cartil. 13 (7) (2005) 632–641. [53] K.R. Brodkin, A.J. Garcia, M.E. Levenston, Chondrocyte phenotypes on different
[30] J.D. Lambert, C.S. Yang, Mechanisms of cancer prevention by tea constituents, J. extracellular matrix monolayers, Biomaterials 25 (28) (2004) 5929–5938.
Nutr. 133 (10) (2003) 3262S–3267S. [54] W.S. Toh, T.C. Lim, M. Kurisawa, M. Spector, Modulation of mesenchymal stem cell
[31] G.L. Tipoe, T.M. Leung, E.C. Liong, T.Y. Lau, M.L. Fung, A.A. Nanji, chondrogenesis in a tunable hyaluronic acid hydrogel microenvironment,
Epigallocatechin-3-gallate (EGCG) reduces liver inflammation, oxidative stress and Biomaterials 33 (15) (2012) 3835–3845.
fibrosis in carbon tetrachloride (CCl4)-induced liver injury in mice, Toxicology 273 [55] R. Jin, L.S. Moreira Teixeira, P.J. Dijkstra, C.A. van Blitterswijk, M. Karperien,
(1–3) (2010) 45–52. J. Feijen, Chondrogenesis in injectable enzymatically crosslinked heparin/dextran
[32] D. Liu, J.T. Perkins, B. Hennig, EGCG prevents PCB-126-induced endothelial cell hydrogels, J. Control. Release 152 (1) (2011) 186–195.
inflammation via epigenetic modifications of NF-kappaB target genes in human [56] D.J. Leong, M. Choudhury, R. Hanstein, D.M. Hirsh, S.J. Kim, R.J. Majeska,
endothelial cells, J. Nutr. Biochem. 28 (2016) 164–170. M.B. Schaffler, J.A. Hardin, D.C. Spray, M.B. Goldring, N.J. Cobelli, H.B. Sun, Green
[33] O. Krupkova, S.J. Ferguson, K. Wuertz-Kozak, Stability of (−)-epigallocatechin tea polyphenol treatment is chondroprotective, anti-inflammatory and palliative in
gallate and its activity in liquid formulations and delivery systems, J. Nutr. a mouse post-traumatic osteoarthritis model, Arthritis Res. Ther. 16 (6) (2014) 508.
Biochem. 37 (2016) 1–12. [57] V. Natarajan, B. Madhan, M.L. Tiku, Intra-articular injections of polyphenols protect
[34] Z. Rasheed, N. Rasheed, H.A. Al-Shobaili, Epigallocatechin-3-O-gallate up-regulates articular cartilage from inflammation-induced degradation: suggesting a potential
microRNA-199a-3p expression by down-regulating the expression of cycloox- role in cartilage therapeutics, PLoS One 10 (6) (2015) e0127165.
ygenase-2 in stimulated human osteoarthritis chondrocytes, J. Cell. Mol. Med. 20 [58] S. Emoto, H. Yamaguchi, T. Kamei, H. Ishigami, T. Suhara, Y. Suzuki, T. Ito,
(12) (2016) 2241–2248. J. Kitayama, T. Watanabe, Intraperitoneal administration of cisplatin via an in situ
[35] T. Hussain, S. Gupta, V.M. Adhami, H. Mukhtar, Green tea constituent epigalloca- cross-linkable hyaluronic acid-based hydrogel for peritoneal dissemination of gas-
techin-3-gallate selectively inhibits COX-2 without affecting COX-1 expression in tric cancer, Surg. Today 44 (5) (2014) 919–926.
human prostate carcinoma cells, Int. J. Cancer 113 (4) (2005) 660–669.

11

You might also like