Sie sind auf Seite 1von 8

International Immunopharmacology 64 (2018) 238–245

Contents lists available at ScienceDirect

International Immunopharmacology
journal homepage: www.elsevier.com/locate/intimp

Isofraxidin inhibits interleukin-1β induced inflammatory response in human T


osteoarthritis chondrocytes
Jian Lin1, Xiaobin Li1, Weihui Qi, Yingzhao Yan, Kai Chen, Xinghe Xue, Xinxian Xu,

Zhenhua Feng, Xiaoyun Pan
Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
The Second School of Medicine, Wenzhou Medical University, China

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

Keywords: Osteoarthritis (OA) is the most prevalent disease of knee especially in the aged people. Isofraxidin (IF) is a
Osteoarthritis coumarin compound refined from traditional Chinese medicines with potential anti-inflammatory ability. This
Inflammation study aimed to evaluate protective anti-inflammatory effects of IF in human OA chondrocytes. The chondrocytes
Isofraxidin were isolated from OA patients and pretreated with IF before treatment with IL-1β. The results showed that IF
Chondrocytes
blocked IL-1β-stimulated production of NO and PGE2. In addition, IF inhibited the expression of COX-2, iNOs,
NF-κB
MMP-1, MMP-3, MMP-13, ADAMTS-4 and ADAMTS-5, and increased the levels of aggrecan and collagen-II.
Mechanistically, IF suppressed IL-1β-induced IκB-α degradation and NF-κB activation. In conclusion, our results
demonstrate that IF inhibits inflammation in OA via the regulation of NF-κB signaling, and suggest that IF may
be a potential therapeutic agent for OA.

1. Introduction compound isolated from traditional Chinese herbs usually used for anti-
tumor, anti-oxidant, anti-bacterial, and anti-inflammatory treatments
Osteoarthritis (OA) is a common deteriorating joint pathologic [11–14]. IF has multifarious biological activities, including anti-oxi-
process caused by joint instability and it is estimated that 12.1% of U.S. dant, anti-bacterial and anti-inflammatory actions [15–17]. IF reduced
population aged 25 and older have OA. OA could cause serious joint the levels of iNOS, COX-2, and tumor necrosis factor-α (TNF-α) by
pain and irreversible loss of joint movement, and OA is characterized by regulating inflammatory modulators, such as inhibitory kappa B (IκBα)
erosion of the articular cartilage, synovium inflammation, subchondral and nuclear factor-kappa B (NF-κB) [18]. IF has been reported to ab-
bone sclerosis and osteophyte formation [1,2]. Recent studies showed rogate LPS-induced oxidative stress and inflammation by inhibiting
that inflammation and inflammatory factors play important role in the TNF-α production and MAPK pathway in mouse peritoneal macro-
progression of OA [3,4]. Interleukin-1β (IL-1β), the major proin- phages [19]. In addition, IF exerted anti-inflammatory effect via NF-κB
flammatory cytokine, could be involved in the destruction of cartilage suppression in vivo [20]. However, anti-inflammatory effect of IF in OA
[5,6]. IL-1β could induce the production of matrix metalloproteinases remains unknown. Therefore, in this study we evaluated the anti-in-
(MMPs) and a disintegrin and metalloproteinase with thrombospondin flammatory effects and explored the possible mechanism of IF on IL-1β-
motifs (ADAMTS), which are responsible for the downregulation of type induced inflammation in human OA chondrocytes in vitro.
II collagen and proteoglycans in articular cartilage [7,8]. Furthermore,
treatment of chondrocytes with IL-1β could stimulate the release of 2. Materials and methods
inducible nitric oxide synthase (iNOS) and cyclooxygenase-2(COX-2),
leading to the production of nitric oxide (NO) and prostaglandin E2 2.1. Reagents and antibodies
(PGE2) [9,10]. Therefore, these inflammatory mediators may be ef-
fective therapeutic targets for OA. Isofraxidin (purity > 98%), collagenase type II, recombinant human
Isofraxidin (IF, 7-hydroxy-6, 8-dimethoxycoumarin) is a coumarin IL-1β, and dimethylsulfoxide (DMSO) were purchased from Sigma


Corresponding author at: Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou
325000, China.
E-mail address: panxy9046@163.com (X. Pan).
1
Jian Lin and Xiaobin Li contribute equally to this work.

https://doi.org/10.1016/j.intimp.2018.09.003
Received 4 June 2018; Received in revised form 1 September 2018; Accepted 5 September 2018
1567-5769/ © 2018 Elsevier B.V. All rights reserved.
J. Lin et al. International Immunopharmacology 64 (2018) 238–245

Table 1
Primers used in this study.
Gene Forward primer Reverse primer

COX-2 5′-GAGAGATGTATCCTCCCACAGTCA-3′ 5′-GACCAGGCACCAGACCAAAG-3′


iNOS 5′-CCTTACGAGGCGAAGAAGGACAG-3′ 5′-CAGTTTGAGAGAGGAGGCTCCG-3′
MMP-1 5′-GGGAATAAGTACTGGGCTGTTCAG-3′ 5′-CCTCAGAAAGAGCAGCATCGATATG-3′
MMP-3 5′-CTGGCCTGCTGGCTCATGCTT-3′ 5′-GCAGGGTCCTTGGAGTGGTCA-3′
MMP-13 5′-CCAGAACTTCCCAACCAT-3′ 5′-ACCCTCCATAATGTCATACC-3′
ADAMTS-4 5′-CCTGGCAAGGACTATGATGCTGA-3′ 5′-GGGCGAGTGTTTGGTCTGG-3′
AMAMTS-5 5′-GCAGAACATCGACCAACTCTACTC-3′ 5′-CCAGCAATGCCCACCGAAC-3′
Collagen-II 5′-CTCAAGTCGCTGAACAACCA-3′ 5′-GTCTCCGCTCTTCCACTCTG-3′
Aggrecan 5′-AAGTGCTATGCTGGCTGGTT-3′ 5′-GGTCTGGTTGGGGTAGAGGT-3′
GAPDH 5′-TCTCCTCTGACTTCAACAGCGAC-3′ 5′-CCCTGTTGCTGTAGCCAAATTC-3′

Fig. 1. Effect of IF on human OA chondrocyte viability. The cells were treated


with IF (0, 1, 10, 50, 100 μM) for 24 h. The cell viability was determined by
CCK-8 assay. The values are mean ± SD of three independent experiments.
*p < 0.05 compared with control group.

Chemical Co. (St. Louis, MO, USA). Primary antibodies against iNOs,
COX-2, and collagen-II were acquired from Abcam (Cambridge, MA,
USA). Primary antibodies against p65, p-p65, IkBα, and p-IkBα were
purchased from Cell Signaling Technology (Beverly, MA, USA). Cell-
Counting Kit-8 (CCK-8) was obtained from Dojindo (Kumamo, Japan).
TRIzol reagent was purchased from Invitrogen (Carlsbad, CA, USA).
QuantiTect Reverse Transcription Kit was purchased from Qiagen
(Valencia, CA, USA). Fetal bovine serum (FBS), bovine serum albumin
(BSA), and Dulbecco's modified Eagle's medium (DMEM)/F12 medium
were purchased from Gibco (Life Technologies Corp., Carlsbad, CA,
USA). SYBR Green Master Mix was purchased from Bio-Rad
Laboratories (CA, USA). ELISA kits of PGE2, MMP-1, MMP-3, MMP-13,
ADAMTS-4, and ADAMTS-5 were purchased from R&D Systems
(Minneapolis, MN, USA). Griess reagent was purchased from Beyotime
Institute of Biotechnology (Shanghai, China).

Fig. 2. Effect of IF on IL-1β-induced NO and PGE2 production in human OA


2.2. Primary human chondrocyte culture chondrocytes. Chondrocytes were pretreated with various concentrations of IF
(1, 10, 50 μM) and then treated with or without IL-1β (10 ng/ml) for 24 h. NO
Tissue collection was according to the terms of the Medical Ethical levels were determined by Griess reaction (a), and the levels of PGE2 were
Committee of the Second Affiliated Hospital, Wenzhou Medical assessed by ELISA (b). The values are mean ± SD of three independent ex-
University and following the guidelines of the Declaration of Helsinki. periments. #p < 0.05 compared with control group. *p < 0.05, **p < 0.01
compared with IL-1β group.
Human cartilage samples were obtained from four OA patients (aged
56–66 years, two men and two women) who were diagnosed as os-
teoarthritis [21]. All patients underwent total knee replacement surgery passage 1 to 2 was used in our study to avoid the phenotype loss.
at the Second Affiliated Hospital of Wenzhou Medical University, and
provided ethical consent. Cartilage tissues were isolated from bone and
connective tissues, cut into small pieces, washed three times with PBS, 2.3. Cell viability
digested with 0.25% trypsin–EDTA solution, and then incubated with
2 mg/ml (0.1%) collagenase II for 4 h at 37 °C. After centrifuged at Cell viability was tested using CCK-8 kit according to the manu-
1000 rpm for 5 min, the digested cartilage tissues were suspended and facturer's instructions. Briefly, human OA chondrocytes were seeded in
seeded into tissue culture flasks. The chondrocytes were cultured in 96-well plates (6000 cells/well) and treated with or without IF (1, 10,
DMEM/F12 with 10% FBS and 1% antibiotic mixture in the incubator 50, 100 μM) for 24 h. After that, 10 uL CCK-8 was added to each well
with 5% CO2 at 37 °C. The medium was changed every two days, and and incubated at 37 °C for 4 h. The absorbance of wells was then
the cells were passaged by using 0.25% Trypsin-EDTA solution. Only measure using a microplate reader (Leica Microsystems, Germany).

239
J. Lin et al. International Immunopharmacology 64 (2018) 238–245

Fig. 3. Effect of IF on IL-1β-induced iNOS and COX-2 expression in human OA chondrocytes. Human OA chondrocytes were pretreated for 2 h with various
concentrations of IF (1, 10, 50 μM) and then stimulated or not stimulated with IL-1β (10 ng/ml) for 24 h. The mRNA expression levels of COX-2 (a) and iNOS (b) were
assessed by qRT-PCR. The protein expression levels of iNOS and COX-2 were determined by Western blot analysis (c–e). The values are mean ± SD of three
independent experiments. #p < 0.05 compared with control group. *p < 0.05, **p < 0.01 compared with IL-1β group.

Fig. 4. Effect of IF on IL-1β-induced MMP-1, MMP-3 and MMP-13 expression in human OA chondrocytes. Human OA chondrocytes were pretreated for 2 h with
various concentrations of IF (1, 10, 50 μM) and then stimulated or not stimulated with IL-1β (10 ng/ml) for 24 h. The mRNA expression levels of MMP-1 (a), MMP-3
(b), and MMP-13 (c) were assayed by qRT-PCR. The protein levels of MMP-1, MMP-3, and MMP-13 in culture medium were determined by ELISA (d-f). The values are
mean ± SD of three independent experiments. #p < 0.05 compared with control group. *p < 0.05, **p < 0.01 compared with IL-1β group.

2.4. Griess reaction and ELISAs strand cDNA was synthesized using 1 μg of total RNA and the
QuantiTect Reverse Transcription kit. Quantitative real-time PCR
The content of NO in culture medium was measured by Griess re- (qPCR) was performed using CFX96 Real-Time PCR System (Bio-Rad
agent as previously described [22]. The levels of PGE2, MMP-1, MMP-3, Laboratories, CA, USA) under the following conditions: 10 min 95 °C,
MMP-13, ADAMTS-4, and ADAMTS-5 in culture medium were eval- followed by 40 cycles of 15 s 95 °C and 1 min 60 °C. The reaction was
uated using commercial ELISA kit according to the manufacturer's in- performed in a total volume of 10 μl, containing 4.5 μl diluted cDNA,
structions. 0.25 μl forward primer, 0.25 μl reverse primer and 5 μl SYBR Green
Master Mix. The level of target mRNA was normalized to the level of
2.5. qRT-PCR GAPDH and compared with control. Data were analyzed using the
2−ΔΔCT method [23]. The primers were designed using GenScript Pcr
Total RNA was extracted from cells using TRIzol reagent. First- Primer Design online program and the sequences were listed in Table 1.

240
J. Lin et al. International Immunopharmacology 64 (2018) 238–245

Fig. 5. Effect of IF on IL-1β-induced


ADAMTS-4 and ADAMTS-5 expression in
human OA chondrocytes. Human OA
chondrocytes were pretreated for 2 h with
various concentrations of IF (1, 10, 50 μM)
and then stimulated or not stimulated with
IL-1β (10 ng/ml) for 24 h. The mRNA ex-
pression levels of ADAMTS-4 (a), and
ADAMTS-5 (b) were assayed by qRT-PCR.
The protein levels of ADAMTS-4, and
ADAMTS-5 in culture medium were de-
termined by ELISA (c, d). The values are
mean ± SD of three independent experi-
ments. #p < 0.05 compared with control
group. *p < 0.05, **p < 0.01 compared
with IL-1β group.

2.6. Western blot analysis expressed as mean ± standard deviation (SD). Statistical analyses were
performed using SPSS statistical software program 20.0. Statistical
The total protein was extracted from chondrocytes using RIPA lysis significance was analyzed by one-way analysis of variance (ANOVA)
buffer supplemented with 1 mM PMSF (Phenylmethanesulfonyl followed by Tukey's post-hoc test if necessary. P < 0.05 was con-
fluoride) on the ice for 10 min followed by 15 min centrifugation at sidered statistically significant.
12,000 rpm at 4 °C, and then protein concentration was measured using
the BCA protein assay kit (Beyotime). 40 μg of total protein were re- 3. Results
solved on 12% SDS-PAGE and transferred to PVDF membranes.
Membranes were blocked with 5% nonfat milk for 2 h at room tem- 3.1. Effect of IF on human OA chondrocyte viability
perature and immediately washed three times for 5 min in TBS with
Tween-20 (TBST). Next, the membranes were incubated with primary The cytotoxic effect of IF on chondrocytes was determined at var-
antibody against COX-2, iNOs, p65, p-p65, IκB-α, p-IκB-α or β-actin ious concentrations (1, 10, 50, 100 μM) using CCK-8. As shown in
(dilution 1:1000) overnight at 4 °C, and washed three times with TBST Fig. 1, 100 μM IF significantly reduced cell viability after treatment for
for 5 min. The membranes were incubated subsequently with respective 24 h, but cell viability was not significantly affected by IF at con-
secondary antibodies for 2 h at room temperature. After washing with centrations up to 50 μM. Thus we chose IF at concentrations of 1, 10
TBST, the blots were visualized by electrochemiluminescence plus re- and 50 μM in subsequent experiments.
agent (Invitrogen). Finally, the intensity of the blots was quantified
with Image Lab 3.0 software (Bio-Rad).
3.2. Effect of IF on NO and PGE2 production in human OA chondrocytes

2.7. Immunofluorescence Chondrocytes were pretreated with various concentration of IF (1,


10, 50 μM) for 2 h, then treated with IL-1β (10 ng/ml) for 24 h. As
The chondrocytes were seeded in glass plated in a six-well plate and shown in Fig. 2, the levels of NO and PGE2 in cell culture medium
incubated for 24 h. Glass coverslips with chondrocyte monolayers were increased significantly in IL-1β treated group compared to control
rinsed three times in PBS before fixation in 4% paraformaldehyde and group, but IF significantly reduced IL-1β-stimulated production of NO
permeation in 0.1% Triton X-100 diluted in PBS for 15 min. Next, the and PGE2 in a concentration-dependent manner. IF alone had no sig-
cells were blocked with 5% goat serum albumin for 1 h at 37 °C, rinsed nificant effects on NO and PGE2 production in the chondrocytes
with PBS and incubated with primary antibody for collagen-II or p65 (Supplemental Fig. 1).
(1:200) in a humid chamber overnight at 4 °C. On the next day, cells
were incubated with fluorescein-conjugated goat anti-rabbit IgG anti- 3.3. Effect of IF on iNOS and COX-2 expression in human OA chondrocytes
body (1:500) for 1 h at 37 °C. Afterwards, cells were washed three times
with PBS and then labeled with DAPI (Invitrogen) for 1 min. Finally, Next we detected the expression of iNOS and COX-2 in chon-
slides were observed under a fluorescence microscope (Olympus Inc., drocytes. We found that IL-1β stimulation led to the upregulation of
Tokyo, Japan). iNOS and COX-2 mRNA expression compared with the control group.
However, IF decreased the expression of iNOS and COX-2 at mRNA
2.8. Statistical analysis level dose-dependently (Fig. 3a, b). Similarly, protein expression levels
of iNOS and COX-2 were significantly stimulated by IL-1β but decreased
All experiments were at least performed triplicately. Data are by IF pretreatment (Fig. 3c–e). IF alone had no significant effects on

241
J. Lin et al. International Immunopharmacology 64 (2018) 238–245

Fig. 6. Effect of IF on IL-1β-induced aggrecan and collagen-II expression in human OA chondrocytes. Human OA chondrocytes were pretreated for 2 h with various
concentrations of IF (1, 10, 50 μM) and then stimulated or not stimulated with IL-1β (10 ng/ml) for 24 h. The expression of collagen-II was visualized by im-
munofluorescence (a). The mRNA expression levels of aggrecan (b) and collagen-II (c) were assayed by qRT-PCR. The values are mean ± SD of three independent
experiments. #p < 0.05 compared with control group. *p < 0.05, **p < 0.01 compared with IL-1β group.

iNOS and COX-2 expression in the chondrocytes (Supplemental Fig. 2). alone had no significant effects on ADAMTS-4 and ADAMTS-5 expres-
sion in the chondrocytes (Supplemental Fig. 3).
3.4. Effect of IF on MMP-1, MMP-3 and MMP-13 expression in human OA
chondrocytes 3.6. Effect of IF on aggrecan and collagen-II expression in human OA
chondrocytes
As shown in Fig. 4a–c, IL-1β significantly upregulated mRNA ex-
pression of MMP-1, MMP-3 and MMP-13 in chondrocytes compared Immunofluorescence analysis showed that IL-1β stimulation led to
with control group. IF remarkably decreased mRNA levels of MMP-1, the downregulation of collagen-II in chondrocytes, but IF pretreatment
MMP-3 MMP-13 in chondrocytes stimulated with IL-1β. ELISA assay inhibited IL-1β-induced downregulation of collagen-II (Fig. 6a). Con-
showed that the secretion of MMP-1, MMP-3 and MMP-13 in culture sistent with these observations, PCR analysis showed that mRNA ex-
medium stimulated by IL-1β was significantly inhibited by IF pression of aggrecan and collagen-II decreased significantly after IL-1β
(Fig. 4d–f). IF alone had no significant effects on MMP-1, MMP-3 and stimulation but recovered in chondrocytes pretreated with IF in a dose-
MMP-13 expression in the chondrocytes (Supplemental Fig. 3). dependent manner (Fig. 6b, c). IF alone had no significant effects on
aggrecan and collagen-II expression in the chondrocytes (Supplemental
Fig. 4).
3.5. Effect of IF on ADAMTS-4 and ADAMTS-5 expression in human OA
chondrocytes
3.7. Effect of IF on NF-κB activation in human OA chondrocytes
As shown in Fig. 5a, b, IL-1β significantly upregulated mRNA ex-
pression of ADAMTS-4 and ADAMTS-5 in chondrocytes compared with To further explore the mechanism underlying the anti-inflammation
control group. IF remarkably decreased mRNA levels of ADAMTS-4 and effect of IF, we focused on NF-κB signaling in chondrocytes. As shown
ADAMTS-5 in chondrocytes stimulated with IL-1β. ELISA assay showed in Fig. 7a–c, IL-1β remarkably increased the phosphorylation of NF-κB
that the secretion of ADAMTS-4 and ADAMTS-5 in culture medium p65 and IκB-α in chondrocytes. In addition, IL-1β led to notable de-
stimulated by IL-1β was significantly inhibited by IF (Fig. 5c, d). IF gradation of IκB-α. In contrast, IF blocked IL-1β-stimulated

242
J. Lin et al. International Immunopharmacology 64 (2018) 238–245

Fig. 7. Effect of IF on NF-κB activation in


human OA chondrocytes. The protein levels
of p-p65, p65, p-IκB, IκB were determined
by Western blot analysis (a–c). The nuclear
translocation of p65 was detected by im-
munofluorescence staining. DAPI stained
the nuclei. In control chondrocytes, p65
was mainly localized in the cytoplasm (d).
Chondrocytes stimulated with IL-1β alone
demonstrated nuclear translocation of p65
(e), but was attenuated in chondrocytes
pretreated with IF (f). All experiments were
repeated three times. The values are
mean ± SD of three independent experi-
ments. #p < 0.05 compared with control
group. *p < 0.05, **p < 0.01 compared
with IL-1β group.

phosphorylation of NF-κB p65 and IκB-α and the degradation of IκB-α. associated with the degeneration of articular cartilage by damaging
Then we investigated the effect of IF on IL-1β-induced NF-κB p65 ECM and inhibiting the proliferation of chondrocytes [27,28]. Several
nuclear translocation by immunofluorescence staining. In chondrocytes studies have indicated that the attenuation of inflammatory mediators
of control group the majority of p65 was located in the cytoplasm such as NO and PGE2 could weaken the progression of OA [29]. In this
(Fig. 7d). In chondrocytes stimulated by IL-1β, we observed enhanced study, we found that IF blocked the production of NO and PGE2, and
nuclear staining of p65 (Fig. 7e). However, IF inhibited IL-1β-induced downregulated COX-2 and iNOS expression at both mRNA and protein
nuclear translocation of p65 in chondrocytes (Fig. 7f). IF alone had no levels. Our results are consistent with previous study that IF blocked the
significant effects on IκB, p-IκB, p65, and p-p65 protein levels in the production of PGE2 and COX-2 in lipopolysaccharide (LPS) induced
chondrocytes (Supplemental Fig. 2). acute lung injury in mice [30].
MMPs are considered as the main enzymes that regulate tissue re-
modeling and the degradation of ECM such as aggrecan and collagens
4. Discussion [31,32]. MMP-13 has been shown to target collagen-II which composes
the main structure of ECM [33]. Furthermore, ADAMTS members play
In this study we showed that IF decreased IL-1β-stimulated over- major role in cartilage degradation in OA, especially ADAMTS-4 and
production of PGE2, NO, COX-2, iNOS, MMP-1, MMP-3, MMP-13, ADAMTS-5, which are responsible for cleaving aggrecan [4,7]. There-
ADAMTS-4 and ADAMTS-5, while recovered IL-1β-stimulated down- fore, targeting ADMATS-5 has been a major strategy in the treatment of
regulation of collagen-II and aggrecan in chondrocytes isolated from OA [34]. Using small interfering RNA (siRNA) to block ADAMTS-5
OA patients, and these effects are correlated with the inhibition of IL- could restrain aggrecan loss from human OA cartilage explants [35]. In
1β-induced NF-κB activation by IF. These findings suggest that IF in- this study, we found that IF inhibited IL-1β-stimulated MMP-1, MMP-3,
hibits IL-1β-stimulated inflammatory response in OA progression. MMP-13, ADAMTS-4, and ADAMTS-5 production at both mRNA and
Elevated IL-1β and TNF-α levels were found in synovial fluid of OA protein levels in human OA chondrocytes. Thus we speculated that IF
patients [24,25]. In addition, IL-1β induced the expression of iNOS and may exert anti-inflammation action by decreasing the production and
COX-2, which contributed to the production of NO and PGE2 [26]. activation of MMPs and ADAMTS in the progression of OA.
Recent studies reported that NO increased the level of PGE2, which is

243
J. Lin et al. International Immunopharmacology 64 (2018) 238–245

NF-κB signaling is a key pathway associated with OA pathogenesis and proinflammatory effects on articular cartilage and synergizes with interleukin-
[36]. NF-κB is known to regulate the expression of inflammatory factors 1, Arthritis Rheumatol. (Hoboken, N.J.) 66 (7) (2014) 1779–1788.
[7] R. Kelwick, I. Desanlis, G.N. Wheeler, D.R. Edwards, The ADAMTS (A Disintegrin
including iNOS, COX-2 and MMPs [37]. Normally, NF-κB is located in and Metalloproteinase with Thrombospondin motifs) family, Genome Biol. 16
the cytoplasm combined with IκB, which is an inhibitory protein. Upon (2015) 113.
stimulation by IL-1β, IκB would be phosphorylated and degraded, fol- [8] M.B. Goldring, M. Otero, Inflammation in osteoarthritis, Curr. Opin. Rheumatol. 23
(5) (2011) 471–478.
lowed by the dissociation of NF-κB p65 from IκB. NF-κB p65 would then [9] S.R. Goldring, M.B. Goldring, The role of cytokines in cartilage matrix degeneration
be translocated into the nucleus to activate inflammatory related genes in osteoarthritis, Clin. Orthop. Relat. Res. (427 Suppl) (2004) S27–S36.
[38]. Previous studies showed that NF-κB inhibitor decreased the ex- [10] S. Ahmed, A. Rahman, A. Hasnain, M. Lalonde, V.M. Goldberg, T.M. Haqqi, Green
tea polyphenol epigallocatechin-3-gallate inhibits the IL-1 beta-induced activity and
pression of MMP-3 and MMP-13 stimulated by IL-1β [39]. Additionally, expression of cyclooxygenase-2 and nitric oxide synthase-2 in human chondrocytes,
NF-κB p65 blocked the levels of COX-2, iNOS and MMP-9 in the de- Free Radic. Biol. Med. 33 (8) (2002) 1097–1105.
velopment of OA [40]. Therefore, targeted downregulation of NF-κB [11] S. Khan, N. Riaz, N. Afza, A. Malik, R. Aziz ur, L. Iqbal, M. Lateef, Antioxidant
constituents from Cotoneaster racemiflora, J. Asian Nat. Prod. Res. 11 (1) (2009)
might be effective for treating OA. In this study, we found that IF re-
44–48.
markably blocked IL-1β-induced NF-κB activation and IκB-α degrada- [12] T. Yamazaki, T. Tokiwa, Isofraxidin, a coumarin component from Acanthopanax
tion in OA chondrocytes. Furthermore, IF restrained nuclear translo- senticosus, inhibits matrix metalloproteinase-7 expression and cell invasion of
cation of NF-κB p65 stimulated by IL-1β. Li et al. reported that IF human hepatoma cells, Biol. Pharm. Bull. 33 (10) (2010) 1716–1722.
[13] A. Simaratanamongkol, K. Umehara, H. Noguchi, P. Panichayupakaranant,
suppressed p65 translocation into the nucleus and inhibited iNOS Identification of a new angiotensin-converting enzyme (ACE) inhibitor from Thai
production in high-fat-induced hepatic lipid metabolism disorders in edible plants, Food Chem. 165 (2014) 92–97.
mice [41]. In addition, IF inhibited TNF-α production and decreased [14] P. Li, Q.L. Zhao, L.H. Wu, P. Jawaid, Y.F. Jiao, M. Kadowaki, T. Kondo, Isofraxidin,
a potent reactive oxygen species (ROS) scavenger, protects human leukemia cells
phospho-p38 in LPS-stimulated peritoneal macrophages [19]. Taken from radiation-induced apoptosis via ROS/mitochondria pathway in p53-in-
together, our data suggest that IF inhibits NF-κB activation in the pro- dependent manner, Apoptosis: An International Journal on Programmed Cell Death
gression of OA. Nevertheless, further studies are needed to understand 19 (6) (2014) 1043–1053.
[15] P. Shen, H.G. Wang, M.M. Li, Q.Y. Ma, C.W. Zhou, F. Pan, R. Xie, Isofraxidin in-
the detailed mechanism by which IF inhibits NF-κB activation during hibited proliferation and induced apoptosis via blockage of Akt pathway in human
OA progression. colorectal cancer cells, Biomedicine & Pharmacotherapy = Biomedecine &
In conclusion, our study is the first to evaluate potential effects of IF Pharmacotherapie 92 (2017) 78–85.
[16] T. Kamino, T. Shimokura, Y. Morita, Y. Tezuka, M. Nishizawa, K. Tanaka,
on human OA chondrocytes. Our results demonstrate that IF inhibits IL- Comparative analysis of the constituents in Saposhnikoviae Radix and Glehniae
1β-induced ECM degradation and inflammation via the regulation of Radix cum Rhizoma by monitoring inhibitory activity of nitric oxide production, J.
NF-κB signaling, and suggest that IF may be a potential therapeutic Nat. Med. 70 (2) (2016) 253–259.
[17] T. Yamazaki, S. Shimosaka, H. Sasaki, T. Matsumura, T. Tukiyama, T. Tokiwa,
agent for OA.
(+)-Syringaresinol-di-O-beta-D-glucoside, a phenolic compound from
Acanthopanax senticosus Harms, suppresses proinflammatory mediators in SW982
Acknowledgements human synovial sarcoma cells by inhibiting activating protein-1 and/or nuclear
factor-kappaB activities, Toxicology In Vitro: An International Journal Published in
Association With BIBRA 21 (8) (2007) 1530–1537.
The authors thank the staff of the Laboratory of the Orthopaedic [18] T. Nakamura, N. Kodama, M. Oda, S. Tsuchiya, Y. Arai, T. Kumamoto, T. Ishikawa,
Research Institute and the Scientific Research Center of the Second K. Ueno, S. Yano, The structure-activity relationship between oxycoumarin deri-
Affiliated Hospital of Wenzhou Medical University. This work was vatives showing inhibitory effects on iNOS in mouse macrophage RAW264.7 cells,
J. Nat. Med. 63 (1) (2009) 15–20.
supported by Zhejiang Province Public Welfare Technology Application [19] X. Niu, W. Xing, W. Li, T. Fan, H. Hu, Y. Li, Isofraxidin exhibited anti-inflammatory
Research Project, China (No. 2015C33209). effects in vivo and inhibited TNF-alpha production in LPS-induced mouse peritoneal
macrophages in vitro via the MAPK pathway, Int. Immunopharmacol. 14 (2) (2012)
164–171.
Compliance with ethical standards [20] L. Liu, Q. Mu, W. Li, W. Xing, H. Zhang, T. Fan, H. Yao, L. He, Isofraxidin protects
mice from LPS challenge by inhibiting pro-inflammatory cytokines and alleviating
The study was in accordance with the Declaration of Helsinki and histopathological changes, Immunobiology 220 (3) (2015) 406–413.
[21] B. Palmieri, D. Lodi, S. Capone, Osteoarthritis and degenerative joint disease: local
Tokyo.
treatment options update, Acta Bio-medica: Atenei Parmensis 81 (2) (2010)
94–100.
Conflict of interest [22] R.Y. Au, T.K. Al-Talib, A.Y. Au, P.V. Phan, C.G. Frondoza, Avocado soybean un-
saponifiables (ASU) suppress TNF-alpha, IL-1beta, COX-2, iNOS gene expression,
and prostaglandin E2 and nitric oxide production in articular chondrocytes and
The authors declare that they have no conflict of interest. monocyte/macrophages, Osteoarthr. Cartil. 15 (11) (2007) 1249–1255.
[23] M.W. Pfaffl, A new mathematical model for relative quantification in real-time RT-
Appendix A. Supplementary data PCR, Nucleic Acids Res. 29 (9) (2001) e45.
[24] F. Eymard, A. Pigenet, D. Citadelle, C.H. Flouzat-Lachaniette, A. Poignard,
C. Benelli, F. Berenbaum, X. Chevalier, X. Houard, Induction of an inflammatory
Supplementary data to this article can be found online at https:// and prodegradative phenotype in autologous fibroblast-like synoviocytes by the
doi.org/10.1016/j.intimp.2018.09.003. infrapatellar fat pad from patients with knee osteoarthritis, Arthritis Rheumatol.
(Hoboken, N.J.) 66 (8) (2014) 2165–2174.
[25] R.K. Chaganti, E. Purdue, T.P. Sculco, L.A. Mandl, Elevation of serum tumor ne-
References crosis factor alpha in patients with periprosthetic osteolysis: a case-control study,
Clin. Orthop. Relat. Res. 472 (2) (2014) 584–589.
[26] C. Stratz, J. Anakwue, H. Bhatia, S. Pitz, B.L. Fiebich, Anti-inflammatory effects of
[1] R.F. Loeser, Aging and osteoarthritis: the role of chondrocyte senescence and aging
5-HT3 receptor antagonists in interleukin-1beta stimulated primary human chon-
changes in the cartilage matrix, Osteoarthr. Cartil. 17 (8) (2009) 971–979.
drocytes, Int. Immunopharmacol. 22 (1) (2014) 160–166.
[2] W.K. Aicher, B. Rolauffs, The spatial organisation of joint surface chondrocytes:
[27] X. Bai, J. Wang, Y. Guo, J. Pan, Q. Yang, M. Zhang, H. Li, L. Zhang, J. Ma, F. Shi,
review of its potential roles in tissue functioning, disease and early, preclinical
W. Shu, Y. Wang, J. Leng, Prostaglandin E2 stimulates beta1-integrin expression in
diagnosis of osteoarthritis, Ann. Rheum. Dis. 73 (4) (2014) 645–653.
hepatocellular carcinoma through the EP1 receptor/PKC/NF-kappaB pathway, Sci.
[3] M. Kobayashi, G.R. Squires, A. Mousa, M. Tanzer, D.J. Zukor, J. Antoniou, U. Feige,
Rep. 4 (2014) 6538.
A.R. Poole, Role of interleukin-1 and tumor necrosis factor alpha in matrix de-
[28] V.K. Podichetty, The aging spine: the role of inflammatory mediators in inter-
gradation of human osteoarthritic cartilage, Arthritis Rheum. 52 (1) (2005)
vertebral disc degeneration, Cell. Mol. Biol. (Noisy-le-Grand) 53 (5) (2007) 4–18.
128–135.
[29] N. Li, M.A. Rivera-Bermudez, M. Zhang, J. Tejada, S.S. Glasson, L.A. Collins-Racie,
[4] M. Kapoor, J. Martel-Pelletier, D. Lajeunesse, J.P. Pelletier, H. Fahmi, Role of
E.R. Lavallie, Y. Wang, K.C. Chang, S. Nagpal, E.A. Morris, C.R. Flannery, Z. Yang,
proinflammatory cytokines in the pathophysiology of osteoarthritis, Nat. Rev.
LXR modulation blocks prostaglandin E2 production and matrix degradation in
Rheumatol. 7 (1) (2011) 33–42.
cartilage and alleviates pain in a rat osteoarthritis model, Proc. Natl. Acad. Sci. U. S.
[5] S.B. Abramson, M. Attur, A.R. Amin, R. Clancy, Nitric oxide and inflammatory
A. 107 (8) (2010) 3734–3739.
mediators in the perpetuation of osteoarthritis, Curr. Rheumatol. Rep. 3 (6) (2001)
[30] X. Niu, Y. Wang, W. Li, Q. Mu, H. Li, H. Yao, H. Zhang, Protective effects of
535–541.
Isofraxidin against lipopolysaccharide-induced acute lung injury in mice, Int.
[6] O. Alvarez-Garcia, N.H. Rogers, R.G. Smith, M.K. Lotz, Palmitate has proapoptotic
Immunopharmacol. 24 (2) (2015) 432–439.

244
J. Lin et al. International Immunopharmacology 64 (2018) 238–245

[31] M. Wang, E.R. Sampson, H. Jin, J. Li, Q.H. Ke, H.J. Im, D. Chen, MMP13 is a critical of effect of Wogonin, a natural flavonoid, on the viability and activation of NF-
target gene during the progression of osteoarthritis, Arthritis Res. Ther. 15 (1) kappaB and MAPKs in IL-1beta-stimulated human OA chondrocytes, Data Brief 12
(2013) R5. (2017) 150–155.
[32] J.J. Li, J. Nahra, A.R. Johnson, A. Bunker, P. O'Brien, W.S. Yue, D.F. Ortwine, [38] A. Kumar, Y. Takada, A.M. Boriek, B.B. Aggarwal, Nuclear factor-kappaB: its role in
C.F. Man, V. Baragi, K. Kilgore, R.D. Dyer, H.K. Han, Quinazolinones and pyrido health and disease, J. Mol. Med. (Berlin, Germany) 82 (7) (2004) 434–448.
[3,4-d]pyrimidin-4-ones as orally active and specific matrix metalloproteinase-13 [39] A. Liacini, J. Sylvester, W.Q. Li, M. Zafarullah, Inhibition of interleukin-1-stimu-
inhibitors for the treatment of osteoarthritis, J. Med. Chem. 51 (4) (2008) 835–841. lated MAP kinases, activating protein-1 (AP-1) and nuclear factor kappa B (NF-
[33] P.S. Burrage, K.S. Mix, C.E. Brinckerhoff, Matrix metalloproteinases: role in ar- kappa B) transcription factors down-regulates matrix metalloproteinase gene ex-
thritis, Front Biosci 11 (2006) 529–543. pression in articular chondrocytes, Matrix Biology: Journal of the International
[34] S. Takeda, ADAM and ADAMTS family proteins and snake venom metalloprotei- Society for Matrix Biology 21 (3) (2002) 251–262.
nases: a structural overview, Toxins 8 (5) (2016). [40] C. Lianxu, J. Hongti, Y. Changlong, NF-kappaBp65-specific siRNA inhibits expres-
[35] C. Cooper, S. Snow, T.E. McAlindon, S. Kellingray, B. Stuart, D. Coggon, sion of genes of COX-2, NOS-2 and MMP-9 in rat IL-1beta-induced and TNF-alpha-
P.A. Dieppe, Risk factors for the incidence and progression of radiographic knee induced chondrocytes, Osteoarthr. Cartil. 14 (4) (2006) 367–376.
osteoarthritis, Arthritis Rheum. 43 (5) (2000) 995–1000. [41] J. Li, X. Li, Z. Li, L. Zhang, Y. Liu, H. Ding, Isofraxidin, a coumarin component
[36] S. Rigoglou, A.G. Papavassiliou, The NF-kappaB signalling pathway in osteoar- improves high-fat diet induced hepatic lipid homeostasis disorder and macrophage
thritis, Int. J. Biochem. Cell Biol. 45 (11) (2013) 2580–2584. inflammation in mice, 8 (8) (2017) 2886–2896.
[37] N.M. Khan, A. Haseeb, M.Y. Ansari, P. Devarapalli, S. Haynie, T.M. Haqqi, Dataset

245

Das könnte Ihnen auch gefallen