Accepted Manuscript A novel FGFR1-binding peptide attenuates the degeneration of articular cartilage in adult mice Qiaoyan Tan, MD, Bo Chen, MS, Quan Wang, MD, Wei Xu, MD, Yuanqiang Wang, PhD, Zhihua Lin, PhD, Fengtao Luo, MD, Shuo Huang, PhD, Ying Zhu, MD, Nan Su, MD, Min Jin, MD, Can Li, BS, Liang Kuang, MD, Huabing Qi, PhD, Zhenghong Ni, MD, Zuqiang Wang, MD, Xiaoqing Luo, MS, Wanling Jiang, MS, Hangang Chen, BS, Shuai Chen, BS, Fangfang Li, BS, Bin Zhang, BS, Junlan Huang, BS, Ruobin Zhang, BS, Kexin Jin, BS, Xiaoling Xu, PhD, Chuxia Deng, PhD, Xiaolan Du, MS, Yangli Xie, MD, Lin Chen, MD. PhD PII:
S1063-4584(18)31434-1
DOI:
10.1016/j.joca.2018.08.012
Reference:
YJOCA 4300
To appear in:
Osteoarthritis and Cartilage
Received Date: 29 November 2017 Revised Date:
13 August 2018
Accepted Date: 28 August 2018
Please cite this article as: Tan Q, Chen B, Wang Q, Xu W, Wang Y, Lin Z, Luo F, Huang S, Zhu Y, Su N, Jin M, Li C, Kuang L, Qi H, Ni Z, Wang Z, Luo X, Jiang W, Chen H, Chen S, Li F, Zhang B, Huang J, Zhang R, Jin K, Xu X, Deng C, Du X, Xie Y, Chen L, A novel FGFR1-binding peptide attenuates the degeneration of articular cartilage in adult mice, Osteoarthritis and Cartilage (2018), doi: 10.1016/ j.joca.2018.08.012. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT 1
Running title: FGFR1-binding peptide and Osteoarthritis
2
Title: A novel FGFR1-binding peptide attenuates the degeneration of articular
4
cartilage in adult mice
RI PT
3
5
Authors: Qiaoyan Tan1†, MD, Bo Chen1†, MS, Quan Wang1, MD, Wei Xu1, MD,
7
Yuanqiang Wang2, PhD, Zhihua Lin2, PhD, Fengtao Luo1, MD, Shuo Huang1, PhD,
8
Ying Zhu1, MD, Nan Su1, MD, Min Jin1, MD, Can Li1, BS, Liang Kuang1, MD,
9
Huabing Qi1, PhD, Zhenghong Ni1, MD, Zuqiang Wang1, MD, Xiaoqing Luo1, MS,
10
Wanling Jiang1, MS, Hangang Chen1, BS, Shuai Chen, BS1, Fangfang Li1, BS, Bin
11
Zhang1, BS, Junlan Huang1, BS, Ruobin Zhang1, BS, Kexin Jin1, BS, Xiaoling Xu3,
12
PhD, Chuxia Deng3, PhD, Xiaolan Du1*, MS, Yangli Xie1**, MD, Lin Chen1***, MD,
13
PhD
14 1
TE D
M AN U
SC
6
Institution:
Department
of Rehabilitation
16
Rehabilitation of Traumatic Injuries, State Key Laboratory of Trauma, Burns and
17
Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital,
18
Third Military Medical University, Chongqing 400042, China.
19
Bioengineering, Chongqing Institute of Technology, Chongqing 400050, China.
20
3
AC C
EP
15
Laboratory for the
2
College of
Faculty of Health Sciences, University of Macau, Macau SAR 00853, China.
21 22
Medicine,
†These authors contributed equally to this work.
ACCEPTED MANUSCRIPT 23
***
24
Medicine, Laboratory for the Rehabilitation of Traumatic Injuries, State Key
25
Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research
26
Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing
27
400042, China.
28
Tel: 86-23-68757041. Fax: 86-23-68702991. E-mail:
[email protected].
29
**
30
Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of
31
Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery,
32
Daping Hospital, Third Military Medical University, Chongqing 400042, China.
33
Tel: 86-23-68757042. Fax: 86-23-68702991.E-mail:
[email protected].
34
*
35
Laboratory for the Rehabilitation of Traumatic Injuries, State Key Laboratory of
36
Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery,
37
Daping Hospital, Third Military Medical University, Chongqing 400042, China.
38
Tel: 86-23-68757042. Fax: 86-23-68702991.E-mail:
[email protected].
RI PT
M AN U
SC
Corresponding authors. Address: Y. Xie, Department of Rehabilitation Medicine,
EP
TE D
Corresponding authors. Address: X. Du, Department of Rehabilitation Medicine,
AC C
39
Corresponding authors. Address: L. Chen, Department of Rehabilitation
40
Abstract
41
Objective: We previously reported that genetic ablation of Fgfr1 in knee cartilage
42
attenuates the degeneration of articular cartilage in adult mice, which suggests that
43
FGFR1 is a potential targeting molecule for osteoarthritis (OA). Here, we identified
44
R1-P1, an inhibitory peptide for FGFR1 and investigated its effect on the
ACCEPTED MANUSCRIPT pathogenesis of OA in mice induced by destabilization of medial meniscus (DMM).
46
Design: Binding ability between R1-P1 and FGFR1 protein was evaluated by ELISA
47
and molecular docking. Alterations in cartilage were evaluated histologically. The
48
expression levels of molecules associated with articular cartilage homeostasis and
49
FGFR1
50
immunohistochemistry. The chondrocyte apoptosis was detected by TUNEL assay.
51
Results: R1-P1 had highly binding affinities to human FGFR1 protein, and efficiently
52
inhibited ERK1/2 pathway in mouse primary chondrocytes. In addition, R1-P1
53
attenuated the IL-1β induced significant loss of proteoglycan in full-thickness
54
cartilage tissue from human femur head. Moreover, this peptide can significantly
55
restore the IL-1β mediated loss of proteoglycan and type II collagen (Col II) and
56
attenuate the expression of matrix metalloproteinase-13 (MMP13) in mouse primary
57
chondrocytes. Finally, intra-articular injection of R1-P1 remarkably attenuated the
58
loss of proteoglycan and the destruction of articular cartilage and decreased the
59
expressions of ECM degrading enzymes and apoptosis in articular chondrocytes of
60
mice underwent DMM surgery. Conclusions: R1-P1, a novel inhibitory peptide for
61
FGFR1, attenuates the degeneration of articular cartilage in adult mice, which is a
62
potential leading molecule for the treatment of OA.
64
by
qRT-PCR,
Western
blotting
and
EP
TE D
M AN U
SC
analyzed
Key Words: FGFR1; Peptide; Osteoarthritis; Mice
65 66
were
AC C
63
signaling
RI PT
45
Introduction
ACCEPTED MANUSCRIPT Osteoarthritis (OA) is one of the most prevalent chronic joint diseases with
68
progressive cartilage destruction and insufficient extracellular matrix synthesis.
69
Currently, there are no effective biological therapies to prevent or treat OA, the most
70
common final therapeutic option is the total joint replacement.
71
In recent years, a variety of molecules and signaling pathways, such as mTOR, IHH
72
and Wnt/β-catenin signaling have been found to be involved in cartilage homeostasis
73
and OA development (1,2). Fibroblast growth factors (FGF) and their receptors
74
(FGFRs) regulate the maintenance of cartilage and therefore, play a crucial role in
75
joint homeostasis and OA development (3,4,5). FGF2 promotes cartilage catabolism
76
via FGFR1-Ras-Raf-MEK1/2-ERK1/2 axis in human articular chondrocytes (6).
77
FGFR1 and FGFR3 are highly expressed in human articular chondrocytes, and the
78
expression ratio of FGFR1 to FGFR3 was significantly increased in OA chondrocytes
79
compared to normal chondrocytes (7), which suggests the potential role of FGF
80
signaling in cartilage homeostasis. FGF18 can promote chondrogenesis through
81
FGFR3 and is a well-established anabolic growth factor for articular cartilage (8).
82
Surgical DMM in mice is a well-established OA model (9,10), which is commonly
83
used to screen biological and pharmacological agents for OA treatment (11,12).
84
FGFR3 deficiency mice exhibited abnormal cartilage metabolism and early signs of
85
OA, including increased cellular hypertrophy and loss of proteoglycan with
86
upregulated expressions of MMP13, type X collagen (Col X) (13,14). Regarding to
87
FGFR1, our previous study showed that conditional knockout (cKO) of Fgfr1 in
88
cartilage attenuates articular cartilage degeneration, which is associated with
AC C
EP
TE D
M AN U
SC
RI PT
67
ACCEPTED MANUSCRIPT down-regulation of MMP13 and increased proteoglycan synthesis. Thus, we expected
90
that pharmacological down-regulation of the activity of FGFR1 or its downstream
91
pathways may attenuate the development of OA. Since we previously found a short
92
FGFR1-binding peptide (GPPDWHWKAMTH, named as R1-P1) using phage display
93
and demonstrated that it can blunt the activity of ERK1/2 MAPK pathway. We thus
94
tested whether it may exert a protective effect on OA in mouse model. Our results
95
demonstrated that R1-P1 can attenuate the severity of OA induced by DMM in mice.
SC
RI PT
89
96
Materials and Methods
98
Sequence analysis and molecular docking
99
Sequence analysis and molecular docking were carried out according to previous
100
protocols (15,16), The peptide sequence was analyzed with Bioedit and ProtParam
101
programs (http://au.expasy.org/tools/protparam.html). The crystal structure of FGFR1
102
was accessed from RCSB database (ID: 5AM6) and prepared as receptor with
103
biopolymer package of crystal. We determined the binding site based on the crystal
104
structure of FGFR1. Then, the peptide was docked into receptor by surflex-dock
105
method and the pose with highest score was used to generate the peptide/FGFR1
106
complex. Molecular dynamics simulation was performed as following steps: (1)
107
Minimization (1000 steps, restraint receptor); (2) Heating (NVE, 50 ps, 0K-300 K,
108
restraint receptor); (3) Density adjustment (NPT, 50 ps, 300 K, constraint receptor); (4)
109
Equilibrium (NPT, 500 ps, 300 K); (5) Production (NPT, 120 ns, 300 K). The
110
molecular dynamics simulation was performed by amber.
AC C
EP
TE D
M AN U
97
ACCEPTED MANUSCRIPT 111
Peptide synthesis
113
The peptide was synthesized at Shanghai China Peptides Co., Ltd. by solid-phase
114
synthesis and purified by high-performance liquid chromatography (HPLC). Peptide
115
with high purity grade (95.43%) was obtained after purification. The synthetic
116
product was further confirmed by mass spectrometry analysis.
RI PT
112
SC
117
ELISA assay for binding ability
119
Binding affinities were checked by ELISA using previous protocol (17). Briefly,
120
microtiter plates were coated with 20 mg/ml FGFR1 overnight at 4°C. After being
121
blocked at 37°C for 1 h, the plates were washed six times. R1-P1 at different
122
concentrations (0,1,5,10,15,25,50,100 mM) were added and incubated at room
123
temperature for 1 h. After the plates were washed six times, anti-FGFR1 antibody
124
were added and incubated at 37°C for 1 h. The plates were washed six times. Goat
125
anti-rabbit immunoglobulin G (IgG)-HRP was then added and incubated at 37°C for
126
30 min. The plates were washed, and color development was initiated with addition of
127
3,3’,5,5’-tetramethylbenzidine (TMB; 100 ml/well of TMB). The reaction was
128
terminated 20 min later by adding 20 µl stopping buffer, and the absorbance was
129
measured at 450 nm.
AC C
EP
TE D
M AN U
118
130 131
Cell culture
132
Mouse embryonic carcinoma-derived cell line ATDC5 cells were cultured in 12-well
ACCEPTED MANUSCRIPT plates until 90% confluence, then cells were pre-incubated with FGF2 (10 ng/ml) for
134
30 min, followed by treatment with peptide R1-P1 (0, 1, 10, 25 or 50mM) for 2 hours.
135
PD173074 (Selleck), an inhibitor of FGFR1, was used as the positive control. Mouse
136
primary chondrocytes were isolated as previously described (18). Chondrocytes were
137
cultured in 12-well plates until 90% confluence and were pre-incubated with R1-P1
138
(25 or 50 mM) alone for 2 hours, before treatment with IL-1β (10 ng/ml) for 24 hours.
139
SC
RI PT
133
Culture of human articular cartilage explants
141
This project received approval from the Ethical and Protocol Review Committee of
142
Daping Hospital, Chongqing, and the culture were carried out according to the
143
previous protocols (19). Briefly, human full-thickness femur head cartilage tissue was
144
cut into 4 mm diameters, following 48 hours culture, explants were treated with IL-1β
145
(20 ng/ml) and peptide R1-P1 (25 or 50 mM) every 3 days for 14 days under
146
serum-free conditions (plus mini-ITS™ + Premix). Following 14 days of incubation,
147
the medium was collected, and all explants were fixed in 4% paraformaldehyde
148
overnight, followed by paraffin embedding.
TE D
EP
AC C
149
M AN U
140
150
Dimethylmethylene Blue (DMMB) assay
151
DMMB dye solution and sample digestion solution were prepared following previous
152
protocol (20). Briefly, 250 µl of DMMB reagent was added to 40 µl of culture
153
medium and the absorbance was measured at 525 nm immediately. Chondroitin
154
sulfate (Sigma-Adrich) with several concentrations was used to plot a standard curve,
ACCEPTED MANUSCRIPT 155
which was used to measure the amount of aggrecan released into culture medium.
156
Aggrecan released into the medium was normalized as mass of GAG per milliliter (ml)
157
of culture medium.
RI PT
158
RNA extraction and quantitative real-time PCR (qPCR)
160
Total RNA of treated mouse primary chondrocytes was isolated by TRIZOL reagent
161
(Invitrogen). Real-time PCR was repeated at least three times using Mx3000P PCR
162
machine (Stratagene) and SYBR Premix Ex TaqTM kit (Takara). The primers for
163
genes measured were listed in Table 1. Expression values were normalized to
164
Cyclophilin A.
M AN U
SC
159
165
Western blotting
167
Protein was extracted using ice-cold RIPA lysis buffer containing protease inhibitors
168
(Roche). Equal amount of protein samples (30 ug) were dissolved by 10%
169
SDS-PAGE gels and transferred onto a polyvinylidene difluoride membrane
170
(Millipore). After being blocked with 8% nonfat milk in tris buffered saline tween
171
buffer, the membrane was probed with antibody specific to ERK1/2 (1:1000 dilution;
172
Cell Signaling Technology (CST)), p-ERK1/2 (1:1000 dilution; CST), MMP13
173
(1:1000 dilution; Millipore), Aggrecan (1:1000 dilution; Abcam) and β-Actin
174
(1:10000 dilution; Sigma) followed by chemiluminescent (Pierce) detection. All
175
antibodies were solvated with antibody diluent (Beyotime). Intensity values were
176
analyzed with the Image J software and were normalized to those of β-Actin. Each
AC C
EP
TE D
166
ACCEPTED MANUSCRIPT 177
sample was analyzed three times and the mean gray values of immunoblot band were
178
calculated (21).
179
Surgical model of OA in mice
181
All mice were maintained in the Animal Facility (specific pathogen free) of the
182
Daping Hospital (Chongqing, China). All procedures were approved by the
183
Institutional Animal Care and Use Committee of Daping Hospital (Chongqing, China).
184
8-week-old male C57BL/6J mice (25-30 g) were purchased from the Beijing HFK
185
Bioscience Co. Ltd.. According to previous protocol (22), DMM surgery was
186
performed on the right knee joints, sham surgery was performed with medial
187
capsulotomy only on the left knee joints of mice. Mice divided into groups at
188
randomisation. After 1 week, mice in treatment group received R1-P1 (once a week,
189
15µg/week) postoperatively via articular cavity injection for 4, 8 and 12 weeks. In
190
control mice, their articular cavities were injected with physiological saline only.
SC
M AN U
TE D
EP
191
RI PT
180
Histologic assessment
193
Microscopic scoring of mouse cartilage degeneration was performed in accordance
194
with the recommendations of the Osteoarthritis Research Society International
195
(OARSI) (23). Briefly, the joints were fixed in 4% paraformaldehyde, then decalcified
196
in 20% formic acid, and embedded in paraffin. Then serial sagittal sections were
197
obtained across the entire knee joint. Histologic grading of cartilage degeneration was
198
performed using the OARSI recommended subjective scoring system (on a scale of
AC C
192
ACCEPTED MANUSCRIPT 199
0-6) (23). In addition, cartilage aggrecan depletion was scored (on a scale of 0-5) as a
200
complement measure of cartilage degeneration
201
agreement between the two blinded observers (Quan Wang and Wei Xu).
(24). There was a high level of
RI PT
202
Synovitis score
204
Mouse articular joint sections were stained with hematoxylin and eosin (H&E) to
205
assess the grade of synovitis. Scoring was performed by two blinded observers (25).
206
Three features of synovitis (enlargement of lining cell layer, cellular density of
207
synovial stroma, leukocytic infiltrate) were semiquantitatively evaluated (from 0,
208
absent to 3, strong) and each feature was graded separately. The sum provided the
209
synovitis score, which is interpreted as follows: 0-1, no synovitis; 2-4, low-grade
210
synovitis; 5-9, high grade synovitis.
TE D
211
M AN U
SC
203
Immunohistochemistry (IHC)
213
IHC were carried out using SP-9000 Histostain-Plus kits (Zsgb Bio) as previous
214
protocols (14). Incubated with antibody specific to Aggrecan (1:200 dilution; Abcam),
215
Col II (1:400 dilution; Chondrex), Adamts5 (1:200 dilution; Abcam), MMP13 (1:200
216
dilution; Abcam), Col X (1:200 dilution; Millipore) and FGFR3 (1:200 dilution; Santa
217
Cruz Biotechnology) followed by diaminobenzidine (DAB) kit detection, all
218
antibodies were solvated with antibody diluent (Origene).
AC C
EP
212
219 220
TUNEL assay
221
Apoptotic cells in articular cartilage were detected by TdT mediated dUTP-X nick
ACCEPTED MANUSCRIPT 222
end labeling (TUNEL) assay using in situ cell death detection kit (peroxidase (POD))
223
of Roche according to the manual.
224
Statistical analysis
226
We used 5-10 animals in each group for in vivo studies and three independent
227
biological replicates for in vitro studies. Individual cell experiments were also
228
performed in triplicate as technical replicates. The sample sizes for the groups of
229
interest were big enough based on a power of 80% and p < 0.05 using power and
230
sample size calculation online software (http://powerandsamplesize.com/Calculators/)
231
(Supplementary Table 1). All tested variables were tested for normality and
232
homogeneity using Shapiro-Wilk test and Levine’s test, respectively. Quantitative
233
data of histologic grading of cartilage degeneration, IHC analysis of MMP13, ColX,
234
FGFR3 and TUNEL assay were compared by using the non-parametric
235
Mann-Whitney U test. For the other data, all the tested variables did not violate the
236
assumptions of normality and equal variance. Differences between two groups were
237
evaluated using Student’s unpaired t-test, and comparisons of multiple groups were
238
evaluated using analysis of variance (ANOVA) followed by Tukey’s test. All data
239
were analyzed using GraphPad Prism v.6.01 software and depicted in univariate
240
scatter plots as previously described (26). P < 0.05 was considered to be statistically
241
significant.
SC
M AN U
TE D
EP
AC C
242
RI PT
225
243
Results
244
R1-P1 binds to human FGFR1 protein and inhibits MAPK/ERK signaling
245
pathway
ACCEPTED MANUSCRIPT All 11 clones with high binding affinity to FGFR1 protein shared the
247
“GPPDWHWKAMTH” amino acid sequences, which was named as peptide R1-P1.
248
ProtParam programs (http://web.expasy.org/protparam/) were then applied to analyze
249
the sequence and predict its properties. The molecular weight of the R1-P1 is 1462.65,
250
theoretical isoelectric point of the peptide is 6.92, grand average of hydropathicity of
251
the R1-P1 is -1.35 (Table 2).
252
To evaluate the binding ability between peptide R1-P1 and the extracellular region of
253
FGFR1 (5AM6) protein, we first determined the binding site based on the crystal
254
structure of FGFR1. Results indicate that there was a large groove in the surface of
255
FGFR1 (Figure 1A), which could accommodate the peptide. Then the peptide was
256
docked into binding site by surface-dock. The best pose, total score and C score were
257
7.87 and 5 respectively, was combined with FGFR1 to generate FGFR1/pep complex.
258
To achieve a stable structure, a 20 nanoseconds MD simulation was performed.
259
According to the RMSD plot (Figure 1B), the RMSD was fluctuated close to 2.5Å
260
after 70 nanoseconds, which means the structure of complex was reasonable. The
261
peptide could bind to FGFR1 stably (Figure 1C) relied on hydrogen bonds,
262
electrostatic and hydrophobic interaction (Figure 1D). Figure 1D shows that there
263
were 8 hydrogen bonds between peptide with residue Arg493, Arg512, Asp513, Leu484,
264
Gly490, Asp527, Glu571, and Asn568.
265
We examined the binding affinity of peptide R1-P1 at different concentrations
266
(0,1,5,10,15,25,50,100 mM) to FGFR1, ELISA result showed that R1-P1 can bind to
267
FGFR1 protein in a dose-response manner, and the binding affinities were not further
AC C
EP
TE D
M AN U
SC
RI PT
246
ACCEPTED MANUSCRIPT increased from 50 to 100 mM (Figure 1E). In addition, ELISA results demonstrated
269
that R1-P1 strongly bound to FGFR1, although it also slightly bound to FGFR2 and
270
FGFR3 (Figure 1F). FGFs/FGFRs exert their diverse biological effects through
271
multiple intracellular signaling pathways, among which the MAPK pathway has been
272
found to inhibit chondrocyte differentiation and matrix synthesis (6). We examined
273
whether R1-P1 could affect the protein level of total ERK1/2 and phosphorylated
274
ERK1/2 induced by FGF2. Furthermore, western blotting result showed that
275
FGF2-mediated ERK1/2 phosphorylation were partially blocked by R1-P1 at 25 and
276
50mM concentration (Figure 1G), indicating that R1-P1 inhibited ERK1/2 MAPK
277
signaling pathway, a classic downstream signaling pathway of FGFR1 in
278
chondrocytes.
M AN U
SC
RI PT
268
TE D
279
R1-P1 attenuates IL-1β induced proteoglycan loss in adult human articular
281
cartilage
282
To determine the effect of R1-P1 on proteoglycan loss, we treated cultured healthy
283
human femoral head cartilage explant with IL-1β (20 ng/ml) with or without the
284
R1-P1 (25 or 50 mM) for 14 days. Safranin O staining showed that IL-1β induced
285
significant proteoglycan loss, which was partially blunted by peptide R1-P1 treatment
286
(Figure 2A).
287
The GAG released into the culture medium was significantly increased after IL-1β
288
treatment (P < 0.001), which were markedly decreased after being incubated with 25
289
or 50 mM R1-P1 (all P < 0.001, Figure 2B), respectively. These data were consistent
AC C
EP
280
ACCEPTED MANUSCRIPT 290
with the results of Safranin O staining. Together, these results indicate that R1-P1 can
291
attenuate the catabolic effect of IL-1β on chondrocytes.
292
R1-P1 inhibits the phosphorylation of ERK1/2 and rescues IL-1β induced
294
dysregulated ECM metabolism in mouse primary chondrocytes
295
To explore the mechanisms for the anti-catabolic effects of R1-P1 on chondrocytes,
296
real-time qPCR and western blotting were performed to examine the effects of R1-P1
297
on the synthesis and breakdown of extracellular matrix in mouse primary
298
chondrocytes. The mRNA expressions of Adamts5 (P < 0.001) and MMP13 (P <
299
0.001) were significantly increased in mouse primary chondrocytes after IL-1β
300
treatment. R1-P1 treatment resulted in a marked reduction in the expressions of these
301
two catabolic markers (P < 0.001 and P = 0.006, respectively). Meanwhile, the
302
mRNA expressions of Aggrecan (P < 0.001) and Col II (P < 0.001) were decreased
303
after IL-1β treatment, which were partially recovered (P < 0.001 and P < 0.001,
304
respectively) by peptide R1-P1 (Figure 3A).
305
Western blotting results showed that following the stimulation of IL-1β, ERK1/2
306
MAPK signaling pathway was activated, the protein levels of Aggrecan was
307
decreased and MMP13 was increased (all P < 0.001). Treatment with 25 or 50 mM
308
R1-P1 significantly decreased the protein levels of MMP13 and phosphorylated
309
ERK1/2, while up-regulated the protein levels of Aggrecan in mouse primary
310
chondrocytes (all P < 0.001, Figure 3B and 3C). Taken together, these results
311
demonstrate that R1-P1 may help to maintain the homeostasis of mouse primary
AC C
EP
TE D
M AN U
SC
RI PT
293
ACCEPTED MANUSCRIPT 312
chondrocytes.
313
R1-P1 delays articular cartilage degradation in a mouse OA model
315
We determined the effect of R1-P1 on the development of OA in mice induced by
316
DMM. There is no remarkable toxicity observed in the R1-P1 treatment group, as
317
evidenced by no signs of abnormal gross phenotypes including weight loss during
318
observation period. Histologic examination at 4 weeks after DMM surgery revealed
319
the loss of proteoglycan content (data not shown). Furthermore, 8 weeks after DMM
320
mice showed an early OA-like manifestations including loss of proteoglycan content
321
and cartilage tissue (Figure 4A-B). The OA-like phenotype became more profound in
322
mice at 12 weeks after DMM surgery, which exhibited as more severe destruction of
323
the articular cartilage associated with greater loss of proteoglycan content (Figure
324
5A-B). However, intra-articular injection of R1-P1 significantly decreased the
325
cartilage destruction and proteoglycan loss in mouse knee joint after DMM surgery
326
(Figure 4A-B and 5A-B).
327
Accordingly, the OARSI scores were significantly increased in mice at 8 and 12
328
weeks after DMM surgery compared to those of sham operation mice (all P < 0.001,
329
Figure 4C-F and 5C-F). R1-P1 treated mice had a significantly lower sum score and
330
maximal score in medial femoral condyle and medial tibial plateau than those in the
331
vehicle treated mice at 8 weeks (P = 0.001, P = 0.014, P = 0.002 and P = 0.008,
332
respectively, Figure 4C-F) and 12 weeks (P < 0.001, P = 0.002, P = 0.021 and P =
333
0.006, respectively, Figure 5C-F) following DMM surgery. In addition, H&E
AC C
EP
TE D
M AN U
SC
RI PT
314
ACCEPTED MANUSCRIPT staining and scoring of synovitis showed that treatment with R1-P1 attenuated the
335
severity of synovitis at 8 weeks (P = 0.002, Figure 4G-H) and 12 weeks (P < 0.001,
336
Figure 5G-H) following DMM surgery compared with vehicle. Taken together, these
337
results demonstrate that intra-articular injection of R1-P1 partially delayed OA
338
development.
RI PT
334
339
R1-P1 attenuates ECM loss and chondrocyte hypertrophy and apoptosis in knee
341
joints of DMM mice
342
To explore the cellular mechanisms underlying the delayed progression of cartilage
343
degradation in peptide R1-P1 treated mice with DMM, we performed IHC to examine
344
the expressions of Aggrecan, Col II, Col X, Adamts5 and MMP13. The results
345
showed that the expressions of Adamts5, MMP13 were significantly increased and the
346
expressions of Aggrecan, Col II were decreased in the articular cartilage after DMM
347
surgery (Figure 6A-D). In addition, after DMM surgery, the ratio of cells with
348
positive Col X in the articular cartilage was significantly increased (Figure 6E).
349
However, peptide R1-P1 largely lowered the ratio of Adamts5, MMP13, Col X
350
positive cells and significantly increased the ratio of Aggrecan, Col II positive cells in
351
the articular cartilage after DMM surgery compared to vehicle treatment (all P <
352
0.001, Figure 6H). Furthermore, the ratio of FGFR3 positive cells was
353
down-regulated in the articular cartilage when OA was induced by DMM surgery.
354
However, inhibition of FGFR1 activity led to increased ratio of FGFR3 positive cells
355
in the articular cartilage (P < 0.001, Figure 6F and Figure 6H).
AC C
EP
TE D
M AN U
SC
340
ACCEPTED MANUSCRIPT Since it has been reported that chondrocyte apoptosis is strongly related to the OA
357
development (27,28). We performed TUNEL assay to analyze the apoptosis of
358
chondrocytes in knee joints. Our results showed that peptide R1-P1 treatment largely
359
decreased the apoptosis of chondrocytes in the articular cartilage 8 weeks after DMM
360
surgery compared to vehicle treatment (as shown in red arrows, P < 0.001, Figure 6G
361
and Figure 6H). Taken together, these results demonstrate that local intra-articular
362
injection of R1-P1 reduced articular cartilage damage, at least in part, through its
363
inhibition on the hypertrophy and apoptosis process of articular chondrocytes.
M AN U
SC
RI PT
356
364
Discussion
366
OA is a degenerative disease characterized with cartilage degradation, synovial
367
inflammation and dysregulated subchondral bone remodeling. The mechanisms
368
underlying OA is not well understood. Currently, there is no effective biological
369
treatment for OA. Finding targeting cells and/or molecules for effective therapies of
370
OA is urgently needed.
371
Decades of studies have demonstrated that FGFs/FGFRs play vital roles in cartilage
372
homeostasis and OA development (11). We previously demonstrated that conditional
373
deletion of Fgfr1 in mouse articular chondrocytes delays the progression of cartilage
374
degeneration (29), suggesting that pharmacological down-regulation of FGFR1
375
activity is a potential therapy for OA. Over the past decades, several FGFR inhibitors,
376
such as PD173074, SU5402 and PD166866 have been developed as candidates for the
377
treatment of FGF signaling related diseases (30). Because of the low-specificity and
AC C
EP
TE D
365
ACCEPTED MANUSCRIPT toxicity, majority of them failed to enter clinical application. Peptides can be easily
379
synthesized and have low immunogenicity. Furthermore, peptide drugs have been
380
successfully used in clinic such as a1-thymosin (31). Here, we, for the first time,
381
provide evidence showing that a novel FGFR1 antagonist, peptide R1-P1, has high
382
binding affinity to FGFR1 protein and can alleviate cartilage degradation in a mouse
383
model of OA.
384
The major characteristic of OA is an imbalance between anabolic effects mediated
385
extracellular matrix synthesis and catabolic effects regulated matrix degradation (32).
386
We demonstrated that R1-P1 inhibited the proteoglycan degradation induced by IL-1β
387
in cultured human cartilage explants. Furthermore, we demonstrated that
388
R1-P1attenuated the IL-1β induced up-regulation of the mRNA levels of Adamts5 and
389
MMP13 and down-regulation of Aggrecan and Col II mRNA levels, in mouse articular
390
chondrocytes. We speculate that the mechanism for the protective effect of R1-P1 on
391
OA is mainly via its inhibition on the activities of ERK1/2 MAPK signaling pathway.
392
Chondrocyte hypertrophy, which can result in increased metabolic activity of articular
393
chondrocytes and trigger unbalanced cartilage homeostasis favoring degenerative
394
changes (33). We found that R1-P1 treatment reduced the expressions of MMP13 and
395
Col X, markers for hypertrophic chondrocytes, in mouse OA model induced by DMM
396
(12,34). These findings revealed that intra-articular injection of R1-P1 prevented
397
articular chondrocytes from hypertrophy, which may contribute to the inhibitory
398
effect of R1-P1 on OA development. The inhibitory effect of R1-P1 on chondrocyte
399
hypertrophy is consistent with our previous findings showing that Fgfr1 deficiency in
AC C
EP
TE D
M AN U
SC
RI PT
378
ACCEPTED MANUSCRIPT chondrocytes decreased chondrocyte hypertrophy in articular cartilage (29). FGF
401
signaling pathway is highly associated with chondrocyte apoptosis (35).
402
Gain-of-function mutation of Fgfr3 promotes chondrocyte apoptosis in thanatophoric
403
dysplasia (TD) mice (36). Fgf2 transgenic mice exhibit chondrodysplasic phenotype
404
resulting from both reduced proliferation and increased apoptosis of growth plate
405
chondrocytes (37). FGF18 markedly reduces chondrocyte apoptosis and enhances the
406
repair response of cartilage following cartilage insult (38). In current study, we
407
showed that pharmacologically inhibiting FGFR1 activity by R1-P1 decreased
408
chondrocyte apoptosis in mouse OA model, indicating that R1-P1 positively
409
maintains cartilage homeostasis partially through downregulating chondrocyte
410
apoptosis.
411
During OA development, the expression ratio of FGFR1 to FGFR3 was up-regulated
412
in articular chondrocytes from patients with OA (7), suggesting that down-regulation
413
of FGFR3 is at least partially responsible for the development of OA in FGFR1
414
deficiency related OA. We found that intra-articular injection of R1-P1 can increase
415
ratio of FGFR3 positive cells in mouse OA model.
416
Whether R1-P1 have the same effects in other animal model need to be studied. In
417
addition, inflammation also plays essential role in the pathogenesis of OA by inducing
418
catabolic effects on ECM, etc. Inhibition of inflammation leads to attenuated OA
419
severity and progression (39,40). As a classic signaling pathway in development and
420
homeostasis of a variety of tissues/organs, FGF signaling appears to have crosstalk
421
with inflammation related signaling pathways. For example, inhibition of FGF23
AC C
EP
TE D
M AN U
SC
RI PT
400
ACCEPTED MANUSCRIPT might serve as a novel anti-inflammatory strategy in chronic obstructive pulmonary
423
disease (COPD) (41). So, we speculate that R1-P1 and anti-inflammatory drug may
424
have overlapping but different effects on attenuating OA progression.
425
In summary, we found a novel inhibitory peptide, R1-P1, for FGFR1 that can
426
maintain the ECM of chondrocytes through its down-regulation of matrix-degrading
427
enzymes and prevent articular chondrocytes from hypertrophy and apoptosis, which
428
will facilitate the development of effective therapeutic agents for OA.
SC
RI PT
422
M AN U
429
Author contributions
431
All authors listed have read and approved all versions of the manuscript.
432
Conception and design: Qiaoyan Tan, Xiaolan Du, Yangli Xie, Lin Chen.
433
Analysis and interpretation of the data: Qiaoyan Tan, Quan Wang, Wei Xu, Yuanqiang
434
Wang, Huabing Qi, Yangli Xie.
435
Drafting of the article: Qiaoyan Tan, Yangli Xie, Lin Chen.
436
Critical revision of the article for important intellectual content: Chuxia Deng, Yangli
437
Xie, Lin Chen.
438
Final approval of the article: Qiaoyan Tan, Xiaolan Du, Yangli Xie, Lin Chen.
439
Provision of study materials or patients: Qiaoyan Tan, Shuo Huang, Zhenhong Ni,
440
Liang Kuang, Nan Su, Xiaolan Du.
441
Statistical expertise: Qiaoyan Tan, Liang Kuang, Wei Xu.
AC C
EP
TE D
430
ACCEPTED MANUSCRIPT Administrative, technical, or logistic support: Yin Zhu, Wanling Jiang, Hangang Chen,
443
Shuai Chen, Fangfang Li, Bin Zhang, Junlan Huang, Ruobing Zhang, Kexin Jin, Can
444
Li, Min Jin, Xiaolan Du.
445
Collection and assembly of data: All authors.
RI PT
442
446
Fundings
448
This work was supported by Special Funds for Major State Basic Research Program
449
of China (973 program) (No.2014CB942904), National Natural Science Foundation
450
of China (No. 81530071, No.81472074), State Key Laboratory of Trauma, Burn and
451
Combined Injury, China (No.201601).
M AN U
SC
447
452
Conflict of interest
454
The authors declare that they have no competing interests.
TE D
453
456
References
457 458 459 460 461 462 463 464 465 466 467 468 469 470
1.
EP
455
Ma, B., van Blitterswijk, C. A., and Karperien, M. (2012) A Wnt/beta-catenin negative
AC C
feedback loop inhibits interleukin-1-induced matrix metalloproteinase expression in human articular chondrocytes. Arthritis and rheumatism 64, 2589-2600
2.
Zhang, Y., Vasheghani, F., Li, Y. H., Blati, M., Simeone, K., Fahmi, H., Lussier, B., Roughley,
P., Lagares, D., Pelletier, J. P., Martel-Pelletier, J., and Kapoor, M. (2015) Cartilage-specific deletion of mTOR upregulates autophagy and protects mice from osteoarthritis. Annals of the rheumatic diseases 74, 1432-1440
3.
Ellman, M. B., Yan, D., Ahmadinia, K., Chen, D., An, H. S., and Im, H. J. (2013) Fibroblast growth factor control of cartilage homeostasis. Journal of cellular biochemistry 114, 735-742
4.
Ornitz, D. M. (2000) FGFs, heparan sulfate and FGFRs: complex interactions essential for development. BioEssays : news and reviews in molecular, cellular and developmental biology 22, 108-112
5.
Ornitz, D. M., and Marie, P. J. (2015) Fibroblast growth factor signaling in skeletal development and disease. Genes & development 29, 1463-1486
ACCEPTED MANUSCRIPT 6.
Yan, D., Chen, D., and Im, H. J. (2012) Fibroblast growth factor-2 promotes catabolism via FGFR1-Ras-Raf-MEK1/2-ERK1/2 axis that coordinates with the PKCdelta pathway in human articular chondrocytes. Journal of cellular biochemistry 113, 2856-2865
7.
Yan, D., Chen, D., Cool, S. M., van Wijnen, A. J., Mikecz, K., Murphy, G., and Im, H. J. (2011) Fibroblast growth factor receptor 1 is principally responsible for fibroblast growth factor 2-induced catabolic activities in human articular chondrocytes. Arthritis research & therapy 13, R130 Davidson, D., Blanc, A., Filion, D., Wang, H., Plut, P., Pfeffer, G., Buschmann, M. D., and
RI PT
8.
Henderson, J. E. (2005) Fibroblast growth factor (FGF) 18 signals through FGF receptor 3 to promote chondrogenesis. The Journal of biological chemistry 280, 20509-20515 9.
Ellman, M. B., Yan, D., Ahmadinia, K., Chen, D., An, H. S., and Im, H. J. (2013) Fibroblast growth factor control of cartilage homeostasis. Journal of cellular biochemistry 114, 735-742 Davidson, D., Blanc, A., Filion, D., Wang, H. F., Plut, P., Pfeffer, G., Buschmann, M. D., and
SC
10.
Henderson, J. E. (2005) Fibroblast growth factor (FGF) 18 signals through FGF receptor 3 to promote chondrogenesis. Journal of Biological Chemistry 280, 20509-20515 11.
Sharif, M., Whitehouse, A., Sharman, P., Perry, M., and Adams, M. (2004) Increased apoptosis
M AN U
in human osteoarthritic cartilage corresponds to reduced cell density and expression of caspase-3. Arthritis and rheumatism 50, 507-515 12.
Weng, T., Xie, Y., Yi, L., Huang, J., Luo, F., Du, X., Chen, L., Liu, C., Chen, D., and Chen, L. (2014) Loss of Vhl in cartilage accelerated the progression of age-associated and surgically induced murine osteoarthritis. Osteoarthritis Cartilage 22, 1197-1205
13.
Tang, J., Su, N., Zhou, S., Xie, Y., Huang, J., Wen, X., Wang, Z., Wang, Q., Xu, W., Du, X., Chen, H., and Chen, L. (2016) Fibroblast Growth Factor Receptor 3 Inhibits Osteoarthritis
14.
TE D
Progression in the Knee Joints of Adult Mice. Arthritis & rheumatology 68, 2432-2443 Valverde-Franco, G., Binette, J. S., Li, W., Wang, H., Chai, S., Laflamme, F., Tran-Khanh, N., Quenneville, E., Meijers, T., Poole, A. R., Mort, J. S., Buschmann, M. D., and Henderson, J. E. (2006) Defects in articular cartilage metabolism and early arthritis in fibroblast growth factor receptor 3 deficient mice. Human molecular genetics 15, 1783-1792 Bunney, T. D., Wan, S., Thiyagarajan, N., Sutto, L., Williams, S. V., Ashford, P., Koss, H.,
EP
15.
Knowles, M. A., Gervasio, F. L., Coveney, P. V., and Katan, M. (2015) The Effect of Mutations on Drug Sensitivity and Kinase Activity of Fibroblast Growth Factor Receptors: A Combined Experimental and Theoretical Study. EBioMedicine 2, 194-204
AC C
471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514
16.
Chen, H., Huang, Z., Dutta, K., Blais, S., Neubert, T. A., Li, X., Cowburn, D., Traaseth, N. J.,
and Mohammadi, M. (2013) Cracking the molecular origin of intrinsic tyrosine kinase activity through analysis of pathogenic gain-of-function mutations. Cell reports 4, 376-384
17.
Jin, M., Yu, Y., Qi, H., Xie, Y., Su, N., Wang, X., Tan, Q., Luo, F., Zhu, Y., Wang, Q., Du, X.,
Xian, C. J., Liu, P., Huang, H., Shen, Y., Deng, C. X., Chen, D., and Chen, L. (2012) A novel FGFR3-binding peptide inhibits FGFR3 signaling and reverses the lethal phenotype of mice mimicking human thanatophoric dysplasia. Human molecular genetics 21, 5443-5455
18.
Wang, X., Qi, H., Wang, Q., Zhu, Y., Wang, X., Jin, M., Tan, Q., Huang, Q., Xu, W., Li, X., Kuang, L., Tang, Y., Du, X., Chen, D., and Chen, L. (2015) FGFR3/fibroblast growth factor receptor 3 inhibits autophagy through decreasing the ATG12-ATG5 conjugate, leading to the delay of cartilage development in achondroplasia. Autophagy 11, 1998-2013
19.
Xu, W., Xie, Y., Wang, Q., Wang, X., Luo, F., Zhou, S., Wang, Z., Huang, J., Tan, Q., Jin, M.,
ACCEPTED MANUSCRIPT Qi, H., Tang, J., Chen, L., Du, X., Zhao, C., Liang, G., and Chen, L. (2016) A novel fibroblast growth factor receptor 1 inhibitor protects against cartilage degradation in a murine model of osteoarthritis. Scientific reports 6, 24042 20.
Stanton, H., Golub, S. B., Rogerson, F. M., Last, K., Little, C. B., and Fosang, A. J. (2011) Investigating ADAMTS-mediated aggrecanolysis in mouse cartilage. Nature protocols 6, 388-404
21.
Ranstam, J. (2012) Repeated measurements, bilateral observations and pseudoreplicates, why 473-475
22.
RI PT
does it matter? Osteoarthritis and cartilage / OARS, Osteoarthritis Research Society 20, Glasson, S. S., Blanchet, T. J., and Morris, E. A. (2007) The surgical destabilization of the medial meniscus (DMM) model of osteoarthritis in the 129/SvEv mouse. Osteoarthritis and cartilage / OARS, Osteoarthritis Research Society 15, 1061-1069
Glasson, S. S., Chambers, M. G., Van Den Berg, W. B., and Little, C. B. (2010) The OARSI
SC
23.
histopathology initiative - recommendations for histological assessments of osteoarthritis in the mouse. Osteoarthritis and cartilage / OARS, Osteoarthritis Research Society 18 Suppl 3, S17-23
Little, C. B., Meeker, C. T., Golub, S. B., Lawlor, K. E., Farmer, P. J., Smith, S. M., and
M AN U
24.
Fosang, A. J. (2007) Blocking aggrecanase cleavage in the aggrecan interglobular domain abrogates cartilage erosion and promotes cartilage repair. The Journal of clinical investigation 117, 1627-1636 25.
Krenn, V., Morawietz, L., Burmester, G. R., Kinne, R. W., Mueller-Ladner, U., Muller, B., and Haupl, T. (2006) Synovitis score: discrimination between chronic low-grade and high-grade synovitis. Histopathology 49, 358-364
Weissgerber, T. L., Milic, N. M., Winham, S. J., and Garovic, V. D. (2015) Beyond bar and
TE D
26.
line graphs: time for a new data presentation paradigm. PLoS biology 13, e1002128 27.
Kim, H. A., and Blanco, F. J. (2007) Cell death and apoptosis in osteoarthritic cartilage. Curr Drug Targets 8, 333-345
28.
Weng, T., Xie, Y., Huang, J., Luo, F., Yi, L., He, Q., Chen, D., and Chen, L. (2014)
EP
Inactivation of Vhl in osteochondral progenitor cells causes high bone mass phenotype and protects against age-related bone loss in adult mice. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 29, 820-829 29.
Weng, T., Yi, L., Huang, J., Luo, F., Wen, X., Du, X., Chen, Q., Deng, C., Chen, D., and Chen,
AC C
515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558
L. (2012) Genetic inhibition of fibroblast growth factor receptor 1 in knee cartilage attenuates the degeneration of articular cartilage in adult mice. Arthritis and rheumatism 64, 3982-3992
30.
Wang, Y., Cai, Y., Ji, J., Liu, Z., Zhao, C., Zhao, Y., Wei, T., Shen, X., Zhang, X., Li, X., and
Liang, G. (2014) Discovery and identification of new non-ATP competitive FGFR1 inhibitors with therapeutic potential on non-small-cell lung cancer. Cancer letters 344, 82-89
31.
Safieh-Garabedian, B., Dardenne, M., Pleau, J. M., and Saade, N. E. (2002) Potent analgesic and anti-inflammatory actions of a novel thymulin-related peptide in the rat. Br J Pharmacol 136, 947-955
32.
Davidson, R. K., Jupp, O., de Ferrars, R., Kay, C. D., Culley, K. L., Norton, R., Driscoll, C., Vincent, T. L., Donell, S. T., Bao, Y., and Clark, I. M. (2013) Sulforaphane represses matrix-degrading proteases and protects cartilage from destruction in vitro and in vivo. Arthritis and rheumatism 65, 3130-3140
ACCEPTED MANUSCRIPT D'Lima, D., Hermida, J., Hashimoto, S., Colwell, C., and Lotz, M. (2006) Caspase inhibitors reduce severity of cartilage lesions in experimental osteoarthritis. Arthritis and rheumatism 54, 1814-1821 34.
Ornitz, D. M., and Marie, P. J. (2002) FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes & development 16, 1446-1465
35.
Sahni, M., Raz, R., Coffin, J. D., Levy, D., and Basilico, C. (2001) STAT1 mediates the
RI PT
increased apoptosis and reduced chondrocyte proliferation in mice overexpressing FGF2. Development 128, 2119-2129 36.
Gigout, A., Lindemann, S., and Guehring, H. (2014) Sprifermin (Rhfgf18) Has an Anabolic Effect on Human Osteoarthritic Chondrocytes Involving Fgfr3 and Erk1/2 but Not P38 Alpha and Jnks. Osteoarthr Cartilage 22, S164-S164
Liang, G., Li, X., Chen, L., Yang, S., Wu, X., Studer, E., Gurley, E., Hylemon, P. B., Ye, F.,
SC
37.
and Li, Y. (2008) Synthesis and anti-inflammatory activities of mono-carbonyl analogues of curcumin. Bioorganic & medicinal chemistry letters 18, 1525-1529 38.
Li, X., Ellman, M. B., Kroin, J. S., Chen, D., Yan, D., Mikecz, K., Ranjan, K. C., Xiao, G.,
M AN U
Stein, G. S., Kim, S. G., Cole, B., van Wijnen, A. J., and Im, H. J. (2012) Species-specific biological effects of FGF-2 in articular cartilage: implication for distinct roles within the FGF receptor family. Journal of cellular biochemistry 113, 2532-2542 39.
Chen, L. X., Lin, L., Wang, H. J., Wei, X. L., Fu, X., Zhang, J. Y., and Yu, C. L. (2008) Suppression of early experimental osteoarthritis by in vivo delivery of the adenoviral vector-mediated NF-kappaBp65-specific siRNA. Osteoarthritis Cartilage 16, 174-184
40.
Chen, Q., Wu, S., Wu, Y., Chen, L., and Pang, Q. (2018) MiR-149 suppresses the
TE D
inflammatory response of chondrocytes in osteoarthritis by down-regulating the activation of TAK1/NF-kappaB. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 101, 763-768 41.
Krick, S., Grabner, A., Baumlin, N., Yanucil, C., Helton, S., Grosche, A., Sailland, J., Geraghty, P., Viera, L., Russell, D. W., Wells, J. M., Xu, X., Gaggar, A., Barnes, J., King, G. D., Campos, M., Faul, C., and Salathe, M. (2018) Fibroblast Growth Factor 23 and Klotho
EP
589
33.
Contribute to Airway Inflammation. The European respiratory journal
AC C
559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588
590
Figure Legends
591
Figure 1. Specific binding of the R1-P1 to FGFR1. (A) The binding site base
592
crystal structure of FGFR1. (B) R1-P1 was docked into binding site by surface-dock.
593
(C) R1-P1 could bind to FGFR1 stably. (D) Showed there were 8 hydrogen bonds
594
between peptide with residue. (E) Binding affinities of R1-P1 at different
595
concentrations (0,1,5,10,15,25,50,100 mM) to FGFR1 revealed by ELISA (n = 3). (F)
ACCEPTED MANUSCRIPT 596
Binding affinities of R1-P1 to FGFRs measured by ELISA (n = 6). (G) Results
597
showed peptide R1-P1 inhibited phosphorylation of ERK1/2 induced by FGF2 (n = 3).
598
Data in were expressed as the mean ± 95% confidence intervals.
RI PT
599
Figure 2. Effects of R1-P1 on proteoglycan loss in adult human articular
601
cartilage. Full thickness cartilage samples from human femur were cultured in the
602
absence or presence of IL-1β (20 ng/ml) and in the presence or absence of R1-P1 (25
603
or 50 mM) for 14 days. (A) Sections were stained with Safranin O to identify the
604
cartilage proteoglycan loss (n = 3). (B) Culture medium were collected, and the
605
amount of GAG released into the medium were quantified by DMMB assay. GAG
606
released into the medium was normalized as mass of GAG per ml of culture medium
607
(n = 9). Scale bar: 100 µm. Data were expressed as the mean ± 95% confidence
608
intervals.
M AN U
TE D
609
SC
600
Figure 3. Effects of R1-P1 on mRNA levels of MMP13, Adamts5, Col II and
611
Aggrecan and on the protein levels of MMP13, Aggrecan and p-ERK1/2 in IL-1β
612
treated mouse primary chondrocytes. (A) Total RNA was isolated, and levels of
613
mRNA of the catabolic markers (MMP13 and Adamts5) and chondrocyte markers
614
(Col II and Aggrecan) were detected by Real-time qPCR (n = 9). (B) Cell lysates
615
were analyzed by western blotting using antibodies specific for MMP13, Aggrecan
616
and p-ERK1/2 (n = 3). (C) The signal intensities of MMP13, Aggrecan and
617
phospho-ERK1/2 were quantitatively analyzed (n = 3). Data were expressed as the
AC C
EP
610
ACCEPTED MANUSCRIPT 618
mean ± 95% confidence intervals.
619
Figure 4. Histologic features of mouse articular at 8 weeks following DMM
621
surgery. (A) The articular cartilage was stained with Safranin O-fast green at 8 weeks
622
after surgery to assess the extent of articular cartilage degeneration. (B) Expansion of
623
the region occupied by articular cartilage. (C-F) OARSI scoring system showed more
624
severe articular cartilage destruction in the medial femur and tibia of mice at 8 weeks
625
following DMM surgery compared with sham operation (n = 8 or 10 mice per group).
626
(G-H) H&E staining and scoring of synovitis showed that treatment with R1-P1 can
627
attenuate the severity of synovitis after DMM surgery compared with vehicle (n = 8
628
or 10 mice per group). MFC: medial femoral condyle; MTP: medial tibial plateau.
629
Scale bar: 100 µm. Data were expressed as the mean ± 95% confidence intervals.
SC
M AN U
TE D
630
RI PT
620
Figure 5. Histologic features of mouse articular cartilage at 12 weeks following
632
DMM surgery. (A) The articular cartilage was stained with Safranin O-fast green at
633
12 weeks after surgery to assess the extent of articular cartilage degeneration. (B)
634
Expansion of the region occupied by articular cartilage. (C-F) OARSI scoring system
635
showed more severe articular cartilage destruction in the medial femur and tibia of
636
mice at 12 weeks following DMM surgery compared with sham operation (n = 5 mice
637
per group). (G-H) H&E staining and scoring of synovitis showed that treatment with
638
peptide R1-P1 can attenuate the severity of synovitis after DMM surgery compared
639
with vehicle (n = 5 mice per group). MFC: medial femoral condyle; MTP: medial
AC C
EP
631
ACCEPTED MANUSCRIPT 640
tibial plateau. Scale bar: 100 µm. Data were expressed as the mean ± 95% confidence
641
intervals.
642
Figure 6. Effects of peptide R1-P1 on cartilage ECM levels and degradation
644
enzymes and chondrocyte apoptosis in mice at 8 weeks after DMM. IHC analysis
645
of Aggrecan (A), Col II (B), Adamts5 (C), MMP13 (D), Col X (E), FGFR3 (F)
646
protein expression in articular cartilage of mice from either sham operation group or
647
surgery group at 8 weeks after DMM surgery with intra-articular injection of peptide
648
R1-P1 or vehicle (n = 3). (G) TUNEL assay was performed on knee joints to measure
649
chondrocyte apoptosis either sham operation or at 8 weeks after DMM surgery with
650
intra-articular injection of peptide R1-P1 or vehicle (as shown in red arrows) (n = 3).
651
(H) The percentage of cells that are positive for MMP13, Col X, FGFR3 and TUNEL
652
in the articular cartilage were calculated (n = 3). Scale bar: 100 µm. Data were
653
expressed as the mean ± 95% confidence intervals.
AC C
EP
TE D
M AN U
SC
RI PT
643
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT Table 1. Primers and sequences for real-time PCR analysis. Forward primer
5'-CTGTGGAGGTCACTGTAGACT-3'
Adamts5
5'-GGAGCGAGGCCATTTACAAC-3'
5'-CGTAGACAAGGTAGCCCACTTT-3'
Col II
5'-CTGGTGGAGCAGCAAGAGCAA-3'
5'-CAGTGGACAGTAGACGGAGGAAAG-3'
Aggrecan
5'-CCTGCTACTTCATCGACCCC-3'
5'-AGATGCTGTTGACTCGAACCT-3'
Cyclophilin A
5'-CGAGCTCTGAGCACTGGAGA-3'
5'-TGGCGTGTAAAGTCACCACC-3'
RI PT
5'-CTTCTTCTTGTTGAGCTGGACTC-3'
SC
MMP13
Reverse primer
Table 2. Properties of peptides displayed by positive phages
Clones
Sequence
Molecular weight
Theoretical pI a
GRAVY b
R1-P1
11
GPPDWHWKAMTH
1462.65
6.92
-1.35
PI, Isoelectric Point.
b
GRAVY, Grand Average of Hydropathicity.
AC C
EP
TE D
a
M AN U
Heptapeptide