Accepted Manuscript + Toll-like receptor-4 Signaling Improved the Migration of Sca-1 stem/progenitor Cells Ying Huang, Chunya Zhang, Jinghua Shen, Xiaogang Zhang, Jianqing Du, Daifu Zhang PII:
S0890-5096(16)31397-8
DOI:
10.1016/j.avsg.2017.07.022
Reference:
AVSG 3514
To appear in:
Annals of Vascular Surgery
Received Date: 26 December 2016 Revised Date:
0890-5096 0890-5096
Accepted Date: 18 July 2017
Please cite this article as: Huang Y, Zhang C, Shen J, Zhang X, Du J, Zhang D, Toll-like receptor-4 + Signaling Improved the Migration of Sca-1 stem/progenitor Cells, Annals of Vascular Surgery (2017), doi: 10.1016/j.avsg.2017.07.022. 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
Toll-like receptor-4 Signaling Improved the Migration of Sca-1+stem/progenitor
2
Cells
3
Ying Huang1, Chunya Zhang1, Jinghua Shen, Xiaogang Zhang, Jianqing Du, Daifu Zhang*
4
1
RI PT
Dept of Cardiology, Pudong New Area People' Hospital, Shanghai, 200120, China
M AN U
SC
2
5
1
6
*Address correspondence to:
7
Daifu Zhang, MD
8
Dept of Cardiology, Pudong New Area People' Hospital, Shanghai, 200120, China
9
Phone: +86(021) 66345628
TE D
equal contribution to this paper
E-mail:
[email protected]
11
There is no conflict of Interest.
AC C
EP
10
1
ACCEPTED MANUSCRIPT
Abbreviation:
13
TLR, toll-like receptor; SPC, stem/progenitor cell; oxLDL, oxidized low density
14
lipoprotein; MMP, Matrix metalloproteinase; PRR, pattern recognition receptor;
15
NF-kB, nuclear factor kB; IRF, IFN-regulatory factor; PMB, polymyxin B.
AC C
EP
TE D
M AN U
SC
RI PT
12
2
ACCEPTED MANUSCRIPT 16
Abstract Atherosclerosis is considered as a chronic inflammatory process, during which
18
macrophages and smooth muscle cells migrate into the vascular wall intima to initiate
19
atherogenesis. Stem/progenitor cells (SPCs) have been demonstrated as a new source
20
of monocytes/macrophages and smooth muscle cells (SMCs). However, the regulation
21
of SPCs migration into atherosclerotic initiation sites to produce macrophages and
22
SMCs in-situis not fully elucidated. As toll-like receptor (TLRs) signaling plays
23
essential role in migration of immunocytes, we wanted to explore the expression and
24
function of TLRs in SPCs in detail. For this we isolated Sca-1+ bone marrow cells as
25
SPCs, and TLRs were detected with quantitative RT-PCR and Western blotting. Our
26
data show that multiple TLRs expressed on SPCs, with TLR4 as the most prominent
27
one, and activation of TLR4 in SPCs is required for migration as assessed by
28
conventional wound healing and transwell assays. Knockdown of TLR4 expression
29
abrogates the migration of SPCs significantly, and re-expression restores it, further
30
confirming functions of TLR4 in regulating SPCs biological behavior.
EP
TE D
M AN U
SC
RI PT
17
In summary, our study shows the essential function of TLR4 signaling in SPC
32
migration, strongly suggesting of its role in atherosclerotic lesion formation. Our data
33
provide new insight into the biological regulators in SPC migration, with potential
34
consequences for therapeutics of atherosclerosis.
35
Key words: toll-like receptor 4 (TLR 4); stem/progenitor cell (SPC); migration
AC C
31
36
3
ACCEPTED MANUSCRIPT 37
Introduction Atherosclerosis has been considered a chronic inflammatory process complicated
39
by multiple genetic and non-genetic factors [1-5]. The atherosclerotic lesion is
40
characterized by cellular infiltration of immunocytes, such as macrophages, smooth
41
muscle cells (SMCs) and accumulation of non-cellular substances, such as lipids, in
42
the artery wall. The process is enhanced with deposition of extracellular matrix
43
composed mostly of collagens to form a fibrous cap[6].
SC
RI PT
38
It has been assumed that oxidized low density lipoprotein (oxLDL), as well as
45
turbulence of blood flow activates local endothelial cells, which express diverse
46
adhesion molecules and chemokines, leading to transmigration of monocytes into the
47
vessel wall intima. Monocytes in the intima, upon local chemokine-cytokine
48
stimulation, differentiate into macrophages, which take up oxLDL and eventually
49
become foam cells. Meanwhile, cytokines from activated endothelial cells lead to
50
SMC migration into the intima, and proliferation to produce a variety of extracellular
51
matrix components [2-4]. However, increasing evidence shows that stem/progenitor
52
cells (SPCs) represent an alternative source of monocytes/macrophages and SMCs
53
[7-9] to participate in the pathophysiology of atherosclerosis.
TE D
EP
AC C
54
M AN U
44
Studies have demonstrated identified both bone marrow–derived SPCs and local
55
SPCs are involved in atherosclerotic lesions, respectively, and SPCs from both
56
sources are necessary for atherosclerosis to produce macrophages and SMCs [9, 10].
57
Consequently, the regulators of SPC migration are of intensive interests also as
58
potential sources of new therapeutics.
4
ACCEPTED MANUSCRIPT 59
The innate immune system is responsible for constant scanning and early detection
61
pathogen-associated molecular patterns from microbes via a number of pattern
62
recognition receptors (PRRs) [11-14]. Recent evidence, however, indicates that PRRs
63
recognize damage-associated molecular patterns from injured tissue and play essential
64
roles in diverse biological events, such as inflammation, tissue repair, and
65
development [15-18]. Toll-like receptors (TLRs) comprise a major group of such
66
PRRs. To date, 13 TLRs have been identified in mammalian cells; 10 of which (TLR1
67
to TLR10) are expressed inhuman cells. These TLRs are located either on the cell
68
surface or in intracellular compartments and detect diverse, distinct signals [19-21].
69
Upon binding of respective ligands, TLR interacts with the TIR domain-containing
70
adapters to initiate NF-kB signaling cascade and/or IFN-regulatory factor (IRF)
71
signaling cascade. This leads to the production of proinflammatory cytokines and type
72
I interferons, thus contributing to [4, 22, 23]. Expression of TLRs has been reported
73
on immunocytes, such as macrophages and dendritic cells during atherogenic plaque
74
generation.
invading
microorganisms
through
recognizing
diverse
SC
M AN U
TE D
EP
AC C
75
of
RI PT
60
However, the expression and function of TLRs in SPCs remain to be determined.
76
In this study we report the expression pattern of different TLRs in SPCs, and explore
77
the TLRs in regulating the biological behavior of SPCs. Our data show that TLR4 is
78
particularly expressed in SPCs, and blocking of TLR4 signaling decrease such
79
migration capabilities, suggesting of potential therapeutic effect in atherosclerosis.
80
Materials and Methods 5
ACCEPTED MANUSCRIPT 81
Lentiviral vectors construction pLVX-shRNA-zsGreen was used to construct the recombinant lentivirus
83
containing TLR4 or control shRNA sequences from Invivogen (San Diego,
84
USA).TLR4 was cloned into pLVX-IRES-ZsGreen vector to construct the
85
recombinant lentivirus. Recombinant lentiviruses were produced by transfecting
86
HEK293T cells (ATCC, USA) with pMD.2G, psPAX2 (Takara, Japan). An MOI
87
(Multiplicity of Infection) of 10 was used to inoculate SPCs, and the efficiency was
88
checked 48 hours later with fluorescence microscopy.
89
Isolation of Sca-1+ stem/progenitor cells
M AN U
SC
RI PT
82
Sca-1+SPCs were isolated from bone marrow of mice by fluorescence cell sorter
91
assay as described previously [10]. In brief, bone marrow was isolated from the
92
femurs and tibias of mice 6-12 weeks of age. The bone marrow was flushed out and
93
subjected a 70µm nylon mesh cell strainer, and was resuspended in DMEM (Gibco
94
BRL Co.Ltd.,USA) supplemented with 2%Fetal Calf Serum (FCS, Gibco BRL
95
Co.Ltd.,USA). After red blood cells lysis, the bone marrow mononuclear cells were
96
washed three times, and resuspended in DMEM with 5%Bovine Serum Albumin
97
(BSA, Gibco BRL Co.Ltd.,USA) at 1x106cells/100µl. Next, the single cells were
98
stained with control FITC-IgG(Cell Signal Technology,USA)or FITC-Sca-1 antibody
99
(Cell Signal Technology ,USA), followed by fluorescence cell sorting assay using
100
flow cytometry (Becton Dickinson, Heidelberg, Germany). Sca-1+ cells were
101
collected in DMEM with 5% BSA and kept on ice for other experiments.
102
Stem/progenitor cell migration assay
AC C
EP
TE D
90
6
ACCEPTED MANUSCRIPT Transwell assay was used for the cell migration assay as previously described
104
[24]. Briefly, Sca-1+SPCs were trypsinization with 0.25% trypsin and 0.02% EDTA in
105
PBS, counted and suspended at a density of 1×106cells/ml in 1% BSA DMEM. The
106
underside of the inserts was pre-coated with 20 g/ml laminin1 (Sigma St. Louis,
107
USA), or pre-grown with monolayer of endothelial cells (The cells were purchased
108
from ATCC and grown in DMEM supplemented with 15% FBS).1.0 × 105cells in
109
300 l serum-free medium were added into each inserts in a 24-well tissue culture
110
plate and 500 l of DMEM medium containing 1% Fetal Bovine Serum (FBS, Gibco
111
BRL Co.Ltd., USA),and SDF-1 (100ng/ml, Sigma St. Louis, USA), were added to the
112
lower chamber. The cells in the Transwell plates were incubated at 37°C for24 h. The
113
cells that had migrated into the bottom well with the cells on the lower surface were
114
quantified. Migration activity was displayed as the percentage (%) of migrated cells to
115
total cells. All experiments were performed in three independent experiments in
116
triplicate.
TE D
M AN U
SC
RI PT
103
For the wound-healing assay, cells were grown in 24-well plate, and with or
118
without pretreated with PMB (20ug/ml, Sigma St. Louis, USA), the cell monolayers
119
were incubated with lipopolysaccharide (LPS, Sigma St. Louis, USA), for 4 hours,
120
then were scraped by the sterile 26G needle to produce a wound to confluent cell layer.
121
After washes with PBS, the wells were added new medium. After overnight culture at
122
37°C, the cells were fixed and images were taken.
123
Quantitative real-time PCR
124
AC C
EP
117
Total RNA was prepared from SPC cells and was reverse-transcribed using 7
ACCEPTED MANUSCRIPT qRT-PCR kit (Invitrogen, San Diego, Ca) Quantitative real-time PCR was carried out
126
on an ABI Prism 7900HT (Applied Biosystems, USA). The program conditions were
127
as follows:2-minute incubation at 50°C, then 95°C for 10 minutes, and followed with
128
95°C for15 seconds and 60°C for 60 seconds for 40 cycles. We used GAPDN as an
129
internal control to normalize for differences in the amount of total RNA in each
130
sample. The primers used in this study were purchased from Invitrogen.
131
Western blotting
SC
RI PT
125
Cells were collected and lysed with a lysis buffer containing 1% NP-40. After
133
brief vortexing and rotation, the cell lysates were separated with SDS-PAGE and
134
transferred to nitrocellulose membranes (Amersham Biosciences, Piscataway,
135
NJ).After 30 min blocking with blocking solution (50 mM Tri-HCl, 150 Mm NaCl,
136
5% (w/v) non-fat dry milk and 0.1% Tween-20), the membrane was incubated with
137
primary antibody, and then with HRP-conjugated secondary antibody(Cell Signal
138
Technology, USA). After washes, the protein bands were visualized with ECL plus
139
immune-blotting detection reagents (Pierce Biotechnology, Rockford, IL).
140
Immunofluorescent staining
TE D
EP
AC C
141
M AN U
132
Cells grown on cover slips were fixed in 4% freshly made paraformaldehyde for
142
15 minutes, and were incubated with FITC-TLR4 antibody (Cell Signal Technology,
143
USA) for 1 hour at room temperature. Images were taken with Nikon C-HGFI
144
fluorescent microscope (Nikon, Japan).
145
Statistics
8
ACCEPTED MANUSCRIPT 146
The 2-tailed Student’st-test or one-way ANOVA was used for statistical analysis in this research. When P<0.05, the difference was defined as statistically significant.
148
Results
149
Expression of TLRs on SPC
RI PT
147
As reported previously, we used Sca-1+ bone marrow cells as SPCs, which are
151
shown to have characteristics of progenitor cells [9]. In order to study the expression
152
of TLRs on SPC, we extracted total RNA from SPCs, and used quantitative RT-PCR
153
to detect the expression level of TLR1 to 9, which are well-studied and have
154
well-defined ligands. Our results showed that TLR2-4 and 7-9 were expressed on
155
SPCs, with prominence of TLR4 (Figure 1A). This suggests that TLR4 is potentially
156
more important than the other TLRs. Accordingly we mainly focused to study TLR4
157
influence on SPCs.
TE D
M AN U
SC
150
Next we explored the respective protein levels using western blotting, and our
159
data showed that TLR4 expressed in CD34+-BM, Sca-1+-BM, SMC, and macrophages
160
was roughly similar, while substantially lower on endothelial cells (Figure 1B). These
161
results unequivocally demonstrated that TLR4 is expressed on both terminally
162
differentiated cells and progenitor cells such as SPCs.
163
Activation of TLR4 enhances SPCs migration
AC C
EP
158
164
Since TLR4 was highly expressed on SPCs, so we assumed TLR4 signaling is
165
involved in SPCs migration, which is essential for SPC recruitment to atherosclerotic
166
lesion. Wound healing and transwell assays are two most common methods to assess
9
ACCEPTED MANUSCRIPT 167
cellular migration, so we used both to evaluate the SPCs migration upon TLR4
168
activation. Lipopolysaccharide (LPS) is a well-defined and specific TLR4 ligand, so we
170
stimulated SPCs with different concentrations of LPS. We found 200ng/ml to be the
171
optimal concentration for SPCs migration (Figure 2A) in our assay. As shown in
172
Figures 2B and C, LPS stimulation dramatically increases the migration of SPCs. The
173
effect was proven specific, as abrogation of LPS binding to TLR4 with polymyxin B
174
(PMB), the migration of SPCs was reversed.
175
Deficiency of TLR4 reduces migration of SPCs
M AN U
SC
RI PT
169
Although we observed the effects of TLR4 on migration of SPCs, it could be
177
other unknown mechanism of LPS on SPCs. So next we used specific shRNA to
178
knockdown the TLR4 expression and to verify the role of TLR4.
TE D
176
As the efficacy of transfection of SPCs with common reagents such as
180
lipofectamine is very low, we produced recombinant lentivirus containing specific
181
shRNAor TLR4 sequence for knockdown and over-expression, respectively. The
182
efficacy of transfection was confirmed by ZsGreen expressed by the lentivirus in
183
fluorescence microscopy (data not shown). The knockdown and over-expression of
184
TLR4 was confirmed using immune-blotting. The results showed that knockdown of
185
TLR4 expression was successful (Figure 3A and B).
AC C
EP
179
186
Next we used SPCs, TLR4 knockdown SPCs and TLR4 rescued SPCs to perform
187
migration experiment. The results showed that the migration of SPCs with TLR4
188
knockdown was greatly decreased (<50%, Figure 3C and D). When the knockdown 10
ACCEPTED MANUSCRIPT of TLR4 was rescued by TLR4-lentivirus, the migration of SPCs was restored (Figure
190
3C and D), confirming the specific effects of TLR4 on SPCs migration. Taken
191
together, our data revealed that the TLR4 are required for SPCs migration and thus
192
most likely are important in atherosclerosis.
193
Discussion
RI PT
189
Increasingly amount of studies have demonstrated that the immunological
195
system plays a paramount role in the pathogenesis of atherosclerosis [1, 3, 5, 25].
196
Both innate and adaptive immunity effects are represented on atherosclerotic lesions.
197
This includes involvement of cells such as macrophages, dendritic cells, B and T
198
cells.
M AN U
SC
194
SMCs are another major cell type in atherosclerotic plaques and appear to play
200
key role in plaque formation and progression. Mounting evidence indicates that
201
hematopoietic SPCs are an alternative source of both macrophages and SMCs, which
202
are present in atherosclerotic lesions. For both cell types, the migration of SPCs is
203
central for their recruitment to the atherosclerotic plaque. Earlier studies have
204
suggested that some molecules are of pivotal importance in the process, such as
205
Matrix metalloproteinases 8 (MMP8) and NEDD-9. As more attention focused on the
206
mechanism of SPCs in atherogenesis, more regulators of SPC migration are most
207
certainly going to be discovered.
AC C
EP
TE D
199
208
TLR is an evolutionarily conserved family of receptors, while PRR being the
209
most studied and has been shown to have diverse functions beyond those in innate
11
ACCEPTED MANUSCRIPT immunity accordingly [26]. Besides immunocytes, such as macrophages and dendritic
211
cells, TLRs are expressed on multiple cell types, such as endothelial and epithelial
212
cells [3, 13]. Recently, it has been reported that TLRs are expressed on stem cells,
213
including on embryonic and mesenchymal stem cells [27, 28].
RI PT
210
Here our data demonstrated that different TLRs are expressed in SPCs, with
215
TLR4 as the most prominent one. TLR4 is known to use both MyD88-dependent and
216
–independent cascades leading to NF- B, IRF5, and mitogen-associated protein
217
kinase (MAPK) signaling. This may lead to multiple biological events, such as
218
migration, proliferation and differentiation[29]. During atherosclerosis, SPCs have to
219
migrate from bone marrow or vascular wall to the atherosclerotic lesion. TLR4
220
activation leads to production of a variety of chemokines, including monocyte
221
chemotactic protein-1 (MCP-1), and Macrophage Inflammatory Protein-1, which are
222
essential for diverse cell migration events. It has been reported that MMP8 was
223
important during SPCs migrating into atheromas [10], and TLR4 signaling did result
224
in production of MMP8, suggesting the possible pathway.
EP
TE D
M AN U
SC
214
In summary, our study shows that TLR4is expressed on SPCs, and the essential
226
function of its signaling is for SPC migration, thus contributing to atherosclerotic
227
lesion formation. Our data provide new insight into the biological regulators and
228
mechanisms involved in the regulation of SPC migration, and may potentially assist
229
in finding new therapeutics for atherosclerosis.
AC C
225
12
ACCEPTED MANUSCRIPT 230
References
231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272
1.
Glass CK, Witztum JL: Atherosclerosis. the road ahead. Cell 2001, 104(4):503-516.
2.
Keaney JF, Jr.: Immune modulation of atherosclerosis. Circulation 2011, 124(22):e559-560.
3.
Hansson GK, Hermansson A: The immune system in atherosclerosis. Nat Immunol 2011, 12(3):204-212. Weber C, Noels H: Atherosclerosis: current pathogenesis and therapeutic options. Nat Med
5.
Lusis AJ: Atherosclerosis. Nature 2000, 407(6801):233-241.
6.
Hansson GK: Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005,
2011, 17(11):1410-1422.
352:1685-1695.
RI PT
4.
Hu Y: Smooth Muscle Cells in Transplant Atherosclerotic Lesions Are Originated From
7.
Recipients, but Not Bone Marrow Progenitor Cells. Circulation 2002, 106(14):1834-1839.
SC
Febbraio M, Guy E, Silverstein RL: Stem cell transplantation reveals that absence of
8.
macrophage CD36 is protective against atherosclerosis. Arterioscler Thromb Vasc Biol 2004, 24(12):2333-2338.
M AN U
Hu Y, Zhang Z, Torsney E, Afzal AR, Davison F, Metzler B, Xu Q: Abundant progenitor cells in
9.
the adventitia contribute to atherosclerosis of vein grafts in ApoE-deficient mice. The Journal of Clinical Investigation 2004, 113(9):1258-1265. 10.
Xiao Q, Zhang F, Lin L, Fang C, Wen G, Tsai T-N, Pu X, Sims D, Zhang Z, Yin X et al: Functional Role of Matrix Metalloproteinase-8 in Stem/Progenitor Cell Migration and Their Recruitment Into Atherosclerotic Lesions. Circ Res 2013, 112(1):35-47.
11.
Blander JM, Sander LE: Beyond pattern recognition: five immune checkpoints for scaling the
12.
TE D
microbial threat. Nat Rev Immunol 2012, 12(3):215-225.
Mogensen TH: Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses. Clin Microbiol Rev 2009, 22(2):240-273.
13.
Takeuchi O, Akira S: Pattern Recognition Receptors and Inflammation. Cell 2010,
14.
Xiang M, Fan J: Pattern recognition receptor-dependent mechanisms of acute lung injury.
EP
140(6):805-820.
Mol Med 2010, 16(1-2):69-82. 15.
Chen K, Huang J, Gong W, Iribarren P, Dunlop NM, Wang JM: Toll-like receptors in infection
and
cancer.
International
Immunopharmacology
2007,
AC C
inflammation,
7(10):1271-1285.
16.
Creagh EM, O Neill LAJ: TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol 2006, 27(8):352-357.
17.
Kawai T, Akira S: TLR signaling. Seminars in Immunology 2007, 19(1):24-32.
18.
Lee CC, Avalos AM, Ploegh HL: Accessory molecules for Toll-like receptors and their function.
19.
Moynagh PN: TLR signalling and activation of IRFs: revisiting old friends from the
Nat Rev Immunol 2012, 12(3):168-179. NF-[kappa]B pathway. Trends in Immunology 2005, 26(9):469-476. 20.
Takeda K, Kaisho T, Akira S: TOLL-LIKE RECEPTORS. Annual Review of Immunology 2003, 21(1):335-376.
21.
Tobias PS, Curtiss LK: Toll-like receptors in atherosclerosis. Biochem Soc Trans 2007, 35(Pt 6):1453-1455. 13
ACCEPTED MANUSCRIPT 22.
Tobias PS, Curtiss LK: TLR2 in murine atherosclerosis. Semin Immunopathol 2008, 30(1):23-27.
23.
Zheng Y, Gardner SE, Clarke MC: Cell death, damage-associated molecular patterns, and sterile inflammation in cardiovascular disease. Arterioscler Thromb Vasc Biol 2011, 31(12):2781-2786.
24.
Xu C, Zhang YH, Thangavel M, Richardson MM, Liu L, Zhou B, Zheng Y, Ostrom RS, Zhang XA: CD82 endocytosis and cholesterol-dependent reorganization of tetraspanin webs and lipid
RI PT
rafts. FASEB J 2009, 23(10):3273-3288.
Hansson GK: Atherosclerosis--an immune disease: The Anitschkov Lecture 2007. Atherosclerosis 2009, 202(1):2-10.
26.
Theofilopoulos AN: TLRs and IFNs: critical pieces of the autoimmunity puzzle. J Clin Invest 2012, 122(10):3464-3466.
27.
Delarosa O, Dalemans W, Lombardo E: Toll-like receptors as modulators of mesenchymal stem cells. Frontiers in immunology 2012, 3:182.
28.
SC
25.
Megias J, Yanez A, Moriano S, O'Connor JE, Gozalbo D, Gil ML: Direct Toll-like receptor-mediated stimulation of hematopoietic stem and progenitor cells occurs in vivo
29.
M AN U
and promotes differentiation toward macrophages. Stem Cells 2012, 30(7):1486-1495. Zanoni I, Ostuni R, Marek Lorri R, Barresi S, Barbalat R, Barton Gregory M, Granucci F, Kagan Jonathan C: CD14 Controls the LPS-Induced Endocytosis of Toll-like Receptor 4. Cell 2011,
EP
TE D
147(4):868-880.
AC C
273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293
14
ACCEPTED MANUSCRIPT
Figure Legends
295
Fig. 1 TLRs expression in SPCs. (A) Total RNA was prepared from SPCs and was
296
reverse-transcribed using qRT-PCR kit. Different types of TLR were detected by real
297
time PCR and * stands for p<0.05. The relative expression of TLR4 was significantly
298
higher than others as the next target protein. (B) CD34+-BM, Sca-1+-BM, SMC,
299
macrophages and endothelial cells were collected to determine the expression of
300
TLR4 using western blot analysis. (C) Sca-1+-BM, macrophages and HEK293 cells
301
were grown on cover slips, stained with FITC-TLR4 and subjected to fluorescent
302
microscopy.
303
Fig. 2 Activation of TLR4 enhances SPCs migration. (A) SPCs treated with LPS at
304
different concentrations for 4 hours were used for the transwell assay and then
305
transmigrated cells were counted. The SPCs treated with LPS at 200ug/ml migrated
306
significantly more suitable for inducing TLR4. (B) Three SPCs were assayed using
307
transwell, including SPC control, SPC treated with LPS (200ug/ml) and SPC treated
308
both LPS (200ug/ml) and PMB. The cells in the transwell plates were incubated at
309
37°C for 12h. Transmigrated cells were counted. (C) Wound-healing assay. Treated
310
cells as indicated scraped, and after overnight culture at 37°C, the cells were fixed and
311
images were taken.
312
Fig. 3 Deficiency of TLR4 reduces migration of SPCs. SPCs were inoculated with
313
lentivirus containing specific shRNA sequence against TLR4 (for knockdown,
314
MOI=20) (A) or lentivirus containing TLR4 sequence (for over-expression, MOI=20)
AC C
EP
TE D
M AN U
SC
RI PT
294
15
ACCEPTED MANUSCRIPT (B), and after 48 hours, cell lysates were separated with SDS-PAGE and
316
immunoblotted using TLR4 antibody. A representative image of immune-blotting
317
from 3 independent experiments. (C) SPCs with TLR4 knockdown and re-expression
318
subjected to transwell assay to assess the migration abilities. Transmigrated cells were
319
quantified. The data is shown as Mean ±SD (n=3). (D) SPCs with TLR4 knockdown
320
and re-expression subjected to wound-healing assay. Treated cells as indicated
321
scraped, and after overnight culture at 37°C, the cells were fixed and images were
322
taken.
AC C
EP
TE D
M AN U
SC
RI PT
315
16
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