Accepted Manuscript Synthetic Niche to Modulate Regenerative Potential of MSCs and Enhance Skeletal Muscle Regeneration Matthias Pumberger, Taimoor H. Qazi, M. Christine Ehrentraut, Martin Textor, Janina Kueper, Gisela Stoltenburg-Didinger, Tobias Winkler, Philipp v. Roth, Simon Reinke, Cristina Borselli, Carsten Perka, David J. Mooney, Georg N. Duda, Sven Geißler PII:
S0142-9612(16)30176-4
DOI:
10.1016/j.biomaterials.2016.05.009
Reference:
JBMT 17491
To appear in:
Biomaterials
Received Date: 8 March 2016 Revised Date:
2 May 2016
Accepted Date: 4 May 2016
Please cite this article as: Pumberger M, Qazi TH, Ehrentraut MC, Textor M, Kueper J, StoltenburgDidinger G, Winkler T, Roth Pv, Reinke S, Borselli C, Perka C, Mooney DJ, Duda GN, Geißler S, Synthetic Niche to Modulate Regenerative Potential of MSCs and Enhance Skeletal Muscle Regeneration, Biomaterials (2016), doi: 10.1016/j.biomaterials.2016.05.009. 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
Title: Synthetic Niche to Modulate Regenerative Potential of MSCs and Enhance Skeletal Muscle Regeneration
† Authors contributed equally as first authors ‡ Authors contributed equally as senior authors * Corresponding author
SC
Affiliations:
RI PT
Matthias Pumberger1,2,3 †, Taimoor H. Qazi1,2 †, M. Christine Ehrentraut1, Martin Textor1,2,3, Janina Kueper1, Gisela Stoltenburg-Didinger4, Tobias Winkler1,2,3, Philipp v. Roth1,2,3, Simon Reinke1,2,3, Cristina Borselli2,5, Carsten Perka1,2,3, David J. Mooney2,5, Georg N. Duda 1,2,3,5‡, Sven Geißler1,2,3‡ *
1
M AN U
Julius Wolff Institute, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
2
Berlin-Brandenburg Center for Regenerative Therapies & Berlin-Brandenburg School for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
3
Center for Musculoskeletal Surgery, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany 4
5
TE D
Institute of Cell Biology and Neurobiology, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
EP
Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, MA 02115 (USA); John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, MA 02138 (USA).
AC C
*Correspondence: E-mail:
[email protected], Fax: +49-30-450559969, Telephone: +49-30-450659539
Key words: Engineered cell microenvironments, Biomaterial based stem cell niche, Trophic factor secretion, Muscle regeneration
ACCEPTED MANUSCRIPT
Abstract: Severe injury to the skeletal muscle often results in the formation of scar tissue, leading to a decline in functional performance. Traditionally, tissue engineering strategies for muscle
RI PT
repair have focused on substrates that promote myogenic differentiation of transplanted cells. In the current study, the reported data indicates that mesenchymal stromal cells (MSCs) transplanted via porous alginate cryogels promote muscle regeneration by secreting bioactive
SC
factors that profoundly influence the function of muscle progenitor cells. These cellular functions, which include heightened resistance of muscle progenitor cells to apoptosis,
M AN U
migration to site of injury, and prevention of premature differentiation are highly desirable in the healing cascade after acute muscle trauma. Furthermore, stimulation of MSCs with recombinant growth factors IGF-1 and VEGF165 was found to significantly enhance their paracrine effects on muscle progenitor cells. Multifunctional alginate cryogels were then utilized as synthetic niches that facilitate local stimulation of seeded MSCs by providing a
TE D
sustained release of growth factors. In a clinically relevant injury model, the modulation of MSC paracrine signaling via engineered niches significantly improved muscle function by remodeling scar tissue and promoting the formation of new myofibers, outperforming
AC C
EP
standalone cell or growth factor delivery.
ACCEPTED MANUSCRIPT
1. Introduction
2
Muscle regeneration after minor trauma is a natural process of adaptation to excessive
3
stressing and is accomplished by multiple cell types that arrive at the site of injury to begin
4
repair [1]. Beyond a certain injury severity threshold this inherent regeneration potential
5
proves insufficient [2], leading to fatty degeneration and fibrotic scar tissue formation, which
6
compromise muscle function and structural integrity [3]. Such severe muscle damages
7
resulting from direct trauma, iatrogenic injuries during surgical procedures, muscle tears,
8
ischemia, or myopathies are common [4]. Current treatment options are limited and plagued
9
with lack of allogeneic donor tissue or donor site morbidity associated with autograft
10
transplantations [5]. Moreover, they often result in unsatisfying functional outcome,
11
substantiating the clear medical demand for novel therapeutic approaches [6, 7].
12
Strategies aiming to improve muscle regeneration via the local delivery of cells, growth
13
factors, or a combination thereof are currently under investigation [8, 9]. Growth factor
14
delivery has been shown to be a useful approach to treat ischemic muscle tissues in animal
15
models [10], but their effectiveness in the repair of severe injuries is yet to be demonstrated.
16
Moreover, the large number of complex biological processes necessary to induce repair of
17
injured muscle tissues may require the spatiotemporal delivery of several distinct growth
18
factors, which poses a significant challenge and renders this strategy suboptimal [11, 12].
19
Since the failure of direct myoblast delivery in earlier clinical trials [13, 14], adult progenitor
20
cells, e.g. muscle satellite cells or mesenchymal stromal cells (MSCs), have emerged as
21
promising cell types to enhance muscle regeneration [15]. MSCs are highly proliferative
22
multipotent cells that are known to modulate injury responses through paracrine signaling
23
[16-18]. For instance, MSCs serve as a source of cytokines and proteinases essential to
24
angiogenesis and tissue regeneration, such as VEGF, MMPs, IGF-1, HGF, TGF-β, and bFGF
25
[19]. The regenerative capability of MSCs has been validated in several animal models of
AC C
EP
TE D
M AN U
SC
RI PT
1
ACCEPTED MANUSCRIPT
muscular dystrophy, myocardial infarction as well as muscle trauma [20-24]. Collectively,
27
these studies provide evidence that MSC administration reduces the local inflammation and
28
enhances tissue function, not as a result of differentiation into muscle cells and fusion with
29
existing myofibers, but apparently due to paracrine effects via the secretion of cytokines and
30
other growth factors [25, 26]. Despite potential efficacy, bolus cell transplantation frequently
31
suffers from lack of tissue engraftment and very low cell survival at the site of injury,
32
compelling researchers to inject higher cell numbers to achieve significant improvements [27,
33
28]. Thus, it appears essential for an effective cell therapy to ensure localization of
34
transplanted cells at the site of injury, for example by using biomaterial scaffolds.
35
A biomaterial driven approach could overcome the problems associated with bolus cell
36
injection by ensuring a localized delivery of signaling factors while shielding cells from
37
harmful environmental effects of the injury site [29, 30]. Various biomaterials have been
38
employed to transplant cells that could undergo myogenic differentiation and fuse with
39
existing fibers of the injured tissue [31-34]. The success of such a strategy hinges on a number
40
of critical factors including migration of cells from the biomaterial substrate towards the
41
injured tissue, successful engraftment and differentiation, and requires a large number of cells
42
in case of volumetric muscle loss or severe trauma. However, novel approaches stipulate
43
biomaterials that present multiple cues for optimization of cell function [35]. We
44
hypothesized that a more effective approach would be to support the endogenous regenerative
45
mechanisms of the skeletal muscle by harnessing desirable cytokines and growth factors
46
secreted by autologous MSCs and ensuring their local delivery at the site of injury. A suitable
47
porous scaffold with drug release capability would provide an appropriate structural and
48
chemical environment, in essence mimicking a niche that is conducive to MSC paracrine
49
function. It was our aim to show that MSCs need not engraft onto the host tissue and undergo
50
differentiation, but could induce muscle regeneration by secreting factors from the niche. For
51
this reason, we chose not to use an injectable in-situ forming hydrogel to deliver the cells
AC C
EP
TE D
M AN U
SC
RI PT
26
ACCEPTED MANUSCRIPT
intramuscularly, instead choosing to place the MSC loaded niche adjacent to the injured
53
muscle. Moreover, in line with clinical observations, the injured muscle is normally exposed
54
and in this case an injectable hydrogel, deliverable in a minimally invasive manner, may not
55
offer an advantage over a macroporous scaffold.
56
We therefore engineered a porous (70-150 µm) multifunctional alginate cryogel that acts as a
57
synthetic niche for MSCs, supports their adhesion and spreading while simultaneously
58
boosting their secretory function via local stimulation with IGF-1 and VEGF165. The
59
transplantation of this synthetic niche in a clinically relevant crush muscle trauma model
60
significantly improved muscle strength, reduced fibrosis, and increased muscle fiber density
61
and the number of regenerated fibers. This work shows that without any genetic manipulation,
62
MSC’s paracrine factor secretion can be upregulated, the released factors being more potent at
63
influencing myoblast behavior and orchestrating muscle regeneration than standalone MSC or
64
growth factor delivery.
65
2. Materials and Methods
66
Animals: 4 month old female Sprague Dawley rats (Charles River laboratories), weighing
67
between 200-250 g were used for this study with each group being representative for at least 8
68
animals. All animal experiments were carried out in compliance with the policies and
69
principles established by the Animal Welfare Act, the NIH Guide for Care and Use of
70
Laboratory Animals, and the national animal welfare guidelines. The study was approved by
71
the local legal representative (Landesamt für Gesundheit und Soziales, Berlin: Registration
72
number: G0119/12)
73
MSC isolation and cell culture: Bone marrows were aspirated from rat tibiae, and cultured in
74
DMEM containing 10 % FBS and 1 % penicillin/streptomycin. MSC phenotype and
75
differentiation potential was confirmed as described earlier [36]. Briefly, cell surface marker
76
expression was validated using flow cytometry. MSC populations were positive (≥ 95% of
AC C
EP
TE D
M AN U
SC
RI PT
52
ACCEPTED MANUSCRIPT
total counts) for CD29, CD44, CD105, CD73, CD166, CD90, and RT1A and negative (≥
78
98%) for CD45, CD34 and RT1B. Differentiation potential upon stimulation with adipogenic
79
or osteogenic media was confirmed via Nile red and Alizarin Red staining, respectively.
80
Unless otherwise stated, MSCs from at least four different donors were used in the in vitro
81
assay described below.
82
The C2C12 cell line was purchased from ATCC (Catalog # ATCC® CRL-1772™).
83
Expansion media for C2C12 myoblasts contained 10 % FCS, which was reduced to 5 % to
84
induce differentiation.
85
MSC conditioned medium: MSCs between passage 3 and 5 were cultured in 6 well plates at a
86
density of 3 x 105 cells per well, and allowed to attach. After overnight incubation, cells were
87
washed twice with calcium free PBS and 1.5 mL/well of fresh serum free media was added.
88
For MSC+GF-CM, the serum free media was supplemented with 5 ng/mL of IGF-1 and
89
VEGF165. After a further 24 hour incubation period, the conditioned media was collected and
90
centrifuged at 700 rcf for 8 minutes to remove cell debris, and passed through a 0.2 µm sterile
91
filter. The conditioned media was then concentrated 20x using Amicon ultracentrifugal filter
92
units (10 kDa) and re-diluted in fresh serum free media to its original volume. For
93
differentiation studies involving C2C12 myoblasts, supplementary FCS (5% v/v) was added
94
to the conditioned media. For growth factor neutralization experiments, conditioned media
95
containing IGF-1 and VEGF165 were incubated with anti-human VEGF antibody (R&D
96
systems, Clone # 26503, 1:10000) and anti-human IGF antibody (R&D systems, catalog #
97
AF-291-NA, 1:500) for 4 hours before use.
98
Myoblast proliferation & survival: C2C12 myoblasts were seeded in 48 well plates at a
99
seeding density of 3 x 103 per well and allowed to attach. After overnight incubation, media
100
was removed and the cells were washed twice with calcium free PBS before adding either (1)
101
serum free cell culture media (blank), (2) serum free media containing growth factors at a
AC C
EP
TE D
M AN U
SC
RI PT
77
ACCEPTED MANUSCRIPT
concentration of 5 ng/mL each (+GF), (3) conditioned media from MSCs (MSC-CM), or (4)
103
conditioned media from MSCs stimulated with 5 ng/mL growth factors (MSC+GF-CM).
104
After 48 hours, media was aspirated and the well plates were frozen at -80 °C. CyQuant® cell
105
proliferation assay kit (Invitrogen) was used to determine cell proliferation. Myoblast survival
106
was quantified by first seeding 3 x 104 C2C12 cells/well of a 48 well plate. After overnight
107
attachment, a scratch was made to mimic injury, washed twice with PBS, and then the
108
appropriate media was added. After a further 24 hours, cells were trypsinized after washing
109
with PBS. The cell suspension was mixed with trypan blue and viability was determined by
110
counting cells in a hemocytometer.
111
Myoblast apoptosis rate: Apoptosis was evaluated with Caspase-Glo® 3/7 assay (Promega)
112
according to the manufacturer’s instructions. Briefly, myoblasts were seeded on white-walled
113
96 well plates with a cell density of 1 x 104/well. After overnight attachment a scratch was
114
made and appropriate media were added. After a further 24 hours, a 1:1 mixture of the
115
Caspase-Glo® buffer and substrate was added into the wells and luminescence was detected
116
using a plate reader. Measurements were normalized to cell number determined by
117
CyQuant®. Myoblast survival was quantified by a similar seeding method, but 24 hours after
118
application of respective media, cells were trypsinized after washing with PBS. The cell
119
suspension was mixed with trypan blue and viability was determined by counting cells in a
120
hemocytometer.
121
Migration assay: The migration behavior of C2C12 myoblasts in response to different media
122
was investigated using a scratch wound healing assay. 1.5 x 105 myoblasts were seeded in
123
each well of a 24 well plate and allowed to attach overnight to form a confluent monolayer. A
124
200 µL pipette tip was used to create a scratch; wells were washed twice with PBS before
125
adding indicated conditioned media. Images were taken every 30 minutes over a period of 24
126
hours using an inverted microscope (DMI6000B, Leica, Germany) with a live cell imaging
AC C
EP
TE D
M AN U
SC
RI PT
102
ACCEPTED MANUSCRIPT
system. The images were analyzed using T-scratch [37], and the kinetics of wound closure
128
were determined for the different treatment groups.
129
Myoblast differentiation (Western Blot): To monitor the influence of MSC’s conditioned
130
media on myoblast differentiation behavior, 4 x 105 C2C12 cells were seed in each well of a 6
131
well plate. After overnight attachment and the formation of a confluent layer, the appropriate
132
conditioned media were added. Four days after induction of differentiation, C2C12 cells were
133
washed with PBS, and lysates were generated using SDS lysis buffer. Protein content was
134
determined using DC protein assay (Bio-Rad). The Novex NuPAGE® system (Invitrogen)
135
was used according to manufacturer’s instructions followed by semi-dry transfer. Primary
136
antibodies for Myosin heavy chain (Clone # MF20, R&D Systems, Catalog # MAB4470)
137
(1:500), Myogenin (Clone # F5D, Abcam, Catalog # ab1835) (1:500), MyoD (Clone # 5.8A,
138
Thermo Scientific, Catalog # MA1-41017) (1:500), and the house keeping gene GAPDH
139
(Clone # 6C5, Abcam, Catalog # ab8245) (1:10000) were used with TBST+5% milk. Anti-
140
mouse horseradish peroxidase (GE Healthcare) (1:7000) was utilized as the secondary
141
antibody. Using ECL substrate (GE Healthcare) pictures were acquired using the G:BOX
142
imager (Syngene). Analysis was performed using band intensities normalized to GAPDH and
143
quantified by NIH ImageJ software package (http://rsb.info.nih.gov/ij/).
144
Myoblast differentiation (Fluorescence microscopy): Cells were seeded at a density of 1 x
145
105/well of a 24 well plate. Four days after inducing differentiation in C2C12 myoblast
146
cultures, cells were washed with PBS, fixed with 4 % paraformaldehyde, permeabilized with
147
0.1 % saponin in 3 % BSA solution, and stained with anti-Myosin heavy chain (1:200), and
148
DAPI (1:1000). Cells were imaged with an inverted fluorescence microscope (Leica).
149
Alginate processing: Ultrapure alginates were purchased from Novamatrix (Oslo, Norway).
150
MVG alginate was used as the high molecular weight component (250 kDa) and LVG
151
alginate (50 kDa) was used as the low molecular weight component to prepare gels. Both
AC C
EP
TE D
M AN U
SC
RI PT
127
ACCEPTED MANUSCRIPT
alginate polymers were diluted to 1 % w/v in double-distilled H2O, and 1 % of the sugar
153
residues in the polymer chains were oxidized with sodium periodate (Sigma-Aldrich, St
154
Louis, MO) by maintaining solutions in the dark for 17 hours at room temperature under
155
constant stirring, as previously described [38]. An equimolar amount of ethylene glycol
156
(Fischer, Pittsburgh, USA) was added to stop the reaction, and the solution was subsequently
157
dialyzed (MWCO 3500 Da, Spectra/Por®) over three days, frozen at -20° C, and lyophilized.
158
Both alginates were modified with G4RGDSP peptides (Commonwealth Biotechnology,
159
Richmond, USA) at a degree of substitution of 10 (10 peptide molecules per alginate chain)
160
using carbodiimide chemistry, as previously described [39]. The modified alginate solutions
161
were dialyzed again, mixed with activated charcoal, sterile filtered, lyophilized, and stored at -
162
20° C until further use.
163
Scaffold Fabrication: Equal amounts of lyophilized MVG and LVG alginates were mixed,
164
and reconstituted in high glucose containing DMEM (Sigma-Aldrich, Germany) to obtain a
165
4 % w/v polymer solution. Using two syringes coupled with a syringe connector, the alginate
166
mixture was ionically cross-linked using aqueous calcium sulfate slurry (40 µL of CaSO4 per
167
mL of alginate mixture) by vigorously mixing the contents of the two syringes, to produce
168
gels with a final concentration of 2 % w/v. Where appropriate, recombinant human IGF-1 and
169
recombinant human VEGF165 (R&D systems, USA) were mixed (at a concentration of 60 µg
170
per mL of alginate solution) with the alginate mixture prior to crosslinking. The mixture was
171
poured between sterile glass plates separated by 2 mm spacers, and allowed to gel for 30
172
minutes. Gels measuring 8 mm x 3 mm x 2 mm were cut out using a punch and frozen at -
173
80°C for 24 hours. Thereafter, the frozen gels were lyophilized to induce porosity. For in vivo
174
transplantation, 50 µL of a 20 x 106 MSCs/mL cell suspension was pipetted on top of the
175
freeze dried porous scaffold and incubated for 20 minutes, before adding excess DMEM.
AC C
EP
TE D
M AN U
SC
RI PT
152
ACCEPTED MANUSCRIPT
Viability of MSCs: The viability of MSCs inside the scaffolds was evaluated over a period of
177
14 days using a Live/Dead cell imaging kit (Invitrogen). Briefly, cell seeded scaffolds (1 x 105
178
MSCs/scaffold) were rinsed thrice in calcium free PBS before dissociation in a 50 mM EDTA
179
solution. The cell suspension was mixed with a mixture of Calcein AM and Ethidium
180
homodimer and imaged with a fluorescence microscope after a 15 minute incubation period.
181
The number of live (green) and dead (red) cells were counted using NIH ImageJ software
182
package.
183
MSCs outward migration: To determine outward migration, cell seeded scaffolds (1 x 105
184
MSCs/scaffold) were placed in separate wells of a 24 well plate. After every 24 hours, the
185
scaffolds were shifted to new wells, and the number of cells that had colonized the wells over
186
the previous 24 hours was quantified using the CyQuant® cell proliferation assay kit
187
(Invitrogen).
188
ELISA: For release kinetics, growth factor containing scaffolds (n=3) were immersed in 1 mL
189
of DMEM and incubated for 30 days. At different time points (days 1, 3, 7, 14, 21, and 28),
190
supernatants were collected and fresh media was added. The amount of IGF-1 and VEGF165 in
191
the supernatants was quantified using Quantikine™ human-specific ELISA kits (R&D
192
systems, Germany). Quantitative concentrations of MSC secreted Hgf, bFgf, Igf-1, and Vegf
193
in conditioned media were determined using commercially available rat-specific ELISA kits
194
(R&D systems) and used according to the manufacturer’s instructions. Cross reactivity with
195
human recombinant growth factors was determined (Supplementary information).
196
Animal experiments: Animals were randomly allocated into the following treatment groups
197
(n=8): (1) empty alginate scaffold, (2) alginate scaffold with incorporated growth factors (3
198
µg each of IGF-1 and VEGF165), (3) alginate scaffold seeded with autologous MSCs (50 µL
199
of a 20 x 106 MSCs/mL cell suspension), and (4) alginate scaffold with incorporated growth
200
factors and seeded with autologous MSCs. Independent from their final group allocation, all
AC C
EP
TE D
M AN U
SC
RI PT
176
ACCEPTED MANUSCRIPT
animals underwent a bone marrow biopsy to harvest autologous MSCs. Bone marrow
202
aspirates were taken from both tibiae as described in [40]. Three weeks later, the left soleus
203
muscles of the animals were bluntly crushed (monolateral surgery) and the appropriate
204
scaffold was immediately transplanted during the same surgery. At day 7, 28, and 56 after
205
transplantation, contraction forces of the injured and the contralateral (uninjured) soleus
206
muscle were measured. Subsequently, the animals were then sacrificed and the muscles were
207
harvested for histological analysis. For the 56 day time point, only groups 1 (Alg.), 3
208
(Alg.+MSC), and 4 (Alg.+GF+MSC) were transplanted based on the legal specification of the
209
responsible authorities, who only permitted us to monitor the long term effects of treatments
210
showing significant difference to the untreated controls at day 28.
211
Muscle trauma model and scaffold transplantation: The animals were anesthetized by
212
inhalation of isoflurane, administered Rimadyl® subcutaneously, and the lower left limbs
213
were shaved. An injury model consisting of standardized blunt crush traumas to the left soleus
214
muscle was employed using procedures described earlier [40]. On the day of surgery,
215
autologous MSCs were trypsinized and 50 µL of a cell suspension containing 20 x 106
216
MSCs/mL was seeded onto each scaffold (total: 1 x 106 MSCs/scaffold). After an initial 20
217
minute incubation period, 500 µL of full culture media was added to the well containing the
218
cell seeded scaffold. The construct was incubated for a further 4 hours to allow for complete
219
infiltration of the cell suspension and to facilitate cell attachment. The scaffold was then
220
immediately transplanted into the recipient animal by placing it along the length of the injured
221
soleus muscle in a fascia pocket lying in between the soleus and the gastrocnemius muscles.
222
No sutures were required to immobilize the scaffold. Thereafter, the superficial muscle and
223
skin were closed by suturing, and the wound was cleaned with saline solution.
224
Muscle force measurement: After an initial isoflurane narcosis, rats were anesthetized by
225
intraperitoneal injection of a mixture of xylazine and ketamine in saline before proceeding
AC C
EP
TE D
M AN U
SC
RI PT
201
ACCEPTED MANUSCRIPT
with the muscle force measurement and muscle harvesting. Fast twitch and slow twitch
227
(tetanic) forces of the treated and contralateral uninjured muscles were measured as described
228
earlier [28]. Briefly, after anesthetizing the animals, the sciatic nerve and the soleus muscle
229
were exposed whilst care was taken to protect the neuromuscular junctions and the
230
neurovascular bundle. The Achilles tendon was severed and the lower extremity was
231
connected to the muscle force measuring device using a silk suture. Subsequently, the sciatic
232
nerve was electrically stimulated and the contraction forces were recorded using a force
233
transducer. After the muscle force measurements, the animals were sacrificed.
234
Histology: Soleus muscles embedded in Tissue-Tek® O.C.T.™ compound (Sakura), were
235
divided into 9 segments along the length of the muscle, and 10 consecutive slices (7 µm
236
sections) from each segment were sectioned and fixed onto glass slides (Marienfeld). Sections
237
undergoing H&E and Picrosirius red staining were post-fixed with 4 % formaldehyde. To
238
identify collagenous scar tissue (Sirius red), the sections were stained with Direct Red 80
239
(Fluka, Germany) by incubation in Sirius red solution (1% w/v in saturated picric acid) for 60
240
minutes. Differentiation was reached with two washes in diluted acetic acid. Dehydration was
241
carried out by immersion in a graded series of ethanol before being incubated in xylol (T. J.
242
Baker) twice for 5 minutes. The amount of fibrosis in muscle sections was quantified using a
243
custom built macro in NIH ImageJ software package. The fibrotic area in the treated muscle
244
was expressed as a percentage of the total section area, and normalized to the percentage of
245
connective tissue in the uninjured control. Muscle sections were stained with Hematoxylin
246
and Eosin (Merck, Germany) for visualization of myofiber nuclei and cytoplasm. For nuclei
247
staining, sections were incubated in Hematoxylin for 7 minutes then washed twice in distilled
248
water. Differentiation was achieved by shortly placing the sections in 0.25 % HCl-ethanol
249
solution (Merck, Germany) followed by washing under tap water for 10 minutes. For
250
cytoplasm staining, sections were placed in 2 % Eosin (CHROMA) for up to 3 seconds and
251
dehydrated in graded series of ethanol and finally incubated in xylol twice, each time for a
AC C
EP
TE D
M AN U
SC
RI PT
226
ACCEPTED MANUSCRIPT
duration of 5 minutes. The number of fibers with centrally located nuclei was counted
253
manually and expressed as a percentage of the total section area. Muscle fibers expressing
254
slow myosin were detected with monoclonal anti-myosin (skeletal, slow) antibody (1:10000)
255
(Sigma-Aldrich, clone # NOQ7.5.4D, catalog # M8421). Blood vessels were detected with
256
CD31 monoclonal antibody (1:50) (abcam, clone # TLD-3A12, catalog # ab64543). For both
257
stainings, air dried sections were fixed in cold acetone for 20 minutes and washed in 10 %
258
PBS twice for 5 minutes each. Sections were blocked in 2 % horse serum (Biozol) diluted in
259
2 % BSA-PBS for 30 minutes at room temperature. The primary antibodies were then applied
260
with overnight incubation at 4 °C. After washing twice with PBS, the secondary antibody
261
(Biotinylated Horse Anti-Mouse IgG Antibody, rat adsorbed, Biozol) was applied (1:50) for
262
30 minutes. The sections were washed twice with PBS and the avidin-biotin complex was
263
applied for 50 minutes at room temperature (AP-Standard kit AK 5000; Vector). After
264
washing, sections were then incubated with Alkaline Phosphatase substrate (Red Alkaline
265
Substrate Kit I, SK-5100, Vector) for up to 8 minutes. Finally, sections were counter-stained
266
following the Mayers Haemalum method. Slow fibers were quantified using a custom built
267
macro in NIH ImageJ software package. For quantification, positively stained blood vessels
268
were manually counted from regions of interest generated randomly from the entire muscle
269
section. For all stained sections, images were captured using a light microscope (Leica
270
Microsystems, Germany) equipped with a digital camera (AxioCam MRc, Carl Zeiss
271
MicroImaging, Germany).
272
Statistical analysis: The IBM® SPSS® 22.0 software package (IBM Corp., Chicago, IL,
273
USA) was used for statistical evaluation. Results are presented as mean ± standard error of the
274
mean (s.e.m). Unless otherwise stated, all in vivo samples are representative of at least n = 8
275
animals (per group & time-point, range: 8-10) and all in vitro results are representative of at
276
least four biological replicates analyzed in two independent experiments. Sample size was
277
selected based on previous experience [28, 40] to minimize the number of animals needed
AC C
EP
TE D
M AN U
SC
RI PT
252
ACCEPTED MANUSCRIPT
while obtaining a valid statistically significant result (in accordance with the 3R principles).
279
The Shapiro-Wilk test was used to test normality and the Levene test was used to assess the
280
homogeneity of variances of the data for the indicated groups. Independent and non-normal
281
distributed data were analyzed using the Mann-Whitney U test (two groups) or the Kruskal-
282
Wallis one-way analysis of variance (>two groups). If normality could be assumed, unpaired
283
Student’s t-test (two groups) or the corresponding one-way analysis of variance (>two groups)
284
was chosen to determine statistical significance. P-values were adjusted using Bonferroni’s
285
(equal variances) or Dunnett T3 (unequal variances) p-value adjustment multiple comparison
286
procedure. Dependent sample data were analyzed using Wilcoxon signed rank and paired
287
Student’s t-test (two samples), respectively, or Friedman’s two-way ANOVA by ranks (> two
288
groups). All tests were analyzed two-sided and P<0.05 was regarded as significant. Detailed
289
information about error bars, sample sizes per group, and statistical analysis are included in
290
all figure legends. The investigators were not blinded to group allocation during the
291
experiments because the described treatments displayed obvious differences.
292
3. Results
293
3.1. Effect of MSC derived paracrine factors on myoblast function
294
The potential effects of bioactive factors derived from rat MSCs on muscle progenitor cells
295
was examined by exposing skeletal myoblasts (C2C12) to conditioned media generated from
296
unstimulated (MSC-CM), and recombinant growth factor (IGF-1 & VEGF165) stimulated
297
MSCs (MSC+GF-CM). Assessment of the growth dynamics of myoblasts via the number of
298
population doublings (PD) showed higher proliferation rates for C2C12 cells grown in MSC-
299
CM or MSC+GF-CM compared to corresponding cultures in serum free media without
300
(blank) or with recombinant growth factors (+GF) (Figure 1A). (PD relative to blank:
301
PD+GF=0.97±0.03,
302
PDMSC+GF-CM=1.49±0.05, Pvs.blank<0.001, Pvs.+GF<0.001). The anti-apoptotic benefits of MSC
AC C
EP
TE D
M AN U
SC
RI PT
278
Pvs.blank=1.000;
PDMSC-CM=1.43±0.03,
Pvs.blank<0.001,
Pvs.+GF<0.001;
ACCEPTED MANUSCRIPT
derived paracrine factors were validated under serum starvation. This analysis revealed
304
significantly improved survival of cells cultured in media +GF (1.7-fold, P<0.001), MSC-CM
305
(2.2-fold, P<0.001), and MSC+GF-CM (2.3-fold, P<0.001) compared to their counterparts
306
grown in blank media. We tested whether improved survival correlated with lower apoptosis
307
rates by determining caspase-3/7 activity as a surrogate marker for apoptotic cell death.[30]
308
Caspase-3/7 activity of C2C12 cells cultured in MSC+GF-CM was 39% (P<0.001) lower than
309
in the parallel cultures with blank media with or w/o GF. Notably, the anti-apoptotic effect of
310
MSC+GF-CM was also significantly higher compared to CM of untreated MSCs (P=0.005),
311
whereas no significant differences in apoptosis rates were observed between MSC-CM and
312
both control groups.
313
For further investigation of the paracrine effects of MSCs on myoblast function, we compared
314
the migration and differentiation potential of C2C12 cells depending on the culture
315
conditions. Employing a scratch wound healing assay to mimic injury, it was found that the
316
supplementation of human GF as well as MSC-CM significantly improved myoblast cell
317
migration (Pvs.blank<0.001; Pvs.+GF<0.001), and that this effect was further enhanced when
318
MSC+GF-CM was applied (Pvs.blank<0.001; Pvs.+GF<0.001; Pvs.CM<0.034) (Figure 1B and C).
319
In contrast, MSC-CM and MSC+GF-CM significantly inhibited myoblast differentiation
320
compared to control media with and without human GF. Cells cultivated in MSC-CM or
321
MSC+GF-CM showed a significantly lower expression of myosin heavy chain (Myh2) and
322
myogenin compared to corresponding control cultures (Figure 1D and E). Accordingly,
323
qualitative analysis by indirect immunofluorescence showed that C2C12 cells underwent
324
terminal differentiation (fusion into multinucleated myotubes and accumulation of Myh2) in
325
the presence of differentiation media independent from GF supplementation, while
326
differentiation was significantly reduced in the presence of MSC-CM or MSC+GF-CM,
327
respectively (Figure 1G).
AC C
EP
TE D
M AN U
SC
RI PT
303
ACCEPTED MANUSCRIPT
These results indicate that paracrine factors secreted from unstimulated and stimulated MSCs
329
promote the proliferation, survival, and migration of C2C12 myoblasts, while preventing their
330
cell cycle withdrawal and subsequent differentiation into myotubes. Previous studies have
331
suggested that promotion of myoblast proliferation and migration, and delaying their
332
differentiation represent favorable conditions for in vivo muscle repair [41, 42].
333
3.2. Modulation of MSC secretion pattern and therapeutic effects by stimulation with
334
recombinant IGF-1 and VEGF165
335
Since GF-stimulation of MSCs enhanced their paracrine actions on myoblasts, we wondered
336
whether this reflected an additive or synergistic effect. Hence, we examined whether
337
depletion of the human GFs from the MSC+GF-CM alters its effects on myoblast
338
proliferation, survival, and migration (Figure 2A, B and C). IGF-1 and VEGF165 were
339
depleted by using human-specific neutralization antibodies and this procedure efficiently
340
reduced levels of the human cytokines in the conditioned media (Figure S1). In the presence
341
of the neutralization antibodies, the positive effects of blank media supplemented with human
342
GF on C2C12 viability and migration were completely abolished. In contrast, the enhanced
343
proliferation, survival, and migration in response to MSC+GF-CM remained significant,
344
confirming that recombinant growth factors do not have an additive effect, but in fact
345
contribute primarily by stimulating MSCs potentially by modulating their paracrine secretion
346
pattern. This was verified by using rat-specific immunosorbent assays (ELISA) to detect
347
cytokines known to play a vital role in muscle regeneration. Conditioned media from GF-
348
stimulated MSCs was found to contain increased levels of basic fibroblast growth factor
349
[bFgf] (1.4-fold, P=0.045), Vegf (1.5-fold, P=0.001), and hepatocyte growth factor [Hgf]
350
(2.4-fold, P=0.026) compared to MSC-CM, while Igf-1 levels were not altered (P=0.277)
351
(Figure 2D). Vegf, bFgf, Hgf, and Igf represent factors originating from rat MSCs, as opposed
352
to recombinant human IGF-1 and VEGF165. Apart from these specific cytokines, the total
AC C
EP
TE D
M AN U
SC
RI PT
328
ACCEPTED MANUSCRIPT
amount of secreted protein was also found to be clearly increased in CM of GF-stimulated
354
MSCs (Pvs.CM=0.006). This might indicate the amplified secretion of other cytokines and
355
growth factors in the conditioned media of stimulated MSCs. Taken together, growth factor
356
stimulation enhances MSC’s paracrine activities and the resulting release of bioactive factors
357
may exert a strong effect on the cells of the injured muscle tissue.
358
RI PT
353
3.3. Porous alginate cryogel supports MSC attachment and spreading
360
Subsequently, we engineered a multifunctional alginate scaffold that could be utilized as an
361
artificial niche for MSCs at the site of injury (Figure 3A). The niche should allow adhesion
362
and spreading of transplanted MSCs (Figure 3C), protect them during transplantation in an
363
initially inflammatory environment, and allow to boost their paracrine signaling by local
364
release of incorporated human growth factors (IGF-1 & VEGF165). The fabrication of alginate
365
scaffolds consisted of several steps including oxidation of alginate and RGD peptide
366
conjugation. The resulting biomaterial was combined with IGF-1 and VEGF165 before being
367
ionically crosslinked using a calcium source, and lyophilized to produce macroporous
368
scaffolds (Figure 3B). Scaffolds provided a sustained release of incorporated growth factors
369
over a period of 30 days after an initial burst release (Figure 3D). Independent from GF
370
incorporation, MSCs were found to adhere to the walls of the RGD-peptide conjugated
371
alginate scaffold and maintain high viability for at least 7 days (Figure 3E). Although the
372
biomaterial exhibited a porous microstructure (pore size 70-150 µm) only a small percentage
373
(3-6 %) of the seeded cells migrated out of the scaffold and GF incorporation even further
374
reduced this outward migration compared to the blank biomaterial (P<0.001 at all time-points
375
investigated; Figure 3F). Such a scaffold would allow us to test the hypothesis that
376
transplanted MSCs are not required to physically engraft onto the injured tissue to achieve
AC C
EP
TE D
M AN U
SC
359
ACCEPTED MANUSCRIPT
377
regeneration, but could orchestrate the regeneration process by secreting bioactive factors in
378
the injury environment.
379
3.4. Transplanted synthetic niche enhances contraction forces of injured muscles
381
For in vivo investigations, an established, clinically relevant rat model of severely crushed
382
soleus muscle tissue was used [24]. Animals were randomly assigned to different groups and
383
were treated with either (1) blank scaffold (Alg.), (2) scaffold containing growth factors
384
(Alg.+GF), (3) scaffold seeded with autologous MSCs (Alg.+MSC), or (4) scaffold
385
containing both MSCs and growth factors (Alg.+GF+MSC). Quantitative measurements of
386
fast twitch and tetanic muscle forces were performed after 7, 28, and 56 days post injury using
387
a custom made set up that stimulated the sciatic nerve and recorded muscle contractions via a
388
force transducer, as described earlier [28]. Two additional regimes were tested to determine
389
their functional benefit: (a) bolus intramuscular injection of growth factors (inj. GF) and (b)
390
bolus intramuscular injection of MSCs and growth factors (inj. MSCs+GF); data shown in
391
supplementary information (Figure S2). All muscle forces were normalized to the uninjured
392
contralateral measurements.
393
At day seven, we found a comparable reduction of the fast twitch (~57±2%) and tetanic
394
(~46±2%) muscle force between all injured muscles (Figure 4A). At day 28, the fast twitch
395
muscle force progressively increased from Alg. (58±4%), Alg.+GF (64±3%), Alg.+MSC
396
(74±4%, PAlg.=0.024), to the highest value detected in the Alg.+GF+MSC group (83±3%,
397
PAlg.<0.001, PAlg.+GF=0.003). We observed a similar trend toward increased tetanic muscle
398
force, however, this tendency only reached statistical significance between Alg.+GF+MSC
399
and the alginate control (P=0.018) (Figure 4B). In order to investigate the long lasting effects
400
of the most promising groups (Alg.+MSC & Alg.+GF+MSC) relative to control (Alg.), we
401
additionally determined their muscle forces after 56 days post injury. Fast twitch and tetanic
AC C
EP
TE D
M AN U
SC
RI PT
380
ACCEPTED MANUSCRIPT
402
forces in the Alg.+GF+MSC group were significantly higher compared to the Alg. control,
403
whereas the tetanic contraction were significantly elevated in both the Alg.+MSC and
404
Alg.+GF+MSC groups in relation to the Alg. control.
RI PT
405
3.5. Effect of transplanted niche on regeneration of muscle fibers
407
To determine which structural aspects are affected by the paracrine signaling and contribute to
408
functional benefit, the muscle fiber density and number of regenerated muscle fibers were
409
quantified in multiple histological cross sections distributed over the entirety of the muscle
410
tissues.
411
In line with the data from the muscle force measurement, no difference in the total number of
412
fibers per section area (muscle fiber density) was observed seven days after injury (Figure 5
413
A; Figure S4). However, the fiber densities in the Alg. (-44%, P<0.001) and the Alg.+GF
414
( 26%, P=0.016) groups were significantly decreased at day 28 compared to day seven,
415
whereas no significant change was observed in the Alg.+MSC (-14%) and Alg.+GF+MSC
416
(+17%) group. Accordingly, treatment of muscles with Alg.+MSC or Alg.+GF+MSC, led to a
417
1.7-fold (P<0.001) and 2-fold (P<0.001) higher fiber density, respectively, compared to the
418
blank alginate group at day 28. Moreover, the fiber density in the Alg.+GF+MSC group was
419
also significantly higher compared to Alg.+GF group (1.5-fold, P=0.001). At day 56, the
420
combination of Alg.+GF+MSC showed a significantly higher muscle fiber density compared
421
to both the Alg. and the Alg.+MSC groups. These results suggest that transplantation of the
422
MSC and GF loaded synthetic niche most likely enhances the survival of the muscle
423
progenitor cells at early phases of the healing process, and may promote fusion and muscle
424
fiber formation at later time points [43].
425
To validate this hypothesis, we investigated the number of regenerated fibers identified by
426
centrally located nuclei within the muscle cross sections. Indeed, the number of regenerated
AC C
EP
TE D
M AN U
SC
406
ACCEPTED MANUSCRIPT
fibers per section area was quantitatively higher in muscles treated with the Alg. and Alg.+GF
428
groups compared to the Alg.+MSC and Alg.+GF+MSC groups at day seven (Figure 5B). At
429
day 28 however, we observed a trend towards increased number of regenerated fibers in
430
muscles treated with Alg.+MSC and Alg.+GF+MSC combinations, but this trend did not
431
reach statistical significance. However, the muscles treated with the combination of
432
Alg.+GF+MSC showed a significantly increased number of regenerated fibers compared to
433
the two other groups at day 56 (Figure 5C).
SC
RI PT
427
434
3.6. Stimulation of angiogenesis and remodeling of scar tissue
436
Next to muscle tissue changes during regeneration, we quantified the amount of fibrotic scar
437
tissue as well as the blood vessel density in the treated muscle tissues. The amount of fibrotic
438
scar tissue was quantitatively lower in muscles treated with the Alg.+GF+MSC combination
439
compared with both control groups (Alg. or Alg.+GF) at all time points investigated (Figure
440
6A and B). In particular, at day seven after injury the amount of fibrotic scar tissue
441
progressively decreased from Alg.+GF (23.8±1.4%), Alg. (20.3±1.0%), Alg.+MSC
442
(18.7±1.7%), to the lowest value detected in the Alg.+GF+MSC group (14.8±1.3%, =0.014,
443
PAlg.+GF<0.001). At day 28, muscles treated with Alg.+GF+MSC (14.3±1.3%) showed
444
significantly lower amounts of fibrotic scar tissue than the Alg. group (22.4±1.6%, P=0.003)
445
as well as the Alg.+GF group (22.3±1.6%, P=0.003). A similar tendency could be observed
446
between the Alg.+MSC (17.5±2.4%) and the control groups, but did not reach statistical
447
significance. The positive effect of the Alg.+GF+MSCs therapy seems to be long-lasting,
448
since we also observed significantly lower fibrosis in that group (11.7±1.3%) compared to
449
Alg.+MSC group (19.5±1.5%, PAlg.+MSC+GF=0.001) as well as the Alg. group (25.4±1.5%,
450
PAlg.+MSC+GF>0.001, PAlg.+MSC=0.027) at 56 days after injury (Figure S5).
451
AC C
EP
TE D
M AN U
435
ACCEPTED MANUSCRIPT
To test whether improved muscle force induced by the combined administration of
453
Alg.+GF+MSC also corresponded to increased vascularization, the total number of vessels
454
per section area was quantified using CD31 staining (Figure 6C and D). We found a
455
significantly higher number of vessels in muscles treated with MSCs or GF alone as well as
456
with the combination of both, compared to the blank alginate control at all time points
457
investigated (Figure S6).
RI PT
452
SC
458
Interestingly, we noted the appearance of new muscle fibers and blood vessels in regions
460
predominantly composed of scar tissue even at day 7 after injury (Figure 7A and B). In line
461
with the reduced amount of scar tissue, vessels and muscle fiber formation were markedly
462
higher in animals that received the Alg.+GF+MSC or the Alg.+MSC treatment (Figure S7 and
463
S8). At 56 days after transplantation, the scar tissue had been largely mitigated as the greater
464
tissue area was remodeled by mature, regenerated myofibers in the Alg.+GF+MSC group.
465
TE D
M AN U
459
4. Discussion
467
The data presented here illustrates the superior effect of an engineered niche that modulates
468
MSC’s paracrine secretion, leading to reduced scar formation, increased angiogenesis, faster
469
reconstitution of muscle structure and improved functional outcome after severe muscle
470
trauma.
AC C
471
EP
466
472
MSC-based bolus transplantation strategies represent a viable approach to improve muscle
473
regeneration, but an unfortunate reality of this approach is the dramatically low percentage of
474
transplanted cells that survive the hostile injury environment and an even lower percentage of
475
cells that eventually differentiate into the desired lineage [44]. Despite these drawbacks,
ACCEPTED MANUSCRIPT
studies report beneficial effects of direct MSC injection, leading to the speculation that the
477
observed outcomes are due to MSC’s trophic, paracrine or immunomodulatory functions [45,
478
46]. Exploring the ability of MSCs delivered in macroporous scaffolds to reinforce
479
endogenous muscle regeneration by influencing the activity of muscle progenitor cells, our
480
study focuses on MSC paracrine effects independent of their physical engraftment to host
481
tissue, myogenic differentiation, or immune modulation.
SC
482
RI PT
476
We identified key modulators of MSC paracrine signaling that indirectly facilitate control
484
over myoblast behavior and function in vitro, and transferred this concept to an in vivo
485
validation experiment in a multiple crush trauma model. In contrast to other frequently used in
486
vivo models, including lacerations, needle punctures or toxin-induced injuries, this model is
487
representative of muscle injuries frequently encountered in the clinic [47]. Similar to human
488
data [48], this preclinical model shows not only a loss in muscle function, but also structural
489
degradation of muscle fibers, formation of scar tissue and disruption of the inherent vascular
490
network. In this context, the outcomes of our study represent a significant advancement in the
491
field of muscle regeneration and has the potential for translation into the clinic.
TE D
EP
492
M AN U
483
Animals in all treatment groups exhibited a 50-60% loss in fast twitch and 40-50% loss in
494
slow twitch tetanic muscle forces 7 days post-trauma relative to the contralateral side. The
495
relatively poor functional outcome of the empty alginate group indicates the inability of the
496
natural healing processes to meet the demands created by severe trauma. In stark contrast,
497
exogenous delivery of MSCs recorded significantly higher muscle contraction forces in the
498
long term. Most importantly, co-delivery of MSCs and growth factors resulted in the fastest
499
and largest recovery of muscle forces. These improved functional outcomes were likely a
500
consequence of significant structural remodeling of the treated tissues. In contrast to earlier
AC C
493
ACCEPTED MANUSCRIPT
studies that employed an ischemic muscle injury model [10], the GF delivery alone did not
502
lead to significant functional or structural improvements. The positive influence of growth
503
factors on tissue regeneration is frequently described in animal models that involve ischemic
504
injury. In those cases, growth factors like VEGF, Ang1, and FGF play a major role in
505
restoring blood vasculature in the injured tissue [49]. However in our injury model, apart from
506
disruption of blood supply, other issues include the onset of fibrosis, loss of cells, and
507
necrosis of muscle fibers. Despite being delivered in large quantities, it seems that VEGF and
508
IGF were unable to do much in the injured tissue except inducing angiogenesis (Figure 6C,
509
D). On the other hand, the MSCs, being a potent source of multiple cytokines, could
510
orchestrate the numerous biological processes necessary to reverse the effects of severe
511
injury, in addition to angiogenesis.
512
M AN U
SC
RI PT
501
In vitro, we found that physiologically relevant indicators of an active regeneration process,
514
such as myoblast proliferation, migration, and survival in harsh conditions, were enhanced
515
after culture in MSC-CM and MSC+GF-CM, in contrast to standalone growth factor
516
supplementation that registered modest improvements. By employing neutralization
517
antibodies, we confirmed that recombinant growth factors do not have an additive effect, but
518
in fact contribute primarily by stimulating MSCs, upregulating their paracrine secretion
519
pattern, and consequently improving myoblast function. Correspondingly, analysis of
520
supernatants revealed that GF-stimulation significantly increased the overall protein content
521
secreted by the MSCs and particularly upregulated the secretion of Hgf, Vegf, and bFgf in
522
comparison to MSC-CM. The growth factor release kinetics outcome suggested that most of
523
the incorporated VEGF165 is released from the cryogel within the first 10 days, whereas the
524
release of IGF-1 persists for 30 days. Together with in vitro studies which revealed that MSCs
525
demonstrate enhanced factor secretion after being stimulated with growth factors for only 24
AC C
EP
TE D
513
ACCEPTED MANUSCRIPT
526
hours, this may indicate that long-term stimulation of MSCs (i.e. more than 10 days) is not
527
necessary to achieve the desired regeneration observed in the Alg.+GF+MSC group.
528
MSCs secrete many factors, which enhance cellular functions including cell division and
530
motility, and provide protection from apoptosis under challenging conditions [25, 50]. For
531
instance HGF, bFGF, GM-CSF, IGF-1, and VEGF have not only been implicated in rendering
532
anti-apoptotic effects [45, 51], but also in the regulation of myoblast cell cycle withdrawal
533
[42, 52-55]. Our results from the Caspase 3/7 activity measurement as well as the
534
differentiation assays are in agreement with the detection of some of these factors in MSC-
535
CM and MSC+GF-CM. In the context of major muscle trauma which results in massive cell
536
loss, our results suggest that MSC-derived factors would likely contribute towards a)
537
preventing further cell loss in the compromised tissue, b) promoting proliferation to increase
538
cell numbers, c) supporting the migration of these proliferating cells to the site of injury, and
539
d) inhibiting the formation of prematurely differentiated muscle fibers. Accordingly, it has
540
been shown that the modulation of the injury environment via soluble factors or anti-fibrotic
541
agents (e.g. TGF-β antagonists) could improve in vivo muscle regeneration by extending
542
myoblast proliferation, increasing their migration and concurrently delaying their
543
differentiation [15, 41, 56].
544
This correlates with our in vivo observation that animals that received Alg.+MSC+GF
545
treatment exhibited the lowest ratio of regenerated fibers at the earliest time point, but
546
recorded a progressive increase thereafter with a significant difference to all other groups at
547
day 56. In accordance with the functional outcome, this treatment also resulted in the highest
548
fiber density of all groups at days 28 and 56. In contrast, animals that received Alg. or
549
Alg.+GF showed quantitatively the highest number of regenerated fibers at the earliest time
550
point, which decreased thereafter. Although no differences in myofiber density were evident
AC C
EP
TE D
M AN U
SC
RI PT
529
ACCEPTED MANUSCRIPT
between all groups at the early time point, we noted that the myofiber density in the Alg. and
552
Alg.+GF group significantly decreased between days 7 and 28. This indicates that the
553
endogenous healing process leads to formation of new fibers, likely due to the action of
554
resident satellite cells, but these fibers fail to reach maturity and become necrotic over time.
555
To the best of our knowledge, there exist no studies investigating the effect of conditioned
556
media from stromal cells on the function of primary satellite cells. This is likely because long-
557
term in vitro culture of satellite cells remains a challenge. Thus, the regenerated fibers in
558
Alg.+GF+MSC group most likely matured over time and participated in the skeletal muscle’s
559
contractile machinery. Additionally, this treatment resulted in a faster restoration of the
560
predominantly slow fiber muscle phenotype, which is also the composition of the native
561
soleus muscle (Figure S3).
562
M AN U
SC
RI PT
551
Re-establishment of a vascular network is an essential criterion for successful muscle healing
564
[57], as cells are normally unable to survive if a nutrient source is located more than 150 µm
565
away. Analysis of muscle sections stained with CD31 revealed a significantly lower amount
566
of blood capillaries in the Alg. group relative to all other treatments. This suggests that
567
improved vessel formation might not be the leading cause for the improved muscle function
568
after MSCs transplantation. However, the absence of a functioning vasculature may also
569
contribute to lower fiber densities and consequently lower muscle contraction forces. Perhaps
570
more importantly, we also observed the presence of blood vessels in regions predominantly
571
composed of scar tissue. Even at the earliest time point, the number of vessels within the scar
572
tissue was notably higher in animals treated with Alg.+MSC+GF or Alg.+MSC. Scar tissue is
573
thought to establish a permanent presence in injured tissues, but the existence of a vascular
574
network within fibrotic regions pointed towards imminent remodeling. We observed that
575
while the area of scar tissue was significantly reduced over time in Alg.+MSC+GF treated
AC C
EP
TE D
563
ACCEPTED MANUSCRIPT
animals, there was a gradual increase in the Alg. group. This could imply that a) the natural
577
healing process is unable to curb or mitigate the progression of scar tissue formation, and b)
578
the availability of soluble factors secreted from MSCs even at very early time points induces a
579
dramatic remodeling of the scar tissue in the long-term. For example, the secretion of HGF
580
was significantly enhanced in MSCs upon their stimulation with growth factors; this has been
581
previously described to play an important role in scar tissue remodeling [58, 59]. To the best
582
of our knowledge, there are no existing reports on the effect of MSC derived paracrine factors
583
on the function of ECM secreting fibroblasts or native fibro/adipogenic progenitor cells
584
(FAPs), despite the fact that MSCs secrete a number cytokines with anti-scarring and matrix
585
remodeling properties. It is probable that in addition to influencing myoblast function, MSCs
586
also guide the behavior of fibroblasts and modulate their collagen deposition.
M AN U
SC
RI PT
576
587
5. Conclusion
589
Collectively, our results suggest that MSC mediated paracrine signaling can support
590
endogenous muscle healing processes in a robust manner, and that the functional and
591
structural outcomes of regeneration can be further improved by enhancing the secretion
592
pattern of MSCs via local growth factor stimulation. The synthetic niche approach described
593
in this study is an important example of enabling in situ regeneration of tissues without
594
physical engraftment of transplanted cells, and will likely be an approach that is applicable in
595
the treatment of a number of compromised tissue functions.
EP
AC C
596
TE D
588
597
Supporting Information
598
Supporting Information accompanies this paper.
599 600
Acknowledgements
ACCEPTED MANUSCRIPT
We would like to acknowledge Gabriela Korus and Mario Thiele for providing technical
602
assistance with histology, and ImageJ macros, respectively. Funding: This work was
603
supported by grants from the BCRT and BSRT through funding by the German Federal
604
Ministry of Education and Research (BMBF), by a grant (GE2512/1-2) from the German
605
Research Foundation (DFG) to SG, and by the award of the Einstein fellowship to DJM. THQ
606
wishes to acknowledge funding from the Friede Springer Foundation and the BSRT. The
607
funders had no role in study design, data collection and analysis, decision to publish, or
608
preparation of the manuscript.
AC C
EP
TE D
M AN U
SC
RI PT
601
ACCEPTED MANUSCRIPT
References
[1] Charge SB, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004;84:209-38.
RI PT
[2] Huard J, Li Y, Fu FH. Muscle injuries and repair: current trends in research. J Bone Joint Surg Am. 2002;84-A:822-32. [3] Gharaibeh B, Chun-Lansinger Y, Hagen T, Ingham SJ, Wright V, Fu F, et al. Biological approaches to improve skeletal muscle healing after injury and disease. Birth defects research Part C, Embryo today : reviews. 2012;96:82-94.
SC
[4] Klein O, Strohschein K, Nebrich G, Oetjen J, Trede D, Thiele H, et al. MALDI imaging mass spectrometry: discrimination of pathophysiological regions in traumatized skeletal muscle by characteristic peptide signatures. Proteomics. 2014;14:2249-60.
M AN U
[5] Huard J, Li Y, Fu FH. Muscle Injuries and Repair: Current Trends in Research. The Journal of Bone & Joint Surgery. 2002;84:822-32. [6] Cezar CA, Mooney DJ. Biomaterial-based delivery for skeletal muscle repair. Advanced drug delivery reviews. 2015;84:188-97. [7] Turner NJ, Badylak SF. Regeneration of skeletal muscle. Cell Tissue Res. 2012;347:75974.
TE D
[8] Fishman JM, Tyraskis A, Maghsoudlou P, Urbani L, Totonelli G, Birchall MA, et al. Skeletal muscle tissue engineering: which cell to use? Tissue Eng Part B Rev. 2013;19:50315. [9] Menetrey J, Kasemkijwattana C, Day CS, Bosch P, Vogt M, Fu FH, et al. Growth factors improve muscle healing in vivo. J Bone Joint Surg Br. 2000;82:131-7.
EP
[10] Borselli C, Storrie H, Benesch-Lee F, Shvartsman D, Cezar C, Lichtman JW, et al. Functional muscle regeneration with combined delivery of angiogenesis and myogenesis factors. Proc Natl Acad Sci U S A. 2010;107:3287-92.
AC C
[11] Browne S, Pandit A. Multi-modal delivery of therapeutics using biomaterial scaffolds. Journal of Materials Chemistry B. 2014;2:6692-707. [12] Santo VE, Gomes ME, Mano JF, Reis RL. Controlled release strategies for bone, cartilage, and osteochondral engineering--Part II: challenges on the evolution from single to multiple bioactive factor delivery. Tissue Eng Part B Rev. 2013;19:327-52. [13] Mendell JR, Kissel JT, Amato AA, King W, Signore L, Prior TW, et al. Myoblast transfer in the treatment of Duchenne's muscular dystrophy. N Engl J Med. 1995;333:832-8. [14] Tremblay JP, Malouin F, Roy R, Huard J, Bouchard JP, Satoh A, et al. Results of a triple blind clinical study of myoblast transplantations without immunosuppressive treatment in young boys with Duchenne muscular dystrophy. Cell transplantation. 1993;2:99-112. [15] Briggs D, Morgan JE. Recent progress in satellite cell/myoblast engraftment -- relevance for therapy. The FEBS journal. 2013;280:4281-93.
ACCEPTED MANUSCRIPT
[16] Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143-7. [17] Gang EJ, Jeong JA, Hong SH, Hwang SH, Kim SW, Yang IH, et al. Skeletal myogenic differentiation of mesenchymal stem cells isolated from human umbilical cord blood. Stem Cells. 2004;22:617-24.
RI PT
[18] Gunetti M, Tomasi S, Giammo A, Boido M, Rustichelli D, Mareschi K, et al. Myogenic potential of whole bone marrow mesenchymal stem cells in vitro and in vivo for usage in urinary incontinence. PLoS One. 2012;7:e45538. [19] Philippou A, Maridaki M, Theos A, Koutsilieris M. Cytokines in muscle damage. Advances in clinical chemistry. 2012;58:49-87.
SC
[20] De Bari C, Dell'Accio F, Vandenabeele F, Vermeesch JR, Raymackers JM, Luyten FP. Skeletal muscle repair by adult human mesenchymal stem cells from synovial membrane. The Journal of cell biology. 2003;160:909-18.
M AN U
[21] Krampera M, Pizzolo G, Aprili G, Franchini M. Mesenchymal stem cells for bone, cartilage, tendon and skeletal muscle repair. Bone. 2006;39:678-83. [22] Amado LC, Saliaris AP, Schuleri KH, St John M, Xie JS, Cattaneo S, et al. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A. 2005;102:11474-9. [23] Choi YH, Kurtz A, Stamm C. Mesenchymal stem cells for cardiac cell therapy. Human gene therapy. 2011;22:3-17.
TE D
[24] Matziolis G, Winkler T, Schaser K, Wiemann M, Krocker D, Tuischer J, et al. Autologous bone marrow-derived cells enhance muscle strength following skeletal muscle crush injury in rats. Tissue Eng. 2006;12:361-7.
EP
[25] Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine & growth factor reviews. 2009;20:419-27. [26] Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circulation research. 2008;103:1204-19.
AC C
[27] Fan Y, Maley M, Beilharz M, Grounds M. Rapid death of injected myoblasts in myoblast transfer therapy. Muscle & nerve. 1996;19:853-60. [28] Winkler T, von Roth P, Matziolis G, Mehta M, Perka C, Duda GN. Dose-response relationship of mesenchymal stem cell transplantation and functional regeneration after severe skeletal muscle injury in rats. Tissue Eng Part A. 2009;15:487-92. [29] Qazi TH, Mooney DJ, Pumberger M, Geissler S, Duda GN. Biomaterials based strategies for skeletal muscle tissue engineering: existing technologies and future trends. Biomaterials. 2015;53:502-21. [30] Moshaverinia A, Chen C, Xu X, Ansari S, Zadeh HH, Schricker SR, et al. Regulation of the Stem Cell-Host Immune System Interplay Using Hydrogel Coencapsulation System with an Anti-Inflammatory Drug. Advanced functional materials. 2015;25:2296-307.
ACCEPTED MANUSCRIPT
[31] Borselli C, Cezar CA, Shvartsman D, Vandenburgh HH, Mooney DJ. The role of multifunctional delivery scaffold in the ability of cultured myoblasts to promote muscle regeneration. Biomaterials. 2011;32:8905-14.
RI PT
[32] Rossi CA, Flaibani M, Blaauw B, Pozzobon M, Figallo E, Reggiani C, et al. In vivo tissue engineering of functional skeletal muscle by freshly isolated satellite cells embedded in a photopolymerizable hydrogel. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2011;25:2296-304. [33] Merritt EK, Cannon MV, Hammers DW, Le LN, Gokhale R, Sarathy A, et al. Repair of traumatic skeletal muscle injury with bone-marrow-derived mesenchymal stem cells seeded on extracellular matrix. Tissue Eng Part A. 2010;16:2871-81.
SC
[34] Natsu K, Ochi M, Mochizuki Y, Hachisuka H, Yanada S, Yasunaga Y. Allogeneic bone marrow-derived mesenchymal stromal cells promote the regeneration of injured skeletal muscle without differentiation into myofibers. Tissue Eng. 2004;10:1093-112.
M AN U
[35] Place ES, Evans ND, Stevens MM. Complexity in biomaterials for tissue engineering. Nature materials. 2009;8:457-70. [36] Geisler S, Textor M, Schmidt-Bleek K, Klein O, Thiele M, Ellinghaus A, et al. In serum veritas[mdash]in serum sanitas[quest] Cell non-autonomous aging compromises differentiation and survival of mesenchymal stromal cells via the oxidative stress pathway. Cell death & disease. 2013;4:e970.
TE D
[37] Geback T, Schulz MM, Koumoutsakos P, Detmar M. TScratch: a novel and simple software tool for automated analysis of monolayer wound healing assays. BioTechniques. 2009;46:265-74. [38] Kong HJ, Smith MK, Mooney DJ. Designing alginate hydrogels to maintain viability of immobilized cells. Biomaterials. 2003;24:4023-9. [39] Rowley JA, Madlambayan G, Mooney DJ. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials. 1999;20:45-53.
EP
[40] Winkler T, von Roth P, Radojewski P, Urbanski A, Hahn S, Preininger B, et al. Immediate and delayed transplantation of mesenchymal stem cells improve muscle force after skeletal muscle injury in rats. J Tissue Eng Regen Med. 2012;6 Suppl 3:s60-7.
AC C
[41] Bencze M, Negroni E, Vallese D, Yacoub-Youssef H, Chaouch S, Wolff A, et al. Proinflammatory macrophages enhance the regenerative capacity of human myoblasts by modifying their kinetics of proliferation and differentiation. Molecular therapy : the journal of the American Society of Gene Therapy. 2012;20:2168-79. [42] Clegg CH, Linkhart TA, Olwin BB, Hauschka SD. Growth factor control of skeletal muscle differentiation: commitment to terminal differentiation occurs in G1 phase and is repressed by fibroblast growth factor. The Journal of cell biology. 1987;105:949-56. [43] Webster Micah T, Manor U, Lippincott-Schwartz J, Fan C-M. Intravital Imaging Reveals Ghost Fibers as Architectural Units Guiding Myogenic Progenitors during Regeneration. Cell Stem Cell.18:243-52.
ACCEPTED MANUSCRIPT
[44] von Bahr L, Batsis I, Moll G, Hagg M, Szakos A, Sundberg B, et al. Analysis of tissues following mesenchymal stromal cell therapy in humans indicates limited long-term engraftment and no ectopic tissue formation. Stem Cells. 2012;30:1575-8.
RI PT
[45] Togel F, Weiss K, Yang Y, Hu Z, Zhang P, Westenfelder C. Vasculotropic, paracrine actions of infused mesenchymal stem cells are important to the recovery from acute kidney injury. American journal of physiology Renal physiology. 2007;292:F1626-35. [46] Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Experimental & molecular medicine. 2013;45:e54.
SC
[47] Shin EH, Caterson EJ, Jackson WM, Nesti LJ. Quality of healing: defining, quantifying, and enhancing skeletal muscle healing. Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society. 2014;22 Suppl 1:18-24.
M AN U
[48] Sicari BM, Rubin JP, Dearth CL, Wolf MT, Ambrosio F, Boninger M, et al. An acellular biologic scaffold promotes skeletal muscle formation in mice and humans with volumetric muscle loss. Sci Transl Med. 2014;6:234ra58. [49] Yla-Herttuala S, Alitalo K. Gene transfer as a tool to induce therapeutic vascular growth. Nat Med. 2003;9:694-701. [50] Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98:1076-84.
TE D
[51] Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, et al. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109:1292-8.
EP
[52] Anastasi S, Giordano S, Sthandier O, Gambarotta G, Maione R, Comoglio P, et al. A natural hepatocyte growth factor/scatter factor autocrine loop in myoblast cells and the effect of the constitutive Met kinase activation on myogenic differentiation. The Journal of cell biology. 1997;137:1057-68.
AC C
[53] Leshem Y, Spicer DB, Gal-Levi R, Halevy O. Hepatocyte growth factor (HGF) inhibits skeletal muscle cell differentiation: a role for the bHLH protein twist and the cdk inhibitor p27. J Cell Physiol. 2000;184:101-9. [54] Linkhart TA, Clegg CH, Hauschka SD. Control of mouse myoblast commitment to terminal differentiation by mitogens. Journal of supramolecular structure. 1980;14:483-98. [55] Allen RE, Boxhorn LK. Regulation of skeletal muscle satellite cell proliferation and differentiation by transforming growth factor-beta, insulin-like growth factor I, and fibroblast growth factor. J Cell Physiol. 1989;138:311-5. [56] Burks TN, Cohn RD. Role of TGF-beta signaling in inherited and acquired myopathies. Skelet Muscle. 2011;1:19. [57] Jung S, Kleinheinz J. Angiogenesis - The Key to Regeneration. In: Andrades JA, editor. Regenerative Medicine and Tissue Engineering: InTech; 2013.
ACCEPTED MANUSCRIPT
[58] Xiao Z, Xi C. Hepatocyte growth factor reduces hypertrophy of skin scar: in vivo study. Advances in skin & wound care. 2013;26:266-70.
AC C
EP
TE D
M AN U
SC
RI PT
[59] Jackson WM, Nesti LJ, Tuan RS. Mesenchymal stem cell therapy for attenuation of scar formation during wound healing. Stem cell research & therapy. 2012;3:20.
ACCEPTED MANUSCRIPT
M AN U
SC
RI PT
Figure 1. Paracrine factors in conditioned media influence myoblast function in vitro. (A) Conditioned media from unstimulated and stimulated MSCs enhance myoblast proliferation (CyQuant®), survival (Trypan Blue), and prevent their apoptosis (Caspase-Glo® 3/7) [n=10,*P<0.05, ANOVA with Bonferroni correction]. (B) Assessment of migration behavior of C2C12 cells in a scratch wound healing assay revealed significantly faster migration in response to MSC+GF-CM. Graphs illustrate the kinetics of scratch repopulation over time (left), and the corresponding single point evaluation at 20h (right). [n=8,*P<0.05, ANOVA with Dunnett correction]. (C) Representative images of the migration assay taken using an inverted live cell-imaging microscope 20h after scratching the cell monolayer. (D-E) Myoblast differentiation quantified using Western blot analysis of Myosin Heavy Chain (Myh2), MyoD, and Myogenin. Equal protein loading was confirmed by detecting GAPDH. The results were quantified by densitometry and are shown in the corresponding diagram. [n=4, *P<0.05, ANOVA with Dunnett correction]. (F) Indirect immunofluorescence of Myh2 (green) and DAPI (blue) showed that C2C12 cells cultivated in blank and blank+GF groups highly expressed Myh2 and formed multinucleated myotubes, while fusion was inhibited in the presence of MSC-CM or MSC+GF-CM. Error bars in all panels represent standard error of the mean.
TE D
Figure 2. Growth factors indirectly influence myoblast function by altering secretion pattern of MSCs. Depletion of IGF-1 and VEGF165 did not alter (A) proliferation, (B) survival, or (C) migration of C2C12 cells cultured in MSC-CM or MSC+GF-CM, whereas positive effects of blank media with GF (+GF) receded. (D) MSCs upregulated their protein and cytokine secretion in response to GF stimulation. MSC+GF-CM contained higher levels of Hgf, bFgf, and Vegf. (For [A - D] n=5, *P<0.05, [A & B] ANOVA with Bonferroni correction, [C] ANOVA with Dunnett correction, [D] paired two-sample t-test). Error bars in all panels represent standard error of the mean.
AC C
EP
Figure 3. An engineered synthetic niche provides MSCs with a structural and chemical environment that is optimal for paracrine secretion. (A) The schematic illustrates the strategy employed in this study. A porous scaffold containing MSCs and growth factors was placed adjacent to the injured muscle. MSCs in the porous niche are stimulated by the sustained release of growth factors, resulting in the secretion of paracrine factors that promote muscle regeneration. (B) Representative SEM image showing the macroporous structure of the alginate scaffold. (C) Representative fluorescent image of rat bone marrow derived MSCs (stained with CellTracker™ and DAPI) 24 hours after seeding on the scaffold. (D) Release kinetics of IGF-1 and VEGF165 from the alginate scaffolds over a period of 30 days monitored using ELISA. (E) Independent from GF incorporation, MSCs remained highly viable for at least 7 days in vitro. (F) Emigration of cells from the scaffolds was quantified using CyQuant® assay. The results indicated that a very low percentage of cells inside the scaffold migrated out over a period of 7 days. (For [D - F] n=5, [E & F] ANOVA with Bonferroni correction). Error bars in all panels represent standard error of the mean.
ACCEPTED MANUSCRIPT
RI PT
Figure 4. Recovery of contraction forces in treated muscles. Functional regeneration was assessed by measuring (A) fast twitch, and (B) tetanic contraction forces at three time points post injury. Forces were normalized to uninjured contralateral muscles, and the graphs show the relative fast twitch and tetanic forces. Animals that received the Alg.+GF+MSC transplantation exhibited the largest recovery of contraction forces. [n ≥ 8, *p<0.05, ANOVA with Bonferroni correction]. Error bars in all panels represent standard error of the mean.
M AN U
SC
Figure 5. Structural repair accomplished via new myofiber formation. (A) Muscle fiber densities were calculated by normalizing the total number of fibers to section area, and revealed higher mature fibers in animals treated with Alg.+GF+MSC in the long term. (B) The number of regenerated fibers (identified by their central nuclei) corroborated the fiber density measurements in the different groups over time. The results indicate a continuous remodeling of the muscle tissue structure. (C) Representative images of myofibers (crosssectional view) over time. Scar tissue is visibly present in the Alginate group even 56 days after injury. This is notable reduced (Alginate+MSC) or absent (Alg.+GF+MSC) in the other treatment groups. The growth factor treated group (Alg.+GF) was not proceeded to 56 days, and is shown in Figure S9 and S10. (n ≥ 8, *p<0.05, [A] ANOVA with Bonferroni correction, [B] Kruskal-Wallis Test with adjustment for multiple comparison). Error bars in all panels represent standard error of the mean.
AC C
EP
TE D
Figure 6. Blood vessels that infiltrate fibrotic areas and facilitate myofiber formation expedite scar tissue remodeling. (A) Fibrotic scar tissue area was quantified over time and revealed dramatic remodeling in animals treated with Alg.+GF+MSC. (B) Representative postprocessed histological images of muscle sections showing scar tissue regions (dark red). Sections from the Alg.+GF group are shown in Figure S9. (C) Vascularization of injured tissues was analyzed with CD31 staining that revealed significantly low number of blood vessels in the control (Alg.) group at all time points. (D) Representative images of vascularization in injured tissues after different treatments (blood vessels stained dark red). Sections from the Alg.+GF group are shown in Figure S9. (For [A, C] n ≥ 8, *p<0.05, [A] ANOVA with Dunett T3 correction, [C] ANOVA with Bonferroni correction). Error bars in all panels represent standard error of the mean. Figure 7. Presence of regenerated muscle fibers and blood vessels in the scar regions of the injured tissues (pale pink = scar tissue, bright pink = muscle fibers). (A) Representative images of scar tissue remodeling by new myofibers indicate structural benefits of MSC derived paracrine factors which are notably absent in the control (Alg.) group even after 56 days. (f) We propose this remodeling of scar tissue was facilitated by infiltration of blood vessels into the fibrotic regions (blood vessels stained dark red).
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
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT