Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Biomaterials 140 (2017) 103e114

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Biomaterials that promote cell-cell interactions enhance the paracrine


function of MSCs
Taimoor H. Qazi a, b, David J. Mooney c, Georg N. Duda a, b, Sven Geissler a, b, *
a
Julius Wolff Institute, Charit €tsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
e Universita
b
Berlin-Brandenburg Center for Regenerative Therapies & Berlin-Brandenburg School for Regenerative Therapies, Charit €tsmedizin Berlin,
e Universita
Augustenburger Platz 1, 13353 Berlin, Germany
c
John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, MA 02138, USA

a r t i c l e i n f o a b s t r a c t

Article history: Mesenchymal stromal cells (MSCs) secrete paracrine factors that play crucial roles during tissue
Received 6 June 2017 regeneration. Whether this paracrine function is influenced by the properties of biomaterials in general,
Accepted 16 June 2017 and those used for cell delivery in particular, largely remains unexplored. Here, we investigated if three-
Available online 18 June 2017
dimensional culture in distinct microenvironments - nanoporous hydrogels (mean pore size ~5 nm) and
macroporous scaffolds (mean pore size ~120 mm) - affects the secretion pattern of MSCs, and conse-
Keywords:
quently leads to differential paracrine effects on target progenitor cells such as myoblasts. We report that
Trophic secretion
compared to MSCs encapsulated in hydrogels, scaffold seeded MSCs show an enhanced secretion profile
N-cadherin
Muscle regeneration
and exert beneficial paracrine effects on various myoblast functions including migration and prolifera-
Stem cell therapy tion. Additionally, we show that the heightened paracrine effects of scaffold seeded cells can in part be
3D culture attributed to N-cadherin mediated cell-cell interactions during culture. In hydrogels, this physical
Paracrine signaling interaction between cells is prevented by the encapsulating matrix. Functionally blocking N-cadherin
Cryogel negatively affected the secretion profile and paracrine effects of MSCs on myoblasts, with stronger effects
Hydrogel observed for scaffold seeded compared to hydrogel encapsulated cells. Together, these findings
demonstrate that the therapeutic potency of MSCs can be enhanced by biomaterials that promote cell-
cell interactions.
© 2017 Elsevier Ltd. All rights reserved.

1. Introduction secrete a wide range of bioactive cytokines and growth factors that
can influence nearby cells via paracrine signaling [5,6]. For instance,
Inefficient natural healing of several debilitating musculoskel- MSCs have been documented to be a generous source of bioactive
etal injuries such as volumetric tissue loss and severe muscle factors including VEGF, FGF, HGF, IGF, PDGF, ILs, and MMPs [6e9].
trauma has led to an unmet clinical demand for potent therapies. These secreted factors can in turn orchestrate various biological
Strategies that embody the principles of tissue engineering and processes that may be desirable for tissue regeneration such as
regenerative medicine, such as autologous and allogeneic stem cell angiogenesis, immune modulation, and cell recruitment and dif-
therapies, have gained considerable traction in the past decade ferentiation [10,11]. An increasing body of evidence suggests that
[1e3]. As witnessed by their extensive application in registered the therapeutic benefits observed following MSC transplantation
clinical trials, mesenchymal stromal cells (MSCs) in particular have may largely be attributed to the paracrine and trophic function of
attracted widespread interest as safe and effective candidates for these cells, rather than their differentiation potential [12]. For this
the regeneration of injured tissues [4]. reason, research efforts by a number of groups have focused on
In addition to their well-known multi-lineage differentiation strategies that can enhance the secretory function of MSCs. These
potential, an intriguing characteristic of MSCs is their ability to include preconditioning in hypoxic conditions, genetic manipula-
tion to overexpress certain growth factors, and cytokine stimula-
tion [13,14]. For example, in a previous study we reported that the
paracrine effects of MSCs stimulated with soluble VEGF and IGF-1
 Universita
* Corresponding author. Julius Wolff Institute, Charite €tsmedizin Berlin,
enhanced myoblast migration, proliferation, and survival in vitro,
Augustenburger Platz 1, 13353 Berlin, Germany.
E-mail address: sven.geissler@charite.de (S. Geissler). and led to robust muscle regeneration in vivo [15]. Recent studies

http://dx.doi.org/10.1016/j.biomaterials.2017.06.019
0142-9612/© 2017 Elsevier Ltd. All rights reserved.
104 T.H. Qazi et al. / Biomaterials 140 (2017) 103e114

have highlighted the importance of cell-cell interactions in regu- the role of substrate microenvironment mediated cell-cell in-
lating the paracrine activity of MSCs by reporting that 3D multi- teractions in regulating the paracrine effects of MSCs. Our data
cellular spheroids show superior secretion profiles compared to indicate that changes in biomaterial microenvironment can elicit
similar number of MSCs in monolayer culture. For instance, Lee strong paracrine responses from MSCs by promoting local cell-cell
et al. reported that MSC spheroid formation via calcium dependent interactions.
E-cadherin interactions led to improved paracrine activity and
therapeutic efficacy in a rat myocardial infarction model [16]. 2. Materials and methods
Similar paracrine effects have been reported for spheroids formed
using adipose derived [17], umbilical cord derived [18], and bone 2.1. Alginate processing and substrate fabrication
marrow derived MSCs [19].
Researchers have also explored the feasibility of applying MSC Ultrapure low and high molecular weight alginates (LVG, MVG)
derived conditioned medium (MSC-CM) to stimulate tissue were purchased from Pronova. A 1:1 (w/w) mixture of the alginates
regeneration. For instance, Osugi et al. reported enhanced in vivo was reconstituted to a concentration of 1% w/v, and functionalized
bone formation in rat calvarial defects after the application of MSC- with the cell adhesive peptide motif G4RGDSP (Peptide 2.0, USA)
CM that contained high quantities of IGF-1 and VEGF [20]. Others using standard carbodiimide chemistry to a final degree of substi-
have used this cell-free approach in spinal cord and cardiac injury tution of 20, as previously described [35]. Briefly, NHS, EDC, and the
models [21,22]. While the transplantation of such a cytokine rich peptide motifs were added sequentially to the reconstituted algi-
cocktail avoids the cost and labor intensive process of cell har- nate, and allowed to react for 20 h. The reaction was stopped using
vesting and subsequent expansion, these cytokines may quickly hydroxyl amine hydrochloride, and the alginates were dialyzed
lose bioactivity in vivo due to short half-lives. Moreover, complete (MWCO 10000, Spectra/Por®) over three days in distilled water
recovery from severe injuries may require a prolonged delivery of with a decreasing salt concentration. The dialyzed alginate was
paracrine factors which necessitates MSC transplantation. In the mixed with activated charcoal, sterile filtered, frozen, and lyophi-
vast majority of pre-clinical and clinical studies, MSCs are admin- lized until use.
istered in a bolus manner either systemically (via the circulatory Hydrogels containing encapsulated cells were prepared as fol-
system) or locally at the site of injury [23,24]. A number of draw- lows. A 2.5% w/v solution of alginate was mixed with a cell sus-
backs are associated with these modes of cell delivery. These pension containing 2  106 freshly trypsinized MSCs and 4% v/v of
include rapid cell death [25], accumulation in non-targeted tissues calcium sulfate slurry (1.22 M in dH2O) using two syringes con-
[26], and diffusion of the cell suspension away from the site of nected with a Luer-lock syringe connector. The components were
injury due to anoikis [27]. By providing a temporary matrix that can mixed rapidly to a yield a cell containing alginate mixture (conc. 2%
facilitate cell attachment, viability, and growth, biomaterials can w/v) and cast between two glass plates separated by a 1 mm spacer.
potentially be used as local delivery depots secreting paracrine The mixture was allowed to ionically crosslink for 45 min before
signaling molecules at sites of tissue damage. Tuning the physical gels were punched out (diameter: 8 mm) and placed in 24 well
properties of such biomaterial depots may offer the possibility to plates with full cell culture media. Scaffolds were prepared in a
elicit desirable MSC activity and thereby steer the regenerative similar manner, but without mixing with cells. The punched out
function of target progenitor cells. gels (~50 mm3) were frozen at 80  C and lyophilized to create a
A wide range of biomaterials with varying structures, chemis- porous structure. The interconnected porous structure was
tries, and mechanical properties have been used for in vivo cell confirmed with scanning electron microscopy, and pore size (short
delivery [28,29]. From a clinical translation perspective, nano- axis) was manually determined using ImageJ. Freshly trypsinized
porous hydrogels may be considered attractive materials for cell MSCs were then seeded onto the 3D porous scaffolds (4  106 cells/
delivery because they can be injected near the site of injury in a cm3) and incubated for 20 min before adding full culture media.
minimally invasive manner and adapt to the size and shape of the
defect [30,31]. Commonly used hydrogels such as alginate, poly 2.2. Mechanical characterization
ethylene glycol (PEG), and hyaluronic acid offer the advantage of
tunable chemistries and mechanical properties, but prevent Hydrogel and scaffold stiffness was determined in the wet state
changes in the morphology of encapsulated cells. On the other using a bench-top PIUMA nanoindenter (Optics 11, Amsterdam). A
hand, pre-formed 3D scaffolds fabricated via 3D printing, electro- spherical indentation tip (radius 48.5 mm) was used to probe
spinning, or freeze drying, may not be conveniently injectable but samples immersed in PBS up to an indentation depth of 10 mm
offer macroporous microenvironments that facilitate cell (sample thickness ~1000 mm). Matrix scans (3  3 points, step size
spreading, migration, and establishment of cell-cell contacts [32]. in each direction ¼ 100 mm) were carried out at several different
While a great deal of research effort has focused on using bio- locations on a number of hydrogel and scaffold samples. Using the
materials to direct stem cell fate, there is a lack of sufficient Oliver and Pharr model, the PIUMA nanoindenter estimated the
knowledge on how biomaterial properties influence the paracrine samples' Young's modulus from the slope of the unloading curve in
function of MSCs. the region 65%e85% of maximum load.
We hypothesized that compared to nanoporous hydrogels that
are used for cell encapsulation and have pores in the range of 2.3. Cell culture
5e70 nm [33,34], macroporous scaffolds with pore sizes in the
range of 10e200 mm may facilitate both cell-cell and cell-matrix MSCs isolated from the tibiae of Sprague Dawley rats via bone
interactions and lead to enhanced paracrine effects of MSCs. To marrow biopsies were used in this study. Cell culture was carried
address this hypothesis, we evaluated the secretion profile of MSCs out in full culture media i.e. low glucose DMEM containing fetal calf
cultured in 3D alginate based substrates that had similar mechan- serum (10%), penicillin/streptomycin (1%), and Glutamax™ (1%).
ical and chemical properties but distinct structural microenviron- Cells were passaged at approximately 70% confluency, and were not
ments. The secreted factors were characterized, and their potential used beyond passage 5. C2C12 myoblasts (ATCC) were cultured in
implication for muscle regeneration was investigated by analyzing full culture media and were not used beyond passage 10. To induce
the behavior and function of myoblasts cultured in conditioned myogenic differentiation, the serum concentration was reduced to
media. Functional blocking experiments were used to determine 5%.
T.H. Qazi et al. / Biomaterials 140 (2017) 103e114 105

2.4. MSC viability numbers recorded earlier for each substrate condition.

MSCs were isolated from hydrogels or scaffolds by alginate 2.8. Myoblast migration
dissolution using 50 mM EDTA in PBS. Cells were stained with
calcein AM (1:1000) and propidium iodide (1:2500) in culture Migration was determined using a scratch wound healing assay,
media for 15 min. The stained cell suspension was imaged using an as previously described [15]. Briefly, C2C12 myoblasts were seeded
inverted fluorescent microscope. Viability was calculated by in 24 well plates at a density of 1.4  105 per well and allowed to
quantifying the number of live and dead cells in at least 5 regions of reach confluency. A 200 mL pipette tip was used to make a scratch
interest. across the monolayer. Wells were washed with PBS and appropriate
conditioned media was then added. Selected regions of interest
2.5. Conditioned media (CM) were imaged every 30 min using a live cell microscope (Leica,
Germany). Scratch area was determined using T-Scratch software
To generate CM from tissue culture plastic (TCP), MSCs were [38]. Cell velocity, distance, displacement, and directionality were
seeded at a density of 1  104 cells/cm2 on 6 well plates. To generate determined by using the Manual Tracking tool in ImageJ to track
Hydrogel-CM, freshly trypsinized MSCs were mixed with alginate single cells in independent wells, and the data was analyzed using
at a density of 2  106 cells/mL alginate and the mixture was the Chemotaxis and Migration Tool (ibidi®).
subsequently crosslinked. To generate Scaffold-CM, trypsinized
MSCs were seeded onto scaffolds at a density of 4  106 cells/cm3. A 2.9. Myoblast proliferation
high cell density was used for the scaffolds to account for the cell
leakage (~40e60%) associated with the macroporosity. MSCs Proliferation was determined using the CyQUANT® cell prolif-
cultured on TCP, hydrogels, or scaffolds were washed with PBS eration assay kit, as previously described [15]. Briefly, C2C12
before adding 1.5 mL serum-free media. For hydrogel encapsulated myoblasts were seeded in 48 well plates at a density of 2.5  104
and scaffold seeded MSCs, collection of CM started 3 days after per well and allowed to attach overnight. After washing with PBS,
initial cell seeding/encapsulation. After 24 h, the conditioned media appropriate serum free conditioned media was then added to the
was collected, centrifuged to pellet cellular debris, and the super- wells, and cultured over two days. At t ¼ 0, 24 h, and 48 h, media
natants were sterile filtered and stored at 80  C until use. At the was aspirated, cells were washed with PBS, and frozen at 80  C for
time of CM collection, cells on the different substrates were coun- up to 7 days. For quantification, cells were thawed, and CyQUANT
ted using the CASY® cell counter. Cells on TCP were trypsinized, dye (400 mM) and lysis buffer (20 mM) in distilled water were
whereas those on alginate substrates were isolated by using 50 mM added to each well followed by incubation for 7.5 min in the dark.
EDTA to dissociate the scaffolds/gels. Cell numbers were then used Fluorescence values were measured using a plate reader.
to normalize concentrations and intensities obtained in ELISA and
cytokine array experiments. 2.10. Myoblast survival

2.6. N-cadherin blocking Cell survival was determined using the Trypan Blue dye to
calculate cell viability. After 24 h culture in appropriate media
Functional blockade of cell-cell interactions was carried out (control, or CM), cells were washed with PBS, trypsinized, and the
according to methods described in previously published reports cell suspension was mixed with Trypan blue dye (1:1). The number
[36,37]. Briefly, trypsinized MSCs were pelleted and re-suspended of live and dead cells were quantified by counting using a
in media containing 40 mg/mL anti-N cadherin antibody (GC-4, hemocytometer.
Sigma-Aldrich) and incubated for 45 min. The cells were then
washed twice with PBS and seeded onto the scaffolds or encapsu- 2.11. Myoblast differentiation
lated in hydrogels. After 3 days in culture, MSCs were treated with
4 mM EDTA in PBS for 5 min to disrupt any calcium dependent cell- To assess myotube formation, C2C12 myoblasts were seeded in
cell interactions, washed twice with PBS, and placed in fresh serum 24 well plates at a density of 1  105 per well and allowed to attach
free media supplemented with an additional 10 mg/mL of anti-N overnight. To induce differentiation, appropriate serum free
cadherin antibody. The additional application of N-cadherin anti- conditioned media was exogenously supplemented with 5% v/v FCS
body was carried out to prevent the re-establishment of cell-cell before being added to the cells. Media was refreshed on alternate
contacts that were disrupted using EDTA. days. Myogenic differentiation was evaluated using immunofluo-
rescence imaging as described earlier. Myotube lengths and density
2.7. ELISA and cytokine arrays were quantified using ImageJ. Further quantification was carried
out using Western Blot analysis. Differentiated myoblasts were
Rat specific ELISAs for VEGF (cat. # RRV00), FGF (cat. # MFB00), lysed using SDS lysis buffer. Protein content was determined using
HGF (cat. # MHG00), and IGF (cat. # MG100) were purchased from the DC protein assay (Bio-Rad). The Novex NuPAGE® system (Invi-
R&D systems, and used according to the manufacturer's in- trogen) was used according to manufacturer's instructions followed
structions to detect concentrations in conditioned media. Rat spe- by semi-dry transfer. Primary antibodies for Myosin heavy chain
cific ELISA for LIF was purchased from LSBio (cat. # LS-F13295), and (Clone # MF20, R&D Systems, Catalog # MAB4470) (1:500), Myo-
used according to the manufacturer's instructions. Rat specific genin (Clone #F5D, Abcam, Catalog # ab1835) (1:500), and the
cytokine arrays were purchased from Ray Biotech (cat. # AAR-BLM- house keeping gene GAPDH (Clone # 6C5, Abcam, Catalog #
1-4-RB and cat. # AAR-CYT-2-8-RB), and used according to the ab8245) (1:10000) were used with TBSTþ5% non-fat milk. Anti-
manufacturer's instructions. Images were acquired using a G:BOX mouse horseradish peroxidase (GE Healthcare) (1:7000) was uti-
imager (Syngene). Intensities were determined using the Protein lized as the secondary antibody. Using ECL substrate (GE Health-
Array Analyzer plugin in ImageJ. To account for inter-array differ- care) pictures were acquired using the G:BOX imager (Syngene).
ences, all detected intensities were normalized to positive controls Analysis was performed using band intensities normalized to
within each array. The intensities were then normalized to cell GAPDH and quantified by ImageJ. Myoblast fusion index was
106 T.H. Qazi et al. / Biomaterials 140 (2017) 103e114

quantified by expressing as a percentage the number of nuclei in groups) was used. Levels of statistical significance were set at
myotubes divided by the total number of nuclei in myotubes and *p < 0.05, **p < 0.01, ***p < 0.001.
myoblasts per region of interest (ROI).
3. Results
2.12. Immunofluorescent staining
3.1. MSC culture in distinct substrate microenvironments
Cells were fixed with 4% paraformaldehyde for 10 min, per-
In addition to TCP wells, primary rat bone marrow derived MSCs
meabilized with 0.1% saponin (myotubes) or 0.1% Triton-X and
were cultured in alginate based 3D substrates. RGD modified algi-
Tween-20 (MSCs), blocked in 1% BSA-PBS containing 3% FCS, and
nate was ionically crosslinked to fabricate nanoporous hydrogels
incubated with the appropriate primary antibody (N-Cadherin:
and freeze-dried macroporous (mean pore size: 122 ± 29 mm)
1:100, cat. # H-63, Santa Cruz Biotechnology (a kind gift from Prof.
scaffolds (Fig. S1). Although both are hydrogels, we will refer to the
Sigmar Stricker); Myosin heavy chain: 1:100, cat. # MAB4470,
nanoporous and macroporous alginates as hydrogels and scaffolds,
Thermo Scientific) overnight on a shaker at 4  C. Cells were then
respectively. MSCs on TCP exhibited a typical spread morphology
incubated with an appropriate secondary antibody (for N-Cadherin:
with prominent actin filaments (Fig. 1 a). When encapsulated in
Goat anti-rabbit IgG Alexa Fluor® 546, cat. # A11035, Invitrogen; for
hydrogels, MSCs were homogenously distributed but adopted a
Myosin heavy chain: Goat anti-mouse IgG Alexa Fluor® 488, cat. #
round morphology (Fig. 1 b). On scaffolds, MSCs were observed to
A11029, Invitrogen) for 1 h at room temperature. DAPI (1:1000 of
spread on and across the interconnected pore walls where they
1 mg/mL initial concentration in PBS) was added for 10 min at room
established physical contacts with neighboring cells (Fig. 1 c).
temperature. Cells were washed with PBS, and visualized using an
Despite the changes in morphology, MSCs maintained high viability
inverted microscope (DMI6000B, Leica, Germany). Actin filaments
for at least seven days in static in vitro culture conditions in all
were stained with Alexa Fluor® 488 Phalloidin (cat. # A12379,
groups (Fig. 1 d). While TCP is known to be non-physiologically stiff,
Thermo Scientific) according to the manufacturer's instructions.
the alginate based hydrogel and scaffold substrates had similar
mean stiffness values of ~20 kPa (Fig. 1 e).
2.13. qPCR
3.2. Modifying biomaterial microenvironment alters MSCs'
For gene expression analysis, cell containing scaffolds or gels secretory profile
were mechanically homogenized in a tube containing Trizol® re-
agent (Life Technologies), and RNA was isolated according to the The conditioned media of MSCs cultured on the three substrates
manufacturer's instructions. RNA concentration was determined (hereafter referred to as: TCP-CM, Hydrogel-CM, and Scaffold-CM)
using a NanoDrop spectrophotometer. The iScript™ Reverse Tran- were characterized using cytokine arrays and ELISA kits to assess
scription Supermix was used to transcribe RNA into cDNA. SYBR whether changes in substrate microenvironment influenced MSC
Green dye was used to detect fluorescence. The amplification pro- secretion profile. Cytokine array analysis (Fig. S3) using MSCs from
file was assessed using a LightCycler® 480 (Roche, Germany). Gene four biological donors showed striking differences in the secretion
expression was quantified using the DDCt method and fold change profile depending on the culture substrate (Fig. 2 a). In general, high
was calculated using the formula 2-DDCt. Values for the genes of concentrations of almost all cytokines were detected in Scaffold-
interest were normalized to the housekeeping gene (Eef1a). The CM, whereas lower concentrations (represented by darker in-
following primers were used for qPCR analysis: tensities on the heatmap) were observed for TCP-CM and Hydrogel-

Gene Forward Primer (50 /30 ) Reverse Primer (50 /30 )

CDH1 (E-Cadherin) ACCCCCTGTTGGCGTTTTCA CATCACGGAGGTTCCTGGAAGAG


CDH2 (N-Cadherin) GGAGCCGATGAAGGAACCACA TGAAGATGCCCGTTGGAGGC
Hgf (Hepatocyte growth factor) CCCCCATGAACACAGCTTTTTG GCTTTCACCGTTGCAGGTCA
Vegfa (Vascular endothelial growth factor A) CTTCAAGCCGTCCTGTGTGC GGCTCACAGTGATTTTCTGGCT
Fgf2 (Fibroblast growth factor 2) AAGAGCGACCCACACGTCAAA CTGCCCAGTTCGTTTCAGTGC
Mmp13 (Matrix metalloproteinase 13) ACAAGCAGCTCCAAAGGCTACA GCTGGGTCACACTTCTCTGGT
Tgfb1 (Transforming growth factor beta-1) TGGACCGCAACAACGCAAT ACTCAGGCGTATCAGTGGGG
Eef1a (Elongation factor) [Housekeeping gene] CCCTGTGGAAGTTTGAGACC CTGCCCGTTCTTGGAGATAC
GAPDH (Glyceraldehyde 3-phosphate dehydrogenase) ATGGGAAGCTGGTCATCAAC GTGGTTCACACCCATCACAA

2.14. Statistical analysis CM. Some cytokines such as brain-derived neurotrophic factor
(BDNF) and matrix metalloproteinase-13 (MMP-13) were present
All values are depicted as mean ± standard deviation. Experi- in high concentrations in all CM without a dependency on culture
ments were performed with MSCs from four biological replicates, substrate. Interestingly, platelet derived growth factor (PDGF-AA)
and were repeated independently in some cases. This information and interleukin-2 (IL-2) were highly secreted only in TCP-CM,
is included in figure legends. Statistical analysis was carried out in whereas high concentrations of interleukin-13 (IL-13) was detec-
GraphPad Prism 7.0 (GraphPad Software Inc., USA). The Shapiro- ted only in Hydrogel-CM. In stark contrast, relatively higher con-
Wilk test was used to test normality. If the data was normally centrations of most cytokines were detected in Scaffold-CM. These
distributed, two-tailed student's t-test (two groups) or one-way included, among others, basic fibroblast growth factor (b-FGF),
ANOVA with Tukey's post-hoc test (>two groups) was used. If interleukin-1 (IL-1), and transforming growth factor (TGF-b).
normality could not be confirmed, the Mann Whitney U test (two ELISA kits were used to more precisely quantify the secretion of
groups) or the Kruskal-Wallis test with Tukey's post-hoc test (>two bioactive factors that are desirable during tissue regeneration
T.H. Qazi et al. / Biomaterials 140 (2017) 103e114 107

Fig. 1. Schematic depiction and corresponding morphological evaluation of MSCs cultured in different substrate microenvironments: (a) TCP well plate, (b) encapsulated in an
alginate hydrogel, and (c) seeded on a macroporous alginate scaffold. MSCs were stained with DAPI (nuclei ¼ blue) and Phalloidin (F-Actin ¼ green) [Scale bar: a-b ¼ 100 mm,
c ¼ 50 mm]. (d) MSCs maintained long-term viability on the three substrates, as determined by LIVE/DEAD staining on days 1, 3, and 7. (e) Alginate hydrogels and scaffolds showed
similar mechanical properties, as determined by micro-indentation [n ¼ 15, two-tailed student's t-test]. (For interpretation of the references to colour in this figure legend, the
reader is referred to the web version of this article.)

Fig. 2. Substrate microenvironment alters MSC paracrine factor secretion. (a) Heatmap showing differences in the secretion profile of MSCs cultured on TCP (TCP-CM), in hydrogels
(Hydrogel-CM), and on scaffolds (Scaffold-CM) [mean intensities of n ¼ 4 biological replicates]. (b) Of the three substrates, Scaffold-CM contained significantly higher concentrations
of growth factors desirable for tissue regeneration such as VEGF, FGF2, IGF, HGF, and LIF [n ¼ 4 biological replicates, one-way ANOVA with Tukey's post-hoc test].

(Fig. 2 b). Vascular endothelial growth factor (VEGF) is a potent cells secreted significantly lower amounts. Concentrations of
stimulator of angiogenesis and is known to be secreted by many cell secreted basic fibroblast growth factor (FGF2), insulin-like growth
types including MSCs. We found that MSCs on TCP and scaffolds factor (IGF), hepatocyte growth factor (HGF), and leukemia inhibi-
secreted the highest concentrations of VEGF, whereas encapsulated tory factor (LIF) were significantly higher in Scaffold-CM compared
108 T.H. Qazi et al. / Biomaterials 140 (2017) 103e114

to the other groups. To exclude the possibility of secreted factors CM was comparable to the positive control (non-conditioned
getting entrapped within the nanoporous hydrogel matrix, mRNA medium þ 5% serum), whereas Scaffold-CM inhibited myotube
expression was performed for multiple cytokines, and growth formation. Quantification of the acquired images revealed signifi-
factor concentrations were determined after hydrogel lysis. cantly lower mean myotube lengths and densities in the Scaffold-
Together, these investigations revealed that MSCs in scaffolds show CM group compared to all others (Fig. 4 b,c). These results were
a several fold higher mRNA expression of multiple cytokines confirmed by Western blot analysis (Fig. 4 d). The normalized
(Fig. S4a), and that insignificant amounts of cytokines were protein expression levels of MHC and MyoG were significantly
detected after hydrogel and scaffold lysis (Fig. S4b). These results lower in Scaffold-CM myoblasts cultures compared to all other
indicate that biomaterial microenvironment can strongly modulate groups (Fig. 4 e,f). In summary, our in vitro data provide evidence
MSC secretion, and that despite chemical and mechanical similar- that the ability of MSCs to modulate myoblasts functions via
ities, 3D macroporous scaffolds promoted significantly enhanced paracrine mechanisms may strongly depend on the biomaterial
cytokine and growth factor secretion by MSCs compared to microenvironment.
hydrogels.
3.4. N-cadherin mediated cell-cell interactions enhance MSC
3.3. Secreted factors differentially influence myoblast functions paracrine effects

The impact of altered MSC secretion was investigated in the We tested the hypothesis that enhanced cell-cell interactions
context of muscle regeneration by analyzing the behavior and facilitated by the interconnected porous structure of scaffolds
function of C2C12 myoblasts in the presence of CM from different played a causal role in the observed modulation of secretion profile
substrates. Successful skeletal muscle regeneration after trauma and paracrine effects of MSCs. Cadherins are a family of cell mem-
requires muscle progenitor cells to survive the harsh injury envi- brane proteins that promote cell-cell adhesion. E-Cadherin and N-
ronment, undergo proliferation, migrate towards the site of tissue Cadherin in particular have been implicated in the formation of
damage, and at later stages undergo myogenic differentiation to multicellular spheroids that lead to enhanced secretion by MSCs.
form contractile fibers. Therefore, in vitro assays were used to study Therefore, we first performed immunofluorescent staining on
how MSC derived paracrine factors could influence these functions hydrogel encapsulated and scaffold seeded MSCs with an anti-N-
in myoblasts. Cadherin antibody. Whereas only a proportion of isolated MSCs in
In a survival assay designed to evaluate the anti-apoptotic ef- hydrogels expressed N-Cadherin, a much more intense staining was
fects of MSC-CM, we found that Scaffold-CM promoted significantly observed for MSCs cultured in scaffolds (Fig. 5 a). This difference was
higher viability of C2C12 myoblasts in injury mimicking conditions, confirmed by performing qPCR analysis, which revealed that scaf-
compared to CM from other substrates (Fig. 3 a). In the absence of fold seeded MSCs showed an almost 10-fold higher expression of
CM, cell viability dropped to ~50%, which was the lowest among all the N-Cadherin gene CDH2 relative to encapsulated MSCs (Fig. 5 b).
tested groups. Next, we tested the ability of myoblasts to proliferate To determine if selectively blocking N-Cadherin in MSCs would alter
in serum free conditions over 48 h (Fig. 3 b). Despite the lack of their secretion profile, MSCs were treated with an N-Cadherin
serum, which negatively affected cell proliferation (control), blocking antibody (þnAb) prior to encapsulation or seeding on the
serum-free CM stimulated cell growth. After 48 h, significantly 3D substrates (Fig. S1). Conditioned media from untreated and nAb
greater cell numbers were detected in the presence of Scaffold-CM treated MSCs were collected and characterized using representative
compared to CM from TCP and hydrogel substrates. The normalized cytokine arrays. The fold difference in intensities with nAb treat-
metabolic activity was similar in all groups after 24 h, but was ment relative to untreated MSCs was quantified (Fig. 5 c). The results
significantly higher in the scaffold and hydrogel CM groups after showed that blocking N-Cadherin in scaffold seeded MSCs down-
48 h (Fig. 3 c). regulated the secretion of all cytokines on the array (red striped
A scratch wound healing assay was used to quantify the bars). However, blocking N-Cadherin in hydrogel encapsulated
migratory behavior of myoblasts exposed to CM from different MSCs led to a downregulation in a fraction of the cytokines (green
substrates. The analysis revealed that myoblasts exposed to striped bars), and fold-changes in secretion were in general, less
Scaffold-CM showed faster kinetics of scratch closure during the pronounced compared to the scaffold group.
experiment and had repopulated a significantly greater scratch area Next, we investigated the effects of CM from nAb treated MSCs
at the end of the experiment (Fig. 3 def). No differences in repo- on myoblast proliferation, migration, and differentiation. Interest-
pulated scratch area or the rates of scratch closure were observed ingly, while after 24 h a significantly lower number of proliferated
between TCP-CM and Hydrogel-CM. These readouts provide infor- cells was observed in Hydrogel-CM þ nAb compared to Hydrogel-
mation on collective cell migration. To investigate single cell CM and no difference was observed in the scaffold groups (þ/
behavior, we performed single cell tracking and analyzed mean nAb), the proliferation of myoblasts after 48 h in the presence of
migration velocity, total migration distance, displacement, and CM þ nAb dropped significantly in the scaffold group, but was not
directionality of a large number of individual myoblasts. The ana- significantly affected in the hydrogel group (Fig. 5 d). In terms of
lyses revealed that Scaffold-CM stimulated myoblasts to migrate collective cell migration, the kinetics of scratch closure and the
faster (Fig. 3 g) and farther (Fig. 3 h,i) compared to TCP-CM and repopulated scratch area were significantly reduced by N-Cadherin
Hydrogel-CM. The directionality remained unaffected by the CM blocking in the scaffold group, but remained unaffected in Hydro-
(Fig. 3 j). gel-CM þ nAb cultures (Fig. 5 eeg). Analysis of single cell tracking
Next, the influence of MSC secreted factors on myoblast fusion during the scratch migration assay revealed that N-Cadherin
and in vitro myotube formation was studied (Fig. 4). Under differ- blocking reverses the positive effects of Scaffold-CM on myoblasts'
entiation inducing conditions (low serum concentrations), myo- migration velocity and their total traversed distance to the values of
blasts fuse together to form long, multinucleated myotubes and Hydrogel-CM, which remained unaffected by nAb treatment (Fig. 5
express differentiation markers such as myosin heavy chain (MHC) h,i). The displacement was more strongly affected in Scaffold-
and myogenin (MyoG). Differentiation was first assessed by CM þ nAb, whereas directionality was adversely influenced in both
immunofluorescent staining of MHC in myoblasts cultured in CM substrate groups (Fig. 5 j,k). The presence of nAb in control media
supplemented with 5% serum (Fig. 4 a). Over the same time dura- alone did not significantly affect myoblast function (Fig. S5).
tion, myotube formation in the presence of TCP-CM and Hydrogel- Multinucleated myotube formation by C2C12 myoblasts in the
T.H. Qazi et al. / Biomaterials 140 (2017) 103e114 109

Fig. 3. Substrate microenvironment dependent MSC paracrine effects on myoblast function. (a) Myoblast survival in injury mimicking conditions [n ¼ 8, one-way ANOVA with
Tukey's post-hoc test]. Assessment of myoblast (b) proliferation, and (c) metabolic activity over 48 h [n ¼ 12, one-way ANOVA with Tukey's post-hoc test]. Migration behavior of
myoblasts was analyzed in a scratch wound healing assay; graphs show (d) repopulated scratch area after 20 h, and (e) the corresponding kinetics of scratch closure [n ¼ 4 biological
replicates, one-way ANOVA with Tukey's post-hoc test]. (f) Representative images of myoblasts migrating into empty scratch area; dashed lines indicate scratch contours at t ¼ 0.
Single cell tracking provided insights into (g) migration velocity, (h) total migrated distance, (i) displacement, and (j) directionality of myoblasts [n ¼ 68e120, Kruskal-Wallis test
with Dunn's multiple comparison test].

presence of Hydrogel-CM (þ/-nAb) was similar to the positive properties can be used to control MSC differentiation [39,40]. In a
control (differentiation media). The inhibition of myotube forma- similar manner, gaining the ability to control or enhance the
tion observed in the presence of Scaffold-CM group was apparently paracrine properties of these cells using cues presented from bio-
reversed in the presence of Scaffold-CM þ nAb, although this did materials can be a promising strategy to improve the therapeutic
not reach statistical significance (p ¼ 0.06) (Fig. 5 l,m). outcomes of MSC based cell therapies.
Together, these results provide evidence to support the hy- Although it is known that dynamic, 3D culture significantly al-
pothesis that differences in N-Cadherin mediated cell-cell in- ters the secretion profile of MSCs [41,42], the vast majority of
teractions between hydrogel encapsulated and scaffold seeded studies investigating MSC secretome have been carried out with
MSCs regulates the paracrine effects on myogenic progenitor cells. conditioned media from cells seeded on tissue culture plastic
substrates under static conditions [43]. A discrepancy may there-
fore arise between the secretion profile characterized in vitro on
4. Discussion
non-physiologically stiff 2D culture substrates and what the MSCs
may secrete in vivo. This is especially plausible because MSCs are
A significant body of work has demonstrated that biomaterial
110 T.H. Qazi et al. / Biomaterials 140 (2017) 103e114

Fig. 4. Substrate microenvironment dependent MSC paracrine effects on myoblast differentiation. (a) Representative immunofluorescent images of differentiated myoblasts stained
with DAPI (nuclei ¼ blue) and the differentiation marker Myosin heavy chain (MHC ¼ green) [Scale bar ¼ 100 mm]. Images from several regions of interest were used to determine
(b) mean myotube length [n ¼ 60, Kruskal-Wallis test with Dunn's multiple comparison test], and (c) mean myotube density [n ¼ 5, one-way ANOVA with Tukey's post-hoc test]. (d)
Western blot analysis was carried out to quantify expression of the differentiation markers MHC and Myogenin; protein loading was determined by the detection of GAPDH. Blots
were quantified using densitometry and corresponding graphs show expression of (e) MHC, and (f) Myogenin [n ¼ 3 biological replicates, one-way ANOVA with Tukey's post-hoc
test]. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

known to be responsive to environmental stimuli [44], and could Scaffold-CM where the MSCs were cultured on relatively compliant
encounter a wide spectrum of in vivo environments with different (~20 kPa) substrates. Other factors including HGF, FGF, IGF, and LIF
stiffness (e.g. fibrotic tissues), chemical composition (e.g. variation were highly secreted by scaffold seeded MSCs compared to corre-
in ECM composition), and shear stresses (e.g. after systemic de- sponding cultures on TCP and in hydrogels. These growth factors
livery). Similarly, cells may encounter unique 3D microenviron- may especially be desirable for skeletal muscle regeneration where
ments in biomaterials used for transplantation, which may affect their utility is well documented [50,51]. For example, LIF is known
their function [45,46]. In agreement with this line of thought, our for its role in maintaining the pluripotency of embryonic stem cells,
results show a remarkable variation between the secretion profile but has also been implicated in the modulation of myoblast pro-
of TCP cultured and 3D substrate cultured MSCs. liferation and differentiation [52]. Similarly, IGF is a potent
Previous studies carried out in 3D substrates report that the myogenic factor that stimulates satellite cells, promotes their
secretion behavior of MSCs can vary substantially with changes in myogenic differentiation, increases muscle mass and strength, and
the substrate's chemical (conjugated peptides, material composi- has been implicated in the timely onset of the anti-inflammatory
tion) and mechanical (stiffness) properties [47e49]. Therefore in phase after injury [53]. Although we confirmed that secreted sol-
this study, similar ionic crosslinker concentrations were used with uble factors were not entrapped inside hydrogels in significant
a common batch of RGD modified alginate to yield structurally concentrations (Fig. S4), the diffusivity of exosomes or other cell-
different 3D substrates with similar chemical and mechanical derived vesicles (in the nm size range) through the nanoporous
properties. Although the freezing and subsequent lyophilization hydrogels has not been investigated, and may potentially be
processes employed to produce macroporous scaffolds may lead to affected by the pore size of the encapsulating hydrogel. However,
a variability of local stiffness values (Fig. S2), our data indicate that this seems not to have influenced the paracrine effects of MSCs on
this did not lead to significant differences compared to hydrogels. myoblast function, as TCP-CM and Hydrogel-CM induce similar
High concentrations of secreted VEGF were detected in TCP-CM, migratory, proliferative, and differentiation behavior in C2C12
which is an expected outcome as some studies have correlated myoblasts. Diffusion of cell derived vesicles through matrices of
VEGF secretion with increased substrate stiffness [46]. However, various pore sizes could be a point of investigation in future studies.
comparably high concentrations of VEGF were also detected in The secreted factors are likely to synergistically contribute
T.H. Qazi et al. / Biomaterials 140 (2017) 103e114 111

Fig. 5. The expression of cell-cell adhesion protein N-Cadherin by MSCs is regulated by 3D substrate microenvironment and plays a key role in MSC paracrine effects on myoblasts.
(a) Representative immunofluorescent images of hydrogel-encapsulated and scaffold-seeded MSCs stained with DAPI (nuclei ¼ blue) and anti-N Cadherin antibody (N-
cadherin ¼ red) [Scale bar: 50 mm]. (b) MSCs in scaffolds showed a significantly higher expression of the N-cadherin gene CDH2, as determined by qPCR [n ¼ 3, two-tailed student's
t-test]. (c) Functional blocking of N-Cadherin in MSCs using a neutralization antibody (nAb) adversely affected the secretion profile of MSCs [CM pooled from n ¼ 3 biological
replicates]. Paracrine effects of nAb-treated and untreated MSCs on myoblast function were compared using migration, proliferation, and differentiation assays. (d) Myoblast
proliferation in the presence of CM from 3D cultured MSCs with (striped bars) or without (solid bars) N-cadherin blocking [n ¼ 4, one-way ANOVA with Tukey's post-hoc test]. (eek)
Myoblast migration was negatively influenced by N-Cadherin blocking of scaffold-seeded MSCs (Scaffold-CM vs. Scaffold-CM þ nAb) but remained unaffected by N-Cadheirn
blocking of hydrogel-encapsulated MSCs (Hydrogel-CM vs. Hydrogel-CM þ nAb). (eeg) Repopulated scratch area and corresponding kinetics of scratch closure [n ¼ 4, one-way
ANOVA with Tukey's post-hoc test]. (hek) Single cell tracking readouts [n ¼ 90e120, one-way ANOVA with Dunn's post-hoc test]. The effects of CM from anti-N cadherin anti-
body treated MSCs on myoblast differentiation was assessed by quantifying myoblast fusion index. (l) Representative immunofluorescent images of multinucleated myotubes
stained with myosin heavy chain (MHC) and DAPI [Scale bar: 100 mm], and (m) the corresponding fusion index values [n ¼ 8, one-way ANOVA with Tukey's post-hoc test]. (For
interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

towards tissue repair and regeneration by modulating the function associated with the long-term ex-vivo culture of satellite cells [56].
of endogenous progenitor cells such as satellite cells [54,55]. In this Moreover, because they are widely used in basic research, C2C12
study, we used C2C12 myoblasts as a representative myogenic myoblasts allow us to compare observations and results with those
progenitor cell primarily due to the unresolved challenges reported in literature [57,58]. Successful skeletal muscle
112 T.H. Qazi et al. / Biomaterials 140 (2017) 103e114

regeneration is characterized by efficient migration, proliferation, morphology does not seem to be required for MSC differentiation
and fusion of satellite cells and their myoblast progenitors to form [70,71], our results show that hydrogels that prevent cell-cell in-
new muscle fibers [59,60]. Our results indicate that MSCs can have teractions and/or changes in morphology leading to cell-cell in-
wide-ranging paracrine effects on various aspects of myoblast teractions may alter or suppress the paracrine properties of MSCs,
function. Compared to other CM groups, Scaffold-CM stimulated indicating that this may be a potentially important variable to
greater proliferation, survival, metabolic activity, and migration of examine in different MSC-based applications. This does not imply
myoblasts. Fusion of individual progenitor myoblasts into elon- that hydrogels in general would suppress the therapeutic potential
gated, multinucleated myotubes marks the beginning of a process of MSCs. Optimization of mechanical and physicochemical prop-
that leads to the formation of new myofibers [61]. Factors such as erties of the encapsulating biomaterial may alter MSC function.
FGF, HGF, and VEGF have been implicated in the regulation of Some work in this area was recently described by Cai et al. who
myoblast proliferation and terminal differentiation [62]. Our showed that engineered hydrogels with a stiffness range of
observation that Scaffold-CM inhibited myotube formation may 200e400 Pa enhance the paracrine secretion of pro-angiogenic
likely be due to the presence of high concentrations of several factors from adipose derived MSCs which were able to spread out
growth factors that regulate myoblast cell-cycle withdrawal [63]. In and proliferate within the gel, possibly also leading to cell-cell
the wake of muscle injury, the primary requirements for regener- contacts [72]. Chaudhuri et al. reported that tuning the viscoelas-
ation involve survival of myoblasts, their migration and sufficient ticity of alginate hydrogels promoted dramatic changes in encap-
proliferation at the site of injury. Once an adequate number of cells sulated cell morphology and proliferation behavior which
are present, differentiation is initiated by the fusion of myoblasts regulated stem cell fate [73]. Similarly, Follin et al. encapsulated
which leads to the formation of contractile myofibers. Accordingly, adipose derived MSCs in alginates with very low molecular weights
previous studies have reported that delayed differentiation in that permitted cell spreading, and led to increased paracrine ac-
conjunction with increased proliferation and migration of myogenic tivity [74]. Together, these examples indicate that cell-cell in-
progenitor cells leads to in vivo muscle regeneration [64]. teractions, and morphological changes that may lead to them, could
We propose that the strong paracrine effects of scaffold seeded be promoted in engineered hydrogel systems while also retaining
MSCs may be attributed to cell-cell interactions that are supported their injectability.
by the macroporous structure of the scaffold, but inhibited by the Taken together, the results described in this study provide
nanoporous structure of the hydrogel (Fig. S6). In mesenchymal strong support for the use of macroporous scaffolds to promote
cells, cell-cell interactions via cadherins are developmentally rele- cell-cell interactions and thereby enhance the paracrine function of
vant because their condensation leads to the onset of endochondral transplanted MSCs. While previously, most work on cell-cell in-
ossification [65]. In a biomaterials based regeneration context, Bian teractions and cytokine secretion has utilized self-organizing
and colleagues have shown that recapitulating mesenchymal spheroid cultures that require minimal matrix contact, our work
condensation in hydrogel encapsulated MSCs by using matrix opens new avenues for exploring the synergistic effects of cell-cell
presented N-cadherin mimicking peptide fragments can enhance and cell-matrix interactions. Future studies could potentially
chondrogenesis [66]. Further, they demonstrated that blocking N- investigate the influence of various physical properties of the
cadherin with an antibody similar to the one used in this study biomaterial matrix on the ability of MSCs to form cell-cell contacts.
abrogated the improved chondrogenic effects. In terms of paracrine Additionally, the impact of cell-cell interactions on the immuno-
effects, researchers have reported cadherin mediated spheroid/ modulatory function of MSCs should be investigated before vali-
aggregate formation that show enhanced secretion of several cy- dating these in vitro findings in a relevant in vivo tissue injury
tokines and growth factors [67]. Although we did not observe model.
spontaneous spheroid formation in scaffolds seeded with MSCs,
physical interaction between the cells was qualitatively evident. 5. Conclusions
Using qPCR and immunofluorescence staining, we confirmed that
scaffold seeded MSCs showed a higher expression of N-cadherin. Paracrine mechanisms allow MSCs to orchestrate a regenerative
Next, we asked whether blocking N-cadherin would negatively response by instructing desirable functions in progenitor cell
affect the secretion profile of scaffold seeded MSCs, and whether populations. That the ability of MSCs to accomplish this effect can
the paracrine effects on myoblasts would only be as effective as be substantially improved using biomaterial properties such as
those of Hydrogel-CM. Indeed, using a neutralization antibody led porosity is an exciting avenue to improve the outcomes of MSC
to a downregulation of all detected cytokines in scaffold seeded based cell therapies. The work reported here provides new insights
MSCs, but the effects were not as strongly apparent for hydrogel into the role of a biomaterial's structural microenvironment in
encapsulated MSCs. The substantially better paracrine effects of promoting cell-cell interactions and thereby enhancing the para-
Scaffold-CM on myoblast migration were abrogated to levels that crine effects of MSCs. These findings are likely to be relevant in
were comparable to Hydrogel-CM. It is unclear why myoblast designing biomaterial based stem cell therapies for a wide range of
proliferation was reduced in Hydrogel-CM þ nAb, but we speculate tissue engineering applications.
that this might be due to a differential modulation in the secretion
of factors that are primary stimulators of proliferation and migra- Acknowledgements
tion. Furthermore, an insignificant yet apparent reversal of the in-
hibition of myotube formation was observed in Scaffold-CM þ nAb. The authors acknowledge Dag Wulsten for assistance with
Whether the differences in the ability of the two CM groups to nanoindentation experiments. This work was supported by grants
support (Hydrogel-CM) or inhibit (Scaffold-CM) myogenic differ- from the BCRT and BSRT through funding by the German Federal
entiation and myotube formation has a tangible impact on the Ministry of Education and Research (BMBF) and the German
regeneration of severely injured skeletal muscle should be a point Research Foundation, respectively. T.H.Q acknowledges funding
of investigation in future studies. from the Friede Springer Stiftung and the BSRT. D.J.M was sup-
Cell encapsulating hydrogels have been suggested to act as a ported by the Einstein fellowship award. G.N.D and S.G were
protective barrier against the potentially harmful effects of in- partially supported by the BMBF (DIMEOS, 01EC1402B) and the
flammatory cytokines that transplanted cells may encounter at the DFG Research Unit (FOR 2165; grant GE2512/2-1). The funders had
site of injury [68,69]. While a change in encapsulated cell no role in study design, data collection and analysis, decision to
T.H. Qazi et al. / Biomaterials 140 (2017) 103e114 113

publish, or preparation of the manuscript. Mesenchymal stem cell injection after myocardial infarction improves
myocardial compliance, Am. J. Physiol. Heart Circ. Physiol. 290 (2006)
H2196eH2203.
Appendix A. Supplementary data [25] Y. Fan, M. Maley, M. Beilharz, M. Grounds, Rapid death of injected myoblasts
in myoblast transfer therapy, Muscle & Nerve 19 (1996) 853e860.
[26] U.M. Fischer, M.T. Harting, F. Jimenez, W.O. Monzon-Posadas, H. Xue,
Supplementary data related to this article can be found at http:// S.I. Savitz, et al., Pulmonary passage is a major obstacle for intravenous stem
dx.doi.org/10.1016/j.biomaterials.2017.06.019. cell delivery: the pulmonary first-pass effect, Stem Cells Dev. 18 (2009)
683e692.
[27] S. Lee, E. Choi, M.-J. Cha, K.-C. Hwang, Cell adhesion and long-term survival of
References transplanted mesenchymal stem cells: a prerequisite for cell therapy,
Oxidative Med. Cell. Longev. 2015 (2015) 9.
[1] A.F. Steinert, L. Rackwitz, F. Gilbert, U. Noth, R.S. Tuan, Concise review: the [28] C.A. Cezar, D.J. Mooney, Biomaterial-based delivery for skeletal muscle repair,
clinical application of mesenchymal stem cells for musculoskeletal regener- Adv. drug Deliv. Rev. 84 (2015) 188e197.
ation: current status and perspectives, Stem Cells Transl. Med. 1 (2012) [29] S.J. Bidarra, C.C. Barrias, P.L. Granja, Injectable alginate hydrogels for cell de-
237e247. livery in tissue engineering, Acta biomater. 10 (2014) 1646e1662.
[2] T.H. Qazi, D.J. Mooney, M. Pumberger, S. Geißler, G.N. Duda, Biomaterials [30] L.M. Marquardt, S.C. Heilshorn, Design of injectable materials to improve stem
based strategies for skeletal muscle tissue engineering: existing technologies cell transplantation, Curr. Stem Cell Rep. 2 (2016) 207e220.
and future trends, Biomaterials 53 (2015) 502e521. [31] J.A. Hunt, R. Chen, T. van Veen, N. Bryan, Hydrogels for tissue engineering and
[3] J. Bashir, A. Sherman, H. Lee, L. Kaplan, J.M. Hare, Mesenchymal stem cell regenerative medicine, J. Mater. Chem. B 2 (2014) 5319e5338.
therapies in the treatment of musculoskeletal diseases, PM R J. Inj. Funct. [32] N. Annabi, J.W. Nichol, X. Zhong, C. Ji, S. Koshy, A. Khademhosseini, et al.,
Rehabilation 6 (2014) 61e69. Controlling the porosity and microarchitecture of hydrogels for tissue engi-
[4] A. Trounson, C. McDonald, Stem cell therapies in clinical trials: progress and neering, Tissue Eng. Part B Rev. 16 (2010) 371e383.
challenges, Cell Stem Cell. 17 (2015) 11e22. [33] T. Boontheekul, H.-J. Kong, D.J. Mooney, Controlling alginate gel degradation
[5] M.F. Pittenger, A.M. Mackay, S.C. Beck, R.K. Jaiswal, R. Douglas, J.D. Mosca, et utilizing partial oxidation and bimodal molecular weight distribution, Bio-
al., Multilineage potential of adult human mesenchymal stem cells, Science materials 26 (2005) 2455e2465.
284 (1999) 143e147. [34] A. Leal-Egana, U.D. Braumann, A. Diaz-Cuenca, M. Nowicki, A. Bader, Deter-
[6] A.I. Caplan, D. Correa, The MSC: an injury drugstore, Cell Stem Cell. 9 (2011) mination of pore size distribution at the cell-hydrogel interface,
11e15. J. Nanobiotechnol. 9 (2011) 24.
[7] P.R. Baraniak, T.C. McDevitt, Stem cell paracrine actions and tissue regenera- [35] J.A. Rowley, G. Madlambayan, D.J. Mooney, Alginate hydrogels as synthetic
tion, Regen. Med. 5 (2010) 121e143. extracellular matrix materials, Biomaterials 20 (1999) 45e53.
[8] S.H. Ranganath, O. Levy, M.S. Inamdar, J.M. Karp, Harnessing the mesenchymal [36] B.D. Cosgrove, K.L. Mui, T.P. Driscoll, S.R. Caliari, K.D. Mehta, R.K. Assoian, et al.,
stem cell secretome for the treatment of cardiovascular disease, Cell Stem Cell. N-cadherin adhesive interactions modulate matrix mechanosensing and fate
10 (2012) 244e258. commitment of mesenchymal stem cells, Nat. Mater. 15 (2016) 1297e1306.
[9] A.J. Salgado, R.L. Reis, N.J. Sousa, J.M. Gimble, Adipose tissue derived stem cells [37] E.J. Lee, E.K. Choi, S.K. Kang, G.H. Kim, J.Y. Park, H.J. Kang, et al., N-cadherin
secretome: soluble factors and their roles in regenerative medicine, Curr. determines individual variations in the therapeutic efficacy of human um-
Stem Cell Res. Ther. 5 (2010) 103e110. bilical cord blood-derived mesenchymal stem cells in a rat model of
[10] F.G. Teixeira, M.M. Carvalho, N. Sousa, A.J. Salgado, Mesenchymal stem cells myocardial infarction, Mol. Ther. J. Am. Soc. Gene Ther. 20 (2012) 155e167.
secretome: a new paradigm for central nervous system regeneration? Cell. [38] T. Geback, M.M. Schulz, P. Koumoutsakos, M. Detmar, TScratch: a novel and
Mol. Life Sci. 70 (2013) 3871e3882. simple software tool for automated analysis of monolayer wound healing
[11] S. Meirelles Lda, A.M. Fontes, D.T. Covas, A.I. Caplan, Mechanisms involved in assays, BioTechniques 46 (2009) 265e274.
the therapeutic properties of mesenchymal stem cells, Cytokine Growth [39] S.W. Lane, D.A. Williams, F.M. Watt, Modulating the stem cell niche for tissue
Factor Rev. 20 (2009) 419e427. regeneration, Nat. Biotechnol. 32 (2014) 795e803.
[12] H.M. Wobma, D. Liu, G. Vunjak-Novakovic, Paracrine Effects of Mesenchymal [40] H.J. Anderson, J.K. Sahoo, R.V. Ulijn, M.J. Dalby, Mesenchymal stem cell fate:
Stromal Cells Cultured in Three-dimensional Settings on Tissue Repair, ACS applying biomaterials for control of stem cell behavior, Front. Bioeng. Bio-
Biomaterials Science & Engineering, 2017. technol. 4 (2016) 38.
[13] C.P. Chang, C.C. Chio, C.U. Cheong, C.M. Chao, B.C. Cheng, M.T. Lin, Hypoxic [41] F.G. Teixeira, K.M. Panchalingam, R. Assuncao-Silva, S.C. Serra, B. Mendes-
preconditioning enhances the therapeutic potential of the secretome from Pinheiro, P. Patricio, et al., Modulation of the mesenchymal stem cell secre-
cultured human mesenchymal stem cells in experimental traumatic brain tome using computer-controlled bioreactors: impact on neuronal cell prolif-
injury, Clin. Sci. (London, England : 1979) 124 (2013) 165e176. eration, survival and differentiation, Sci. Rep. 6 (2016) 27791.
[14] A. Saparov, V. Ogay, T. Nurgozhin, M. Jumabay, W.C.W. Chen, Preconditioning [42] J.D. Glaeser, S. Geissler, A. Ode, C.J. Schipp, G. Matziolis, W.R. Taylor, et al.,
of human mesenchymal stem cells to enhance their regulation of the immune Modulation of matrix metalloprotease-2 levels by mechanical loading of
response, Stem Cells Int. 2016 (2016) 10. three-dimensional mesenchymal stem cell constructs: impact on in vitro tube
[15] M. Pumberger, T.H. Qazi, M.C. Ehrentraut, M. Textor, J. Kueper, G. Stoltenburg- formation, Tissue Eng. Part A 16 (2010) 3139e3148.
Didinger, et al., Synthetic niche to modulate regenerative potential of MSCs [43] T. Schinkothe, W. Bloch, A. Schmidt, In vitro secreting profile of human
and enhance skeletal muscle regeneration, Biomaterials 99 (2016) 95e108. mesenchymal stem cells, Stem Cells Dev. 17 (2008) 199e206.
[16] E.J. Lee, S.J. Park, S.K. Kang, G.-H. Kim, H.-J. Kang, S.-W. Lee, et al., Spherical [44] M.B. Murphy, K. Moncivais, A.I. Caplan, Mesenchymal stem cells: environ-
bullet formation via e-cadherin promotes therapeutic potency of mesen- mentally responsive therapeutics for regenerative medicine, Exp. Mol. Med.
chymal stem cells derived from human umbilical cord blood for myocardial 45 (2013) e54.
infarction, Mol. Ther. 20 (2012) 1424e1433. [45] C. Fischbach, H.J. Kong, S.X. Hsiong, M.B. Evangelista, W. Yuen, D.J. Mooney,
[17] C.L. Rettinger, A.B. Fourcaudot, S.J. Hong, T.A. Mustoe, R.G. Hale, K.P. Leung, Cancer cell angiogenic capability is regulated by 3D culture and integrin
In vitro characterization of scaffold-free three-dimensional mesenchymal engagement, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 399e404.
stem cell aggregates, Cell Tissue Res. 358 (2014) 395e405. [46] A.A. Abdeen, J.B. Weiss, J. Lee, K.A. Kilian, Matrix composition and mechanics
[18] J.M. Santos, S.P. Camoes, E. Filipe, M. Cipriano, R.N. Barcia, M. Filipe, et al., direct proangiogenic signaling from mesenchymal stem cells, Tissue Eng. Part
Three-dimensional spheroid cell culture of umbilical cord tissue-derived A 20 (2014) 2737e2745.
mesenchymal stromal cells leads to enhanced paracrine induction of wound [47] R.A. Marklein, D.E. Soranno, J.A. Burdick, Magnitude and presentation of
healing, Stem Cell Res. Ther. 6 (2015) 90. mechanical signals influence adult stem cell behavior in 3-dimensional
[19] S.S. Ho, K.C. Murphy, B.Y. Binder, C.B. Vissers, J.K. Leach, Increased survival and macroporous hydrogels, Soft Matter 8 (2012) 8113e8120.
function of mesenchymal stem cell spheroids entrapped in instructive algi- [48] D. Thomas, G. Fontana, X. Chen, C. Sanz-Nogue s, D.I. Zeugolis, P. Dockery, et
nate hydrogels, Stem Cells Transl. Med. 5 (2016) 773e781. al., A shape-controlled tuneable microgel platform to modulate angiogenic
[20] M. Osugi, W. Katagiri, R. Yoshimi, T. Inukai, H. Hibi, M. Ueda, Conditioned paracrine responses in stem cells, Biomaterials 35 (2014) 8757e8766.
media from mesenchymal stem cells enhanced bone regeneration in rat cal- [49] S. Jose, M.L. Hughbanks, B.Y. Binder, G.C. Ingavle, J.K. Leach, Enhanced trophic
varial bone defects, Tissue Eng. Part A 18 (2012) 1479e1489. factor secretion by mesenchymal stem/stromal cells with Glycine-Histidine-
[21] D. Cantinieaux, R. Quertainmont, S. Blacher, L. Rossi, T. Wanet, A. Noel, et al., Lysine (GHK)-modified alginate hydrogels, Acta biomater. 10 (2014)
Conditioned medium from bone marrow-derived mesenchymal stem cells 1955e1964.
improves recovery after spinal cord injury in rats: an original strategy to avoid [50] C. Borselli, H. Storrie, F. Benesch-Lee, D. Shvartsman, C. Cezar, J.W. Lichtman,
cell transplantation, PLoS One 8 (2013) e69515. et al., Functional muscle regeneration with combined delivery of angiogenesis
[22] L. Timmers, S.K. Lim, I.E. Hoefer, F. Arslan, R.C. Lai, A.A.M. van Oorschot, et al., and myogenesis factors, Proc. Natl. Acad. Sci. 107 (2010) 3287e3292.
Human mesenchymal stem cell-conditioned medium improves cardiac [51] J. Menetrey, C. Kasemkijwattana, C.S. Day, P. Bosch, M. Vogt, F.H. Fu, et al.,
function following myocardial infarction, Stem Cell Res. 6 (2011) 206e214. Growth factors improve muscle healing in vivo, British Volume, J. Bone Jt.
[23] F. Wang, T. Yasuhara, T. Shingo, M. Kameda, N. Tajiri, W.J. Yuan, et al., Intra- Surg. 82-B (2000) 131e137.
venous administration of mesenchymal stem cells exerts therapeutic effects [52] L.C. Hunt, J. White, The role of leukemia inhibitory factor receptor signaling in
on parkinsonian model of rats: focusing on neuroprotective effects of stromal skeletal muscle growth, injury and disease, Adv. Exp. Med. Biol. 900 (2016)
cell-derived factor-1alpha, BMC Neurosci. 11 (2010) 52. 45e59.
[24] M.F. Berry, A.J. Engler, Y.J. Woo, T.J. Pirolli, L.T. Bish, V. Jayasankar, et al., [53] Y.H. Song, J.L. Song, P. Delafontaine, M.P. Godard, The therapeutic potential of
114 T.H. Qazi et al. / Biomaterials 140 (2017) 103e114

IGF-I in skeletal muscle repair, Trends Endocrinol. Metabol. 24 (2013) Mol. Ther. 20 (2012) 2168e2179.
310e319. [65] F. Long, D.M. Ornitz, Development of the endochondral skeleton, Cold Spring
[54] Fry CS, Kirby TJ, Kosmac K, McCarthy JJ, Peterson CA. Myogenic progenitor Harb. Perspect. Biol. (2013) 5.
cells control extracellular matrix production by fibroblasts during skeletal [66] L. Bian, M. Guvendiren, R.L. Mauck, J.A. Burdick, Hydrogels that mimic
muscle hypertrophy. Cell Stem Cell.20:56e69. developmentally relevant matrix and N-cadherin interactions enhance MSC
[55] S. Hauerslev, J. Vissing, T.O. Krag, Muscle atrophy reversed by growth factor chondrogenesis, Proc. Natl. Acad. Sci. 110 (2013) 10117e10122.
activation of satellite cells in a mouse muscle atrophy model, PLoS One 9 [67] T.J. Bartosh, J.H. Ylostalo, A. Mohammadipoor, N. Bazhanov, K. Coble,
(2014), e100594. K. Claypool, et al., Aggregation of human mesenchymal stromal cells (MSCs)
[56] B.D. Cosgrove, A. Sacco, P.M. Gilbert, H.M. Blau, A home away from home: into 3D spheroids enhances their antiinflammatory properties, Proc. Natl.
challenges and opportunities in engineering in vitro muscle satellite cell Acad. Sci. U. S. A. 107 (2010) 13724e13729.
niches, Differ. Res. Biol. Divers. 78 (2009) 185e194. [68] A. Moshaverinia, C. Chen, X. Xu, S. Ansari, H.H. Zadeh, S.R. Schricker, et al.,
[57] A.J. Engler, M.A. Griffin, S. Sen, C.G. Bo €nnemann, H.L. Sweeney, D.E. Discher, Regulation of the stem cellehost immune system interplay using hydrogel
Myotubes differentiate optimally on substrates with tissue-like stiffness, coencapsulation system with an anti-inflammatory drug, Adv. Funct. Mater.
Pathol. Implic. Soft Stiff Microenviron. 166 (2004) 877e887. 25 (2015) 2296e2307.
[58] C. Sassoli, A. Pini, F. Chellini, B. Mazzanti, S. Nistri, D. Nosi, et al., Bone marrow [69] A. Burdick Jason, L. Mauck Robert, S. Gerecht, To serve and protect: hydrogels
mesenchymal stromal cells stimulate skeletal myoblast proliferation through to improve stem cell-based therapies, Cell Stem Cell. 18 (2016) 13e15.
the paracrine release of VEGF, PLoS One 7 (2012), e37512. [70] N. Huebsch, P.R. Arany, A.S. Mao, D. Shvartsman, O.A. Ali, S.A. Bencherif, et al.,
[59] C.A. Griffin, K.A. Kafadar, G.K. Pavlath, MOR23 promotes muscle regeneration Harnessing traction-mediated manipulation of the cell/matrix interface to
and regulates cell adhesion and migration, Dev. Cell. 17 (2009) 649e661. control stem-cell fate, Nat. Mater. 9 (2010) 518e526.
[60] F. Alwes, C. Enjolras, M. Averof, Live imaging reveals the progenitors and cell [71] S. Khetan, M. Guvendiren, W.R. Legant, D.M. Cohen, C.S. Chen, J.A. Burdick,
dynamics of limb regeneration, eLife 5 (2016), e19766. Degradation-mediated cellular traction directs stem cell fate in covalently
[61] S.M. Abmayr, G.K. Pavlath, Myoblast fusion: lessons from flies and mice, crosslinked three-dimensional hydrogels, Nat. Mater. 12 (2013) 458e465.
Development 139 (2012) 641e656. [72] L. Cai, R.E. Dewi, A.B. Goldstone, J.E. Cohen, A.N. Steele, Y.J. Woo, et al.,
[62] T.A. Linkhart, C.H. Clegg, S.D. Hauschka, Control of mouse myoblast commit- Regulating stem cell secretome using injectable hydrogels with in situ
ment to terminal differentiation by mitogens, J. Supramol. Struct. 14 (1980) network formation, Adv. Healthc. Mater. 5 (2016) 2758e2764.
483e498. [73] O. Chaudhuri, L. Gu, D. Klumpers, M. Darnell, S.A. Bencherif, J.C. Weaver, et al.,
[63] C.H. Clegg, T.A. Linkhart, B.B. Olwin, S.D. Hauschka, Growth factor control of Hydrogels with tunable stress relaxation regulate stem cell fate and activity,
skeletal muscle differentiation: commitment to terminal differentiation oc- Nat. Mater. 15 (2016) 326e334.
curs in G1 phase and is repressed by fibroblast growth factor, J. Cell Biol. 105 [74] B. Follin, M. Juhl, S. Cohen, A.E. Pedersen, M. Gad, J. Kastrup, et al., Human
(1987) 949e956. adipose-derived stromal cells in a clinically applicable injectable alginate
[64] M. Bencze, E. Negroni, D. Vallese, H. Yacoub-Youssef, S. Chaouch, A. Wolff, et hydrogel: phenotypic and immunomodulatory evaluation, Cytotherapy 17
al., Proinflammatory macrophages enhance the regenerative capacity of hu- (2015) 1104e1118.
man myoblasts by modifying their kinetics of proliferation and differentiation,

You might also like