You are on page 1of 10

Biomaterials 164 (2018) 70e79

Contents lists available at ScienceDirect

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

Engineering functional and histological regeneration of vascularized


skeletal muscle
Jordana Gilbert-Honick a, b, Shama R. Iyer e, Sarah M. Somers a, b, Richard M. Lovering e,
Kathryn Wagner f, g, h, Hai-Quan Mao a, b, c, d, Warren L. Grayson a, b, c, d, *
a
Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
b
Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
c
Department of Materials Science & Engineering, Johns Hopkins University School of Engineering, Baltimore, MD, 21218, USA
d
Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
e
Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
f
The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, 21205, USA
g
Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
h
Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA

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

Article history: Tissue engineering strategies to treat patients with volumetric muscle loss (VML) aim to recover the
Received 30 November 2017 structure and contractile function of lost muscle tissue. Here, we assessed the capacity of novel elec-
Received in revised form trospun fibrin hydrogel scaffolds seeded with murine myoblasts to regenerate the structure and function
1 February 2018
of damaged muscle within VML defects to the mouse tibialis anterior muscle. The electrospun fibrin
Accepted 2 February 2018
Available online 20 February 2018
scaffolds provide pro-myogenic alignment and stiffness cues, myomimetic hierarchical structure,
suturability, and scale-up capabilities. Myoblast-seeded scaffolds enabled remarkable muscle regenera-
tion with high myofiber and vascular densities after 2 and 4 weeks, mimicking that of native skeletal
Keywords:
Volumetric muscle loss
muscle, while acellular scaffolds lacked muscle regeneration. Both myoblast-seeded and acellular scaf-
Vascularized skeletal muscle folds fully recovered muscle contractile function to uninjured values after 2 and 4 weeks. Electrospun
Electrospun hydrogels scaffolds pre-vascularized with co-cultured human endothelial cells and human adipose-derived stem
Tissue engineering cells implanted into VML defects for 2 weeks anastomosed with host vasculature and were perfused with
host red blood cells. These data demonstrate the significant potential of electrospun fibrin scaffolds
seeded with myoblasts to fully regenerate the structure and function of volumetric muscle defects and
these scaffolds offer a promising treatment option for patients with VML.
© 2018 Elsevier Ltd. All rights reserved.

1. Introduction decrease positive patient outcomes [2]. Tissue engineered grafts


have great potential to create clinical treatment options for
Volumetric muscle loss (VML) is a defect impacting greater than regeneration of VML.
20% of an individual muscle volume overwhelming muscle's nat- Decellularized extracellular matrix (dECM), both acellular and
ural repair mechanisms and leading to chronic functional deficits cell-seeded, has been extensively investigated as a scaffold to treat
[1]. Current treatments are limited and include transfer of an VML and evaluated in preclinical [3e8] and clinical [9e11] models
autologous free muscle flap [2], muscle transposition [1], or with varied results. Despite some improvements to muscle func-
amputation and power bracing [2], all of which have major limi- tion, results with acellular dECM lack significant myofiber forma-
tations. Donor site morbidity, lack of donor tissue, and the need for tion, while cell-seeded dECM is limited by batch-to-batch
a highly skilled surgical team complicate VML treatment and variability, poor mechanical durability, and potential immunoge-
nicity. Other scaffold materials used in preclinical models to treat
VML include collagen [12], hyaluronic acid [13], fibrin [14e16],
* Corresponding author. Johns Hopkins University, Department of Biomedical poly(lactic acid)/poly(lactic-co-glycolic acid) [17], keratin [18,19],
Engineering, Translational Tissue Engineering Center, 400 North Broadway, Smith gelatin [20,21], and methacrylated gelatin [22]. Despite improve-
Building 5023, Baltimore, MD, 21231, USA. ments to muscle function, formation of regenerating myofibers,
E-mail address: wgrayson@jhmi.edu (W.L. Grayson).

https://doi.org/10.1016/j.biomaterials.2018.02.006
0142-9612/© 2018 Elsevier Ltd. All rights reserved.
J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79 71

and vascular infiltration, there has yet to be a scaffold system that seeding density of 300,000 cells/scaffold.
enables the formation and long-term survival of dense myofibers
and vasculature to mimic that of native skeletal muscle. 2.4. Whole mount immunostaining
Our group has recently demonstrated the capacity to enhance
the bioactivity and biocompatibility of naturally occurring fibrin by Samples fixed in methanol at 20  C overnight were washed
electrospinning it into microfiber bundles with alignment topog- with PBS then blocked and permeabilized for 3 h at 4  C. Scaffolds
raphy, tunable substrate stiffness, and controllable porous archi- were incubated with primary antibodies overnight at 4  C
tecture [23]. Fibrin is an ideal scaffold material because it is FDA- (135 rpm) followed by three 1 h washes. Scaffolds were then
approved, biodegradable, has tunable chemical and mechanical incubated with DyLight 488-conjugated goat anti-mouse or
properties, and has been used previously for tissue engineered DyLight 649-conjugated goat anti-rabbit (1:400; Jackson Immu-
skeletal muscle with promising results [14e16]. Alignment cues noResearch) overnight at 4  C (135 rpm) followed by three 1 h
enhance stem cell myogenic differentiation as well as myocyte washes with DAPI in the second wash (1:2000; Sigma). Samples
fusion [24,25] and substrate stiffness impacts stem cell differenti- were imaged with a confocal microscope (Zeiss).
ation and myogenesis [26,27]. The electrospun hydrogels are also
suturable and can be bundled to offer scale-up capabilities. Here we 2.5. In vitro force measurement
developed axially aligned fibrin hydrogel microfiber bundles with
an elastic modulus similar to native muscle and evaluated their Day 7 C2C12-seeded scaffolds were loaded into a custom
ability to support the development of mature, contractile skeletal bioreactor described previously [28] with one end clamped to a
muscle constructs using C2C12 myoblasts. Moreover, we assessed force sensor. Field stimulation was applied with platinum elec-
the ability of the electrospun hydrogels with and without C2C12s to trodes every 5 s at 5e7 V and 50e100 Hz and the resulting force of
treat VML in a murine model in which 30e50% of the tibialis contraction was measured with the force sensor.
anterior (TA) muscle was removed. We tested the hypothesis that
the combination of C2C12s on electrospun hydrogels promotes 2.6. VML defect model
muscle regeneration and functional recovery compared to scaffold
alone or no repair. In addition, we tested the ability of pre- Animal and surgical procedures were approved by the Institu-
vascularized electrospun hydrogels to provide an implanted tional Animal Care and Use Committee at Johns Hopkins University
vascular source and enable eventual scale-up to larger injury School of Medicine. Bilateral VML defects in female NOD-scid
models. IL2Rgnull (NSG) immunodeficient mice (Jackson Lab) were used
for all experiments. Treatment groups per experiment are detailed
2. Materials and methods in SI. Mice were anesthetized with isofluorane, the TA muscle was
exposed, and approximately 30e50% of the muscle removed. After
2.1. Electrospinning fibrin scaffolds bleeding ceased, scaffolds were placed in the defect site and ligated
on both ends to the remaining TA muscle with non-absorbable
Scaffolds were fabricated as described previously [23]. Briefly, sutures (6-0 Nylon, Express Medical Supplies) (Fig. 2). Surgical
1% fibrinogen (Sigma) or sodium alginate (Sigma) were extruded in glue (Histoacryl, B. Braun) and Reflex wound clips (CellPoint Sci-
parallel with 3e5 kV applied to form hydrogel microfiber bundles. entific) were used to close the skin. Rimadyl was injected subcu-
Fibrinogen remained constant at 4 ml/h while alginate concentra- taneously post-surgery for pain management (5 mg/kg). Mice were
tions and extrusion rates were varied to alter scaffold properties sacrificed by isoflurane overdose and cervical dislocation. Upon
(Fig. S1E). Scaffolds with 15% alginate were utilized for experi- harvest, implanted scaffolds and surrounding TA muscle tissue
ments. Additional details are described in Supplemental were removed and cryopreserved.
Information (SI).
2.7. Histology
2.2. Characterization of scaffold pore size, porosity, and stiffness
Harvested TA muscles were weighed and flash-frozen in OCT
Scaffolds frozen in OCT (Tissue Tek) were sectioned with a with 2-methylbutane (Sigma). Samples were sectioned with a
cryostat (10 mm; Leica). Porosity was quantified using Hematoxylin cryostat (10 mm) and rehydrated in DI water before staining with
& Eosin (H&E, Sigma) stained sections with ImageJ (NIH) and H&E or Masson's Trichrome (Sigma). For immunohistochemistry,
defined as the pore areas divided by total scaffold area (n ¼ 2e6). slides were fixed in methanol or acetone at 20  C for 10 min,
Scaffolds were also loaded into a custom bioreactor and stiffness rinsed with PBS three times, then blocked for 1 h at RT. Slides were
analyzed as described previously [28]. Briefly, scaffolds were incubated with antigen-specific primary antibodies overnight at
adjusted to zero strain then stretched to 10% strain at 1% strain/s as 4  C. After three 15 min washes with PBS, slides were incubated
force sensors generated a stress-strain curve. Stress data was with secondary antibody for 1 h at RT. Slides were washed then
filtered using the Savitzky-Golay filter to remove cyclic noise from mounted with 50% glycerol and imaged on a fluorescence micro-
the motor. scope (Zeiss). Antibody concentrations are detailed in SI. Myofiber
diameters were quantified using SMASH Matlab analysis [43].
2.3. Cell culture and seeding on scaffolds
2.8. In vivo functional testing
C2C12s (ATCC) were used at passage 8. A seeding volume of
40 ml at 7500 cells/ml was pipetted onto scaffolds in 5 ml droplets In vivo assessment of mouse TA muscle contractility was per-
and incubated for 1 h at 37  C, after which C2C12 Growth Medium formed at 2 and 4 weeks as previously described [30]. Briefly, mice
with 30 mg/ml aprotinin (Affymetrix) was added. Samples were were anesthetized with isoflurane and the leg was stabilized in a
differentiated on day 3 of culture. Pre-vascularized scaffolds were custom-made apparatus. The foot was taped to a foot plate con-
seeded with a 1:1 ratio of human umbilical vein endothelial cells nected to a torque sensor and stepper motor and the ankle was
(HUVECs; Lonza) and passage 3 primary human adipose-derived positioned at 20 of plantarflexion, which has been shown to be the
stem cells (ASCs) isolated as previously described [29] for a total optimum length to achieve maximum ankle dorsiflexor torque (in
72 J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79

which the TA muscle accounts for the majority of ankle dorsi- (Fig. 1A). The optimized fibrin scaffolds contain intrinsic alignment
flexion). Electrodes were placed subcutaneously at the fibular head on the micro-scale, have a porous interior, and are suturable
to stimulate the fibular nerve and induce dorsiflexion. Voltage and (Fig. 1BeD). To control porosity and stiffness, fibrin scaffolds were
frequency were optimized for each leg during tetanic contractions electrospun containing various amounts of alginate, which served
(300 ms duration) and the maximal isometric torque was quanti- as a sacrificial polymer to induce porosity (Fig. S1). A 15% alginate
fied per leg at each time point (n ¼ 7). To also assess resistance to composition was selected for subsequent studies due to its myo-
fatigue, each TA was subjected to a series of 50 isometric tetanic mimetic stiffness similar to the 12 kPa stiffness of native skeletal
contractions (one contraction every 2 s) and the resulting drop in muscle [27] (Fig. 1E) and high internal porosity. Topographical
maximal isometric torque and recovery after 2 and 5 min were alignment cues enable alignment of a range of cell types (Fig. S2), a
quantified (n ¼ 7). vital component of functional skeletal muscle constructs that en-
ables uniaxial contraction and has been shown to contribute to
2.9. Statistics myogenesis [24,25].

Statistical analysis was performed using GraphPad Prism 5 3.2. Myoblast-seeded scaffolds form mature contractile muscle
software. Statistical significance for most experiments was deter- constructs in vitro
mined by one-way ANOVA with Dunnet post-test utilizing the
uninjured values as control. Statistical significance for myofiber and To test the suitability of electrospun fibrin scaffolds as a sub-
vascular densities was determined by one-way ANOVA with strate for mature muscle constructs, C2C12 myoblasts were seeded
Tukey's post-test. Error bars represent standard error of the mean and cultured for 7 days. C2C12s formed densely aligned myosin
(SE). *: p < 0.05; **: p < 0.01; ***: p < 0.001; ǂ: not significant. heavy chain positive (MHCþ) myotubes along the scaffolds and
exhibited multinucleation and sarcomeric striations, hallmarks of
3. Results mature muscle (Fig. 1F). In comparison, C2C12s cultured on glass
coverslips for 7 days formed disorganized myotubes lacking similar
3.1. Electrospun scaffold properties tailored for tissue engineered alignment and features. C2C12-seeded scaffolds also exhibited
skeletal muscle spontaneous contractions (Video S1) and contracted in response to
electrical stimulation (Video S2) generating approximately 1 mN of
Electrospun fibrin scaffolds were designed to mimic the hier- force (Fig. 1G). To potentially increase cell uniformity, an alternate
archical structure and native characteristics of skeletal muscle ‘spread’ scaffold structure was fabricated (Fig. S3A,B) and its

Fig. 1. Fabrication of electrospun fibrin scaffolds. (A) Schematic of hierarchical structure of electrospun scaffolds. (B) Scanning EM of microscopic scaffold structure and
alignment. (C) Longitudinal section of 15% alginate scaffold stained with H&E demonstrating internal porosity. (D) Scaffolds bundled and sutured into a muscle defect. (E)
Representative stress-strain curve. (F) (i) MHC (red) and DAPI (blue) staining of aligned, multinucleated myotubes on scaffolds at day 7 with (ii) sarcomeric striations. Inset: C2C12s
on glass substrate. (G) Representative contractile force generation in response to electrical stimulation. (For interpretation of the references to colour in this figure legend, the reader
is referred to the web version of this article.)
J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79 73

myogenic potential assessed. C2C12-seeded spread scaffolds seeded scaffolds exhibited significant volume recovery, muscle
exhibited a similar density of aligned, multinucleated myotubes mass increase, lack of fibrosis, and high myofiber density (Fig. 5). In
compared to ‘bulk’ scaffolds (Fig. S3C-E) and spontaneous con- the C2C12 treatment group, myofibers were primarily centrally-
tractions (Video S3). nucleated and began to approximate the structure of native unin-
Supplementary videos related to this article can be found at jured TA muscle.
https://doi.org/10.1016/j.biomaterials.2018.02.006. Defects treated with acellular scaffolds exhibited disorganized
laminin deposition within the scaffold interior, but had no myo-
fibers present at 2 weeks. In contrast, defects treated with C2C12s
3.3. Regeneration of murine VML defects with myoblast-seeded
had organized circular myofibers with a laminin structure similar
electrospun scaffolds
to uninjured muscle (Fig. 6A). Embryonic myosin, a transient
marker of regenerating skeletal muscle, was evident in C2C12-
An established murine VML defect model [3,7,31] was used to
treated defects at 2 weeks with expression decreasing by 4
test the regenerative potential of C2C12-seeded electrospun scaf-
weeks. Defects treated with acellular scaffolds did not express
folds. Scaffolds were bundled and sutured into the defect (Fig. 2) for
embryonic myosin at 2 weeks, suggesting that the regeneration
2 or 4 weeks. Each mouse was treated with bulk scaffolds in the
seen in C2C12-treated groups was due to the implanted cells
right leg and spread scaffolds in the left leg (n ¼ 3). Both bulk and
(Fig. 6B). Myofibers in C2C12 treatment groups stained positively
spread C2C12-seeded scaffolds resulted in significant levels of
for the slow-twitch myofiber marker BA-D5 at 4 weeks (Fig. 6C).
muscle regeneration at 2 weeks (Fig. 3C) with graft areas densely
Significantly, C2C12 treatment groups also exhibited host neuro-
populated with centrally-nucleated MHCþ myofibers and a robust,
filament within the graft area, suggesting the possibility of func-
dense capillary network. Myofiber density increased from 2 to 4
tional connectivity between the host nerve and implanted muscle
weeks for both groups, approaching that of native skeletal muscle,
constructs (Fig. 6D).
whereas vascular density remained high at both time points
To track implanted cells in C2C12-seeded scaffolds, a subset of
(Fig. 3D). Individual myofiber diameters in all groups averaged
C2C12s was transduced to express mCherry and luciferase and
~23 mm, which is roughly half the diameter of myofibers in native
seeded onto scaffolds. Transduced C2C12s formed aligned
TA muscle (Fig. 3E, S4). Treatment groups also exhibited significant
MHCþ myotubes on the scaffold surface that had similar
volume recovery of the graft areas, with total TA cross-sectional
morphology to non-transduced samples and expressed mCherry
areas in treatment groups mimicking that of native TA muscle
and luciferase (Fig. S5B-E). Bundles of 4 scaffolds containing
(Fig. 3F). As spread scaffolds did not confer any advantage in
transduced C2C12s were implanted into bilateral VML defects and
regenerative outcomes, bulk scaffolds were utilized for future
luciferase expression was quantified. Implanted C2C12s were
experiments.
retained within the defect areas, remained viable, and proliferated
in vivo up to 28 days (Fig. S5A). Vascular perfusion was observed in
3.4. Myoblast-seeded scaffolds enable muscle regeneration but uninjured muscles (Fig. S5F) and C2C12-seeded scaffolds (Fig. S5G)
acellular scaffolds do not via tail vein injection of FITC dextran and confocal imaging.

We directly compared three experimental groups (n ¼ 7): (i)


untreated defects; (ii) acellular scaffolds; or (iii) C2C12-seeded 3.5. Myoblast-seeded and acellular scaffolds both enable functional
scaffolds in the VML model to assess the role of cells and scaf- recovery
folds in regeneration. Approximately 11 ± 3 mg of muscle tissue
was resected per defect and the contralateral leg remained as an The effects of acellular and C2C12-seeded scaffolds on muscle
uninjured control. Bundles of 4 acellular or C2C12-seeded scaffolds contractile function were evaluated by measurement of maximal
were sutured into the defect site in treatment groups and the defect isometric torque of the ankle dorsiflexor muscles (Fig. S6A). Mus-
remained empty for the untreated group. Constructs remained cles containing untreated defects demonstrated ~30% torque deficit
in vivo for 4 weeks. compared to uninjured controls at 2 and 4 weeks post-injury
Untreated defects exhibited loss of muscle tissue at the defect (Fig. 6E). Interestingly, defects treated with both acellular and
site, lacked myofibers, and contained significant fibrotic tissue C2C12-seeded scaffolds demonstrated complete functional recov-
(Fig. 4AeC). Remnants of acellular scaffolds could be observed at 4 ery at both 2 and 4 weeks post-injury, with maximum isometric
weeks, but exhibited varying levels of fibrosis and lacked the torque levels equal to uninjured controls (Fig. 6E). Muscle resis-
presence of any MHC. In stark contrast, defects treated with C2C12- tance to fatigue was also tested via the loss in maximal torque after

Fig. 2. Murine VML defect and scaffold implantation model. Macroscopic depiction of VML defect creation via partial TA resection, an empty VML defect, scaffold implantation,
and morphology of the scaffolds within the defect at 4 weeks post-implantation.
74 J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79

Fig. 3. In vivo regeneration of VML defects. (A, B) Bulk and spread scaffolds. (C) MHC (green), CD31 (red), and DAPI (blue) staining of VML defects treated with C2C12-seeded
scaffolds at 2 and 4 weeks. High densities of centrally nucleated myofibers (arrows) and vasculature are present. Dotted line denotes approximate graft region. H: host muscle
tissue. (D) Myofiber and vascular densities at 2 and 4 weeks (n ¼ 18). Red: 2 weeks; Blue: 4 weeks; Green: native muscle. (E) Myofiber diameters (n ¼ 24) and (F) cross-sectional
areas (n ¼ 11) following treatment. *: p < 0.05; **: p < 0.01; ***: p < 0.001; ǂ: not significant. Error bars are SE. (For interpretation of the references to colour in this figure legend, the
reader is referred to the web version of this article.)

50 isometric contractions. No significant differences in percent fa- there was little host vessel infiltration to the scaffold interior as
tigue were present between groups at both time points (Fig. 6F) well as a complete lack of myofibers within the scaffolds
although untreated muscles were slower to recover (S6C). (Fig. 7EeF). Cross-sections of in vitro pre-vascularized and C2C12-
seeded scaffolds stained with Masson's Trichrome demonstrated
a similar difference in collagen deposition on the scaffold surface by
3.6. Pre-vascularized scaffolds anastomose to host vessels ASC:HUVEC co-cultures compared to C2C12s (Fig. 7G). To test our
hypothesis that the collagen on the scaffold surface was deposited
Although C2C12-seeded scaffolds implanted in VML defects by ASCs and prevents integration with surrounding host tissue,
exhibited a high density of vascular infiltration and myoblast sur- scaffolds seeded with ASCs were implanted into VML defects for 2
vival up to 4 weeks, scale-up to large animal models would likely weeks. ASC-seeded scaffolds had dense collagen boundaries pre-
require an implanted vessel source to meet the increased nutrient venting scaffold degradation and integration with host tissue
and oxygen demands of larger tissues. Therefore, we tested (Fig. S7), similar to those seen in implanted pre-vascularized
whether engineering scaffolds with pre-formed vascular networks scaffolds.
would expedite blood flow throughout the grafts. Electrospun
scaffolds were seeded with human umbilical vein endothelial cells
(HUVECs) and human adipose-derived stem cells (ASCs) in a 1:1 4. Discussion
ratio and cultured for 11 days to form aligned vessels along the
scaffold length (Fig. 7A). Bundles of 4 pre-vascularized scaffolds Electrospun fibrin scaffolds mimic the hierarchical structure and
were implanted into VML defects and harvested after 10 days. mechanical properties of muscle and provide a pro-regenerative
Implanted human vessels successfully anastomosed with host environment for cells. The scaffolds also have the potential to be
vessels and exhibited perfusion by host red blood cells (Fig. 7BeC). used off-the-shelf and can be stored long-term and rehydrated
Interestingly, the implanted pre-vascularized scaffolds exhibi- prior to cell seeding and implantation [23]. Prior reports indicate
ted low levels of degradation and were each surrounded by a thick that treatment with acellular scaffolds relies on muscle stem/pro-
collagen boundary (Fig. 7D). This is in contrast to C2C12-seeded genitor cell and vascular ingrowth from the remaining muscle tis-
scaffolds implanted for a similar amount of time where scaffolds sue and provides demonstrable improvements to muscle
were completely replaced by dense myofibers and host vessels, contractile function, but muscle regeneration has been limited
lacking significant collagen deposition. In addition, although pre- [7,9,10] and the need for inclusion of a myogenic cell source to
vascularized scaffolds retained the implanted human vessels, regenerate muscle tissue following VML has been demonstrated
J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79 75

Fig. 4. Evaluation of muscle morphology. (A) MHC (green) and DAPI (blue) staining at 4 weeks post-injury compared to uninjured muscle (f: acellular fibrin scaffold). Outlines
denote approximate graft region. (B) Masson's Trichrome staining demonstrating low fibrosis and integration of C2C12 samples. (C) H&E staining of cell morphology within the
defects. Arrowheads denote centrally-nucleated myofibers. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this
article.)

Fig. 5. Morphological Characteristics. (A) Mouse body and TA weights at various time points. (B) TAs containing C2C12 constructs weighed significantly more than other groups.
Values are means ± SE (n ¼ 4e5). *p < 0.05.

[6e8,19]. Yet cell-seeded scaffolds have been typically impaired by treat VML with mixed results. Murine TA VML defects treated with
fibrosis, an inability to fully recapitulate the myofiber and vascular fibrin microthreads seeded with C2C12s enabled some muscle
density present in native skeletal muscle, and poor cell viability. regeneration, vascular infiltration, and functional recovery 60 days
This study demonstrates that the pro-regenerative environment post-implantation, but regenerated myofibers were bounded by
created by electrospun scaffolds stimulates remarkable vascular significant collagen deposition and were disorganized despite
and myogenic regeneration with little to no fibrosis at the graft- microthread alignment cues [14]. Fibrin microthreads seeded with
host interface. human muscle cells and used to treat murine TA VML defects
Fibrin has been used previously in skeletal muscle constructs to resulted in high levels muscle regeneration and some collagen
76 J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79

Fig. 6. Morphologic and functional characterization of regenerating muscle. (A) Laminin (red) and DAPI (blue) staining demonstrates differences in myofiber size and
morphology between groups. (B) Embryonic myosin (red) and DAPI (blue) staining. (C) Slow-twitch myosin (green; BA-D5) and DAPI (blue) staining. (D) Neurofilament ingrowth
into the graft region of C2C12 samples at 4 weeks is visible with b-tubulin III (pink) and DAPI (blue) staining. Arrowheads show neurofilament within host muscle. Scale bars:
500 mm, inset- 50 mm. Outlines denote approximate graft region. (E) Maximal isometric torque (n ¼ 7). (F) Percent fatigue (n ¼ 7). *p < 0.05; ǂ: not significant. Error bars are SE. (For
interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

deposition within the defect at 10 weeks post-implantation, but no laminin staining. Myofibers within the defect contained thicker
vascular regeneration was shown [16]. Another study assessed the laminin boundaries and smaller myofiber diameters than native
efficacy of progressive injections of C2C12s or human muscle pro- skeletal muscle, which may explain the overall difference in myo-
genitor cells in fibrin to treat murine TA VML defects, which fiber densities. Vascular density in the grafts was higher than in
resulted in a variable density of vascularized myofiber formation at native skeletal muscle with an average coverage of 5.3% of the field
4 weeks post-implantation with high interior myofiber density but of view. The blood vessels in C2C12-treated defects were signifi-
a low density at the graft outskirts [15]. A recent study where dECM cantly larger in cross-sectional area than those seen in native
seeded with primary muscle cells was used to treat a murine TA muscle, increasing the total area of vessel coverage per field of view
VML defect resulted in high levels of vascularized muscle regen- compared to native muscle. In vivo degradation of electrospun
eration 30 days post-implantation where the number of total blood fibrin scaffolds varied depending on the cell type with which it was
vessels per field mimicked native muscle. However, the number of seeded. We have found that C2C12-seeded scaffolds completely
vessels per myofiber was around 0.2 and was not compared with degraded 2 weeks post-implantation and were replaced with dense
native skeletal muscle [3]. In addition, although myofiber density regenerating muscle and vasculature, while ASC-seeded and pre-
within the defect appeared to be high in histological images, the vascularized scaffolds maintained much of their structure after 2
value was not compared to native skeletal muscle. In the current weeks in vivo and lacked corresponding regeneration. Interestingly,
study, the electrospun fibrin scaffolds provided alignment cues and cross-sections of in vitro C2C12-seeded scaffolds exhibited
unique mechanical properties that overcame these limitations. increased porosity and swelling compared to cross-sections of pre-
The extent of muscle and vascular regeneration following VML vascularized scaffolds, which also contained a dense collagen
injury when treated with C2C12-seeded electrospun scaffolds seen boundary. It is thus likely that differences in cell-biomaterial in-
here is a significant improvement over previous treatment mo- teractions and subsequent scaffold swelling and degradation
dalities. Although regenerating myofibers had roughly half the contribute to variations in regeneration, and that rapid scaffold
diameter of native TA myofibers, the density of myofibers seen as degradation in response to C2C12s enables the robust muscle and
well as the high prevalence of perfusable vasculature within the vascular regeneration seen here.
defect was substantial. Myofiber density within the grafts reached The recovery of volume in C2C12-treated versus acellular or
68% coverage of the field of view with remaining areas composed of untreated defects is likely due to beneficial cell-matrix interactions
blood vessels and connective tissue, as demonstrated by CD31 and coupled with survival and growth of implanted cells. It is unlikely
J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79 77

Fig. 7. Pre-vascularized scaffolds in VML defect. (A) CD31 (green) and DAPI (blue) staining of vasculature on scaffolds. (B) Pre-vascularized scaffolds implanted in VML defects for
10 days contained human vessels (green) that anastomosed with host mouse vessels (red; arrow). (C) Implanted human vessels (green) were perfused with mouse red blood cells
(red; arrows). (D) Masson's Trichrome staining demonstrating differences in muscle regeneration, collagen deposition, and scaffold degradation. (E) Mouse (red) and human (green)
CD31 staining. (F) MHC (green) staining. (G) Masson's Trichrome staining of cross-sections from in vitro pre-vascularized scaffolds and C2C12-seeded scaffolds demonstrating
differences in collagen deposition, scaffold porosity, and swelling. f: fibrin scaffold. (For interpretation of the references to colour in this figure legend, the reader is referred to the
web version of this article.)

to be due to hypertrophy of the remaining host muscle in response the host. This result was unexpected, as ASCs have been utilized for
to injury as this would have been present in defects that were their pro-regenerative paracrine signaling [32e34] and as a
untreated or treated with acellular scaffolds as well. ASC-seeded myogenic cell source both in vitro [35e38] and in vivo [39e41].
scaffolds also demonstrated good volume recovery, but cell- Nevertheless, in a study where pre-vascularized collagen scaffolds
scaffold interactions differed between ASCs and C2C12 myoblasts. containing adipose-derived microvascular fragments and primary
C2C12-seeded scaffolds were completely degraded by 2 weeks myoblasts were implanted in a full-thickness VML defect in the
post-implantation, while pre-vascularized and ASC-seeded scaf- biceps femoris, there was no muscle regeneration and instead
folds were still present after 2 weeks and acellular scaffolds were significant collagen deposition within the defect, supporting our
still present after 4 weeks in vivo. Cross-sections of in vitro C2C12- results [42]. The integrative capacity of scaffolds with cultured ASCs
seeded scaffolds exhibited large internal pores and increased and the contribution of ASCs to collagen deposition within muscle
swelling compared to cross-sections of pre-vascularized scaffolds, defects requires further investigation.
demonstrating that scaffold remodeling exhibits marked differ- Interestingly, both C2C12-seeded and acellular scaffolds recov-
ences depending on cell type. In fact, an interesting result of the ered the contractile function of injured muscle at both 2 and 4
study was the apparent “insulation” of scaffolds seeded with ASCs. weeks despite the inability of acellular scaffolds to support muscle
The increased collagen deposition surrounding pre-vascularized regeneration and the vast difference in myofiber formation be-
scaffolds is unlikely due to an immune response by the host, tween C2C12-seeded and acellular treatment groups. A possible
since immunodeficient animals were used and a similar response explanation for this discrepancy could be the presence of scaffold
was not present when C2C12-seeded scaffolds were implanted. Our mediated functional fibrosis that has been recently described [7],
hypothesis that ASCs were the collagen source was supported by where the implanted scaffold provides a mechanical bridge to
experiments with ASC-seeded scaffolds without HUVECs, which prevent prolonged overload injury to the remaining muscle after
resulted in distinct collagen boundaries surrounding the scaffold as injury. Implanted acellular scaffolds are thus able to transiently
well as a complete lack of scaffold degradation or integration with augment or repair contractile function without a corresponding
78 J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79

presence of regenerating myofibers in the defect area [8]. volumetric muscle loss injury, Tissue Eng. Part A [Internet] 20 (2014)
705e715. Available from: http://online.liebertpub.com/doi/abs/10.1089/ten.
In conclusion, the current study provides a novel construct
tea.2012.0761.
system containing electrospun fibrin scaffolds that, when com- [7] A. Aurora, J.L. Roe, B.T. Corona, T.J. Walters, An acellular biologic scaffold does
bined with a myoblast cell source, enable unprecedented levels of not regenerate appreciable de novo muscle tissue in rat models of volumetric
functional muscle and vascular regeneration. The electrospun muscle loss injury, Biomaterials [Internet] 67 (2015) 393e407. Available from:
http://linkinghub.elsevier.com/retrieve/pii/S0142961215006225.
scaffolds mimic the stiffness of native muscle, contain intrinsic [8] B.T. Corona, X. Wu, C.L. Ward, J.S. McDaniel, C.R. Rathbone, T.J. Walters, The
alignment cues, are suturable, biodegradable, mechanically robust, promotion of a functional fibrosis in skeletal muscle with volumetric muscle
and are made from an FDA-approved biomaterial. Interestingly, loss injury following the transplantation of muscle-ECM, Biomaterials
[Internet] 34 (13) (2013) 3324e3335, https://doi.org/10.1016/
VML defects treated with acellular electrospun scaffolds recovered j.biomaterials.2013.01.061.
the muscle's contractile function similar to C2C12-seeded scaffolds, [9] J. Dziki, S. Badylak, M. Yabroudi, B. Sicari, F. Ambrosio, K. Stearns, et al., An
despite their lack of corresponding regenerating myofibers. The acellular biologic scaffold treatment for volumetric muscle loss: results of a
13-patient cohort study, Nat. Regen Med. [Internet] 1 (2016) 1e12. Available
electrospun scaffolds have scale-up potential to larger animal from: http://www.nature.com/articles/npjregenmed20168.
models and support the formation of implantable vasculature. [10] B.M. Sicari, J.P. Rubin, C.L. Dearth, M.T. Wolf, F. Ambrosio, M. Boninger, et al.,
An acellular biologic scaffold promotes skeletal muscle formation in mice and
humans with volumetric muscle loss, Sci. Transl. Med. [Internet] 6 (234)
Author contributions (2014), 234ra58-234ra58. Available from: http://stm.sciencemag.org/cgi/doi/
10.1126/scitranslmed.3008085.
J.G.H., R.M.L., K.W., H.Q.M., and W.L.G. designed research; J.G.H., [11] V.J. Mase, J.R. Hsu, S.E. Wolf, J.C. Wenke, D.G. Baer, J. Owens, et al., Clinical
application of an acellular biologic scaffold for surgical repair of a large,
S.R.I. and S.M.S. performed research; J.G.H, S.R.I, R.M.L. K.W., H.Q.M, traumatic quadriceps femoris muscle defect, Orthopedics [Internet] 33 (7)
and W.L.G. analyzed data; and J.G.H. and W.L.G. wrote the paper. (2010). Available from: http://www.slackinc.com/doi/resolver.asp?doi¼10.
3928/01477447-20100526-24.
[12] J. Ma, K. Holden, J. Zhu, H. Pan, Y. Li, The application of three-dimensional
Data availability collagen-scaffolds seeded with myoblasts to repair skeletal muscle defects,
J. Biomed. Biotechnol. 2011 (2011) 1e9.
The raw/processed data required to reproduce these findings [13] C.A. Rossi, M. Flaibani, B. Blaauw, M. Pozzobon, E. Figallo, C. Reggiani, et al.,
In vivo tissue engineering of functional skeletal muscle by freshly isolated
cannot be shared at this time due to technical limitations. satellite cells embedded in a photopolymerizable hydrogel, FASEB J [Internet]
25 (7) (2011) 2296e2304. Available from: http://www.fasebj.org/cgi/doi/10.
Conflicts of interest 1096/fj.10-174755.
[14] J.M. Grasman, D.M. Do, R.L. Page, G.D. Pins, Rapid release of growth factors
regenerates force output in volumetric muscle loss injuries, Biomaterials 72
The authors declare no conflict of interest. (2015) 49e60.
[15] J.H. Kim, I.K. Ko, A. Atala, J.J. Yoo, Progressive muscle cell delivery as a solution
for volumetric muscle defect repair, Sci. Rep. [Internet] 6 (1) (2016) 38754.
Acknowledgements Available from: http://www.nature.com/articles/srep38754.
[16] R.L. Page, C. Malcuit, L. Vilner, I. Vojtic, S. Shaw, E. Hedblom, et al., Restoration
Funding was provided by the Maryland Stem Cell Research Fund of skeletal muscle defects with adult human cells delivered on fibrin micro-
threads, Tissue Eng. Part A [Internet] 17 (21e22) (2011) 2629e2640. Available
(2016-MSCRFI-2692) and the Wilmer Core Grant for Vision from: http://www.ncbi.nlm.nih.gov/pubmed/21699414%5Cnhttp://online.
Research, Microscopy and Imaging Core Module (EY001765). Dr. liebertpub.com/doi/pdfplus/10.1089/ten.tea.2011.0024.
Assaf Gilad kindly provided a custom CMV-mCherry-Luciferase [17] Y. Shandalov, D. Egozi, J. Koffler, D. Dado-Rosenfeld, D. Ben-Shimol,
A. Freiman, et al., An engineered muscle flap for reconstruction of large soft
plasmid used to produce lentivirus for the study. We would like tissue defects, Proc. Natl. Acad. Sci. India 111 (16) (2014) 6010e6015. Avail-
to acknowledge Aaron Black and Alexandra Rindone for their able from: http://www.pnas.org/cgi/doi/10.1073/pnas.1402679111.
assistance in performing tail vein injections and C2C12 trans- [18] J.A. Passipieri, H.B. Baker, M. Siriwardane, M. Ellenburg, M. Vadhavkar,
J.M. Saul, et al., Keratin hydrogel enhances in vivo skeletal muscle function in
duction and bioluminescence imaging, respectively.
a rat model of volumetric muscle loss, Tissue Eng. Part A [Internet] 23 (11e12)
(2017) 556e571. Available from: http://online.liebertpub.com/doi/10.1089/
Appendix A. Supplementary data ten.TEA.2016.0458.
[19] H.B. Baker, J.A. Passipieri, M. Siriwardane, M. Ellenburg, M. Vadhavkar,
C.R. Bergman, et al., Cell and growth factor loaded keratin hydrogels for
Supplementary data related to this article can be found at treatment of volumetric muscle loss (VML) in a mouse model, Tissue Eng. Part
https://doi.org/10.1016/j.biomaterials.2018.02.006. A [Internet] 0 (0) (2017). Available from: http://online.liebertpub.com/doi/
abs/10.1089/ten.TEA.2016.0457%0A. http://online.liebertpub.com/doi/pdf/10.
1089/ten.TEA.2016.0457.
References [20] K. Hagiwara, G. Chen, N. Kawazoe, Y. Tabata, H. Komuro, Promotion of muscle
regeneration by myoblast transplantation combined with the controlled and
[1] K. Garg, C.L. Ward, B.J. Hurtgen, J.M. Wilken, D.J. Stinner, J.C. Wenke, et al., sustained release of bFGFcpr, J Tissue Eng Regen Med 10 (2016) 325e333.
Volumetric muscle Loss : persistent functional deficits beyond frank loss of [21] Y.M. Ju, A. Atala, J.J. Yoo, S.J. Lee, In situ regeneration of skeletal muscle tissue
tissue, J. Orthop. Res. 33 (1) (2015) 40e46. through host cell recruitment, Acta Biomater. [Internet] 10 (10) (2014)
[2] B.F. Grogan, J.R. Hsu, Volumetric muscle loss, J. Am. Acad. Orthop. Surg. 19 4332e4339, https://doi.org/10.1016/j.actbio.2014.06.022. Available from:.
(2011) 35e37. [22] V. Hosseini, S. Ahadian, S. Ostrovidov, G. Camci-Unal, S. Chen, H. Kaji, et al.,
[3] M. Quarta, M. Cromie, R. Chacon, J. Blonigan, V. Garcia, I. Akimenko, et al., Engineered contractile skeletal muscle tissue on a microgrooved methacry-
Bioengineered constructs combined with exercise enhance stem cell- lated gelatin substrate, Tissue Eng. Part A [Internet] 18 (23e24) (2012)
mediated treatment of volumetric muscle loss, Nat. Commun. [Internet] 8 2453e2465. Available from: http://online.liebertpub.com/doi/abs/10.1089/
(2017) 15613, https://doi.org/10.1038/ncomms15613%5Cn. Available from: ten.tea.2012.0181.
http://www.nature.com/doifinder/10.1038/ncomms15613. [23] S. Zhang, X. Liu, S.F. Barreto-Ortiz, Y. Yu, B.P. Ginn, N.A. DeSantis, et al.,
[4] B.M. Sicari, V. Agrawal, B.F. Siu, C.J. Medberry, C.L. Dearth, N.J. Turner, et al., Creating polymer hydrogel microfibres with internal alignment via electrical
A murine model of volumetric muscle loss and a regenerative medicine and mechanical stretching, Biomaterials [Internet] 35 (10) (2014) 3243e3251.
approach for tissue replacement, Tissue Eng. Part A [Internet] 18 (19e20) Available from: http://linkinghub.elsevier.com/retrieve/pii/
(2012) 1941e1948. Available from: http://online.liebertpub.com/doi/abs/10. S0142961213015779.
1089/ten.tea.2012.0475. [24] Y.S. Choi, L.G. Vincent, A.R. Lee, K.C. Kretchmer, S. Chirasatitsin, M.K. Dobke, et
[5] M.T. Conconi, P.De Coppi, S. Bellini, G. Zara, M. Sabatti, M. Marzaro, et al., al., The alignment and fusion assembly of adipose-derived stem cells on
Homologous muscle acellular matrix seeded with autologous myoblasts as a mechanically patterned matrices, Biomaterials [Internet] 33 (29) (2012)
tissue-engineering approach to abdominal wall-defect repair, Biomaterials 6943e6951. Available from: http://linkinghub.elsevier.com/retrieve/pii/
[Internet] 26 (15) (2005) 2567e2574. Available from: http://linkinghub. S0142961212007077.
elsevier.com/retrieve/pii/S0142961204006878. [25] M. Chen, Y. Sun, Y. Chen, Electrically conductive nanofibers with highly ori-
[6] B.T. Corona, C.L. Ward, H.B. Baker, T.J. Walters, G.J. Christ, Implantation of ented structures and their potential application in skeletal muscle tissue en-
in vitro tissue engineered muscle repair constructs and bladder acellular gineering, Acta Biomater. [Internet] 9 (3) (2013) 5562e5572, https://doi.org/
matrices partially restore in vivo skeletal muscle function in a rat model of 10.1016/j.actbio.2012.10.024. Available from:.
J. Gilbert-Honick et al. / Biomaterials 164 (2018) 70e79 79

[26] Y.S. Choi, L.G. Vincent, A.R. Lee, M.K. Dobke, A.J. Engler, Mechanical derivation [36] Y. Lin, L. Liu, Z. Li, J. Qiao, L. Wu, W. Tang, et al., Pluripotency potential of
of functional myotubes from adipose-derived stem cells, Biomaterials human adipose-derived stem cells marked with exogenous green fluorescent
[Internet] 33 (8) (2012) 2482e2491. Available from: http://linkinghub. protein, Mol. Cell Biochem. [Internet] 291 (1e2) (2006) 1e10. Available from:
elsevier.com/retrieve/pii/S0142961211014591. http://link.springer.com/10.1007/s11010-006-9188-5.
[27] A.J. Engler, M.A. Griffin, S. Sen, C.G. Bo €nnemann, H.L. Sweeney, D.E. Discher, [37] V. Bayati, Y. Sadeghi, M.A. Shokrgozar, N. Haghighipour, K. Azadmanesh,
Myotubes differentiate optimally on substrates with tissue-like stiffness : A. Amanzadeh, et al., The evaluation of cyclic uniaxial strain on myogenic
pathological implications for soft or stiff microenvironments, J. Cell Biol. 166 differentiation of adipose-derived stem cells, Tissue Cell [Internet] 43 (6)
(6) (2004) 877e887. (2011) 359e366. Available from: http://linkinghub.elsevier.com/retrieve/pii/
[28] C.A. Cook, P.Y. Huri, B.P. Ginn, J. Gilbert-Honick, S.M. Somers, J.P. Temple, et al., S0040816611000863.
Characterization of a novel bioreactor system for 3D cellular mechanobiology [38] F.Y. Meligy, K. Shigemura, H.M. Behnsawy, M. Fujisawa, M. Kawabata,
studies, Biotechnol. Bioeng. 113 (8) (2016) 1825e1837. T. Shirakawa, The efficiency of in vitro isolation and myogenic differentiation
[29] S.G. Dubois, E.Z. Floyd, S. Zvonic, G. Kilroy, X. Wu, S. Carling, et al., Isolation of of MSCs derived from adipose connective tissue, bone marrow, and skeletal
human adipose-derived stem cells from biopsies and liposuction specimens. muscle tissue, Vitr. Cell Dev. Biol. Anim. [Internet] 48 (4) (2012) 203e215.
In: methods in molecular biology, Mesenchymal Stem Cells: Methods and Available from: http://link.springer.com/10.1007/s11626-012-9488-x.
Protocols (2008) 69e79. [39] G. Di Rocco, M.G. Iachininoto, A. Tritarelli, S. Straino, A. Zacheo, A. Germani, et
[30] S.R. Iyer, A.P. Valencia, E.O. Hernandez-Ochoa, R.M. Lovering, In vivo assess- al., Myogenic potential of adipose-tissue-derived cells, J. Cell Sci. [Internet]
ment of muscle contractility in animal studies, Methods Mol. Biol. [Internet] 119 (14) (2006) 2945e2952. Available from: http://jcs.biologists.org/cgi/doi/
1460 (2016) 293e307. Available from: http://link.springer.com/10.1007/978- 10.1242/jcs.03029.
1-4939-3810-0. [40] Y. Liu, X. Yan, Z. Sun, B. Chen, Q. Han, J. Li, et al., Flk-1 þ adipose-derived
[31] K.W. VanDusen, B.C. Syverud, M.L. Williams, J.D. Lee, L.M. Larkin, Engineered mesenchymal stem cells differentiate into skeletal muscle satellite cells and
skeletal muscle units for repair of volumetric muscle loss in the tibialis ameliorate muscular dystrophy in MDX mice, Stem Cells Dev. [Internet] 16 (5)
anterior muscle of a rat, Tissue Eng. Part A 20 (21e22) (2014) 2920e2930. (2007) 695e706. Available from: http://www.liebertonline.com/doi/abs/10.
[32] C.H.J. da Pinheiro, J.C.F. de Queiroz, L. Guimar~ aes-Ferreira, K.F. Vitzel, 1089/scd.2006.0118.
R.T. Nachbar, L.G.O. de Sousa, et al., Local injections of adipose-derived [41] M. Kim, Y.S. Choi, S.H. Yang, H.-N. Hong, S.-W. Cho, S.M. Cha, et al., Muscle
mesenchymal stem cells modulate inflammation and increase angiogenesis regeneration by adipose tissue-derived adult stem cells attached to injectable
ameliorating the dystrophic phenotype in dystrophin-deficient skeletal PLGA spheres, Biochem. Biophys. Res. Commun. [Internet] 348 (2) (2006)
muscle, Stem Cell Rev. Rep. 8 (2) (2012) 363e374. 386e392. Available from: http://linkinghub.elsevier.com/retrieve/pii/
[33] M. Gautam, D. Fujita, K. Kimura, H. Ichikawa, A. Izawa, M. Hirose, et al., S0006291X06015695.
Transplantation of adipose tissue-derived stem cells improves cardiac con- [42] M.T. Li, M. Ruehle, H. Stevens, N. Servies, N. Willett, S. Karthikeyakannan, et
tractile function and electrical stability in a rat myocardial infarction model, al., Skeletal myoblast-seeded vascularized tissue scaffolds in the treatment of
J. Mol. Cell Cardiol. [Internet] 81 (2015) 139e149, https://doi.org/10.1016/ a large volumetric muscle defect in the rat biceps femoris muscle, Tissue Eng.
j.yjmcc.2015.02.012. Available from:. Part A [Internet] (2017) ten.TEA.2016.0523. Available from: http://www.ncbi.
[34] S. Gehmert, C. Wenzel, M. Loibl, G. Brockhoff, M. Huber, W. Krutsch, et al., nlm.nih.gov/pubmed/28372522%0Ahttp://online.liebertpub.com/doi/10.
Adipose tissue-derived stem cell secreted IGF-1 protects myoblasts from the 1089/ten.TEA.2016.0523.
negative effect of myostatin, BioMed. Res. Int. 2014 (2014) 1e9. [43] L.R. Smith, E.R. Barton, SMASH - semi-automatic muscle analysis using seg-
[35] H. Mizuno, P.A. Zuk, D. Ph, M. Zhu, H.P. Lorenz, P. Benhaim, et al., Experi- mentation of histology: a MATLAB application, Skeletal Muscle 4 (1) (2014)
mental myogenic differentiation by human processed lipoaspirate cells, Plast. 21.
Reconstr. Surg. 109 (1) (2002) 199e209.

You might also like