Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Biomaterials 278 (2021) 121173

Contents lists available at ScienceDirect

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

Cells, scaffolds, and bioactive factors: Engineering strategies for improving


regeneration following volumetric muscle loss
Ioannis Eugenis a, d, 1, Di Wu b, d, 1, Thomas A. Rando b, c, d, *
a
Department of Bioengineering, Stanford University, Stanford, CA, USA
b
Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
c
Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
d
Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Severe traumatic skeletal muscle injuries, such as volumetric muscle loss (VML), result in the obliteration of large
Volumetric muscle loss amounts of skeletal muscle and lead to permanent functional impairment. Current clinical treatments are limited
Tissue engineering in their capacity to regenerate damaged muscle and restore tissue function, promoting the need for novel muscle
Skeletal muscle regeneration
regeneration strategies. Advances in tissue engineering, including cell therapy, scaffold design, and bioactive
Biomaterials
Vascularization
factor delivery, are promising solutions for VML therapy. Herein, we review tissue engineering strategies for
Innervation regeneration of skeletal muscle, development of vasculature and nerve within the damaged muscle, and
Immunomodulation achievements in immunomodulation following VML. In addition, we discuss the limitations of current state of the
art technologies and perspectives of tissue-engineered bioconstructs for muscle regeneration and functional re­
covery following VML.

1. Introduction and complications such as infection and necrosis [7,14,15]. Therefore,


the development of regenerative technologies, which can address cur­
Skeletal muscle injury is common following excessive exercise and rent challenges and provide substitutions for autologous muscle for
traumatic incidents, such as motor vehicle accidents, orthopedic sur­ transplantation, is vital for VML treatment. The therapeutic success re­
geries, or combat-related fractures [1–4]. Following minor injuries, quires a large amount of muscle formation to replace the lost tissue,
skeletal muscle has a remarkable ability to regenerate, which relies on sufficient vascularization to supply blood flow, functional innervation to
muscle stem cells (MuSCs), also referred to as “satellite cells”, and their generate action potentials, and effective immunomodulation to reduce
interaction with the surrounding microenvironment [3,5]. However, fibrosis and support tissue regeneration [16–19]. Tissue engineering
volumetric muscle loss (VML) injury overwhelms the endogenous repair approaches have tremendous potential for muscle regeneration and
capacity and leads to an irrecoverable functional impairment [6–10]. functional recovery (Fig. 1). In this review, we will summarize current
Following VML, substantial loss of MuSCs diminishes the innate regen­ engineering strategies for VML treatment that promote myogenesis,
erative ability of the injured muscle. Complete removal of the extra­ vascularization, innervation, and immunomodulation. The advances
cellular matrix results in the disappearance of a scaffolding to support and limitations of tissue engineering technologies, including cell ther­
cell attachment and guide tissue reconstruction [11]. Moreover, VML apy, scaffold design, and bioactive factor delivery will be discussed in
induces chronic inflammation that causes continued tissue damage and each category.
massive fibrosis, impeding muscle regeneration [12,13]. In this context,
VML leads to permanent loss of muscle and life-long disability [14]. 2. Myogenesis
Current clinical strategies for VML treatment are limited to free
functional muscle transfer, an autologous muscle transplanted to the 2.1. Cells
defect area for functional restoration [6,7]. However, autografting is
often associated with donor-site morbidity, limited tissue availability, Cell-based therapies have great potential for treating VML injuries.

* Corresponding author. Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
E-mail address: rando@stanford.edu (T.A. Rando).
1
Authors contribute equally to this work.

https://doi.org/10.1016/j.biomaterials.2021.121173
Received 22 February 2021; Received in revised form 1 August 2021; Accepted 14 August 2021
Available online 1 October 2021
0142-9612/Published by Elsevier Ltd.
I. Eugenis et al. Biomaterials 278 (2021) 121173

Transplanted myogenic cells can form new myofibers as well as repair model yielded de novo muscle fiber formation and a 40% recovery of
damaged existing fibers, thereby restoring muscle mass and function. muscle mass after one month [31]. Despite their proliferative potential,
Several studies have investigated acellular approaches for VML treat­ it is likely that a large number of MuSCs would need to be transplanted
ment, demonstrating improved muscle function but yielding inadequate for an improved therapeutic benefit. However, MuSCs expanded in vitro
muscle regeneration [20–22]. Systematic evaluations of muscle biome­ have a drastic reduction in engraftment and regenerative capacity when
chanics and histology show that decellularized scaffolds alone are transplanted back in vivo, compared to freshly isolated MuSCs [32,33].
insufficient for promoting de novo muscle fiber formation [23–25]. Several groups have developed in vitro culture conditions that enable
Therefore, an exogenous stem cell source is likely to be necessary for MuSC expansion, while maintaining their therapeutic potential [34,35],
effective treatment of VML injuries. Cell types that participate in normal but cells produced by these methods have not been transplanted into a
skeletal muscle regeneration have been explored as candidates for VML model. On the other hand, various proliferating populations of
regenerative medicine and previously reviewed in detail [26,27]. Here, myogenic progenitors and myogenic cells lines, such as L8 and C2C12
we will highlight the cell sources that have shown potential, specifically myoblasts, have been investigated for VML applications because they
for VML therapies, based on their ability to generate new muscle can be easily expanded in vitro [36–38]. In this review, we will refer to
(Fig. 2A). myogenic populations that have not been prospectively derived from
MuSCs are the predominant muscle fiber forming cells in skeletal MuSCs as myogenic progenitor cells (MPCs), even if poorly
muscle tissue. Following injury, MuSCs activate, proliferate, differen­ characterized.
tiate, and fuse to repair damaged myofibers, as well as form new myo­ In vivo, MuSCs are supported by other cells in the tissue [39,40]. A
fibers. As a result, MuSCs have been extensively investigated as cell mixed cell population termed “muscle resident cells” (MRCs), made up
sources for regenerative medicine [28,29]. Despite their scarcity in of endothelial cells, hematopoietic cells, fibro-adipogenic progenitors,
skeletal muscle, MuSCs demonstrate a dramatic proliferation potential and fibroblast-like cells, has been shown to enhance the survival of
in vivo [30]. Freshly isolated MuSCs transplanted into a mouse VML MuSCs both from studies in vitro and in a mouse VML model in vivo [31].

Fig. 1. Volumetric Muscle Loss (VML) results in a significant ablation of skeletal muscle. Tissue engineering strategies combining cells, scaffolds, and bioactive
factors, present promising solutions for VML therapy. Muscle regeneration and functional recovery following VML requires sophisticated approaches to promote
myogenesis, vascularization and innervation, and immunomodulation.

2
I. Eugenis et al. Biomaterials 278 (2021) 121173

Fig. 2. Advances in tissue engineering for enhancing myogenesis following VML. (A) Myogenic cells used for VML bioconstructs are harvested from skeletal muscle
or differentiated from iPSCs. Following muscle digestion, MuSCs are isolated by sorting methods whereas MPCs are purified and expanded in vitro. (B) Scaffold
physiochemical properties play an important role in skeletal muscle regeneration following VML. (C) Bioactive factors can be incorporated into bioconstructs to
modulate different stages of myogenesis.

A combination of MuSCs and MRCs doubled the recovery of muscle implantation in vivo. In a recent study, human iPSC-derived MPCs
mass, reduced fibrosis, increased myogenesis, and significantly transplanted in a mouse VML model yielded an improvement in muscle
improved ex vivo muscle function compared to MuSC-only treatment. In contractility and donor-derived muscle fiber formation [44]. Moreover,
the same study, combinatorial co-culture experiments of MuSCs with in vitro iPSC-derived functional skeletal muscle constructs have been
endothelial cells, MRCs, or MRCs without endothelial cells, revealed that shown to fuse with endogenous myofibers when implanted in vivo
endothelial cells were both necessary and sufficient to sustain MuSC [45–47]. A recent study demonstrated the therapeutic potential of using
viability, expansion, and engraftment in the VML model [31]. Minced multilineage engineering to create artificial skeletal muscles containing
muscle grafts, which include MuSCs and other cells residing in muscle iPSC-derived myofibers, endothelial cells, pericytes, and motor neurons
tissue, have also been explored as cell sources for VML therapy. Minced [48]. Although generating iPSC-derived skeletal muscle constructs, such
muscle grafts transplanted into a rat VML defect yielded a 55% recovery as induced skeletal muscle (iSKM) bundles or other artificial muscles
of functional deficit measured by net torque production [41]. In a larger, [45,48], in vitro and then implanting them in vivo is a promising
porcine model of VML, autologous minced muscle grafts produced a approach for skeletal muscle regeneration, further investigation is
32% increase in strength after 12 weeks [42]. However, few areas of de needed to determine the efficacy of these therapies in a VML model. In
novo muscle regeneration and significant fibrosis were observed within vivo, iSKM bundles have been shown to progressively vascularize when
the defect area. Moreover, clinical limitations, including inadequate implanted subcutaneously and to successfully engraft in a TA muscle,
tissue availability and donor site morbidity, must be addressed as a while maintaining functionality one-week post implantation. Nonethe­
significant volume of autologous minced muscle would be required to less, clinical limitations, including potential for tumorigenicity and
repair a defect at the scale of VML injuries. immunogenicity [49–51], must be addressed to fully realize an
Induced pluripotent stem cells (iPSCs) are attractive cell candidates iPSC-based therapy for VML.
for a variety of tissue engineering applications because they are easily
obtained, capable of robust proliferation in vitro, and can be differenti­
ated into multiple cell types [43]. iPSC-based strategies investigated for 2.2. Scaffolds
skeletal muscle regeneration include direct transplantation of
iPSC-derived MPCs and in vitro engineered skeletal muscle constructs, Since VML injury also results in the removal of native ECM, endog­
where iPSCs are differentiated into myotubes in vitro prior to enous and transplanted cells lack a scaffold that is necessary to regen­
erate organized skeletal muscle [15]. A recent study found that acellular

3
I. Eugenis et al. Biomaterials 278 (2021) 121173

muscle architecture is required to guide MuSCs during regeneration scaffold bioactivity and muscle regeneration. Unlike most natural ma­
[52], underscoring the need for scaffolds that support myogenesis in terials, synthetic materials must be modified to promote the attachment
VML therapies. Scaffolds used for enhancing skeletal muscle regenera­ of transplanted and endogenous cells. Synthetic biomaterials for skeletal
tion have been reviewed extensively [15,53–55]. Biomaterials can muscle applications are generally coated, or mixed, with natural mate­
provide essential biochemical and biophysical cues to support cell rials such as fibrin, laminin, or Matrigel, to promote cell attachment
attachment, survival, proliferation, and myogenic differentiation [53, [72–74]. However, incorporating full-length ECM proteins into syn­
56]. For example, MPCs encapsulated in an in situ casted fibrin hydrogel thetic scaffolds can elicit an inflammatory immune response and result
engrafted and formed new muscle fibers in a murine VML defect [57]. in undesirable changes to scaffold properties, such as surface charge and
Similarly, human MPCs micropatterned on poly lactic-co-glycolic acid topography [75]. Functionalizing scaffolds with small peptides is an
(PLGA) sheets, stacked to form a 3D construct, and transplanted into a attractive alternative to improve bioactivity and overcome the afore­
murine VML defect also showed improved engraftment compared to mentioned limitations associated with full-length proteins. Peptides are
direct cell injection [58]. Here, we will highlight some of the biomaterial able to mimic the bioactivity of their full-length protein counterparts
properties, including composition, stiffness, porosity, and degradation, while also being easier to produce and modify under well-defined con­
that have been shown to improve the regeneration of skeletal muscle ditions. Arginine-glycine-aspartate (RGD) peptides tethered to an algi­
following VML (Fig. 2B). nate scaffold have been shown to enhance MPC viability following
Scaffolds for regenerative medicine can generally be characterized as transplantation into skeletal muscle [76]. In addition to improving cell
natural or synthetic based on their composition [15,59]. As implied by attachment, peptides have also been incorporated into natural and
their name, natural biomaterials are polymers derived from natural synthetic scaffolds to promote a variety of cellular responses, including
sources, such as proteins, polysaccharides, and DNA, and are often survival, proliferation, angiogenesis, and anti-inflammation [70]. RGD
biocompatible, biodegradable, and have low cytotoxicity. Despite their and IKVAV, an isoleucine-lysine-valine-alanine-valine peptide derived
biocompatibility, natural biomaterials generally have weak mechanical from laminin, have been investigated for controlling MPC behavior in
strength and batch-to-batch variability. Common natural biomaterials vitro [77]. When cultured on top of hyaluronic acid hydrogels func­
that have been used in skeletal muscle tissue engineering include tionalized with IKVAV, MPCs demonstrated increased migration and
collagen, fibrin, alginate, and decellularized ECM. Synthetic bio­ expression of Pax7, a transcription factor involved in MuSC proliferation
materials, on the other hand, are more scalable and their physico­ and differentiation, compared to cells presented with RGD.
chemical properties are easily tailored for their application. Unlike their Scaffold properties, such as stiffness, degradation, and porosity, also
natural counterparts, synthetic biomaterials must be functionalized with play critical roles in mediating tissue regeneration. Fabrication methods,
bioactive cues to make them biocompatible. Common synthetic bio­ such as electrospinning, pore generation, and 3D printing, as well as
materials that have been used for skeletal muscle tissue engineering crosslinking techniques can be used to develop scaffolds with physical
include polyethylene glycol (PEG), polyglycolic acid (PGA), poly­ properties that closely mimic the tissue of interest. An ideal scaffold for
caprolactone (PCL), and poly lactic-co-glycolic acid (PLGA). Although VML therapy will likely match the stiffness of skeletal muscle with a
natural and synthetic biomaterials are generally associated with the Young’s modulus of approximately 12 kPa [78]. Scaffold degradation
respective properties and limitations mentioned above, scaffold char­ rate is another key factor that determines the success of a regenerative
acteristics can be altered by modifying the polymer, introducing new medicine therapy. Ideally, the rate at which a material degrades should
crosslinking chemistries, or mixing biomaterials to produce hybrid be proportional to the rate of new tissue formation [79]. Relative to the
scaffolds. rate of new tissue formation, scaffolds with too slow of a degradation are
Decellularized ECM (dECM) is the most widely used natural scaffold prone to fibrosis and encapsulation, whereas too fast of a degradation
for VML applications due to the abundance of biochemical cues that are will not provide enough physical support for cells to regenerate the
critical for tissue regeneration [60–62]. dECM scaffolds sourced from injured tissue [55]. Introducing porosity into scaffolds can allow
muscle, small intestine, and urinary bladder have demonstrated variable endogenous cells to participate in the regeneration process and vascu­
success in VML models [20,24,63,64]. Differences in ECM composition, larization to take place more easily [80]. Porosity becomes increasingly
decellularization protocols, and scaffold processing are all contributing important for larger tissue defects, such as VML, which require rapid
factors to dECM variability. Other natural materials, such as keratin ingrowth of vasculature to deliver essential nutrients and remove waste.
derived from human hair, have also demonstrated potential as scaffolds A recent study investigating the therapeutic potential of a porous scaf­
for VML therapy [65,66]. Compared to bladder dECM, acellular keratin fold in a mouse VML model found that a collagen-GAG sponge promoted
hydrogels yielded increased muscle regeneration, decreased fibrosis, a functional improvement and upregulation of factors involved in
and improved functional recovery following 12 weeks post-implantation wound healing [81]. Moreover, a collagen-gelatin-laminin sponge
in a rat VML model [65]. Another study explored the ability of a hyal­ implanted in another hindlimb VML model supported the recruitment of
uronic acid hydrogel supplemented with laminin-111, an embryonic endogenous cells, including MuSCs, but yielded limited myofiber
isoform of laminin, to support minced muscle grafts in a rat VML model regeneration after 2 weeks [82].
[67]. Hyaluronic acid, a polysaccharide known for its anti-adhesive and In addition to stiffness and composition, the anisotropic, or aligned,
anti-inflammatory properties [68], was chosen as a scaffold to effec­ organization of skeletal muscle can also be mimicked using scaffold
tively shield transplanted cells from the harsh immune environment. fabrication techniques. Aligned scaffolds more closely resemble the ar­
Additionally, laminin was incorporated to selectively promote the chitecture of skeletal muscle and have also been shown to mediate
adhesion, migration, and proliferation of transplanted cells. Consistent myofiber maturation in vitro [83]. Anisotropic materials for skeletal
with previous minced muscle and scaffold strategies [67,69], muscle regeneration have been investigated further and reviewed in
co-delivery of hyaluronic acid, laminin, and minced muscle yielded a detail [84,85]. A recent study demonstrated the importance of align­
functional improvement compared to an acellular hydrogel control. ment in the regenerative response following VML in a rat model by
However, functional recovery was comparable to that achieved with implanting autografts in misaligned orientations [86]. Misaligned au­
minced muscle grafts without any scaffolding [67], indicating that the tografts, rotated at 45- or 90-degree angles, were shown to have
scaffold did not enhance the effect of minced muscle grafts alone. significantly less functional recovery, fewer myofibers, and considerably
Scaffold functionalization with bioactive peptides has gained sig­ more fibrosis compared to aligned autografts. Scaffolds with aligned
nificant attention for improving regeneration in both soft (e.g. skin, architecture for VML applications have generally been fabricated by
muscle, brain) and hard (e.g. bone, cartilage) tissue applications [70, electrospinning and 3D printing. Aligned electrospun PCL/dECM scaf­
71]. To our knowledge, peptide functionalization has not yet been folds demonstrated increased myofiber regeneration compared to the
explored in VML applications but has great potential for enhancing PCL only group but did not yield significant improvements in muscle

4
I. Eugenis et al. Biomaterials 278 (2021) 121173

weights or functional recovery in a mouse VML model [87]. Aligned VML injury, application to other injury models demonstrate potential.
nanofibrillar collagen scaffolds implanted into a mouse VML model For example, Wnt7a is a potent signaling protein that can promote
yielded de novo myofibers with 60% larger cross-sectional areas symmetric MuSC expansion and directional migration of both MuSCs
compared to dECM controls [88]. Recent advances in 3D bioprinting and MPCs [107–109]. In a cryo-injured muscle model, co-delivery of
have drastically enhanced the fabrication of aligned skeletal muscle fi­ Wnt7a and MuSCs in a hydrogel on top of the injury area significantly
bers [89,90]. Human MPCs in a 3D printed scaffold with aligned ar­ increased the migration distance of transplanted MuSCs towards the
chitecture showed increased muscle regeneration, improved functional injury site to form new muscle fibers [110]. Another study that pro­
recovery, and decreased fibrosis compared to non-printed groups moted the survival of transplanted MPCs and induced their migration
following 8 weeks post implantation in a rat TA VML model [91]. The from a scaffold placed on top of the injured tissue was investigated in a
same bioconstruct, implanted into a pelvic floor VML model, maintained muscle laceration model [111]. HGF and FGF2, potent growth factors
muscle volume over 8 weeks and significantly improved muscle function known to increase proliferation and prevent premature differentiation of
compared to acellular bioconstructs [92]. 3D printed bioconstructs myoblasts [112,113], were co-delivered with myoblasts [111]. In
composed of human MPCs in photo-crosslinkable dECM also demon­ response to HGF and FGF2, myoblasts demonstrated increased migra­
strated increased muscle regeneration and functional recovery in a rat tion outward from the scaffold and incorporation with host tissues,
TA VML model [93]. Incorporation of non-myogenic cell types into 3D resulting in a significant reduction of defect area at 30 days post-injury.
printed bioconstructs have further improved myogenesis in VML lesions However, functional recovery was not characterized in this study [111].
[94,95]. Moreover, novel skeletal muscle 3D printing deposition stra­
tegies, including in situ and intravital bioprinting, have recently been 2.3.2. Factors for cell proliferation
explored and show great promise for VML therapy [96,97]. Achieving a large enough number of myogenic cells for regeneration
is critical for VML therapy [15,56]. FGF-2 was utilized to promote the in
2.3. Bioactive factors vivo proliferation of L8 myoblasts [114]. In a crush injury model, L8
myoblasts, which were genetically modified to overexpress FGF-2,
Besides therapeutic cells and scaffolds, bioactive factors, such as demonstrated a 53% reduction in apoptotic cell death and a 1.8-fold
growth factors, cytokines, and signaling molecules, are other key me­ increase in proliferation compared with control myoblasts [114]. Be­
diators of skeletal muscle regeneration [98]. The native regeneration sides promoting the proliferation of transplanted myoblasts, bioactive
process, including cell recruitment, proliferation, and differentiation, is factors have also been used to induce endogenous cell proliferation [65,
orchestrated by different biochemical cues [99]. For instance, HGF has 66,115]. However, direct injection of FGF-2, with or without heparin to
been found to be a critical factor for triggering the activation of quies­ provide a sustained release, to a crush-injured muscle showed no
cent MuSCs in vivo [100]. In addition, FGF-2 has been shown to be a measurable effect on the number of proliferating cells [115]. A negli­
potent inducer of MuSC proliferation [101]. Owing to their pivotal role gible effect of FGF-2 on cell proliferation was also found in VML treat­
in controlling cell behavior, bioactive factors are of great interest in ment by delivering FGF-2 in a keratin hydrogel [65,66]. Nevertheless,
regenerative medicine. Although the delivery of bioactive factors can co-delivery of FGF-2 and IGF-1 demonstrated a synergistic effect on
improve myogenesis, their dosage must be tightly controlled since improving functional recovery following VML [66]. Therefore, the de­
suboptimal dose and improper timing may result in adverse outcomes livery of inductive factors can promote myogenic cell proliferation in
such as increased fibrosis, hypertension, and edema [102,103]. Bio­ vivo and improve muscle functional recovery following VML injury.
materials can enable the spatiotemporal control of bioactive factors to
guide the regenerative process in vivo. Here, we will summarize the 2.3.3. Factors for cell differentiation and fusion
factors that have been used in VML therapies and their delivery strate­ To achieve functional muscle tissue, myogenic progenitors need to
gies (Fig. 2C). Moreover, additional factors, which have been shown to differentiate and fuse together to form mature myofibers. In the native
have therapeutic potential in the other muscle injury models besides repair process, IGF-1 plays a critical role in regulating myogenic cell
VML, will be covered for future exploration. differentiation and fusion [116,117]. C2C12 myoblasts cultured on a
collagen scaffold with IGF-1 in vitro formed myotubes exhibiting a
2.3.1. Factors for cell activation and recruitment 1.5-fold increase in length and 2-fold greater nuclei per myotube
Owing to the remarkable capabilities of MuSCs for muscle regener­ compared with the myoblasts without IGF-1 [118]. The therapeutic
ation [104], recruitment of endogenous MuSCs is an attractive approach potential of IGF-1 was further explored by transplanting the acellular
for in situ muscle regeneration. Bioactive factors have been delivered to IGF-1 loaded scaffold into a mouse VML model. Although the density of
recruit host MuSCs and provide proper guidance for muscle regeneration regenerating fibers in the IGF group increased, there was no statistical
following VML [105,106]. For example, poly(L-lactic acid) (PLLA) significance found compared with scaffold only group [118]. The
scaffolds releasing bioactive factors, including SDF-1α, HGF, IGF-1, and capability of IGF-1 to induce myogenic differentiation was also inves­
FGF-2, were investigated for their ability to improve MuSC recruitment tigated in a rabbit VML model [119]. IGF-1 was sustained released to the
and myogenesis following VML [105]. Only the scaffolds releasing IGF-1 injury site by dECM scaffolds following VML. The immunohistochemical
or FGF-2 demonstrated a significant MuSC enrichment compared to the staining for myosin heavy chain (MHC) illustrated that the dECM scaf­
control scaffold without growth factors. In addition, delivery of IGF-1, fold with IGF-1 yielded a significantly increased number of newly
FGF2, or HGF yielded more newly formed muscle fibers than control formed myofibers and a reduction in collagen deposition compared to a
scaffold. The therapeutic effects of HGF on muscle functional recovery scaffold only group.
following VML was further investigated [106]. A sustained delivery of
HGF for 48 h to mimic its native release kinetics after injury was ach­ 3. Vascularization and innervation
ieved by adsorbing HGF onto fibrin microthreads. About 91% of iso­
metric twitch force was recovered compared with the contralateral Vascularization and innervation are essential processes for muscle
uninjured muscle after 60 days post-injury, while the no intervention regeneration and functional recovery following VML injury. Vascula­
group recovered 70%. In addition, although the HGF group recovered ture, which is highly dense and organized, provides oxygen and nutri­
99% force at tetanic contraction, there was no significance between the ents for skeletal muscle. Although spontaneous revascularization is
treatment groups with and without HGF. triggered in response to muscle injury without any treatment, the
Besides recruiting endogenous cells, bioactive factors can be used to ingrowth of blood vessels is too slow to meet the high metabolic demand
induce activation and promote mobilization of transplanted cells. While of muscle regeneration in large size of defect like VML [16,120,121].
the therapeutic effects of such a strategy have not yet been explored in Bioconstruct vascularization is particularly challenging for VML therapy

5
I. Eugenis et al. Biomaterials 278 (2021) 121173

due to the large defect size and fibrosis at the injury site [122]. In the bioconstructs 21 days post implantation. Currently, both in vitro and
addition, VML injury also results in significant axotomy of motoneurons in vivo tissue engineering approaches using endothelial cells show
and removal of the neuromuscular junctions (NMJs) [123]. Denervation promise for enhancing vascularization of bioconstructs for VML therapy.
of injured muscle results in functional impairment and muscle fiber at­ Using endothelial cells to develop functional vascular networks,
rophy [124]. Therefore, emerging studies have focused on developing whether pre-formed in vitro or via direct cell transplantation in vivo, is
novel approaches to promote vascularization and innervation for muscle often a lengthy process. Microvessel fragments (MVFs) are attractive
regeneration. In this section, we will summarize current strategies to bioconstruct pre-vascularization units because they already have
improve vascularization and innervation of skeletal muscle by cell de­ developed microvessel morphology, rapidly assemble into networks
livery, scaffold design, and bioactive factor delivery (Fig. 3). when transplanted in vivo, and can be easily isolated in large quantities
from adipose tissue [127]. MVFs are inherently more complex than
endothelial cells and have been shown to contain mesenchymal stem
3.1. Cells cells, which contribute to their regenerative potential [128]. Collagen
hydrogels containing MVFs yielded a higher vessel density within a VML
Endothelial cells are known to play an important role in muscle defect compared to hydrogels with adipose derived stem cells after 7
regeneration [125]. Therefore, endothelial cells have been studied days [129]. At 14 days post injury, MVF bioconstructs contained 75% of
extensively for creating vascular networks to enhance VML therapies. In the vessel density of uninjured controls. Moreover, perfusion of the MVF
vitro and in vivo tissue engineering strategies, where endothelial cells are network was observed in just 7 days and was maintained at 14 days post
used to form vasculature prior to implantation or are transplanted injury. Despite evidence of anastomosis with host vasculature, bio­
directly, have both been investigated. Pre-vascularization of bio­ construct perfusion was still low and heterogeneously distributed within
constructs with human umbilical vein endothelial cells (HUVECs) was the defect. More recent work investigated the vascularization potential
investigated in a mouse VML model [126]. Prior to implantation, bio­ of MVFs with or without the addition of myoblasts in a rat VML model
constructs were cultured in vitro to allow co-cultured HUVECs and [130]. Moreover, more small-diameter vessels were observed in both of
C2C12s to form aligned vessels and myotubes, respectively. Ten days the pre-vascularized groups than in acellular hydrogel and autograft
after implantation in vivo, pre-vascularized bioconstructs successfully controls.
anastomosed with host vasculature and were perfused with host red Similar to the aforementioned pre-vascularization strategies, using
blood cells. However, pre-vascularized bioconstructs were surrounded progenitor or differentiated cell populations, pre-innervation ap­
by fibrosis and yielded minimal myofiber formation, unlike their proaches for VML applications have entailed the incorporation of neu­
non-vascularized counterparts. In another study, endothelial cells and rons or neural stem cells (NSCs) into bioconstructs. Addition of motor
MuSCs on a dECM scaffold were directly transplanted, without a neurons to bioconstructs containing differentiated C2C12s demon­
pre-vascularization culture period, into a similar VML model [31]. strated improved myocyte maturation in vitro [131]. Following
Endothelial cells proliferated and contributed to vasculogenesis within

Fig. 3. Enhancing bioconstruct vascularization and innervation for VML therapy. (A) Vascular and neural cells can be incorporated into VML bioconstructs to
support regenerating muscle. (B) Scaffold fabrication and patterning techniques, including 3D bioprinting and aligning nanofibers, recreate complex tissue archi­
tecture and organization. (C) Pro-angiogenic and pro-neurogenic factor delivery can recruit vasculature and nerves to improve skeletal muscle regeneration
and function.

6
I. Eugenis et al. Biomaterials 278 (2021) 121173

implantation into a rat VML model, these pre-innervated bioconstructs non-alignment.


maintained muscle cross-sectional area after 3 weeks, unlike the
non-innervated, acellular, or non-treated control groups. In addition, 3.3. Bioactive factors
pre-innervated bioconstructs enhanced the recruitment of endogenous
MuSCs and the formation of NMJs near the injury area. Compared to 3.3.1. Bioactive factors for vascularization
non-innervated and acellular groups, pre-innervated bioconstructs also Angiogenic factors are well-established therapeutics for improving
promoted vascularization within and outside the injury area. Another tissue vascularization [120,138]. Current strategies of angiogenic factor
promising strategy for pre-innervating bioconstructs that has been delivery in VML therapy have focused on promoting and accelerating
tested in a VML model involves the transplantation of NSCs instead of the ingrowth of pre-existing vessels toward the injury site by recruiting
differentiated neurons. A recent study demonstrated that the addition of endogenous endothelial or progenitor cells. For instance, IGF-1 loaded
human NSCs to human MPCs yielded improved myofiber and NMJ scaffolds can significantly increase the density of perfused microvessels
formation after 8 weeks in a rat VML model, compared to bioconstructs compared to empty scaffolds following VML injury [118]. Improving
containing only MPCs [95]. The addition of NSCs to bioconstructs also muscle angiogenesis using other angiogenic factors has also been
yielded a decrease in fibrosis relative to MPC-only and non-treated investigated in muscle ischemic injury. Sustained release of factors,
controls. In contrast to pre-innervated bioconstructs [131], which including vascular endothelial growth factor (VEGF), SDF-1, FGF-2, and
demonstrated increased vascularization compared with their FGF-9, to ischemic muscle was shown to promote vascular density and
non-innervated counterparts, no significant improvements in vascular­ improve angiogenesis at the injury site [139–144]. These factors can
ization were observed upon the addition of NSCs to MPC bioconstructs serve as potential candidates for VML therapy. Besides delivering
[95]. angiogenic proteins, delivery of genes encoding for angiogenic factors is
another interesting approach for improving vascularization during
3.2. Scaffolds muscle regeneration. For example, mRNA encoding HGF was loaded
into scaffolds along with Lipofectamine transfection agent, which
The arrangement of myofibers, blood vessels, and nerves in native enabled the intracellular delivery of the mRNA [145]. Two weeks after
skeletal muscle is highly organized and essential for proper muscle VML injury, the HGF encoded mRNA resulted in a 1.3-fold increase in
function. 3D bioprinting has emerged as a promising tool for fabricating capillary density compared with luciferase encoded mRNA [145].
complex bioconstructs that mimic the highly organized, aligned struc­ In addition to inducing endogenous vessel infiltration, other appli­
ture of native skeletal muscle tissue [91,132]. Bioconstructs can be cations that have not been explored in VML model, such as improving
pre-vascularized and pre-innervated by spatially patterning a combi­ transplanted cell engraftment and stabilizing newly formed vessels, also
nation of multiple cell types, biomaterials, and bioactive factors. Tech­ have great therapeutic potential for VML. For example, angiogenic
niques for 3D bioprinting vasculature and nerve have both been factors have been integrated into endothelial cell-laden bioconstructs to
extensively reviewed for other tissue applications [121,133]. The role of improve the survival of transplanted cells and stimulate de novo vascu­
spatial patterning in pre-vascularization was specifically evaluated in a larization. HUVECs co-delivered with VEGF exhibited increased survival
rat VML model [94]. Bioconstructs composed of human MPCs and after transplantation into ischemic muscle, leading to improved tissue
HUVECs were 3D printed in two distinct conformations; MPC-HUVEC perfusion and muscle regeneration [146]. In addition to inducing the
hybrid fibers (mixed printing) and MPC fibers, each surrounded by a formation of new vessels, delivery of angiogenic factors can also pro­
layer of HUVECs (coaxial printing). Coaxially printed bioconstructs mote maturation to develop functional vascular networks [120]. The
yielded a significant increase in TA muscle weight compared to mixed combination of factors associated with both vessel formation and
printed bioconstructs. In addition, coaxially printed bioconstructs vas­ maturation has been found to be a more promising approach than the
cularized and anastomosed with host vasculature more than mixed delivery of single factors [147]. Nevertheless, different factors have their
printed and MPC-only groups. Muscles treated with coaxially printed own time window of action during vascularization [148], suggesting
bioconstructs also achieved approximately an 85% recovery of the force that distinct release kinetics are required for each of the factors.
production of uninjured muscle. 3D printed bioconstructs, which have Simultaneous delivery of VEGF and PDGF-BB did not induce stable in­
been pre-innervated by the incorporation of human NSCs, have also crease in blood vessel density [148,149]. In contrast, a sequential de­
shown promise for enhancing skeletal muscle regeneration following livery of VEGF and PDGF-BB to ischemic muscle yielded increased vessel
VML [95]. Bioconstructs containing NSCs yielded a significantly higher density at 2 weeks and maturity at 4 weeks compared with single factor
number of NMJs at 8 weeks post implantation in a rat VML model delivery [149]. This result suggests that delivery of bioactive factor
compared to MPC-only bioconstructs. Moreover, 3D printed bio­ combinations in their physiological sequences may greatly improve the
constructs, with and without NSCs, became highly vascularized within 4 regeneration process. Biomaterials, in this context, serve as a powerful
weeks and contained more blood vessels than uninjured muscle. technology for controlling the release kinetics of different bioactive
In addition to their role in myofiber maturation discussed previously, factors to mimic the native regeneration process.
aligned fibrillar scaffolds have also been investigated for their ability to
enhance vascularization and innervation during skeletal muscle regen­ 3.3.2. Bioactive factors for innervation
eration following VML. Compared to 3D bioprinting, where feature sizes Although bioactive factor delivery is extensively explored in nerve
below several microns are generally unable to be fabricated, electro­ regeneration [150], using bioactive factors to improve innervation for
spinning and shear-based extrusion methods can produce biomaterial VML treatment is still a nascent field. A recent study investigated the
fibers at the micro- and nano-scale [134,135]. Moreover, electrospun therapeutic potential of IGF-1 for the innervation of VML injured muscle
and shear-based extruded fibers can be aggregated in parallel to [118]. Scaffolds loaded with IGF-1 yielded a greater number of mature
generate scaffolds with aligned architecture, more closely mimicking the NMJs compared with empty scaffolds. Another strategy to achieve
architecture of native ECM [136]. Acellular, aligned nanofibrillar innervation for VML therapy is to induce acetylcholine receptor (AChR)
collagen scaffolds transplanted into a VML defect yielded improved clustering in bioconstructs by delivering agrin, a large proteoglycan for
density of perfused microvessels compared to non-aligned scaffolds inducing AChR clustering [151,152]. Muscle-derived cells were seeded
[88]. C2C12s and human endothelial cells co-cultured on the same on scaffolds coated with agrin and cultured for 6 days in vitro [151]. A
aligned scaffold also demonstrated a significant increase in perfused dramatic enhancement of AChR clustering has been found in the com­
vessel density compared to non-aligned scaffolds in a VML model [137]. bination of agrin presence and cyclic stretch, providing a potential
In addition, alignment was also shown to support the formation of approach for VML treatment. The in vivo therapeutic effect for VML was
organized vascular networks with greater anisotropy than further explored by physically adsorbing or chemically tethering agrin

7
I. Eugenis et al. Biomaterials 278 (2021) 121173

to the scaffold prior to implantation [152]. A significant increase of regeneration after 3 months of implantation was limited. Although few
neuromuscular junctions and neural infiltration was found in both transplanted ASCs were found to fuse with host myofibers, mesenchymal
groups containing agrin 4 weeks after VML injury. In addition, the stem cells are unlikely to replace a myogenic cell source.
scaffolds that were chemically tethered with agrin achieved a further
improvement of muscle regeneration compared with the physically 4.2. Scaffolds
adsorbed group due to a prolonged release profile.
Biomaterials generally trigger an immune response in vivo [168,
4. Immunomodulation 169]. Scaffold physicochemical properties play a critical role in the host
immune reaction and can be altered to modulate the immune micro­
The immune system plays a critical role in tissue regeneration. The environment and promote tissue regeneration. The interactions between
innate and adaptive immune systems are both key mediators of the biomaterials and the immune system have been reviewed in detail
injury site microenvironment and are known to influence progenitor cell [170–173]. Immunomodulatory properties of scaffolds used for VML
expansion, survival, and integration [153]. Immediately following therapy have also been investigated. Small intestinal submucosa dECM,
injury, the innate immune response results in a cascade of inflammatory cardiac dECM, and muscle dECM/PCL blended scaffolds yielded an
signals, infiltration of immune cells, and deposition of ECM. During increased M2-macrophage polarization in VML models compared to
normal regeneration, the inflammatory response generally resolves untreated controls [87,165,174]. Although urinary bladder dECM
quickly as tissues begin to repair. However, a prolonged inflammatory implanted into a rat VML model did not recapitulate autograft macro­
response leads to pathological fibrosis, insufficient regeneration, and phage polarization dynamics over 8 weeks, M2-macrophage polariza­
impairment of normal tissue function [154]. Immunomodulation is tion was comparable at the final time point [175]. Mechanistic studies of
therefore critical for promoting tissue regeneration and has been the adaptive immune response in skeletal muscle regeneration following
reviewed extensively [153,155–157]. VML revealed that dECM scaffolds induce a pro-regenerative response
VML injuries result in a dysregulated immune response that causes via T helper 2 cells, which in turn guide macrophage polarization [176].
sustained tissue inflammation, pathological fibrosis, and impaired A direct comparison of natural and synthetic scaffolds implanted into a
muscle regeneration [12,17,18]. In skeletal muscle, the inflammatory VML defect revealed divergent immune responses [177]. Specifically,
response is temporally coupled with myogenesis during regeneration M2-macrophage markers were up-regulated in dECM treated groups and
[158]. Macrophages, which polarize between pro-inflammatory (M1) down-regulated in PEG and poly(ethylene) scaffolds after 3 weeks.
and pro-regenerative (M2) phenotypes during regeneration, play an Synthetic scaffold groups induced a chronic neutrophil infiltrate, which
important role in regulating myogenesis, inflammation, and fibrosis was dependent on material stiffness and size. In addition to increasing
[159,160]. Specifically, macrophages modulate fibro-adipogenic pro­ neutrophil number, increases in scaffold stiffness caused decreased
genitor (FAP) proliferation and differentiation into fibroblasts or adi­ M2-macrophage polarization in synthetic scaffolds [177]. Similar to
pocytes [161,162]. A prolonged inflammatory response leads to an dECM scaffolds, natural biomaterial hydrogels, including elastin and a
increase in the number of FAPs and the production of extracellular fibrin/laminin-111 blend, also demonstrated increased M2 macrophage
matrix. Regulatory T-cells (Treg) have been shown to regulate macro­ polarization in VML models [178,179]. However, muscle regeneration
phage polarization towards the pro-regenerative phenotype and drive was minimal as both approaches lacked a myogenic cell source.
the later stages of muscle regeneration [158]. In this section, we will
highlight the immunomodulatory cells, scaffolds, and bioactive factors 4.3. Bioactive factors
that have been used to enhance skeletal muscle regeneration following
VML. Local delivery of immunomodulatory drugs in conjunction with
bioconstructs is another promising approach for enhancing tissue
4.1. Cells regeneration. FDA approved immunosuppressants have been investi­
gated in regenerative therapies for artery, bone, and nerve tissue
Mesenchymal stem cells (MSCs) exhibit anti-inflammatory and [180–182]. FTY70 and tacrolimus, immunosuppressants used to treat
immunomodulatory effects on both innate and adaptive immune cells multiple sclerosis and organ rejection, respectively, have also been used
[153]. For example, transplanted MSCs promote the transition of mac­ to enhance regeneration following VML injury [183,184]. FTY720
rophages from the M1 to M2 phenotype, inhibit immune cell prolifera­ loaded PLGA films implanted into a VML mouse model yielded an
tion, and induce the formation of Treg cells in vivo [163]. Therefore, increased number of anti-inflammatory monocytes and M2 macro­
MSCs have been used as an immunomodulatory cell source for phages at the injury site, compared to PLGA films alone [183]. In
enhancing regeneration in many tissue engineering applications [164]. addition, FTY720 treatment accelerated vascularization and yielded
MSCs transplanted into a rat VML defect significantly increased increased muscle regeneration. A combination therapy of minced mus­
macrophage polarization to the M2 phenotype after 2 weeks compared cle grafts and systemic delivery of tacrolimus demonstrated increased
to the untreated control [165]. In addition, MSC treatment yielded muscle regeneration and decreased fibrosis in a porcine VML model,
significantly increased muscle fiber regeneration and decreased fibrosis compared to minced muscle grafts alone [184]. In addition, systemic
after 8 weeks compared to the untreated control. Transplanting MSCs delivery of tacrolimus moderately improved muscle function in muscles
with a dECM scaffold demonstrated a synergistic effect, further treated with minced muscle grafts. Although delivery of immunomod­
improving macrophage polarization and muscle regeneration [165]. In a ulatory drugs in VML therapies is still nascent, local and systemic de­
similar VML model, MSCs derived from adipose tissue (adipose stem livery methods are both viable approaches to improve regeneration.
cells (ASCs)) in a collagen hydrogel also promoted an M2 macrophage
transition and decreased inflammatory cytokines [166]. In addition, 5. Summary and future perspectives
ASC treatment demonstrated improved myogenesis, increased blood
flow restoration, and decreased fibrosis compared to the acellular The ultimate goals of VML treatment are to fully regenerate the lost
hydrogel control. Improved myogenesis and vascularization were also muscle and recover tissue function. To achieve these goals, a variety of
observed in muscles treated with ASCs in a collagen hydrogel following tissue engineering approaches, including cell therapy, scaffold design,
a full thickness VML injury [129]. In a more severe VML mouse model, and bioactive factor delivery, have been developed. Although a signifi­
where TA and EDL muscles were removed, ASCs seeded on electrospun cant improvement has been made in the regeneration of vascularized
fibrin scaffolds yielded improved muscle regeneration and decreased and innervated muscle with reduced scar formation, challenges still
fibrosis compared to acellular controls [167]. However, muscle fiber persist in this field. To investigate therapeutic efficacy, multiple animal

8
I. Eugenis et al. Biomaterials 278 (2021) 121173

models of VML have been developed across species and evaluated in [4] T.A.H. Järvinen, T.L.N. Järvinen, M. Kääriäinen, V. Aärimaa, S. Vaittinen,
H. Kalimo, et al., Muscle injuries: optimising recovery, Best Pract. Res. Clin.
different muscles [64]. The critical size of defects among different
Rheumatol. 21 (2007) 317–331, https://doi.org/10.1016/j.berh.2006.12.004.
models ranges from 15% to 40% of muscle mass [8–10]. Besides the size [5] F. Relaix, P.S. Zammit, Satellite cells are essential for skeletal muscle
of defects, there is still a lack of standard criteria in dimensions, location, regeneration: the cell on the edge returns centre stage, Development 139 (2012)
and muscle type. In addition to the characterization of pre-clinical 2845–2856, https://doi.org/10.1242/dev.069088.
[6] B.F. Grogan, J.R. Hsu, Skeletal Trauma Research Consortium, Volumetric muscle
models, strategies for scale-up to yield clinical-grade bioconstructs loss, J. Am. Acad. Orthop. Surg. 19 (Suppl 1) (2011) S35–S37, https://doi.org/
also need to be developed. Resources of therapeutic cells with preferred 10.5435/00124635-201102001-00007.
properties, such as potential for self-renewal, robust regenerative [7] K. Garg, C.L. Ward, B.J. Hurtgen, J.M. Wilken, D.J. Stinner, J.C. Wenke, et al.,
Volumetric muscle loss: persistent functional deficits beyond frank loss of tissue,
capability, and easy accessibility, limit the clinical translation of cell J. Orthop. Res. 33 (2015) 40–46, https://doi.org/10.1002/jor.22730.
therapy. Future materials need to be developed with consistent [8] S.E. Anderson, W.M. Han, V. Srinivasa, M. Mohiuddin, M.A. Ruehle, J.Y. Moon, et
composition and highly tunable properties as synthetic scaffolds and al., Determination of a critical size threshold for volumetric muscle loss in the
mouse quadriceps, Tissue Eng. C Methods 25 (2019) 59–70, https://doi.org/
favorable biological properties as natural materials. A more compre­ 10.1089/ten.TEC.2018.0324.
hensive understanding of the concentrations and combinations of [9] X. Wu, B.T. Corona, X. Chen, T.J. Walters, A standardized rat model of volumetric
cellular and molecular components will facilitate the rational design of muscle loss injury for the development of tissue engineering therapies, Biores
Open Access 1 (2012) 280–290, https://doi.org/10.1089/biores.2012.0271.
the bioconstructs. [10] E.E. Vega-Soto, B.L. Rodriguez, R.E. Armstrong, L.M. Larkin, A 30% volumetric
In addition to the strategies mentioned above, exercise is an muscle loss does not result in sustained functional deficits after a 90-day recovery
emerging approach for improving muscle regeneration. Evidence in­ in rats, Regen Eng Transl Med 6 (2020) 62–68, https://doi.org/10.1007/s40883-
019-00117-2.
dicates that exercise, either as voluntary running or high-intensity in­
[11] C.J. Caldwell, D.L. Mattey, R.O. Weller, Role of the basement membrane in the
terval training, can promote muscle formation with vascularization and regeneration of skeletal muscle, Neuropathol. Appl. Neurobiol. 16 (1990)
innervation as well as reduced fibrosis following the transplantation of 225–238, https://doi.org/10.1111/j.1365-2990.1990.tb01159.x.
bioconstructs [31,118,185]. An immediate rehabilitation regimen post [12] J. Larouche, S.M. Greising, B.T. Corona, C.A. Aguilar, Robust inflammatory and
fibrotic signaling following volumetric muscle loss: a barrier to muscle
bioconstruct transplantation has been shown to promote scaffold regeneration, Cell Death Dis. 9 (2018) 409, https://doi.org/10.1038/s41419-
remodeling, but may hinder regeneration and increase fibrosis [20,31, 018-0455-7.
186]. Beginning exercise 1–2 weeks after transplantation, on the other [13] K. Garg, B.T. Corona, T.J. Walters, Therapeutic strategies for preventing skeletal
muscle fibrosis after injury, Front. Pharmacol. 6 (2015) 87, https://doi.org/
hand, can enhance the muscle regeneration potential of the cell-seeded 10.3389/fphar.2015.00087.
or growth factor-laden scaffold [118,185]. Another important goal is to [14] B.T. Corona, J.C. Rivera, J.G. Owens, J.C. Wenke, C.R. Rathbone, Volumetric
restore the integration of muscle and its neighboring tendon tissues. muscle loss leads to permanent disability following extremity trauma, J. Rehabil.
Res. Dev. 52 (2015) 785–792, https://doi.org/10.1682/JRRD.2014.07.0165.
Creation of the interface between muscle and tendon tissues requires [15] J.M. Grasman, M.J. Zayas, R.L. Page, G.D. Pins, Biomimetic scaffolds for
precise imitation of the different biological and mechanical properties regeneration of volumetric muscle loss in skeletal muscle injuries, Acta Biomater.
between distinct tissues. The development of the 3D bioprinting tech­ 25 (2015) 2–15, https://doi.org/10.1016/j.actbio.2015.07.038.
[16] J. Gilbert-Honick, W. Grayson, Vascularized and innervated skeletal muscle tissue
nologies makes it possible to have precise spatial control of bioconstruct engineering, Adv. Healthc Mater. 9 (2020), e1900626, https://doi.org/10.1002/
architecture at the microscale and build up a complex myotendinous adhm.201900626.
junction structure [187,188]. [17] C.A. Aguilar, S.M. Greising, A. Watts, S.M. Goldman, C. Peragallo, C. Zook, et al.,
Multiscale analysis of a regenerative therapy for treatment of volumetric muscle
In summary, tissue engineering is a promising field that applies
loss injury, Cell Death Dis. 4 (2018) 33, https://doi.org/10.1038/s41420-018-
multidisciplinary strategies to develop substitutes for lost muscle tissue 0027-8.
and restore its function. While a number of achievements have been [18] S.M. Greising, J.C. Rivera, S.M. Goldman, A. Watts, C.A. Aguilar, B.T. Corona,
made in skeletal muscle tissue engineering, VML therapy as a complex Unwavering pathobiology of volumetric muscle loss injury, Sci. Rep. 7 (2017)
13179, https://doi.org/10.1038/s41598-017-13306-2.
process is still challenging and requires systematic and sophisticated [19] S.M. Greising, B.T. Corona, C. McGann, J.K. Frankum, G.L. Warren, Therapeutic
approaches. The research in biology, material science, engineering, and approaches for volumetric muscle loss injury: a systematic review and meta-
medicine need to converge for future clinical success. analysis, Tissue Eng. B Rev. 25 (2019) 510–525, https://doi.org/10.1089/ten.
TEB.2019.0207.
[20] J. Dziki, S. Badylak, M. Yabroudi, B. Sicari, F. Ambrosio, K. Stearns, et al., An
Author credit acellular biologic scaffold treatment for volumetric muscle loss: results of a 13-
patient cohort study, Npj Regen Med 1 (2016) 16008, https://doi.org/10.1038/
npjregenmed.2016.8.
This manuscript was written by IE, DW, and TAR. [21] X.K. Chen, T.J. Walters, Muscle-derived decellularised extracellular matrix
improves functional recovery in a rat latissimus dorsi muscle defect model,
Declaration of competing interest J. Plast. Reconstr. Aesthetic Surg. 66 (2013) 1750–1758, https://doi.org/
10.1016/j.bjps.2013.07.037.
[22] N.J. Turner, A.J. Yates, D.J. Weber, I.R. Qureshi, D.B. Stolz, T.W. Gilbert, et al.,
The authors declare that they have no known competing financial Xenogeneic extracellular matrix as an inductive scaffold for regeneration of a
interests or personal relationships that could have appeared to influence functioning musculotendinous junction, Tissue Eng. 16 (2010) 3309–3317,
https://doi.org/10.1089/ten.TEA.2010.0169.
the work reported in this paper. [23] M. Quarta, M.J. Cromie Lear, J. Blonigan, P. Paine, R. Chacon, T.A. Rando,
Biomechanics show stem cell necessity for effective treatment of volumetric
Acknowledgements muscle loss using bioengineered constructs, Npj Regen Med 3 (2018) 18, https://
doi.org/10.1038/s41536-018-0057-0.
[24] A. Aurora, J.L. Roe, B.T. Corona, T.J. Walters, An acellular biologic scaffold does
This work was supported by grants from the NIH (AG036695, not regenerate appreciable de novo muscle tissue in rat models of volumetric
AR073248) and the Department of Veterans Affairs (I01RX001222, muscle loss injury, Biomaterials 67 (2015) 393–407, https://doi.org/10.1016/j.
biomaterials.2015.07.040.
I01BX002324) to T.A.R. All figures created with BioRender.com. [25] K. Garg, C.L. Ward, C.R. Rathbone, B.T. Corona, Transplantation of devitalized
muscle scaffolds is insufficient for appreciable de novo muscle fiber regeneration
References after volumetric muscle loss injury, Cell Tissue Res. 358 (2014) 857–873, https://
doi.org/10.1007/s00441-014-2006-6.
[26] M.N. Pantelic, L.M. Larkin, Stem cells for skeletal muscle tissue engineering,
[1] C.B. Ebbeling, P.M. Clarkson, Exercise-induced muscle damage and adaptation,
Tissue Eng. B Rev. 24 (2018) 373–391, https://doi.org/10.1089/ten.
Sports Med. 7 (1989) 207–234, https://doi.org/10.2165/00007256-198907040-
TEB.2017.0451.
00001.
[27] B.M. Sicari, C.L. Dearth, S.F. Badylak, Tissue engineering and regenerative
[2] J.D. Cross, J.R. Ficke, J.R. Hsu, B.D. Masini, J.C. Wenke, Battlefield orthopaedic
medicine approaches to enhance the functional response to skeletal muscle
injuries cause the majority of long-term disabilities, J. Am. Acad. Orthop. Surg.
injury, Anat. Rec. 297 (2014) 51–64, https://doi.org/10.1002/ar.22794.
19 (Suppl 1) (2011) S1–S7, https://doi.org/10.5435/00124635-201102001-
[28] R.N. Judson, F.M.V. Rossi, Towards stem cell therapies for skeletal muscle repair,
00002.
Npj Regen Med 5 (2020) 10, https://doi.org/10.1038/s41536-020-0094-3.
[3] N.J. Turner, S.F. Badylak, Regeneration of skeletal muscle, Cell Tissue Res. 347
(2012) 759–774, https://doi.org/10.1007/s00441-011-1185-7.

9
I. Eugenis et al. Biomaterials 278 (2021) 121173

[29] M. Shayan, N.F. Huang, Pre-clinical cell therapeutic approaches for repair of [54] A. Dunn, M. Talovic, K. Patel, A. Patel, M. Marcinczyk, K. Garg, Biomaterial and
volumetric muscle loss, Bioengineering 7 (2020), https://doi.org/10.3390/ stem cell-based strategies for skeletal muscle regeneration, J. Orthop. Res. 37
bioengineering7030097. (2019) 1246–1262, https://doi.org/10.1002/jor.24212.
[30] C.A. Collins, I. Olsen, P.S. Zammit, L. Heslop, A. Petrie, T.A. Partridge, et al., Stem [55] C.A. Cezar, D.J. Mooney, Biomaterial-based delivery for skeletal muscle repair,
cell function, self-renewal, and behavioral heterogeneity of cells from the adult Adv. Drug Deliv. Rev. 84 (2015) 188–197, https://doi.org/10.1016/j.
muscle satellite cell niche, Cell 122 (2005) 289–301, https://doi.org/10.1016/j. addr.2014.09.008.
cell.2005.05.010. [56] J. Liu, D. Saul, K.O. Böker, J. Ernst, W. Lehman, A.F. Schilling, Current methods
[31] M. Quarta, M. Cromie, R. Chacon, J. Blonigan, V. Garcia, I. Akimenko, et al., for skeletal muscle tissue repair and regeneration, BioMed Res. Int. (2018)
Bioengineered constructs combined with exercise enhance stem cell-mediated 1984879, https://doi.org/10.1155/2018/1984879.
treatment of volumetric muscle loss, Nat. Commun. 8 (2017) 15613, https://doi. [57] N. Matthias, S.D. Hunt, J. Wu, J. Lo, L.A. Smith Callahan, Y. Li, et al., Volumetric
org/10.1038/ncomms15613. muscle loss injury repair using in situ fibrin gel cast seeded with muscle-derived
[32] B.D. Cosgrove, P.M. Gilbert, E. Porpiglia, F. Mourkioti, S.P. Lee, S.Y. Corbel, et al., stem cells (MDSCs), Stem Cell Res. 27 (2018) 65–73, https://doi.org/10.1016/j.
Rejuvenation of the muscle stem cell population restores strength to injured aged scr.2018.01.008.
muscles, Nat. Med. 20 (2014) 255–264, https://doi.org/10.1038/nm.3464. [58] L. Boldrin, A. Malerba, L. Vitiello, E. Cimetta, M. Piccoli, C. Messina, et al.,
[33] D. Montarras, J. Morgan, C. Collins, F. Relaix, S. Zaffran, A. Cumano, et al., Direct Efficient delivery of human single fiber-derived muscle precursor cells via
isolation of satellite cells for skeletal muscle regeneration, Science 309 (2005) biocompatible scaffold, Cell Transplant. 17 (2008) 577–584, https://doi.org/
2064–2067, https://doi.org/10.1126/science.1114758. 10.3727/096368908785095980.
[34] P.M. Gilbert, K.L. Havenstrite, K.E.G. Magnusson, A. Sacco, N.A. Leonardi, [59] J. Zhu, R.E. Marchant, Design properties of hydrogel tissue-engineering scaffolds,
P. Kraft, et al., Substrate elasticity regulates skeletal muscle stem cell self-renewal Expet Rev. Med. Dev. 8 (2011) 607–626, https://doi.org/10.1586/erd.11.27.
in culture, Science 329 (2010) 1078–1081, https://doi.org/10.1126/ [60] R. Csapo, M. Gumpenberger, B. Wessner, Skeletal muscle extracellular matrix -
science.1191035. what do we know about its composition, regulation, and physiological roles? A
[35] G.W. Charville, T.H. Cheung, B. Yoo, P.J. Santos, G.K. Lee, J.B. Shrager, et al., Ex narrative review, Front. Physiol. 11 (2020) 253, https://doi.org/10.3389/
vivo expansion and in vivo self-renewal of human muscle stem cells, Stem Cell fphys.2020.00253.
Rep. 5 (2015) 621–632, https://doi.org/10.1016/j.stemcr.2015.08.004. [61] B.N. Brown, S.F. Badylak, Extracellular matrix as an inductive scaffold for
[36] B. Gharaibeh, A. Lu, J. Tebbets, B. Zheng, J. Feduska, M. Crisan, et al., Isolation of functional tissue reconstruction, Transl. Res. 163 (2014) 268–285, https://doi.
a slowly adhering cell fraction containing stem cells from murine skeletal muscle org/10.1016/j.trsl.2013.11.003.
by the preplate technique, Nat. Protoc. 3 (2008) 1501–1509, https://doi.org/ [62] S.F. Badylak, J.L. Dziki, B.M. Sicari, F. Ambrosio, M.L. Boninger, Mechanisms by
10.1038/nprot.2008.142. which acellular biologic scaffolds promote functional skeletal muscle restoration,
[37] B.L. Rodriguez, E.E. Vega-Soto, C.S. Kennedy, M.H. Nguyen, P.S. Cederna, L. Biomaterials 103 (2016) 128–136, https://doi.org/10.1016/j.
M. Larkin, A tissue engineering approach for repairing craniofacial volumetric biomaterials.2016.06.047.
muscle loss in a sheep following a 2, 4, and 6-month recovery, PLoS One 15 [63] A. Urciuolo, L. Urbani, S. Perin, P. Maghsoudlou, F. Scottoni, A. Gjinovci, et al.,
(2020), e0239152, https://doi.org/10.1371/journal.pone.0239152. Decellularised skeletal muscles allow functional muscle regeneration by
[38] K.W. VanDusen, B.C. Syverud, M.L. Williams, J.D. Lee, L.M. Larkin, Engineered promoting host cell migration, Sci. Rep. 8 (2018) 8398, https://doi.org/10.1038/
skeletal muscle units for repair of volumetric muscle loss in the tibialis anterior s41598-018-26371-y.
muscle of a rat, Tissue Eng. 20 (2014) 2920–2930, https://doi.org/10.1089/ten. [64] T.L. Sarrafian, S.C. Bodine, B. Murphy, J.K. Grayson, S.M. Stover, Extracellular
TEA.2014.0060. matrix scaffolds for treatment of large volume muscle injuries: a review, Vet.
[39] C.F. Bentzinger, Y.X. Wang, N.A. Dumont, M.A. Rudnicki, Cellular dynamics in Surg. 47 (2018) 524–535, https://doi.org/10.1111/vsu.12787.
the muscle satellite cell niche, EMBO Rep. 14 (2013) 1062–1072, https://doi.org/ [65] J.A. Passipieri, H.B. Baker, M. Siriwardane, M.D. Ellenburg, M. Vadhavkar, J.
10.1038/embor.2013.182. M. Saul, et al., Keratin hydrogel enhances in vivo skeletal muscle function in a rat
[40] R.W. Ten Broek, S. Grefte, J.W. Von den Hoff, Regulatory factors and cell model of volumetric muscle loss, Tissue Eng. 23 (2017) 556–571, https://doi.org/
populations involved in skeletal muscle regeneration, J. Cell. Physiol. 224 (2010) 10.1089/ten.TEA.2016.0458.
7–16, https://doi.org/10.1002/jcp.22127. [66] H.B. Baker, J.A. Passipieri, M. Siriwardane, M.D. Ellenburg, M. Vadhavkar, C.
[41] B.T. Corona, K. Garg, C.L. Ward, J.S. McDaniel, T.J. Walters, C.R. Rathbone, R. Bergman, et al., Cell and growth factor-loaded keratin hydrogels for treatment
Autologous minced muscle grafts: a tissue engineering therapy for the volumetric of volumetric muscle loss in a mouse model, Tissue Eng. 23 (2017) 572–584,
loss of skeletal muscle, Am. J. Physiol. Cell Physiol. 305 (2013) C761–C775, https://doi.org/10.1089/ten.TEA.2016.0457.
https://doi.org/10.1152/ajpcell.00189.2013. [67] S.M. Goldman, B.T. Corona, Co-delivery of micronized urinary bladder matrix
[42] C.L. Ward, B.E. Pollot, S.M. Goldman, S.M. Greising, J.C. Wenke, B.T. Corona, damps regenerative capacity of minced muscle grafts in the treatment of
Autologous minced muscle grafts improve muscle strength in a porcine model of volumetric muscle loss injuries, PLoS One 12 (2017), e0186593, https://doi.org/
volumetric muscle loss injury, J. Orthop. Trauma 30 (2016) e396–403, https:// 10.1371/journal.pone.0186593.
doi.org/10.1097/BOT.0000000000000673. [68] J.A. Burdick, G.D. Prestwich, Hyaluronic acid hydrogels for biomedical
[43] K.G. Chen, B.S. Mallon, R.D.G. McKay, P.G. Robey, Human pluripotent stem cell applications, Adv Mater Weinheim 23 (2011) H41–H56, https://doi.org/
culture: considerations for maintenance, expansion, and therapeutics, Cell Stem 10.1002/adma.201003963.
Cell 14 (2014) 13–26, https://doi.org/10.1016/j.stem.2013.12.005. [69] B. Kasukonis, J. Kim, L. Brown, J. Jones, S. Ahmadi, T. Washington, et al.,
[44] J. Wu, N. Matthias, S. Bhalla, R. Darabi, Evaluation of the therapeutic potential of Codelivery of infusion decellularized skeletal muscle with minced muscle
human iPSCs in a murine model of VML, Mol. Ther. (2020), https://doi.org/ autografts improved recovery from volumetric muscle loss injury in a rat model,
10.1016/j.ymthe.2020.09.012. Tissue Eng. 22 (2016) 1151–1163, https://doi.org/10.1089/ten.TEA.2016.0134.
[45] L. Rao, Y. Qian, A. Khodabukus, T. Ribar, N. Bursac, Engineering human [70] K. Hosoyama, C. Lazurko, M. Muñoz, C.D. McTiernan, E.I. Alarcon, Peptide-based
pluripotent stem cells into a functional skeletal muscle tissue, Nat. Commun. 9 functional biomaterials for soft-tissue repair, Front Bioeng Biotechnol 7 (2019)
(2018) 126, https://doi.org/10.1038/s41467-017-02636-4. 205, https://doi.org/10.3389/fbioe.2019.00205.
[46] Y. Mizuno, H. Chang, K. Umeda, A. Niwa, T. Iwasa, T. Awaya, et al., Generation of [71] R. Agarwal, A.J. García, Biomaterial strategies for engineering implants for
skeletal muscle stem/progenitor cells from murine induced pluripotent stem cells, enhanced osseointegration and bone repair, Adv. Drug Deliv. Rev. 94 (2015)
Faseb. J. 24 (2010) 2245–2253, https://doi.org/10.1096/fj.09-137174. 53–62, https://doi.org/10.1016/j.addr.2015.03.013.
[47] E. van der Wal, P. Herrero-Hernandez, R. Wan, M. Broeders, S.L.M. In ’t Groen, T. [72] L. Perry, S. Landau, M.Y. Flugelman, S. Levenberg, Genetically engineered human
J.M. van Gestel, et al., Large-scale expansion of human iPSC-derived skeletal muscle transplant enhances murine host neovascularization and myogenesis,
muscle cells for disease modeling and cell-based therapeutic strategies, Stem Cell Commun Biol 1 (2018) 161, https://doi.org/10.1038/s42003-018-0161-0.
Rep. 10 (2018) 1975, https://doi.org/10.1016/j.stemcr.2018.04.002, 90. [73] N. Ziemkiewicz, M. Talovic, J. Madsen, L. Hill, R. Scheidt, A. Patel, et al.,
[48] S.M. Maffioletti, S. Sarcar, A.B.H. Henderson, I. Mannhardt, L. Pinton, L.A. Moyle, Laminin-111 functionalized polyethylene glycol hydrogels support myogenic
et al., Three-Dimensional human iPSC-derived artificial skeletal muscles model activity in vitro, Biomed. Mater. 13 (2018), 065007, https://doi.org/10.1088/
muscular dystrophies and enable multilineage tissue engineering, Cell Rep. 23 1748-605X/aad915.
(2018) 899–908, https://doi.org/10.1016/j.celrep.2018.03.091. [74] Y. Shandalov, D. Egozi, J. Koffler, D. Dado-Rosenfeld, D. Ben-Shimol, A. Freiman,
[49] I. Gutierrez-Aranda, V. Ramos-Mejia, C. Bueno, M. Munoz-Lopez, P.J. Real, et al., An engineered muscle flap for reconstruction of large soft tissue defects,
A. Mácia, et al., Human induced pluripotent stem cells develop teratoma more Proc. Natl. Acad. Sci. U.S.A. 111 (2014) 6010–6015, https://doi.org/10.1073/
efficiently and faster than human embryonic stem cells regardless the site of pnas.1402679111.
injection, Stem Cell. 28 (2010) 1568–1570, https://doi.org/10.1002/stem.471. [75] U. Hersel, C. Dahmen, H. Kessler, RGD modified polymers: biomaterials for
[50] Y. Miyagoe-Suzuki, S. Takeda, Skeletal muscle generated from induced stimulated cell adhesion and beyond, Biomaterials 24 (2003) 4385–4415,
pluripotent stem cells - induction and application, World J. Stem Cell. 9 (2017) https://doi.org/10.1016/s0142-9612(03)00343-0.
89–97, https://doi.org/10.4252/wjsc.v9.i6.89. [76] C. Borselli, C.A. Cezar, D. Shvartsman, H.H. Vandenburgh, D.J. Mooney, The role
[51] T. Zhao, Z.-N. Zhang, Z. Rong, Y. Xu, Immunogenicity of induced pluripotent stem of multifunctional delivery scaffold in the ability of cultured myoblasts to
cells, Nature 474 (2011) 212–215, https://doi.org/10.1038/nature10135. promote muscle regeneration, Biomaterials 32 (2011) 8905–8914, https://doi.
[52] M.T. Webster, U. Manor, J. Lippincott-Schwartz, C.-M. Fan, Intravital imaging org/10.1016/j.biomaterials.2011.08.019.
reveals ghost fibers as architectural units guiding myogenic progenitors during [77] J.M. Silva Garcia, A. Panitch, S. Calve, Functionalization of hyaluronic acid
regeneration, Cell Stem Cell 18 (2016) 243–252, https://doi.org/10.1016/j. hydrogels with ECM-derived peptides to control myoblast behavior, Acta
stem.2015.11.005. Biomater. 84 (2019) 169–179, https://doi.org/10.1016/j.actbio.2018.11.030.
[53] M.M. Smoak, A.G. Mikos, Advances in biomaterials for skeletal muscle [78] A.J. Engler, M.A. Griffin, S. Sen, C.G. Bönnemann, H.L. Sweeney, D.E. Discher,
engineering and obstacles still to overcome, Mater Today Bio 7 (2020) 100069, Myotubes differentiate optimally on substrates with tissue-like stiffness:
https://doi.org/10.1016/j.mtbio.2020.100069.

10
I. Eugenis et al. Biomaterials 278 (2021) 121173

pathological implications for soft or stiff microenvironments, J. Cell Biol. 166 [103] L. Baoge, E. Van Den Steen, S. Rimbaut, N. Philips, E. Witvrouw, K.F. Almqvist, et
(2004) 877–887, https://doi.org/10.1083/jcb.200405004. al., Treatment of skeletal muscle injury: a review, ISRN Orthop (2012) 689012,
[79] H. Zhang, L. Zhou, W. Zhang, Control of scaffold degradation in tissue https://doi.org/10.5402/2012/689012.
engineering: a review, Tissue Eng. B Rev. 20 (2014) 492–502, https://doi.org/ [104] F. Le Grand, M.A. Rudnicki, Skeletal muscle satellite cells and adult myogenesis,
10.1089/ten.TEB.2013.0452. Curr. Opin. Cell Biol. 19 (2007) 628–633, https://doi.org/10.1016/j.
[80] Q.L. Loh, C. Choong, Three-dimensional scaffolds for tissue engineering ceb.2007.09.012.
applications: role of porosity and pore size, Tissue Eng. B Rev. 19 (2013) [105] Y.M. Ju, A. Atala, J.J. Yoo, S.J. Lee, In situ regeneration of skeletal muscle tissue
485–502, https://doi.org/10.1089/ten.TEB.2012.0437. through host cell recruitment, Acta Biomater. 10 (2014) 4332–4339, https://doi.
[81] A.C. Panayi, L. Smit, N. Hays, K. Udeh, Y. Endo, B. Li, et al., A porous collagen- org/10.1016/j.actbio.2014.06.022.
GAG scaffold promotes muscle regeneration following volumetric muscle loss [106] J.M. Grasman, D.M. Do, R.L. Page, G.D. Pins, Rapid release of growth factors
injury, Wound Repair Regen. 28 (2020) 61–74, https://doi.org/10.1111/ regenerates force output in volumetric muscle loss injuries, Biomaterials 72
wrr.12768. (2015) 49–60, https://doi.org/10.1016/j.biomaterials.2015.08.047.
[82] G.J. Haas, A.J. Dunn, M. Marcinczyk, M. Talovic, M. Schwartz, R. Scheidt, et al., [107] F. Le Grand, A.E. Jones, V. Seale, A. Scimè, M.A. Rudnicki, Wnt7a activates the
Biomimetic sponges for regeneration of skeletal muscle following trauma, planar cell polarity pathway to drive the symmetric expansion of satellite stem
J. Biomed. Mater. Res. 107 (2019) 92–103, https://doi.org/10.1002/jbm. cells, Cell Stem Cell 4 (2009) 535–547, https://doi.org/10.1016/j.
a.36535. stem.2009.03.013.
[83] I.-C. Liao, J.B. Liu, N. Bursac, K.W. Leong, Effect of electromechanical stimulation [108] J. von Maltzahn, C.F. Bentzinger, M.A. Rudnicki, Wnt7a-Fzd7 signalling directly
on the maturation of myotubes on aligned electrospun fibers, Cell. Mol. Bioeng. 1 activates the Akt/mTOR anabolic growth pathway in skeletal muscle, Nat. Cell
(2008) 133–145, https://doi.org/10.1007/s12195-008-0021-y. Biol. 14 (2011) 186–191, https://doi.org/10.1038/ncb2404.
[84] S. Jana, S.K.L. Levengood, M. Zhang, Anisotropic materials for skeletal-muscle- [109] C.F. Bentzinger, J. von Maltzahn, N.A. Dumont, D.A. Stark, Y.X. Wang, K. Nhan,
tissue engineering, Adv Mater Weinheim 28 (2016) 10588–10612, https://doi. et al., Wnt7a stimulates myogenic stem cell motility and engraftment resulting in
org/10.1002/adma.201600240. improved muscle strength, J. Cell Biol. 205 (2014) 97–111, https://doi.org/
[85] S. Politi, F. Carotenuto, A. Rinaldi, P. Di Nardo, V. Manzari, M.C. Albertini, et al., 10.1083/jcb.201310035.
Smart ECM-based electrospun biomaterials for skeletal muscle regeneration, [110] W.M. Han, M. Mohiuddin, S.E. Anderson, A.J. García, Y.C. Jang, Co-delivery of
Nanomaterials 10 (2020), https://doi.org/10.3390/nano10091781. Wnt7a and muscle stem cells using synthetic bioadhesive hydrogel enhances
[86] J. Kim, B. Kasukonis, K. Roberts, G. Dunlap, L. Brown, T. Washington, et al., Graft murine muscle regeneration and cell migration during engraftment, Acta
alignment impacts the regenerative response of skeletal muscle after volumetric Biomater. 94 (2019) 243–252, https://doi.org/10.1016/j.actbio.2019.06.025.
muscle loss in a rat model, Acta Biomater. 105 (2020) 191–202, https://doi.org/ [111] E. Hill, T. Boontheekul, D.J. Mooney, Regulating activation of transplanted cells
10.1016/j.actbio.2020.01.024. controls tissue regeneration, Proc. Natl. Acad. Sci. U.S.A. 103 (2006) 2494–2499,
[87] K.H. Patel, M. Talovic, A.J. Dunn, A. Patel, S. Vendrell, M. Schwartz, et al., https://doi.org/10.1073/pnas.0506004103.
Aligned nanofibers of decellularized muscle extracellular matrix for volumetric [112] L.L. Tortorella, D.J. Milasincic, P.F. Pilch, Critical proliferation-independent
muscle loss, J. Biomed. Mater. Res. B Appl. Biomater. 108 (2020) 2528–2537, window for basic fibroblast growth factor repression of myogenesis via the p42/
https://doi.org/10.1002/jbm.b.34584. p44 MAPK signaling pathway, J. Biol. Chem. 276 (2001) 13709–13717, https://
[88] K.H. Nakayama, C. Alcazar, G. Yang, M. Quarta, P. Paine, L. Doan, et al., doi.org/10.1074/jbc.M100091200.
Rehabilitative exercise and spatially patterned nanofibrillar scaffolds enhance [113] K.J. Miller, D. Thaloor, S. Matteson, G.K. Pavlath, Hepatocyte growth factor
vascularization and innervation following volumetric muscle loss, Npj Regen Med affects satellite cell activation and differentiation in regenerating skeletal muscle,
3 (2018) 16, https://doi.org/10.1038/s41536-018-0054-3. Am. J. Physiol. Cell Physiol. 278 (2000) C174–C181, https://doi.org/10.1152/
[89] C. Blake, O. Massey, M. Boyd-Moss, K. Firipis, A. Rifai, S. Franks, et al., Replace ajpcell.2000.278.1.C174.
and repair: biomimetic bioprinting for effective muscle engineering, APL [114] I. Stratos, H. Madry, R. Rotter, A. Weimer, J. Graff, M. Cucchiarini, et al.,
Bioengineering 5 (2021), 031502, https://doi.org/10.1063/5.0040764. Fibroblast growth factor-2-overexpressing myoblasts encapsulated in alginate
[90] P. Zhuang, J. An, C.K. Chua, L.P. Tan, Bioprinting of 3D in vitro skeletal muscle spheres increase proliferation, reduce apoptosis, induce adipogenesis, and
models: a review, Mater. Des. 193 (2020) 108794, https://doi.org/10.1016/j. enhance regeneration following skeletal muscle injury in rats, Tissue Eng. 17
matdes.2020.108794. (2011) 2867–2877, https://doi.org/10.1089/ten.tea.2011.0239.
[91] J.H. Kim, Y.-J. Seol, I.K. Ko, H.-W. Kang, Y.K. Lee, J.J. Yoo, et al., 3D bioprinted [115] C.A. Mitchell, J.K. McGeachie, M.D. Grounds, The exogenous administration of
human skeletal muscle constructs for muscle function restoration, Sci. Rep. 8 basic fibroblast growth factor to regenerating skeletal muscle in mice does not
(2018) 12307, https://doi.org/10.1038/s41598-018-29968-5. enhance the process of regeneration, Growth Factors 13 (1996) 37–55, https://
[92] J.H. Kim, I.K. Ko, M.J. Jeon, I. Kim, M.M. Vanschaayk, A. Atala, et al., Pelvic floor doi.org/10.3109/08977199609034565.
muscle function recovery using biofabricated tissue constructs with [116] A. Philippou, A. Halapas, M. Maridaki, M. Koutsilieris, Type I insulin-like growth
neuromuscular junctions, Acta Biomater. 121 (2021) 237–249, https://doi.org/ factor receptor signaling in skeletal muscle regeneration and hypertrophy,
10.1016/j.actbio.2020.12.012. J. Musculoskelet. Neuronal Interact. 7 (2007) 208–218.
[93] H. Lee, W. Kim, J. Lee, K.S. Park, J.J. Yoo, A. Atala, et al., Self-aligned myofibers [117] S.B.P. Chargé, M.A. Rudnicki, Cellular and molecular regulation of muscle
in 3D bioprinted extracellular matrix-based construct accelerate skeletal muscle regeneration, Physiol. Rev. 84 (2004) 209–238, https://doi.org/10.1152/
function restoration, Appl. Phys. Rev. 8 (2021), 021405, https://doi.org/ physrev.00019.2003.
10.1063/5.0039639. [118] C.A. Alcazar, C. Hu, T.A. Rando, N.F. Huang, K.H. Nakayama, Transplantation of
[94] Y.-J. Choi, Y.-J. Jun, D.Y. Kim, H.-G. Yi, S.-H. Chae, J. Kang, et al., A 3D cell insulin-like growth factor-1 laden scaffolds combined with exercise promotes
printed muscle construct with tissue-derived bioink for the treatment of neuroregeneration and angiogenesis in a preclinical muscle injury model,
volumetric muscle loss, Biomaterials 206 (2019) 160–169, https://doi.org/ Biomater. Sci. 8 (2020) 5376–5389, https://doi.org/10.1039/d0bm00990c.
10.1016/j.biomaterials.2019.03.036. [119] H. Lee, Y.M. Ju, I. Kim, E. Elsangeedy, J.H. Lee, J.J. Yoo, et al., A novel
[95] J.H. Kim, I. Kim, Y.-J. Seol, I.K. Ko, J.J. Yoo, A. Atala, et al., Neural cell decellularized skeletal muscle-derived ECM scaffolding system for in situ muscle
integration into 3D bioprinted skeletal muscle constructs accelerates restoration regeneration, Methods 171 (2020) 77–85, https://doi.org/10.1016/j.
of muscle function, Nat. Commun. 11 (2020) 1025, https://doi.org/10.1038/ ymeth.2019.06.027.
s41467-020-14930-9. [120] J. Rouwkema, N.C. Rivron, C.A. van Blitterswijk, Vascularization in tissue
[96] C.S. Russell, A. Mostafavi, J.P. Quint, A.C. Panayi, K. Baldino, T.J. Williams, et al., engineering, Trends Biotechnol. 26 (2008) 434–441, https://doi.org/10.1016/j.
In situ printing of adhesive hydrogel scaffolds for the treatment of skeletal muscle tibtech.2008.04.009.
injuries, ACS Appl Bio Mater 3 (2020) 1568–1579, https://doi.org/10.1021/ [121] C. Tomasina, T. Bodet, C. Mota, L. Moroni, S. Camarero-Espinosa, Bioprinting
acsabm.9b01176. vasculature: materials, cells and emergent techniques, Materials 12 (2019),
[97] A. Urciuolo, I. Poli, L. Brandolino, P. Raffa, V. Scattolini, C. Laterza, et al., https://doi.org/10.3390/ma12172701.
Intravital three-dimensional bioprinting, Nat. Biomed. Eng. 4 (2020) 901–915, [122] M.E. Carnes, G.D. Pins, Skeletal muscle tissue engineering: biomaterials-based
https://doi.org/10.1038/s41551-020-0568-z. strategies for the treatment of volumetric muscle loss, Bioengineering 7 (2020),
[98] D.E. Discher, D.J. Mooney, P.W. Zandstra, Growth factors, matrices, and forces https://doi.org/10.3390/bioengineering7030085.
combine and control stem cells, Science 324 (2009) 1673–1677, https://doi.org/ [123] B.T. Corona, K.E. Flanagan, C.M. Brininger, S.M. Goldman, J.A. Call, S.
10.1126/science.1171643. M. Greising, Impact of volumetric muscle loss injury on persistent motoneuron
[99] B.C. Syverud, K.W. VanDusen, L.M. Larkin, Growth factors for skeletal muscle axotomy, Muscle Nerve 57 (2018) 799–807, https://doi.org/10.1002/
tissue engineering, Cells Tissues Organs 202 (2016) 169–179, https://doi.org/ mus.26016.
10.1159/000444671. [124] E. Gutmann, J. Zelená, Morphological changes in the denervated muscle, in:
[100] R. Tatsumi, J.E. Anderson, C.J. Nevoret, O. Halevy, R.E. Allen, HGF/SF is present E. Gutmann (Ed.), The Denervated Muscle, Springer US, Boston, MA, 1962,
in normal adult skeletal muscle and is capable of activating satellite cells, Dev. pp. 57–102, https://doi.org/10.1007/978-1-4899-4854-0_3.
Biol. 194 (1998) 114–128, https://doi.org/10.1006/dbio.1997.8803. [125] H. Yin, F. Price, M.A. Rudnicki, Satellite cells and the muscle stem cell niche,
[101] B. Pawlikowski, T.O. Vogler, K. Gadek, B.B. Olwin, Regulation of skeletal muscle Physiol. Rev. 93 (2013) 23–67, https://doi.org/10.1152/physrev.00043.2011.
stem cells by fibroblast growth factors, Dev. Dynam. 246 (2017) 359–367, [126] J. Gilbert-Honick, S.R. Iyer, S.M. Somers, R.M. Lovering, K. Wagner, H.-Q. Mao, et
https://doi.org/10.1002/dvdy.24495. al., Engineering functional and histological regeneration of vascularized skeletal
[102] K. Lee, E.A. Silva, D.J. Mooney, Growth factor delivery-based tissue engineering: muscle, Biomaterials 164 (2018) 70–79, https://doi.org/10.1016/j.
general approaches and a review of recent developments, J. R. Soc. Interface 8 biomaterials.2018.02.006.
(2011) 153–170, https://doi.org/10.1098/rsif.2010.0223. [127] F.S. Frueh, T. Später, C. Scheuer, M.D. Menger, M.W. Laschke, Isolation of murine
adipose tissue-derived microvascular fragments as vascularization units for tissue
engineering, JoVE (2017), https://doi.org/10.3791/55721.

11
I. Eugenis et al. Biomaterials 278 (2021) 121173

[128] J.S. McDaniel, M. Pilia, C.L. Ward, B.E. Pollot, C.R. Rathbone, Characterization regeneration following volumetric muscle loss, Biomaterials 255 (2020) 120154,
and multilineage potential of cells derived from isolated microvascular fragments, https://doi.org/10.1016/j.biomaterials.2020.120154.
J. Surg. Res. 192 (2014) 214–222, https://doi.org/10.1016/j.jss.2014.05.047. [153] J.W. Godwin, A.R. Pinto, N.A. Rosenthal, Chasing the recipe for a pro-
[129] M. Pilia, J.S. McDaniel, T. Guda, X.K. Chen, R.P. Rhoads, R.E. Allen, et al., regenerative immune system, Semin. Cell Dev. Biol. 61 (2017) 71–79, https://doi.
Transplantation and perfusion of microvascular fragments in a rodent model of org/10.1016/j.semcdb.2016.08.008.
volumetric muscle loss injury, Eur. Cell. Mater. 28 (2014) 11–23, https://doi.org/ [154] T.A. Wynn, T.R. Ramalingam, Mechanisms of fibrosis: therapeutic translation for
10.22203/ecm.v028a02, discussion 23. fibrotic disease, Nat. Med. 18 (2012) 1028–1040, https://doi.org/10.1038/
[130] M.-T. Li, M.A. Ruehle, H.Y. Stevens, N. Servies, N.J. Willett, S. Karthikeyakannan, nm.2807.
et al., * skeletal myoblast-seeded vascularized tissue scaffolds in the treatment of [155] A.B. Aurora, E.N. Olson, Immune modulation of stem cells and regeneration, Cell
a large volumetric muscle defect in the rat biceps femoris muscle, Tissue Eng. 23 Stem Cell 15 (2014) 14–25, https://doi.org/10.1016/j.stem.2014.06.009.
(2017) 989–1000, https://doi.org/10.1089/ten.TEA.2016.0523. [156] Z. Julier, A.J. Park, P.S. Briquez, M.M. Martino, Promoting tissue regeneration by
[131] S. Das, K.D. Browne, F.A. Laimo, J.C. Maggiore, M.C. Hilman, H. Kaisaier, et al., modulating the immune system, Acta Biomater. 53 (2017) 13–28, https://doi.
Pre-innervated tissue-engineered muscle promotes a pro-regenerative org/10.1016/j.actbio.2017.01.056.
microenvironment following volumetric muscle loss, Commun Biol 3 (2020) 330, [157] S.J. Forbes, N. Rosenthal, Preparing the ground for tissue regeneration: from
https://doi.org/10.1038/s42003-020-1056-4. mechanism to therapy, Nat. Med. 20 (2014) 857–869, https://doi.org/10.1038/
[132] H.-W. Kang, S.J. Lee, I.K. Ko, C. Kengla, J.J. Yoo, A. Atala, A 3D bioprinting nm.3653.
system to produce human-scale tissue constructs with structural integrity, Nat. [158] J.G. Tidball, Regulation of muscle growth and regeneration by the immune
Biotechnol. 34 (2016) 312–319, https://doi.org/10.1038/nbt.3413. system, Nat. Rev. Immunol. 17 (2017) 165–178, https://doi.org/10.1038/
[133] X. Yu, T. Zhang, Y. Li, 3D printing and bioprinting nerve conduits for neural tissue nri.2016.150.
engineering, Polymers 12 (2020), https://doi.org/10.3390/polym12081637. [159] T.A. Wynn, K.M. Vannella, Macrophages in tissue repair, regeneration, and
[134] N. Bhardwaj, S.C. Kundu, Electrospinning: a fascinating fiber fabrication fibrosis, Immunity 44 (2016) 450–462, https://doi.org/10.1016/j.
technique, Biotechnol. Adv. 28 (2010) 325–347, https://doi.org/10.1016/j. immuni.2016.02.015.
biotechadv.2010.01.004. [160] M.N. Wosczyna, T.A. Rando, A muscle stem cell support group: coordinated
[135] R.D. Pedde, B. Mirani, A. Navaei, T. Styan, S. Wong, M. Mehrali, et al., Emerging cellular responses in muscle regeneration, Dev. Cell 46 (2018) 135–143, https://
biofabrication strategies for engineering complex tissue constructs, Adv Mater doi.org/10.1016/j.devcel.2018.06.018.
Weinheim 29 (2017), https://doi.org/10.1002/adma.201606061. [161] D.R. Lemos, F. Babaeijandaghi, M. Low, C.-K. Chang, S.T. Lee, D. Fiore, et al.,
[136] W.E. Teo, S. Ramakrishna, A review on electrospinning design and nanofibre Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF-
assemblies, Nanotechnology 17 (2006) R89–R106, https://doi.org/10.1088/ mediated apoptosis of fibro/adipogenic progenitors, Nat. Med. 21 (2015)
0957-4484/17/14/R01. 786–794, https://doi.org/10.1038/nm.3869.
[137] K.H. Nakayama, M. Quarta, P. Paine, C. Alcazar, I. Karakikes, V. Garcia, et al., [162] C. Moratal, J. Raffort, N. Arrighi, S. Rekima, S. Schaub, C.A. Dechesne, et al., IL-
Treatment of volumetric muscle loss in mice using nanofibrillar scaffolds 1β- and IL-4-polarized macrophages have opposite effects on adipogenesis of
enhances vascular organization and integration, Commun Biol 2 (2019) 170, intramuscular fibro-adipogenic progenitors in humans, Sci. Rep. 8 (2018) 17005,
https://doi.org/10.1038/s42003-019-0416-4. https://doi.org/10.1038/s41598-018-35429-w.
[138] J. Li, Y.-P. Zhang, R.S. Kirsner, Angiogenesis in wound repair: angiogenic growth [163] Q. Zhao, H. Ren, Z. Han, Mesenchymal stem cells: immunomodulatory capability
factors and the extracellular matrix, Microsc. Res. Tech. 60 (2003) 107–114, and clinical potential in immune diseases, J. Cellular. Immunotherapy 2 (2016)
https://doi.org/10.1002/jemt.10249. 3–20, https://doi.org/10.1016/j.jocit.2014.12.001.
[139] K. Doi, T. Ikeda, A. Marui, T. Kushibiki, Y. Arai, K. Hirose, et al., Enhanced [164] M.F. Pittenger, D.E. Discher, B.M. Péault, D.G. Phinney, J.M. Hare, A.I. Caplan,
angiogenesis by gelatin hydrogels incorporating basic fibroblast growth factor in Mesenchymal stem cell perspective: cell biology to clinical progress, Npj Regen
rabbit model of hind limb ischemia, Heart Ves. 22 (2007) 104–108, https://doi. Med 4 (2019) 22, https://doi.org/10.1038/s41536-019-0083-6.
org/10.1007/s00380-006-0934-0. [165] X. Qiu, S. Liu, H. Zhang, B. Zhu, Y. Su, C. Zheng, et al., Mesenchymal stem cells
[140] H. Layman, M.-G. Spiga, T. Brooks, S. Pham, K.A. Webster, F.M. Andreopoulos, and extracellular matrix scaffold promote muscle regeneration by synergistically
The effect of the controlled release of basic fibroblast growth factor from ionic regulating macrophage polarization toward the M2 phenotype, Stem Cell Res.
gelatin-based hydrogels on angiogenesis in a murine critical limb ischemic model, Ther. 9 (2018) 88, https://doi.org/10.1186/s13287-018-0821-5.
Biomaterials 28 (2007) 2646–2654, https://doi.org/10.1016/j. [166] H. Huang, J. Liu, H. Hao, D. Chen, L. Zhizhong, M. Li, et al., Preferred M2
biomaterials.2007.01.044. polarization by ASC-based hydrogel accelerated angiogenesis and myogenesis in
[141] L. Wang, L. Cao, J. Shansky, Z. Wang, D. Mooney, H. Vandenburgh, Minimally volumetric muscle loss rats, Stem Cell. Int. (2017) 2896874, https://doi.org/
invasive approach to the repair of injured skeletal muscle with a shape-memory 10.1155/2017/2896874.
scaffold, Mol. Ther. 22 (2014) 1441–1449, https://doi.org/10.1038/mt.2014.78. [167] J. Gilbert-Honick, B. Ginn, Y. Zhang, S. Salehi, K.R. Wagner, H.-Q. Mao, et al.,
[142] E.A. Silva, D.J. Mooney, Spatiotemporal control of vascular endothelial growth Adipose-derived stem/stromal cells on electrospun fibrin microfiber bundles
factor delivery from injectable hydrogels enhances angiogenesis, J. Thromb. enable moderate muscle reconstruction in a volumetric muscle loss model, Cell
Haemostasis 5 (2007) 590–598, https://doi.org/10.1111/j.1538- Transplant. 27 (2018) 1644–1656, https://doi.org/10.1177/
7836.2007.02386.x. 0963689718805370.
[143] J. Yamaguchi, K.F. Kusano, O. Masuo, A. Kawamoto, M. Silver, S. Murasawa, et [168] S. Franz, S. Rammelt, D. Scharnweber, J.C. Simon, Immune responses to implants
al., Stromal cell-derived factor-1 effects on ex vivo expanded endothelial - a review of the implications for the design of immunomodulatory biomaterials,
progenitor cell recruitment for ischemic neovascularization, Circulation 107 Biomaterials 32 (2011) 6692–6709, https://doi.org/10.1016/j.
(2003) 1322–1328, https://doi.org/10.1161/01.cir.0000055313.77510.22. biomaterials.2011.05.078.
[144] S.S. Said, H. Yin, M. Elfarnawany, Z. Nong, C. O’Neil, H. Leong, et al., Fortifying [169] D.P. Vasconcelos, A.P. Águas, M.A. Barbosa, P. Pelegrín, J.N. Barbosa, The
angiogenesis in ischemic muscle with FGF9-loaded electrospun poly(Ester Amide) inflammasome in host response to biomaterials: bridging inflammation and tissue
fibers, Adv. Healthc Mater. 8 (2019), e1801294, https://doi.org/10.1002/ regeneration, Acta Biomater. 83 (2019) 1–12, https://doi.org/10.1016/j.
adhm.201801294. actbio.2018.09.056.
[145] T.S. Zaitseva, C. Alcazar, M. Zamani, L. Hou, S. Sawamura, E. Yakubov, et al., [170] L. Chung, D.R. Maestas, F. Housseau, J.H. Elisseeff, Key players in the immune
Aligned nanofibrillar scaffolds for controlled delivery of modified mRNA, Tissue response to biomaterial scaffolds for regenerative medicine, Adv. Drug Deliv. Rev.
Eng. 25 (2019) 121–130, https://doi.org/10.1089/ten.TEA.2017.0494. 114 (2017) 184–192, https://doi.org/10.1016/j.addr.2017.07.006.
[146] S.M. Jay, B.R. Shepherd, J.P. Bertram, J.S. Pober, W.M. Saltzman, Engineering of [171] J.I. Andorko, C.M. Jewell, Designing biomaterials with immunomodulatory
multifunctional gels integrating highly efficient growth factor delivery with properties for tissue engineering and regenerative medicine, Bioeng. Transl. Med.
endothelial cell transplantation, Faseb. J. 22 (2008) 2949–2956, https://doi.org/ 2 (2017) 139–155, https://doi.org/10.1002/btm2.10063.
10.1096/fj.08-108803. [172] K. Sadtler, A. Singh, M.T. Wolf, X. Wang, D.M. Pardoll, J.H. Elisseeff, Design,
[147] J. Li, Y. Wei, K. Liu, C. Yuan, Y. Tang, Q. Quan, et al., Synergistic effects of FGF-2 clinical translation and immunological response of biomaterials in regenerative
and PDGF-BB on angiogenesis and muscle regeneration in rabbit hindlimb medicine, Nat. Rev. Mater. 1 (2016) 16040, https://doi.org/10.1038/
ischemia model, Microvasc. Res. 80 (2010) 10–17, https://doi.org/10.1016/j. natrevmats.2016.40.
mvr.2009.12.002. [173] K. Sadtler, B.W. Allen, K. Estrellas, F. Housseau, D.M. Pardoll, J.H. Elisseeff, The
[148] L.E. Benjamin, I. Hemo, E. Keshet, A plasticity window for blood vessel scaffold immune microenvironment: biomaterial-mediated immune polarization
remodelling is defined by pericyte coverage of the preformed endothelial network in traumatic and nontraumatic applications, Tissue Eng. 23 (2017) 1044–1053,
and is regulated by PDGF-B and VEGF, Development 125 (1998) 1591–1598. https://doi.org/10.1089/ten.TEA.2016.0304.
[149] T.P. Richardson, M.C. Peters, A.B. Ennett, D.J. Mooney, Polymeric system for dual [174] J.L. Dziki, B.M. Sicari, M.T. Wolf, M.C. Cramer, S.F. Badylak, Immunomodulation
growth factor delivery, Nat. Biotechnol. 19 (2001) 1029–1034, https://doi.org/ and mobilization of progenitor cells by extracellular matrix bioscaffolds for
10.1038/nbt1101-1029. volumetric muscle loss treatment, Tissue Eng. 22 (2016) 1129–1139, https://doi.
[150] P.J. Kingham, G. Terenghi, Bioengineered nerve regeneration and muscle org/10.1089/ten.TEA.2016.0340.
reinnervation, J. Anat. 209 (2006) 511–526, https://doi.org/10.1111/j.1469- [175] A. Aurora, B.T. Corona, T.J. Walters, A porcine urinary bladder matrix does not
7580.2006.00623.x. recapitulate the spatiotemporal macrophage response of muscle regeneration
[151] J.B. Scott, C.L. Ward, B.T. Corona, M.R. Deschenes, B.S. Harrison, J.M. Saul, et al., after volumetric muscle loss injury, Cells Tissues Organs 202 (2016) 189–201,
Achieving acetylcholine receptor clustering in tissue-engineered skeletal muscle https://doi.org/10.1159/000447582.
constructs in vitro through a materials-directed agrin delivery approach, Front. [176] K. Sadtler, K. Estrellas, B.W. Allen, M.T. Wolf, H. Fan, A.J. Tam, et al., Developing
Pharmacol. 7 (2016) 508, https://doi.org/10.3389/fphar.2016.00508. a pro-regenerative biomaterial scaffold microenvironment requires T helper 2
[152] J. Gilbert-Honick, S.R. Iyer, S.M. Somers, H. Takasuka, R.M. Lovering, K. cells, Science 352 (2016) 366–370, https://doi.org/10.1126/science.aad9272.
R. Wagner, et al., Engineering 3D skeletal muscle primed for neuromuscular

12
I. Eugenis et al. Biomaterials 278 (2021) 121173

[177] K. Sadtler, M.T. Wolf, S. Ganguly, C.A. Moad, L. Chung, S. Majumdar, et al., Neurosci. 10 (2019) 1411–1419, https://doi.org/10.1021/
Divergent immune responses to synthetic and biological scaffolds, Biomaterials acschemneuro.8b00452.
192 (2019) 405–415, https://doi.org/10.1016/j.biomaterials.2018.11.002. [183] C.L. San Emeterio, C.E. Olingy, Y. Chu, E.A. Botchwey, Selective recruitment of
[178] A. Ibáñez-Fonseca, S. Santiago Maniega, D. Gorbenko Del Blanco, B. Catalán non-classical monocytes promotes skeletal muscle repair, Biomaterials 117
Bernardos, A. Vega Castrillo, Á.J. Álvarez Barcia, et al., Elastin-like recombinamer (2017) 32–43, https://doi.org/10.1016/j.biomaterials.2016.11.021.
hydrogels for improved skeletal muscle healing through modulation of [184] B.T. Corona, J.C. Rivera, J.C. Wenke, S.M. Greising, Tacrolimus as an adjunct to
macrophage polarization, Front Bioeng Biotechnol 8 (2020) 413, https://doi.org/ autologous minced muscle grafts for the repair of a volumetric muscle loss injury,
10.3389/fbioe.2020.00413. J. Exp ORTOP 4 (2017) 36, https://doi.org/10.1186/s40634-017-0112-6.
[179] M. Marcinczyk, A. Dunn, G. Haas, J. Madsen, R. Scheidt, K. Patel, et al., The effect [185] M.R. Izadi, A. Habibi, Z. Khodabandeh, M. Nikbakht, Synergistic effect of high-
of laminin-111 hydrogels on muscle regeneration in a murine model of injury, intensity interval training and stem cell transplantation with amniotic membrane
Tissue Eng. 25 (2019) 1001–1012, https://doi.org/10.1089/ten.TEA.2018.0200. scaffold on repair and rehabilitation after volumetric muscle loss injury, Cell
[180] A.O. Awojoodu, M.E. Ogle, L.S. Sefcik, D.T. Bowers, K. Martin, K.L. Brayman, et Tissue Res. (2020), https://doi.org/10.1007/s00441-020-03304-8.
al., Sphingosine 1-phosphate receptor 3 regulates recruitment of anti- [186] B.M. Sicari, J.P. Rubin, C.L. Dearth, M.T. Wolf, F. Ambrosio, M. Boninger, et al.,
inflammatory monocytes to microvessels during implant arteriogenesis, Proc. An acellular biologic scaffold promotes skeletal muscle formation in mice and
Natl. Acad. Sci. U.S.A. 110 (2013) 13785–13790, https://doi.org/10.1073/ humans with volumetric muscle loss, Sci. Transl. Med. 6 (2014) 234ra58, https://
pnas.1221309110. doi.org/10.1126/scitranslmed.3008085.
[181] E. O’Neill, K. Rajpura, E.J. Carbone, G. Awale, H.-M. Kan, K.W.-H. Lo, [187] T.K. Merceron, M. Burt, Y.-J. Seol, H.-W. Kang, S.J. Lee, J.J. Yoo, et al., A 3D
Repositioning tacrolimus: evaluation of the effect of short-term tacrolimus bioprinted complex structure for engineering the muscle-tendon unit,
treatment on osteoprogenitor cells and primary cells for bone regenerative Biofabrication 7 (2015), 035003, https://doi.org/10.1088/1758-5090/7/3/
engineering, Assay Drug Dev. Technol. 17 (2019) 77–88, https://doi.org/ 035003.
10.1089/adt.2018.876. [188] L. Baldino, S. Cardea, N. Maffulli, E. Reverchon, Regeneration techniques for
[182] Y. Yin, G. Xiao, K. Zhang, G. Ying, H. Xu, B.A.G. De Melo, et al., Tacrolimus- and bone-to-tendon and muscle-to-tendon interfaces reconstruction, Br. Med. Bull.
nerve growth factor-treated Allografts for neural tissue regeneration, ACS Chem. 117 (2016) 25–37, https://doi.org/10.1093/bmb/ldv056.

13

You might also like