You are on page 1of 17

Biomaterial and Stem Cell-Based Strategies for Skeletal

Muscle Regeneration
Andrew Dunn, Muhamed Talovic, Krishna Patel, Anjali Patel, Madison Marcinczyk, Koyal Garg
Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
Received 12 June 2018; accepted 13 December 2018
Published
Published online
online 14 February
in Wiley 2019
Online in Wiley
Library Online Library (wileyonlinelibrary.com).
(wileyonlinelibrary.com). DOI 10.1002/jor.24212
DOI 10.1002/jor.24212

ABSTRACT: Adult skeletal muscle can regenerate effectively after mild physical or chemical insult. Muscle trauma or disease can
overwhelm this innate capacity for regeneration and result in heightened inflammation and fibrotic tissue deposition resulting in loss
of structure and function. Recent studies have focused on biomaterial and stem cell-based therapies to promote skeletal muscle
regeneration following injury and disease. Many stem cell populations besides satellite cells are implicated in muscle regeneration.
These stem cells include but are not limited to mesenchymal stem cells, adipose-derived stem cells, hematopoietic stem cells, pericytes,
fibroadipogenic progenitors, side population cells, and CD133þ stem cells. However, several challenges associated with their isolation,
availability, delivery, survival, engraftment, and differentiation have been reported in recent studies. While acellular scaffolds offer a
relatively safe and potentially off-the-shelf solution to cell-based therapies, they are often unable to stimulate host cell migration and
activity to a level that would result in clinically meaningful regeneration of traumatized muscle. Combining stem cells and biomaterials
may offer a viable therapeutic strategy that may overcome the limitations associated with these therapies when they are used in
isolation. In this article, we review the stem cell populations that can stimulate muscle regeneration in vitro and in vivo. We also
discuss the regenerative potential of combination therapies that utilize both stem cell and biomaterials for the treatment of skeletal
muscle injury and disease. ß 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1246–1262, 2019.

Keywords: Skeletal muscle; stem cells; biomaterials; regeneration

Skeletal muscle is the most abundant tissue in the decline in regenerative capacity.2 Satellite cells reside
body and constitutes approximately 40% of the total in a specialized microenvironment known as the niche,
body mass in humans.1 Each muscle cell (or myofiber) which contains stem cells, stromal cells, soluble
is a multi-nucleated contractile unit surrounded by the factors, ECM components, neural inputs, vascular
basal lamina.2 The basal lamina is a specialized layer networks, and cell adhesion molecules.2,9–12 Satellite
of the extracellular matrix (ECM) between the sarco- cells are located near blood vessels, and their density
lemma of each myofiber and the endomysium which is reported to be higher around neuromuscular and
surrounds each muscle fiber. The basement membrane myotendinous junctions.9,13 The satellite cell niche is
consists of the outer reticular lamina and the inner responsible for protecting the inactive, physiologically
basal lamina. The outer reticular lamina is composed quiescent stem cell population from diminishing and
mainly of fibrillar collagen (type I, III, VI) and also for regulating its activation in response to various
fibronectin whereas the inner basal lamina is com- stressors such as exercise, injury, or disease.9 Once
posed of an interconnected network of nonfibrillar activated, satellite cells migrate out of stem cell niche
collagen (type IV) and laminin. Laminin is a cross- undergo asymmetric division to form myoblast
shaped, heterotrimeric glycoprotein with an a-, b-, and (Pax7þMyoDþ) and satellite cells (Pax7þMyoD) which
g-subunit.3 It is the primary ligand for the dystrophin- can either continue to proliferate or return to a
associated glycoprotein complex (DGC) and a7b1 quiescent state.14 The myoblasts can either fuse with
integrin receptor (Fig. 1).4,5 Both these receptors one another or with existing myofibers to promote
associate with laminin in the extracellular matrix and muscle regeneration or repair, respectively.15 The
actin in the cytoskeleton and are essential for the dynamic interaction between satellite cells and their
transfer of mechanical force from the inside of the niche components is crucial for the maintenance of the
myofiber to the outer ECM during contraction.6,7 Any satellite cell pool and muscle mass. Any perturbations
disruption to the expression of these components can in the cross-talk between the satellite cells and their
impair the sarcolemma and myofiber cytoskeletal niche can result in impaired regeneration and fibrotic
integrity resulting in various forms of muscular dys- tissue deposition.4,14,16,17
trophy.4,5 A variety of stem cell populations have also been
Satellite cells are muscle resident stem cells that identified in skeletal muscle and the satellite cell niche
are located in between the plasma membrane of the including, mesenchymal stem cells,18 pericytes,19 side
muscle fiber and the surrounding basal lamina.3,4,8 population cells,20,21 fibro/adipogenic progenitors
Skeletal muscle’s outstanding regenerative capacity is (FAPs),22,23 and interstitial stem cells.24 These cell
derived from the satellite cells. These cells persist types share many features with one another such as
throughout life, although age and disease can lead to a multipotency and cell-surface marker expression and
are known to proliferate in response to muscle
injury.18 Transplantation studies and in vitro co-
Grant sponsor: Saint Louis University Start up Funds.
Correspondence to: Koyal Garg, (T: 314.977.8200; F: 314.977.8288; culture experiments13,25–29 have shown that these
E-mail: koyal.garg@slu.edu) stem cell populations can influence the self-renewal,
# 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. proliferation, and differentiation of satellite cells.

1246 JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
2019
BIOMATERIALS AND STEM CELLS 1247
3

Figure 1. The structure of (A) sarcolemmal proteins and basal lamina, and the (B) sarcomere. Reproduced with permission from
Rockefeller University Press via copyright clearance center from Fedik Rahimov, and Louis M. Kunkel J Cell Biol 2013;201:499–510.

During muscle repair, satellite cells also interact with factor-a (TNF-a), histamine, and tryptase which ini-
immune cells and endothelial cells. Following injury, tiates de novo synthesis of other cytokines like interleu-
the capillary density in damaged muscle tissue kin-6 (IL-6). Low levels of TNF-a, tryptase, and IL-6
increases initially and then returns to baseline 4 promote the activation and proliferation of satellite
weeks after injury.30,31 Co-culture experiments have cells.33 The initial release of cytokines and chemokines
shown that endothelial cells promote the proliferation attracts circulating granulocytes which consist of neu-
of the satellite cells by secreting mitogenic and anti- trophils and eosinophils.10 Neutrophils recruit mono-
apoptotic factors, such as vascular endothelial growth cytes which differentiate into macrophages.
factor (VEGF). A reciprocal relationship has been Macrophages can be divided into two main phenotypes
observed where differentiating myogenic cells also (M1/M2). There is an initial wave of pro-inflammatory
secrete VEGF which regulates the proangiogenic func- M1 macrophages which is followed by a second wave of
tion of endothelial cells. These findings indicate that anti-inflammatory M2 macrophages.34 M1 and M2
there might be a feed-forward mechanism through macrophages, respectively, represent the early and late
which VEGF modulates myogenesis and angiogenesis stages of myogenesis. Studies show that M1 macro-
in the stem cell niche.10 phages are found close to proliferating myogenic cells
Injury to muscle signals an inflammatory process while M2 macrophages interact with differentiating
that removes damaged cells, coordinates the regenera- myocytes.10 Prolonged and heightened inflammation
tive response, and restores tissue homeostasis.2,9,11,12,32 characterized by the persistent presence of M1 macro-
The immune response involves interactions between phages, often seen in orthopedic trauma, dysregulates
leukocytes and satellite cells. In resting conditions, the regenerative process and impairs satellite cell
multiple leukocyte populations reside in the adult activity.35,36
skeletal muscle, the most abundant being mast cells Therefore, a greater understanding of satellite cells
and macrophages. Mast cells and macrophages are and their niche components- resident stem cells and
resident cell types, and in conjunction with circulatory ECM proteins is of considerable therapeutic value.
monocytes, sense distress and secrete chemoattractive This review highlights the studies that have explored
molecules following muscle injury. Damage activated the functional capacity of stem cells and bioengineered
mast cells instantly begin to secrete tumor necrosis scaffolds for skeletal muscle regeneration.

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1248
2 DUNN ET AL.

STEM CELLS FOR SKELETAL MUSCLE instance, Sacco et al. showed that transplantation of a
REGENERATION single luciferase-expressing muscle stem cell (a7 integ-
Skeletal muscle repair and regeneration is a compli- rinþ CD34þ) into the muscle of mice underwent
cated process which is reliant not only on the muscle extensive proliferation, self-renewal, and differentia-
resident stem cell population but also on a vast tion in vivo.40 In another study, a unique combination
number of other stem and stromal cell populations of cell surface markers (CD45-Sca-1-Mac-1-CXCR4þ
(Fig. 2). The microenvironment of skeletal muscle is b1-integrinþ) was used to purify a distinct population
heterogeneous and contains a diverse population of of stem cells from the satellite cell pool.41 When
cells which are influenced by both structural and transplanted into dystrophic mice, these cells contrib-
biochemical cues. The following section will review uted to muscle regeneration both by fusion with
these cell types and their contributions to muscle existing myofibers and by de novo myogenesis. Resto-
repair in several animal models of disease and ration of dystrophin expression and improvement in
injury.37 muscle function was also observed. Human satellite
cells also display heterogeneity and have been identi-
Satellite Cells fied as NCAMþCD45CD31 cells,42 and also by the
The most important and well-characterized stem cell variable expression of Pax7, c-Met, and Dlk1.43
in the context of muscle repair is the satellite cell.38 Although the transplantation of autologous muscle
The best way to identify satellite cells in skeletal satellite cells may seem like a viable therapeutic
muscle is through their expression of the canonical strategy, this approach has met limited clinical suc-
marker, Pax7.39 However, satellite cells are known to cess. Challenges of satellite cell transplantation in-
exhibit heterogeneity and researchers have isolated clude the reduction of the regenerative capacity of
and transplanted different satellite cell subpopulations cultured cells compared to freshly isolated cells, poor
based on their diverse molecular signatures. For

Figure 2. (A) The relative presence and contribution of non-myogenic cell types in muscle regeneration. (B-D) Immunofluorescence
micrographs of tissue sections from regenerating mouse muscles. Pax7-positive satellite cells (green) are in close proximity to various
non-myogenic cell types (red): (B) CD11b-positive leukocytes; (C) Sca1-positive interstitial cells; and (D) VE-Cad-positive endothelial
cells. ECM is shown in orange and nuclei are labelled with DAPI (blue). Scale bar, 10 mm. Reproduced with permission from John
Wiley & Sons, Inc, from C Florian Bentzinger et al. EMBO Rep. 2013;14:1062–1072.

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019
BIOMATERIALS AND STEM CELLS 1249
3

survivability and migration of the transplanted cells, injury and resulted in improvements in regeneration
and adverse immune reactions.39,44,45 For instance, and muscle function.62 The exact mechanisms of MSC
Montarras et al.46 demonstrated that ex vivo expan- mediated tissue repair are unclear, and some contro-
sion of satellite cells for as little as three days severely versy exists regarding their capacity for myogenic
reduces their proliferative capacity and increases the differentiation.57 For instance, intramuscular trans-
fraction of differentiated cells, resulting in limited plantation of human BMMCs in dystrophic mice
regeneration upon transplantation in vivo. Improved restored dystrophin expression but did not improve
results have been reported by the transplantation of contractile function post engraftment.56 The authors
larger number of cells (105 myoblasts/cm of injection)47 suggested that the presence of dystrophinþ myofibers
and the use of immunosuppressive drugs.48,49 in transplanted mice may result exclusively from the
As a way to circumvent these challenges, many fusion of donor cells with resident myofibers rather
researchers are attempting the delivery of satellite than from the reprogramming of BMMCs into myo-
cells through methods other than direct transplanta- genic progenitors.56
tion. For example, a study that transplanted single, Besides bone marrow, muscle-derived MSCs have
intact myofibers into a radiation-ablated muscle dem- also been investigated in recent studies. Muscle-resi-
onstrated that even as few as seven satellite cells from dent MSCs (Sca1þ/CD45) delivered intramuscularly
the myofibers could regenerate more than 100 new were shown to increase Pax7þ satellite cell quantity,
myofibers with thousands of nuclei.50 This method has myofiber hypertrophy, and arteriogenesis in mouse
also been used in animal models of aging,51 and heart hindlimb muscles damaged by eccentric contraction
failure52 and beneficial effects have been reported. injury.18,25,63–66 These studies have suggested that the
However, even transplanting satellite cells through a primary contribution of MSCs to the repair and
single myofiber has its own challenges. The expertise, regeneration process is the trophic support they pro-
technical skill, and regulatory approval required to vide through the release of soluble mediators such as
implement this process successfully could prevent the growth factors and cytokines.18
clinical translation of this strategy. Adipose-derived stem cells (ASCs) have increasingly
gained popularity in regenerative medicine due to
Mesenchymal Stem Cells their morphological and phenotypic similarities to
Mesenchymal stem cells (MSCs) are the focus of MSCs with the added bonus that they can be readily
several cell-based therapies for a wide range of isolated with minimally invasive procedures.28 These
diseases.53,54 They exhibit features that are attractive cells have been reported to have myogenic potential in
for use in tissue engineering, including self-renewal, vitro, and myogenic progenitors that were ASC derived
multipotency, immunomodulatory properties, and se- have shown to successfully engraft into skeletal mus-
cretion of growth factors and cytokines.18 Further- cle and promote myofibers synthesis in dystrophic
more, these cells can be easily isolated from a variety mice (mdx).67 Studies have reported that compared to
of adult tissues including the bone marrow or adipose bone marrow, MSCs derived from adipose tissue have
tissue. MSCs have been shown to differentiate into a greater tendency for myogenic differentiation.68,69
mesodermal lineages in vitro including bone, cartilage, Similar to MSCs, ASCs can aid in muscle regeneration
and fat.55 indirectly as well, largely through their modulation of
MSCs represent less than 0.01% of the bone marrow inflammation and fibrosis, which has been demon-
mononuclear cell fraction but can be easily cultured strated in mdx mouse models.70 In one study, the
and genetically modified in vitro.56,57 Delivering these delivery of ASCs with an anti-fibrotic medication
bone marrow-derived MSCs (BMMC) intramuscularly called losartan to dystrophic muscles downregulated
has shown to improve muscle function in a dose- TGF-b1, inhibited fibrosis, and also improved muscle
dependent manner in rodent models of crush regeneration and hypertrophy.71 Thus from these
trauma.58,59 When BMMCs were delivered to a rodent studies, it can be seen that ASCs can support muscle
crush model intra-arterially, the muscle function was regeneration either directly or indirectly through
restored. Interestingly, this systemically delivered cell differentiation into myogenic progenitors or via the
population was not observed localized to the injured modulation of inflammation and fibrosis, respectively.
muscle. Thus the authors conjecture that the improve-
ments in function were likely due to the paracrine Hematopoietic Stem Cells
action of the injected BMMCs.60 Corona et al. worked Hematopoietic stem cells (HSCs), originating in adult
with a lineage depleted (Lin-) population of bone bone marrow have been shown to home to injured
marrow cells to increase the stem and progenitor cell muscle sites with guidance from the stromal cell-
populations in a mouse ischemia-reperfusion (I/R) derived factor-1 (SDF-1)/chemokine receptor 4
model. They found that when injected intramuscularly (CXCR4) and chemokine receptor 2 (CCR2) coupling.72
cells survived up to 1-month post-transplantation, but Other studies have also implicated HGF/c-Met signal-
did not improve function.61 However, when injected ing in the homing of HSCs to injured muscle.73
intravenously to avoid damage that can occur during Multipotent bone marrow-derived HSCs were first
intramuscular injection, the cells homed to the site of reported to mediate muscle regeneration in vivo in 1998,

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1250
2 DUNN ET AL.

and since then studies have repeatedly reported sup- osteogenic, and myogenic lineages.19,85–87 These cells
porting evidence of their regenerative capacity.26 For also express canonical markers of MSCs such as
example, one study showed that intravenously injected CD29, CD44, CD73, CD90, and CD105.88
HSCs could contribute to the myogenesis and restore In contrast to satellite cells, pericytes generally do
dystrophin expression in lethally irradiated mdx mice.74 not express Pax7 or MyoD. These cells only express
Another study showed that following irradiation, the myogenic markers post-differentiation, and this capac-
satellite cell niche could be reconstituted by bone ity has been demonstrated both in vitro and in
marrow-derived HSCs that expressed satellite cell vivo.19,83,87,89 Two major subpopulations of pericytes
markers such as a7 integrin, c-Met, and Myf5.75 have been identified: Type 1 (Nestin-NG2þ) and type 2
Furthermore, these bone marrow-derived satellite cells (NestinþNG2þ). While type 1 pericytes are reported to
were demonstrated to undergo myogenic differentiation contribute to fat accumulation, type 2 pericytes are
in vitro and myofiber regeneration in vivo.75 known to support new muscle formation.90 Type 2
While HSCs are quiescent in myofiber repair under pericytes, which are Nestinþ/NG2þ, have been identi-
normal physiological conditions, it has been shown fied in the satellite cell niche and are known to
that in response to damage or injury they do contrib- support the new formation of muscle tissue.90
ute to myogenic events. One such study showed that Depletion of pericytes has resulted in insignificant
the capacity of hematopoietic progenitors to fuse with hypotrophy and a slight increase in total Pax7þ cells.91
myofibers is increased significantly following muscle Co-culture of pericytes and satellite cells resulted in
damage.76 Sacco et al. identified insulin-like growth either myogenic differentiation through the secretion
factor 1 (IGF-1) as a damage related molecule that of insulin-like growth factor (IGF-1) or quiescence
substantially augmented the fusion of bone marrow- through the secretion of angiopoietin 1 (ANGPT1).91
derived cells with adult skeletal muscle.77 In another Another study demonstrated that when transplanted
study, Camargo et al. suggested that the fusion of into immunodeficient dystrophic mice, pericytes can
HSC-derived progenitors with damaged myofibers sup- generate dystrophinþ myofibers.19 Further, this study
ported regeneration following injury.78 Xynos et al. also showed that when delivered systemically the
used a cardiotoxin injury model to demonstrate that pericytes can cross the vessel wall to colonize the
upon interaction with the muscle microenvironment, injured muscle. To support these findings, Lorant
HSCs undergo profound yet incomplete myogenic et al. found that when injecting perivascular stem cells
reprogramming, differentiation, and incorporation into into cryoinjured muscles of scid mice the cells inte-
muscle myofibers to participate in muscle regenera- grated into host tissue and contributed to the produc-
tion.79 However, the ability of these cells to undergo tion of structural proteins associated with
reprogramming post-fusion has been challenged by differentiated myofibers.92 In contrast, when Meng
studies that have reported that myofibers with donor- et al. delivered muscle-derived pericytes intra-arteri-
derived nuclei failed to express sarcoglycan80 or dys- ally to mice, they found that they did not contribute to
trophin81 and only temporarily turn on myogenic the regeneration of muscle.93 These authors believed
genes. Negative results with transplantation of bone the discrepancy in results were due to the differences
marrow-derived cells have also been reported in a rat in experimental techniques, animal models, and out-
model of volumetric muscle loss (VML), where these come measurements. Despite these studies, the mech-
cells only contributed to limited de novo myofiber anisms that regulate the myogenic differentiation of
regeneration and did not show significant changes in pericytes is largely unknown, and to fully understand
muscle function or myogenic gene transcription82 the role of pericytes in muscle regeneration it is
Overall, the efficacy of HSCs in supporting muscle necessary to perform more comprehensive studies.
regeneration is controversial. In addition, it is unclear
if muscle repair is facilitated by HSC derived myogenic Fibro/Adipogenic Progenitors
cells, or if the HSCs themselves undergo reprogram- Skeletal muscle resident PDGFR-aþ Sca-1þ CD34þ
ming after fusion with damaged myofibers. fibro/adipogenic mesenchymal progenitor (FAPs) are
bipotent cells that primarily contribute to fibrotic and
Pericytes fatty tissue deposition in skeletal muscle.22,23,94,95
Pericytes are perivascular stem cells that form According to some reports, the differentiation of these
intimate connections with adjacent capillary endothe- interstitial cells depends on the severity and mecha-
lial cells. These contractile connective tissue cells nism of muscle injury. For instance, Uezumi et al.,
reside beneath the microvascular basal lamina and showed that PDGFR-aþ cells isolated from glycerol
regulate capillary blood flow.83,84 These cells are damaged skeletal muscle did not differentiate into
known to affect migration, proliferation, permeabil- adipocytes when transplanted into cardiotoxin (CTX)
ity, and contractility of endothelial cells. This multi- injured muscle.96 However, PDGFR-aþ cells isolated
potent cell population shares many similarities with from CTX injured muscle readily accumulated into
MSCs. For instance, pericytes have been isolated glycerol damaged areas and differentiated into lipid
from various tissues and are known to possess laden adipocytes (C/EBPaþ PPARgþ).22 Therefore,
differentiation potential for adipogenic, chondrogenic, these results suggest that the differentiation of FAPs

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019
BIOMATERIALS AND STEM CELLS 1251
3

into adipocytes is highly dependent on the muscle also generate three-dimensional (3D) functional skele-
microenvironment. It was revealed that CTX injured tal muscle in vitro.108–111 The breadth of lineages in
muscles show higher numbers of new myofibers com- which iPSCs differentiate through allows for a variety
pared to glycerol injured muscles and the interaction of cell types (neural, vascular, and muscular) to
between regenerating fibers and FAPs may be neces- populate these 3D tissue constructs.108 These func-
sary for the inhibition of fat deposition. Consistent tional tissue constructs were able to survive, vascular-
with CTX injury results, FAPs did not undergo signifi- ize, and function when transplanted in vivo,
cant adipocyte differentiation in response to notexin suggesting that the can replace damaged or dysfunc-
injured muscles. However, they showed extensive tional muscle.111 Additionally, these constructs can be
proliferation in the defect region and were found in used for disease modeling and drug screening applica-
close proximity to the regenerating fibers, indicating tions.108
that FAPs may have a role to play in modulating However, this revolutionary therapy is not without
myogenesis.23 While FAPs do not directly differentiate its flaws. Despite the appeal of autografting, there
through myogenesis, they do produce several factors have been cases of immune rejection and the necessity
such as IL-6, IL-10, IL-33, IGF-1, Wnt 1, Wnt3A, and for immunosuppressive drugs.112 Tumor formation is
Wnt5A that are known to support myogenic activity. also a rare but observable problem with iPSCs trans-
23,97,98
plantation. 113,114
The ability of these cells to accumulate and in
fibrotic areas and express fibrosis related molecules Other Stem Cell Populations
has also been well-documented. In the diaphragm of Several other stem cell populations have been identi-
dystrophic mice and CTX injured skeletal muscle, fied in skeletal muscle, and there has been increasing
FAPs were found co-localized in collagen type 1 rich interest in understanding their role in the process of
regions.96 Studies have identified TGF-b1 and PDGF- muscle regeneration. The muscle side-population (SP)
AA to be potent regulators of FAP mediated fibrosis in is a heterogeneous cell population that is reportedly
skeletal muscle. Exposure to these growth factors in positive for Sca1, ABGC2, Syndecan-4 but negative for
vitro was shown to promote the proliferation and CD45.115 An interesting feature of these cells is their
expression of fibrosis related markers (e.g., Col1a1, ability to migrate and engraft into host myofibers
col3a1, CTGF, TIMP1, and a-SMA) in PDGFR-aþ following systemic delivery.116 The myogenic potential
FAPs.96 of these cells is under investigation. In one study,
Targeting these cells could provide new therapeutic direct injection of the SP cells in injured muscles
strategies for preventing pathological changes in mus- resulted in efficient engraftment into the satellite cell
cle following injury and disease. Genetic and pharma- niche. These cells also gave rise to satellite cells and
cological ablation of FAPs has been associated with donated myonuclei to the regenerating fibers.115 How-
impaired muscle regeneration.99,100 Therefore, modu- ever, in another study, the ABGC2þ SP cells were
lation of FAP activity may hold the key for enhancing reported to increase in muscle after injury, giving rise
muscle regeneration. For instance, by inhibiting TGF- to endothelial cells and pericytes but not contributing
b1, FAPs were shown to become apoptotic in the significantly to myonuclei.117 Similar results were
presence of macrophages, which correlated with de- obtained in a recent study where SP cells derived from
creased fibrosis in vivo.101 Cytokines such as IL-4 and dystrophic or injured muscles gave rise to FAPs but
IL-15 were shown to prevent FAPs from undergoing failed to undergo myogenesis.118 Therefore, more rig-
adipogenic differentiation.102,103 Future studies should orous studies are needed to establish a clear role of
focus on elucidating the diverse differentiation mecha- these cells in skeletal muscle regeneration.
nisms of FAPs. Identification of precise factors and CD133þ stem cells have gained increased popularity
molecules that can modulate FAP activity to promote in recent years. These cells have been tested in both
myogenic regeneration without fibrotic and fatty tissue animal models and clinical trials to repair injured
deposition are likely to offer great therapeutic benefits tissues.119 A major advantage of this cell population
to aging, diseased, or severely injured skeletal muscle. from a clinical standpoint is the ease with which it can
be purified from the peripheral blood. A study com-
Induced Pluripotent Stem Cells pared the regenerative capacity of the human muscle-
Induced pluripotent stem cells (iPSCs) provide several derived CD133þ stem cells with human satellite cell-
attractive features for myogenic research such as derived myoblasts in a mouse model of cryoinjury.120
capacity for self-renewal, suitability for genetic edit- The results showed that the CD133þ stem cells out-
ing, ability to differentiate into multiple cells types performed myoblasts in terms of migration, repopula-
including myogenic cells while maintaining pathologi- tion of the satellite cell niche, and overall regenerative
cal phenotypes.104,105 Myogenic cells have been derived capacity. A recent study demonstrated that human
from iPSCs via genetic reprogramming or addition of CD133þ cells transplanted into mouse muscles are
small molecules. Studies have reported that iPSC functional and can give rise to satellite cells in vitro
derived myogenic cells can fuse with existing myofib- and in vivo.121 However, similar to myoblasts, the
ers following in vivo transplantation105–107 and can prolonged culture of donor CD133þ cells was shown to

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1252
2 DUNN ET AL.

reduce their effectiveness in vivo. In another study, maintain the Pax7þMyoD population when grown on puri-
human adult CD133þ stem cells were mobilized from fied collagen VI, with decreased apoptosis and increased
the peripheral blood by granulocyte colony-stimulating proliferation compared to cultures grown on Matrigel alone.
factor (G-CSF) and intramuscularly injected into lacer- Taken together, these studies show that biomaterials
ated muscles in athymic rats. The injected cells designed for supporting specific cellular functions have the
potential to overcome the limitations associated with cell-
differentiated into endothelial and myogenic cells and
based therapies. However, recent work has suggested that
improved the microenvironment of the injured tissue acellular biomaterial-based therapies have several limita-
by upregulating angiogenesis and downregulating fi- tions of their own as they have failed to recover
brosis.122 Autologous muscle-derived CD133þ cells strength,128,129 recruit endogenous satellite cells to the defect
have also been transplanted in humans with Duch- site,130,131 and regenerate nerves.132,133 Instead of using the
enne muscular dystrophy.123 While the procedure was two strategies in isolation, researchers have investigated cell
well-tolerated and safe, functional benefits were not and biomaterial combination therapies. Combining cells with
observed. candidate biomaterials may overcome issues associated with
Therefore, it is clear that muscle function and acellular scaffolds and cell-only therapies. Such combination
regeneration are impacted not only by myogenic cells therapies have shown promising results in animal models
and may provide a potential therapy in the near future.134
but also by a lot of other stem cell populations. The
The following section describe some of the promising scaffold
heterogeneity of stem cells and their function further
technology that has been developed in combination with
demonstrates the complexity of skeletal muscle main- cellular therapies over the recent years.
tenance and regeneration. Studies that illustrate and
define the mechanism through which these stem cell Satellite Cells and Myoblasts Transplantation with Biomaterials
populations orchestrate muscle repair would be valu- A scaffold-based strategy for satellite cell or myoblast
able for the development of cellular therapies. encapsulation and delivery has been attempted in various
studies. Rossi et al. reported improvement in muscle struc-
BIOMATERIALS FOR SKELETAL MUSCLE ture, the total number of new myofibers as well as muscle
REGENERATION function by delivering satellite cells encapsulated in a
While the quest for an ideal stem cell population for hyaluronan-based photocrosslinkable hydrogel to partially
stimulating muscle regeneration continues, some researchers ablated tibialis anterior muscles.135 At 6 weeks post-injury,
have diverted their attention toward finding an ideal bioma- the cell-loaded hydrogels had extensive and consistent mus-
terial substrate to modulate tissue-resident stem cells, cle regeneration with minimal scar tissue formation, while
deliver therapeutic molecules, and encapsulate cells to the untreated and hydrogel only groups had a large deposi-
protect them from the injured microenvironment and the tion of fibrotic connective tissue and altered morphology.
immune system.124 In a study by Pollot et al., five commonly used natural
As mentioned previously, stem cells often constitute an polymeric materials were used in skeletal muscle tissue
extremely rare population in tissues, and their ex vivo engineering grafts and were characterized for mechanical
expansion is reported to reduce their regenerative capacity and myogenic properties by seeding primary rat satellite
greatly [96–100]. Several studies have shown that freshly cells.136 The five materials were collagen I, agarose, fibrin,
isolated satellite cells or satellite cells derived from trans- collagen-chitosan, and alginate. Overall findings of this study
plantation of one intact myofiber contribute to muscle repair suggested collagen I was supportive of myogenesis, while
more significantly than cultured myoblasts.2,40,125 Gilbert fibrin demonstrated the highest myogenic potential mea-
et al. demonstrated that the stem cell niche supports freshly sured by the quantification of gene expression of MyoD,
isolated satellite cells in their ability to regenerate when myogenin, and myosin heavy chain.
transplanted whereas satellite cells grown on standard tissue Fibrin provides an excellent matrix for myoblast activity
culture plastic lose their stem cell capability by producing and vascularization.137 Marcinczyk et al. studied the efficacy
progenitors with greatly diminished regenerative poten- of laminin-111 enriched fibrin hydrogels for skeletal muscle
tial.125 Furthermore, matrix rigidity was shown to affect the regeneration.3 With the addition of increasing concentrations
quiescence, proliferation, and differentiation of satellite cells. of laminin-111, fibrin hydrogels displayed progressively
The study showed that cultured on poly(ethylene glycol) thinner, interlaced fibers. While lower concentrations of
hydrogels with rigidity similar to native muscle tissue laminin-111 supported myoblast differentiation, a higher
(12 kPa), the satellite cells are capable of self-renewal and concentration of laminin-111 promoted myoblast differentia-
maintenance of regenerative properties. Similar results were tion. Additionally, myoblasts seeded hydrogels displayed
obtained in another study where muscle progenitor cells responsiveness to electromechanical stimulation through
showed increased proliferation on polyacrylamide gels of elongation, alignment, myokine secretion, and sirtuin
lower stiffness and adopted a differentiating phenotype on (SIRT1) expression.
substrates with higher stiffness.126 These results were Fibrin-based scaffolds have also been used in animal
corroborated by Urciuolo et al. who also showed that satellite models of muscle trauma with encouraging results. Page
cells grown on gelatin hydrogels of physiological (12 kPa) et al. fabricated fibrin microthreads to serve as efficient
stiffness maintained a large proportion of Pax7þMyoD cells, delivery vehicles for primary human muscle-derived cells
which was dramatically decreased when satellite cells were that expressed both pluripotent and myogenic markers in
grown on structures of 7 kPa stiffness.127 The authors further culture.138 Cell-seeded fibrin microthreads were implanted in
demonstrated the effect of ECM components on the myogenic a VML defect in immunodeficient mice. The microthreads
potential of satellite cells. Satellite cells derived from colla- were reabsorbed at 1-week post injury, and new muscle
gen VI null mice (Col6a1/) showed a higher ability to fibers were reconstituted at 2-weeks for microthread treated

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019
BIOMATERIALS AND STEM CELLS 1253
3

wound sites. In contrast, the untreated control wounds had satellite cell population in the injured muscles. However,
collagen as the predominant constituent of the healed functional recovery of the VML injured muscle was not
wound. After 4 months, the cell-loaded microthreads signifi- assessed. Taken together, these studies suggest that cell-
cantly improved the recovery of muscle strength compared to loaded fibrin scaffolds can prevent the rapid progression of
untreated controls. fibrosis that impedes muscle regeneration and restore muscle
Gilbert-Honick et al. engineered electrospun fibrin scaf- functionality.
folds to promote muscle regenerations and functional recov- It has also been shown that growth factors coupled with
ery.139 Hydrogel microfiber bundles were created by cell or scaffold-based therapies can improve muscle regenera-
extruding both fibrinogen and alginate in parallel. Acellular tion. Keratin hydrogels have been used for the controlled
or C2C12 myoblast seeded scaffolds were implanted in a release of growth factors in some recent studies, since their
hindlimb VML injury model in immunodeficient mice. While degradation rate can be tailored easily. Tomblyn et al.
the acellular electrospun scaffolds resulted in fibrosis and investigated a keratin hydrogel carrier system for the deliv-
lacked the presence of MHC myofibers, defects treated with ery of exogenous growth factors and muscle progenitor cells
C2C12-seeded electrospun scaffolds showed an increase in (MPCs) to the site of VML injury.141 Two different isomers
muscle mass, lack of fibrosis as well as high myofiber and were studied—oxidatively extracted keratin (or keratose)
vascular density. Interestingly, both acellular and C2C12- and reductively extracted keratin, (or kerateine). Due to the
seeded scaffolds demonstrated maximum torque levels equal presence of covalent disulfide bonds within kerateine, its
to uninjured controls, while untreated defects showed a 30% degradation is slower compared to keratose. This difference
torque deficit at 2 and 4 weeks post-injury. In the absence of in degradation rates resulted in different release profiles of
muscle regeneration, this result could be explained by exogenous growth factors such as vascular endothelial
“functional fibrosis”—a fibrosis mediated mechanical bridg- growth factor (VEGF), insulin-like growth factor 1 (IGF-1)
ing effect that helps in force transmission and protection of and basic fibroblast growth factor (bFGF). Following in vivo
remaining muscle mass from reinjury.140 testing in a subcutaneous mouse model, the study suggested
Another study evaluated the efficacy of muscle-derived that keratose hydrogels maintained the viability of MPCs
stem cells (MDSCs) combined with fibrin gel for VML and the bioactivity of growth factors with minimal inflamma-
repair in immunodeficient mice.134 At 4 weeks post-injury, tory responses. The delivered muscle progenitor cells were
this combined approach showed that MDSCs differentiated reported to form a very primitive muscle-like construct.
into new myofibers and significantly increased muscle In another study, Baker et al. loaded kerateine hydrogels
mass along with a significant decrease in fibrotic tissue with skeletal MPCs with or without IGF-1 or bFGF to treat
deposition. The number of small fibers with centrally located volumetric muscle loss (VML) injury in the latissimus dorsi
nuclei were reported to increase with treatment indicating muscle in a mouse model.142 After 2 months, keratin hydro-
significant increase of regenerative fibers in the engrafted gels loaded with both growth factors but without MPCs
area of treated muscles (Fig. 3). The MDSC seeded fibrin gels yielded a greater recovery of contractile force than the other
also supported increased vessel formation and restored treatment groups and promoted extensive new muscle tissue

Figure 3. Therapeutic efficiency of MDSC/fibrin gel casting method for muscle defect repair. (A) Representative histological images of
muscle cross sections from different experimental groups show different levels of fibrosis versus engraftment. (B) Quantification of
muscle regeneration and fibrosis. Combination therapy of MDSC/fibrin gel significantly increased muscle regeneration and decreased
fibrosis. (C) Quantification of fibers with centrally located nuclei and their cross section area (CSA) indicates significant increase of
regenerative fibers in engrafted area of treated muscles compared to non-engrafted native regions. Reprinted from “Volumetric muscle
loss injury repair using in situ fibrin gel cast seeded with muscle-derived stem cells (MDSCs)” by Matthias et al., Stem Cell Research,
volume 27 pages 65–73, Copyright 2018, with permission from Elsevier.

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1254
2 DUNN ET AL.

formation. These results were unexpected as the inclusion of the observed positive effects to the possibly enhanced para-
MPCs did not provide functional improvement. The authors crine action of ASCs.
speculated that likely reasons for this result include a lower In another study, ASCs and their conditioned medium
cell density of MPCs and lack of preconditioning in a were delivered to the VML defect site in a collagen hydrogel.
bioreactor.143 Similar results were obtained in a subsequent The results showed that this treatment approach accelerated
study conducted in a rat VML model.144 The exact mecha- muscle repair, improved vascularization, and blood perfu-
nism responsible for the enhanced performance of the kera- sion, while simultaneously reducing inflammation and fibro-
tin hydrogel with the addition of growth factors needs to be sis.151 The authors attributed these effects to the enhanced
investigated in future studies. release of anti-inflammatory cytokines by the ASCs which
were able to polarize the macrophages in the defect site
Mesenchymal Stem Cells Delivery with Engineered Matrices toward an M2 phenotype.
Several studies have delivered MSCs via hydrogels to the Aurora et al. combined PEGylated-platelet free plasma
site of muscle injury. In a model of repeated laceration hydrogels with a decellularized muscle ECM scaffold to
injury, BMMC were suspended in Matrigel and transplanted deliver human ASCs in a rat VML injury model.152 At 2
in the soleus muscles.145 Compared to non-treated muscles, weeks post-injury, the study showed that the composite
the BMMC treated muscles showed fewer fibers with a scaffolds are viable cell delivery vehicles as shown by the
centrally located nucleus, and larger muscle fiber cross- increased presence of ASCs in the defect region. These
sectional area, but no differences in fibrotic tissue deposition delivered cells were also reported to home to the perivascular
were reported. In another study, transplantation of BMMC space. The composite treatment resulted in increased vascu-
suspended in a fibrin matrix in a rodent model of muscle larization but did not result in appreciable muscle regenera-
laceration restored muscle function. The cells did not differ- tion. The authors speculated that perhaps vascularization
entiate into myotubes or fuse with existing myofibers, alone is not enough and a myogenic cell source is necessary
suggesting an alternate mechanism for repair.146 In a recent to support muscle regeneration.
study, BMMCs were delivered with recombinant growth
factors in an alginate cryogel to injured soleus muscles, a Co-Delivery of Minced Muscle Grafts with Biomaterials
strategy that enhanced paracrine signaling in MSCs. This An alternative approach to delivering a specific stem cell
approach resulted in improved muscle function, remodeling population is to deliver bundles of minced muscle fibers to
of scar tissue, and increase in new myofiber regeneration.147 transfer not just satellite cells but also other cell types
MSCs have also been used with decellularized ECM in their niche with an intact ECM. Corona et al. have
scaffolds for the regeneration of skeletal muscle. Merritt repeatedly demonstrated appreciable muscle regeneration
et al. injected BMMCs at the site of VML injury 1 week after by transplantation of minced muscle autografts in both
the implantation of muscle-derived ECM.148 After 42 days of rodent130,153,154 and porcine155 models of VML. Since this
recovery, the BMMC injected ECM group showed more blood approach has the obvious limitations of inadequate availabil-
vessels, regenerating skeletal myofibers, and functional ity and potential donor site morbidity, Corona and coworkers
recovery than ECM treatment alone. Qui et al. repaired have investigated various biomaterials to deliver the autolo-
VML injured rat hindlimb muscles with human umbilical gous minced muscle grafts to the VML defect site.156 Since
cord MSCs and decellularized porcine cardiac ECM scaf- collagen type I is the primary skeletal muscle ECM constitu-
folds.149 The treatment groups included decellularized ECM ent, it was evaluated as a potential expansion biomaterial.
powder, MSC aggregates, or the combination of the two. At 8 The VML injured hindlimb muscles received a 25 or 50%
weeks post-injury, the compound group showed improved autologous minced graft suspended in collagen hydrogel,
muscle regeneration and functional recovery along with with the 100% minced muscle graft treatment serving as the
reduced collagen deposition compared to the other treatment positive control. After 8 weeks post-injury, only the 50 and
groups. The study further reported that treatment with 100% minced graft treatment restored promoted significant
either MSCs or ECM scaffold alone was immunomodulatory de novo fiber regeneration and improved muscle function. It
and that this effect was more pronounced when the two was also noted that the regenerated tissue associated with
therapies were combined. The combination of MSCs and the 50% graft had greater fibrotic and fat tissue deposition.
decellularized ECM scaffolds promoted the pro-regenerative In another study, Goldman and Corona found that the
M2 macrophage phenotype, while simultaneously suppress- regenerative capacity of minced muscle grafts used for the
ing the pro-inflammatory M1 phenotype. These promising treatment of VML injuries is impeded with the co-delivery of
results need to be replicated and confirmed by other urinary bladder matrix (MicroMatrixTM, ACell Inc., Colum-
researchers in the field in order to establish the use of MSCs bia, MD).157 MicroMatrixTM was either used alone or in
with biomaterials as a viable therapeutic strategy for trau- conjunction with the minced muscle graft (1:1). After 8 weeks
matized muscle tissue. of treatment, the compound group showed little to no muscle
regeneration but improved functional capacity, possibly due
Co-Administration of Adipose-Derived Stem Cells with Scaffolds to functional fibrosis.140 An aggravated immune response to
Human adipose tissue-derived adult stem cells (ASCs) the decellularized MicroMatrixTM was reported contrary
were co-administered with bFGF loaded gelatin-poly(ethyl- to several previous reports of a favorable immune
ene glycol)-tyramine (GPT) hydrogels in a mouse model of response.132,133,158–162
muscle laceration.150 At 4 weeks post-injury, the combined In a subsequent study, Corona and coworkers investi-
treatment resulted in significant improvements in muscle gated a laminin-111 supplemented PEG-hyaluronic acid-
regeneration, functional recovery, revascularization, and based hydrogel as a delivery vehicle for minced muscle grafts
reinnervation with a concomitant decrease in fibrosis com- for VML injuries.163 At 8 weeks, the hydrogels were found to
pared to other treatment groups (Fig. 4). However, the exact be wholly intact and did not support cellular infiltration into
mechanism of repair was unclear, and the authors attributed the defect site. The lack of cellular infiltration was associated

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019
BIOMATERIALS AND STEM CELLS 1255
3

Figure 4. (A) Representative histological images from Masson’s trichrome staining of lacerated gastrocnemius muscle 4 weeks after
surgery. Experimental treatment groups included saline (C), the h-ADSCs (AD), the bFGF hydrogel (bH), and the bFGF hydrogel
containing h-ADSCs (AD/bH group). (B) Quantified area of muscular fibrosis. AD/bH group revealed significantly lower muscular
fibrosis induced by muscle laceration (scale bars, 100 mm,  p < 0.05 compares with the laceration group; #p < 0.01 compares with the
laceration group). Reprinted from “Combination therapy of human adipose-derived stem cells and basic fibroblast growth factor
hydrogel in muscle regeneration” by Hwang et al., Biomaterials, volume 34 issue 25 pages 6037–6045, Copyright 2013, with permission
from Elsevier.

with impaired muscle regeneration and function. Therefore, and neurogenesis while simultaneously suppressing
an optimal carrier material for the expansion and delivery of fibrotic tissue deposition.
minced muscle grafts needs to be investigated intensively. Decellularized scaffolds have been widely used for
skeletal muscle repair and regeneration.167 Recent
CHALLENGES WITH EXISTING TECHNOLOGIES work has also described encouraging clinical outcomes
AND FUTURE TRENDS following the implantation of decellularized scaffolds
A successful strategy for muscle regeneration cur- in patients with VML injuries (reviewed in37). How-
rently does not exist. The two main obstacles to muscle ever, acellular ECM based therapies have a set of
regeneration following trauma are fibrotic tissue depo- challenges that must be overcome in order to promote
sition and persistent inflammation.36,130,153,157,164–166 clinically relevant levels of skeletal muscle regenera-
A regenerative strategy for traumatic injuries such as tion. A common problem with acellular treatments is
VML must support the migration, proliferation, and that they do not produce a meaningful regeneration of
differentiation of cell populations that can promote muscle tissue and get remodeled into a fibrotic
myofiber synthesis, angiogenesis, immunomodulation, scar.128,131,140,168–170 These scaffolds completely rely on

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1256
2 DUNN ET AL.

the migration and proliferation of host satellite cells to these trials are highlighting the roles these stem cell
infiltrate the defect area and restore function to the populations play in replacing damaged tissue and
injured site. Degradation products of these ECM replenishing trophic and extracellular factors, con-
scaffolds termed as “cryptic peptides” are known to cerns about these treatments have also been
exert chemotactic and mitogenic effects on multipoten- expressed. Several factors must be considered for
tial progenitor cells in vitro171 and in vivo.172 In selecting a suitable stem cell population for skeletal
animal models, implantation of ECM scaffolds has muscle regeneration which include availability and
resulted in the recruitment of several stem cell abundance of the cell source, susceptibility to genetic
populations such as Sca1þ cells,130,158 perivascular manipulation, ease of delivery, and finally the ability
stem cells,173,174 pluripotent adult progenitors to undergo functional myogenesis in vivo.183 Regard-
(Sox2þ),175 CD133þ progenitor cells,132 and neural ing stem cell therapies, uncontrolled differentiation is
stem cells.176 However, several studies have shown a potential concern. For instance, heterotopic ossifica-
that satellite cells or myogenic progenitors fail to tion—an aberrant growth of bone within a soft tissue
substantially repopulate the acellular construct,130,131 has been associated with orthopedic trauma.184,185
resulting in the formation of small muscle fiber islands This debilitating condition has been linked to
(<50 fibers) in the defect region of VML that are MSCs,186,187 pericytes,188 FAPs,189 and decellularized
unable to restore functional use of the injured mus- scaffolds.190 Pre-treating cells to promote their com-
cle.161 Furthermore, each laboratory decellularizes mitment down a particular lineage prior to injection is
tissues using different protocols and processing techni- one avenue that that could be explored to potentially
ques. The variable and heterogeneous composition of evade these issues. Some research has been done to
the resulting matrix not only influences their perfor- “prime” cells prior to injection.191,192 In several stud-
mance in vivo but also makes it difficult to compare ies, preconditioning of myoblasts seeded on 3D scaf-
and conclude published work. Future studies should folds has accelerated muscle tissue regeneration and
strive to meticulously present decellularization meth- function when implanted in vivo.143,193–195 In other
ods as well as the protein and DNA composition of the studies, pre-conditioning MSCs with mechanical strain
ECM scaffolds. has resulted in enhanced myogenesis and secretion of
The capability of stem cells and ECM scaffolds to beneficial growth and immunomodulatory factors in
modulate the immune response at the site of injury is vitro.192,196–198 However, a positive effect on in vivo
another aspect that is crucial to muscle regeneration. myogenesis was not observed following the injection of
Of particular interest, VML injury results in a height- “preconditioned” MSCs in aged muscles.191 The poten-
ened and persistent inflammatory response that over- tial of preconditioning stem cells is immense, and
whelms its innate capacity for regeneration.36,164 In a further research in this field is necessary. Culturing
recent study, it was shown that muscle trauma cells on tissue engineered scaffolds with electrical and
prompts robust and persistent overexpression of in- mechanical stimulation is an avenue currently being
flammatory transcripts, which contributes to fibrotic explored in various studies.3,199
tissue deposition and may impair satellite cell-medi- Recently, the paracrine action of stem cells is being
ated repair.35 It has been suggested that both M1 (pro- investigated via exosomes. Extracellular vesicles, i.e.,
inflammatory) and M2 (anti-inflammatory) macro- exosomes, allow for cell-cell communication and could
phage phenotypes are necessary for muscle regenera- be a key to unlocking many of the challenges associated
tion.33,130 M1 macrophages promote the activation, with the delivery and engraftment of stem cells.
migration, and proliferation of satellite cells.34,160,177 Exosomes can be released from paracrine cells and
M2 macrophages are needed to mitigate the immune myofibers, containing mRNA, miRNA, and proteins
response and support the differentiation and matura- that can influence gene expression and cellular func-
tion of satellite cells into myotubes and myofibers.33 tions after being endocytosed by the host cell.200 These
However, early recruitment of M2 macrophages can nanospheres could be used to trick the host tissue into
dysregulate satellite cell activity and delay regenera- a more pro-regenerative state. Using specific myogenic
tion.160,177 On the other hand, the persistent M1 media, cultured MSCs will release exosomes with a
phenotype is likely to exacerbate muscle damage and specific myogenic load. Exosomes can be collected using
support fibrotic tissue deposition. Therefore, these ultracentrifugation and could supplement the injection
macrophage populations need to be activated for an of myogenic stem cells. C2C12 cells showed increased
optimum duration and at an ideal intensity. Immuno- proliferation and migration when exposed to MSC
genic concerns have been reported with decellularized exosomes.200 Similar results have been seen in myo-
scaffolds,157 poly(ethylene glycol),178 and hyaluronic blast derived exosomes.201 In a murine TA laceration
acid179 based materials. Immunomodulation via immu- model, muscles treated with myoblast-derived exosomes
nosuppressant drugs180 or paracrine action of stem showed increased regeneration and decreased fibrosis
cells181 may be able to regulate the intensity and compared to untreated controls. Despite convincing
duration of macrophage phenotypes. evidence for this cell-free therapeutic approach, the
There are several ongoing clinical trials with stem identification of exact factors in exosomes that play key
cells for a variety of different conditions.182 While roles in muscle regeneration remains unclear.

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019
BIOMATERIALS AND STEM CELLS 1257
3

Overall, it is becoming increasingly clear that tissue 16. Conboy IM, Conboy MJ, Wagers AJ, et al. 2005. Rejuvena-
engineering solutions to muscle trauma can be im- tion of aged progenitor cells by exposure to a young
systemic environment. Nature 433:760–764.
proved by combining bioengineered scaffolds with
17. Brack AS, Conboy MJ, Roy S, et al. 2007. Increased Wnt
stem cells, growth factors, and potentially electrical/ signaling during aging alters muscle stem cell fate and
mechanical stimulation to guide muscle tissue growth. increases fibrosis. Science 317:807–810.
Although a combination therapy of cells and biomate- 18. Boppart MD, De Lisio M, Zou K, et al. 2013. Defining a role
rials seems promising, the complexity of this approach for non-satellite stem cells in the regulation of muscle
is likely to impact regulatory approval, clinical trans- repair following exercise. Front Physiol 4:310.
lation, and off-the-shelf availability to patients. 19. Dellavalle A, Sampaolesi M, Tonlorenzi R, et al. 2007.
Pericytes of human skeletal muscle are myogenic precur-
sors distinct from satellite cells. Nat Cell Biol 9:255–267.
AUTHORS’ CONTRIBUTIONS
20. Challen GA, Little MH. 2006. A side order of stem cells: the
AP drafted the Introduction. AD and MM drafted SP phenotype. Stem Cells 24:3–12.
section 2, Stem cells for skeletal muscle regeneration. 21. Motohashi N, Uezumi A, Yada E, et al. 2008. Muscle CD31
MT and KP drafted section 3, Biomaterials for skeletal () CD45() side population cells promote muscle regener-
muscle regeneration. AD drafted section 4, Challenges ation by stimulating proliferation and migration of myo-
with existing technologies and future trends. KG and blasts. Am J Pathol 173:781–791.
AD reviewed and revised each section. All authors have 22. Uezumi A, Fukada S, Yamamoto N, et al. 2010. Mesenchymal
progenitors distinct from satellite cells contribute to ectopic fat
read and approved the final version of the submitted
cell formation in skeletal muscle. Nat Cell Biol 12:143–152.
manuscript. 23. Joe AW, Yi L, Natarajan A, et al. 2010. Muscle injury
activates resident fibro/adipogenic progenitors that facili-
REFERENCES tate myogenesis. Nat Cell Biol 12:153–163.
1. Tedesco FS, Dellavalle A, Diaz-Manera J, et al. 2010. 24. Cottle BJ, Lewis FC, Shone V, et al. 2017. Skeletal muscle-
Repairing skeletal muscle: regenerative potential of skele- derived interstitial progenitor cells (PICs) display stem
tal muscle stem cells. J Clin Invest 120:11–19. cell properties, being clonogenic, self-renewing, and multi-
2. Pannerec A, Marazzi G, Sassoon D. 2012. Stem cells in the potent in vitro and in vivo. Stem Cell Res Ther 8:158.
hood: the skeletal muscle niche. Trends Mol Med 18:599–606. 25. Valero MC, Huntsman HD, Liu J, et al. 2012. Eccentric
3. Marcinczyk M, Elmashhady H, Talovic M, et al. 2017. exercise facilitates mesenchymal stem cell appearance in
Laminin-111 enriched fibrin hydrogels for skeletal muscle skeletal muscle. PLoS ONE 7:e29760.
regeneration. Biomaterials 141:233–242. 26. Ferrari G, Cusella-De Angelis G, Coletta M, et al. 1998.
4. Garg K, Boppart MD. 2016. Influence of exercise and aging Muscle regeneration by bone marrow-derived myogenic
on extracellular matrix composition in the skeletal muscle progenitors. Science 279:1528–1530.
stem cell niche. J Appl Physiol (1985) 121:1053–1058. 27. Cossu G, Sampaolesi M. 2007. New therapies for Duchenne
5. Garg K. 2017. Laminin Enriched Scaffolds for Tissue muscular dystrophy: challenges, prospects and clinical
Engineering Applications.Talovic M, Marcinczyk M, trials. Trends Mol Med 13:520–526.
Ziemkiewicz N, et al. 2017. Laminin enriched scaffolds for 28. Peault B, Rudnicki M, Torrente Y, et al. 2007. Stem and
tissue engineering applications. Adv Tissue Eng Regen progenitor cells in skeletal muscle development, mainte-
Med 2:194–200. nance, and therapy. Mol Ther 15:867–877.
6. Ervasti JM. 2000–2013. Structure and Function of the 29. Murry CE, Keller G. 2008. Differentiation of embryonic
Dystrophin-Glycoprotein Complex. Madame Curie Biosci- stem cells to clinically relevant populations: lessons from
ence Database: Landes Bioscience. embryonic development. Cell 132:661–680.
7. Liu J, Burkin DJ, Kaufman SJ. 2008. Increasing alpha 30. Luque E, Pe~ na J, Martin P, et al. 1995. Capillary supply
7 beta 1-integrin promotes muscle cell proliferation, during development of individual regenerating muscle
adhesion, and resistance to apoptosis without changing gene fibers. Anat Histol Embryol 24:87–89.
expression. Am J Physiol Cell Physiol 294:C627–C640. 31. Ochoa O, Sun D, Reyes-Reyna SM, et al. 2007. Delayed
8. Kuang S, Kuroda K, Le Grand F, et al. 2007. Asymmetric angiogenesis and VEGF production in CCR2-/- mice during
self-Renewal and commitment of satellite stem cells in impaired skeletal muscle regeneration. Am J Physiol Regul
muscle. Cell 129:999–1010. Integr Comp Physiol 293:R651–R661.
9. Jones DL, Wagers AJ. 2008. No place like home: anatomy and 32. Pillon NJ, Bilan PJ, Fink LN, et al. 2013. Cross-talk
function of the stem cell niche. Nat Rev Mol Cell Biol 9:11. between skeletal muscle and immune cells: muscle-derived
10. Bentzinger CF, Wang YX, Dumont NA, et al. 2013. Cellular mediators and metabolic implications. Am J Physiol Endo-
dynamics in the muscle satellite cell niche. EMBO Rep crinol Metab 304:E453–E465.
14:1062–1072. 33. Tidball JG, Villalta SA. 2010. Regulatory interactions between
11. Judson RN, Zhang RH, Rossi FM. 2013. Tissue-resident muscle and the immune system during muscle regeneration.
mesenchymal stem/progenitor cells in skeletal muscle: Am J Physiol Regul Integr Comp Physiol 298:R1173–R1187.
collaborators or saboteurs?. FEBS J 280:4100–4108. 34. Saclier M, Yacoub-Youssef H, Mackey AL, et al. 2013.
12. Mounier R, Chretien F, Chazaud B. 2011. Blood vessels Differentially activated macrophages orchestrate myogenic
and the satellite cell niche. Curr Top Dev Biol 96:121–138. precursor cell fate during human skeletal muscle regenera-
13. Christov C, Chretien F, Abou-Khalil R, et al. 2007. Muscle tion. Stem Cells 31:384–396.
satellite cells and endothelial cells: close neighbors and 35. Aguilar CA, Greising SM, Watts A, et al. 2018. Multiscale
privileged partners. Mol Biol Cell 18:1397–1409. analysis of a regenerative therapy for treatment of volu-
14. Le Grand F, Rudnicki MA. 2007. Skeletal muscle satellite metric muscle loss injury. Cell Death Discov 4:33.
cells and adult myogenesis. Curr Opin Cell Biol 19:628–633. 36. Hurtgen BJ, Ward CL, Garg K, et al. 2016. Severe muscle
15. Zammit PS, Partridge TA, Yablonka-Reuveni Z. 2006. The trauma triggers heightened and prolonged local musculo-
skeletal muscle satellite cell: the stem cell that came in skeletal inflammation and impairs adjacent tibia fracture
from the cold. J Histochem Cytochem 54:1177–1191. healing. J Musculoskelet Neuronal Interact 16:122–134.

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1258
2 DUNN ET AL.

37. Dunn A, Marcinczyk M, Talovic M, et al. 2018. Role of stem functional regeneration after severe skeletal muscle injury in
cells and extracellular matrix in skeletal muscle regenera- rats. Tissue Eng Part A 15:487–492.
tion. In: Sakuma PK editor. Muscle cell and tissue: 59. Matziolis G, Winkler T, Schaser K, et al. 2006. Autologous
inTechOpen. IntechOpen. bone marrow-derived cells enhance muscle strength following
38. Kuang S, Gillespie MA, Rudnicki MA. 2008. Niche regula- skeletal muscle crush injury in rats. Tissue Eng 12:361–367.
tion of muscle satellite cell self-renewal and differentiation. 60. von Roth P, Duda GN, Radojewski P, et al. 2012. Intra-
Cell Stem Cell 2:22–31. arterial MSC transplantation restores functional capacity
39. Shadrach JL, Wagers AJ. 2011. Stem cells for skeletal muscle after skeletal muscle trauma. Open Orthop J 6:352–356.
repair. Philos Trans R Soc Lond B Biol Sci 366:2297–2306. 61. Corona BT, Wenke JC, Walters TJ, et al. 2013. Intramus-
40. Sacco A, Doyonnas R, Kraft P, et al. 2008. Self-renewal and cular transplantation and survival of freshly isolated bone
expansion of single transplanted muscle stem cells. Nature marrow cells following skeletal muscle ischemia-reperfu-
456:502. sion injury. J Trauma Acute Care Surg 75:S142–S149.
41. Cerletti M, Jurga S, Witczak CA, et al. 2008. Highly 62. Corona BT, Rathbone CR. 2014. Accelerated functional
efficient, functional engraftment of skeletal muscle stem recovery after skeletal muscle ischemia-reperfusion injury
cells in dystrophic muscles. Cell 134:37–47. using freshly isolated bone marrow cells. J Surg Res
42. Meyer GA, Farris AL, Sato E, et al. 2015. Muscle progeni- 188:100–109.
tor cell regenerative capacity in the torn rotator cuff. 63. Zou K, Huntsman HD, Carmen Valero M, et al. 2015.
J Orthop Res 33:421–429. Mesenchymal stem cells augment the adaptive response to
43. Lindstrom M, Pedrosa-Domellof F, Thornell LE. 2010. eccentric exercise. Med Sci Sports Exerc 47:315–325.
Satellite cell heterogeneity with respect to expression of 64. Huntsman HD, Zachwieja N, Zou K, et al. 2013. Mesenchy-
MyoD, myogenin, Dlk1 and c-Met in human skeletal mal stem cells contribute to vascular growth in skeletal
muscle: application to a cohort of power lifters and seden- muscle in response to eccentric exercise. Am J Physiol
tary men. Histochem Cell Biol 134:371–385. Heart Circ Physiol 304:H72–H81.
44. Skuk D, Tremblay JP. 2003. Myoblast transplantation: the 65. Huntsman HD, Rendeiro C, Merritt JR, et al. 2017. The
current status of a potential therapeutic tool for myopa- impact of mechanically stimulated muscle-derived stromal
thies. J Muscle Res Cell Motil 24:285–300. cells on aged skeletal muscle. Exp Gerontol 103:35–46.
45. Skuk D, Tremblay JP. 2011. Intramuscular cell transplanta- 66. De Lisio M, Jensen T, Sukiennik RA, et al. 2014. Substrate
tion as a potential treatment of myopathies: clinical and and strain alter the muscle-derived mesenchymal stem cell
preclinical relevant data. Expert Opin Biol Ther 11:359–374. secretome to promote myogenesis. Stem Cell Res Ther 5:74.
46. Montarras D, Morgan J, Collins C, et al. 2005. Direct 67. Zhang Y, Zhu Y, Li Y, et al. 2015. Long-term engraftment
isolation of satellite cells for skeletal muscle regeneration. of myogenic progenitors from adipose-derived stem cells
Science 309:2064–2067. and muscle regeneration in dystrophic mice. Hum Mol
47. Skuk D, Goulet M, Tremblay JP. 2014. Intramuscular Genet 24:6029–6040.
transplantation of myogenic cells in primates: importance 68. de la Garza-Rodea AS, van der Velde-van Dijke I, Boersma
of needle size, cell number, and injection volume. Cell H, et al. 2012. Myogenic properties of human mesenchymal
Transplant 23:13–25. stem cells derived from three different sources. Cell
48. Kinoshita I, Vilquin JT, Guerette B, et al. 1994. Very Transplant 21:153–173.
efficient myoblast allotransplantation in mice under FK506 69. Mizuno H, Zuk PA, Zhu M, et al. 2002. Myogenic differenti-
immunosuppression. Muscle Nerve 17:1407–1415. ation by human processed lipoaspirate cells. Plast Reconstr
49. Guerette B, Asselin I, Skuk D, et al. 1997. Control of Surg 109:199–209. discussion 210-191.
inflammatory damage by anti-LFA-1: increase success of 70. Pinheiro CH, de Queiroz JC, Guimaraes-Ferreira L, et al.
myoblast transplantation. Cell Transplant 6:101–107. 2012. Local injections of adipose-derived mesenchymal
50. Collins CA, Olsen I, Zammit PS, et al. 2005. Stem cell stem cells modulate inflammation and increase angiogene-
function, self-renewal, and behavioral heterogeneity of cells sis ameliorating the dystrophic phenotype in dystrophin-
from the adult muscle satellite cell niche. Cell 122:289–301. deficient skeletal muscle. Stem Cell Rev 8:363–374.
51. Hall JK, Banks GB, Chamberlain JS, et al. 2010. Preven- 71. Lee EM, Kim AY, Lee EJ, et al. 2015. Therapeutic effects of
tion of muscle aging by myofiber-associated satellite cell mouse adipose-derived stem cells and losartan in the skele-
transplantation. Sci Transl Med 2:57ra83. tal muscle of injured mdx mice. Cell Transplant 24:939–953.
52. Suzuki K, Murtuza B, Heslop L, et al. 2002. Single fibers of 72. Ratajczak MZ, Majka M, Kucia M, et al. 2003. Expression
skeletal muscle as a novel graft for cell transplantation to of functional CXCR4 by muscle satellite cells and secretion
the heart. J Thorac Cardiovasc Surg 123:984–992. of SDF-1 by muscle-derived fibroblasts is associated with
53. Bianco P, Cao X, Frenette PS, et al. 2013. The meaning, the presence of both muscle progenitors in bone marrow
the sense and the significance: translating the science of and hematopoietic stem/progenitor cells in muscles. Stem
mesenchymal stem cells into medicine. Nat Med 19:35–42. Cells 21:363–371.
54. Phinney DG, Galipeau J, Krampera M, et al. 2013. MSCs: 73. Rosu-Myles M, Stewart E, Trowbridge J, et al. 2005.
science and trials. Nat Med 19:812. A unique population of bone marrow cells migrates to
55. Sheng G. 2015. The developmental basis of mesenchymal skeletal muscle via hepatocyte growth factor/c-met axis.
stem/stromal cells (MSCs). BMC Dev Biol 15:44. J Cell Sci 118:4343–4352.
56. Gang EJ, Darabi R, Bosnakovski D, et al. 2009. Engraft- 74. Gussoni E, Soneoka Y, Strickland CD, et al. 1999. Dystro-
ment of mesenchymal stem cells into dystrophin-deficient phin expression in the mdx mouse restored by stem cell
mice is not accompanied by functional recovery. Exp Cell transplantation. Nature 401:390–394.
Res 315:2624–2636. 75. LaBarge MA, Blau HM. 2002. Biological progression from
57. Lee JH, Kosinski PA, Kemp DM. 2005. Contribution of adult bone marrow to mononucleate muscle stem cell to
human bone marrow stem cells to individual skeletal multinucleate muscle fiber in response to injury. Cell
myotubes followed by myogenic gene activation. Exp Cell 111:589–601.
Res 307:174–182. 76. Corbel SY, Lee A, Yi L, et al. 2003. Contribution of
58. Winkler T, von Roth P, Matziolis G, et al. 2009. Dose-response hematopoietic stem cells to skeletal muscle. Nat Med
relationship of mesenchymal stem cell transplantation and 9:1528–1532.

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019
BIOMATERIALS AND STEM CELLS 1259
3

77. Sacco A, Doyonnas R, LaBarge MA, et al. 2005. IGF-I 97. Kuswanto W, Burzyn D, Panduro M, et al. 2016. Poor
increases bone marrow contribution to adult skeletal repair of skeletal muscle in aging mice reflects a defect in
muscle and enhances the fusion of myelomonocytic precur- local, interleukin-33-Dependent accumulation of regulatory
sors. J Cell Biol 171:483–492. T cells. Immunity 44:355–367.
78. Camargo FD, Green R, Capetenaki Y, et al. 2003. Single 98. Lemos DR, Paylor B, Chang C, et al. 2012. Functionally
hematopoietic stem cells generate skeletal muscle through convergent white adipogenic progenitors of different line-
myeloid intermediates. Nat Med 9:1520–1527. ages participate in a diffused system supporting tissue
79. Xynos A, Corbella P, Belmonte N, et al. 2010. Bone regeneration. Stem Cells 30:1152–1162.
marrow-derived hematopoietic cells undergo myogenic dif- 99. Fiore D, Judson RN, Low M, et al. 2016. Pharmacological
ferentiation following a Pax-7 independent pathway. Stem blockage of fibro/adipogenic progenitor expansion and sup-
Cells 28:965–973. pression of regenerative fibrogenesis is associated with
80. Lapidos KA, Chen YE, Earley JU, et al. 2004. Transplanted impaired skeletal muscle regeneration. Stem Cell Res
hematopoietic stem cells demonstrate impaired sarcoglycan 17:161–169.
expression after engraftment into cardiac and skeletal 100. Murphy MM, Lawson JA, Mathew SJ, et al. 2011. Satellite
muscle. J Clin Invest 114:1577–1585. cells, connective tissue fibroblasts and their interactions
81. Wernig G, Janzen V, Schafer R, et al. 2005. The vast are crucial for muscle regeneration. Development
majority of bone-marrow-derived cells integrated into mdx 138:3625–3637.
muscle fibers are silent despite long-term engraftment. 101. Lemos DR, Babaeijandaghi F, Low M, et al. 2015. Nilotinib
Proc Natl Acad Sci U S A 102:11852–11857. reduces muscle fibrosis in chronic muscle injury by promot-
82. Goldman SM, Henderson BEP, Corona BT. 2017. Evalua- ing TNF-mediated apoptosis of fibro/adipogenic progenitors.
tion of bone marrow mononuclear cells as an adjunct Nat Med 21:786–794.
therapy to minced muscle graft for the treatment of 102. Heredia JE, Mukundan L, Chen FM, et al. 2013. Type 2
volumetric muscle loss injuries. Stem Cell Res Ther 8:142. innate signals stimulate fibro/adipogenic progenitors to
83. Birbrair A, Delbono O. 2015. Pericytes are essential for facilitate muscle regeneration. Cell 153:376–388.
skeletal muscle formation. Stem Cell Rev 11:547–548. 103. Kang X, Yang MY, Shi YX, et al. 2018. Interleukin-15
84. Yin H, Price F, Rudnicki MA. 2013. Satellite cells and the facilitates muscle regeneration through modulation of fibro/
muscle stem cell niche. Physiol Rev 93:23–67. adipogenic progenitors. Cell Commun Signal 16:42.
85. Farrington-Rock C, Crofts NJ, Doherty MJ, et al. 2004. 104. Maherali N, Hochedlinger K. 2008. Guidelines and techni-
Chondrogenic and adipogenic potential of microvascular ques for the generation of induced pluripotent stem cells.
pericytes. Circulation 110:2226–2232. Cell Stem Cell 3:595–605.
86. Doherty MJ, Ashton BA, Walsh S, et al. 1998. Vascular 105. Choi IY, Lim H, Estrellas K, et al. 2016. Concordant but
pericytes express osteogenic potential in vitro and in vivo. varied phenotypes among duchenne muscular dystrophy
J Bone Miner Res 13:828–838. patient-Specific myoblasts derived using a human iPSC-
87. Dellavalle A, Maroli G, Covarello D, et al. 2011. Pericytes Based model. Cell Rep 15:2301–2312.
resident in postnatal skeletal muscle differentiate into 106. Mizuno Y, Chang H, Umeda K, et al. 2010. Generation of
muscle fibres and generate satellite cells. Nat Commun skeletal muscle stem/progenitor cells from murine induced
2:499. pluripotent stem cells. FASEB J 4:2245–2253.
88. Murray IR, Baily JE, Chen WCW, et al. 2017. Skeletal and 107. Goudenege S, Lebel C, Huot NB, et al. 2012. Myoblasts
cardiac muscle pericytes: functions and therapeutic poten- derived from normal hESCs and dystrophic hiPSCs effi-
tial. Pharmacol Ther 171:65–74. ciently fuse with existing muscle fibers following transplan-
89. Farup J, De Lisio M, Rahbek SK, et al. 2015. Pericyte tation. Mol Ther 20:2153–2167.
response to contraction mode-specific resistance exercise 108. Maffioletti SM, Sarcar S, Henderson ABH, et al. 2018.
training in human skeletal muscle. J Appl Physiol (1985) Three-Dimensional human iPSC-Derived artificial skeletal
119:1053–1063. muscles model muscular dystrophies and enable multi-
90. Birbrair A, Zhang T, Wang ZM, et al. 2013. Role of lineage tissue engineering. Cell Rep 23:899–908.
pericytes in skeletal muscle regeneration and fat accumula- 109. Chal J, Al Tanoury Z, Hestin M, et al. 2016. Generation
tion. Stem Cells Dev 22:2298–2314. of human muscle fibers and satellite-like cells from
91. Kostallari E, Baba-Amer Y, Alonso-Martin S, et al. 2015. human pluripotent stem cells in vitro. Nat Protoc
Pericytes in the myovascular niche promote post-natal 11:1833–1850.
myofiber growth and satellite cell quiescence. Development 110. Jiwlawat S, Lynch E, Glaser J, et al. 2017. Differentiation
142:1242–1253. and sarcomere formation in skeletal myocytes directly
92. Lorant J, Saury C, Schleder C, et al. 2017. Skeletal muscle prepared from human induced pluripotent stem cells using
regenerative potential of human MuStem cells following a sphere-based culture. Differentiation 96:70–81.
transplantation into injured mice muscle. Mol Ther 111. Rao L, Qian Y, Khodabukus A, et al. 2018. Engineering
26:618–633. human pluripotent stem cells into a functional skeletal
93. Meng J, Adkin CF, Xu SW, et al. 2011. Contribution of muscle tissue. Nat Commun 9:126.
human muscle-derived cells to skeletal muscle regeneration 112. Zhao T, Zhang ZN, Rong Z, et al. 2011. Immunogenicity of
in dystrophic host mice. PLoS ONE 6:e17454. induced pluripotent stem cells. Nature 474:212–215.
94. Pretheeban T, Lemos DR, Paylor B, et al. 2012. Role of 113. Tateno H, Onuma Y, Ito Y, et al. 2015. Elimination of
stem/progenitor cells in reparative disorders. Fibrogenesis tumorigenic human pluripotent stem cells by a recombi-
Tissue Repair 5:20. nant lectin-toxin fusion protein. Stem Cell Rep 4:811–820.
95. Gonzalez D, Contreras O, Rebolledo DL, et al. 2017. ALS 114. Miyagoe-Suzuki Y, Takeda S. 2017. Skeletal muscle gener-
skeletal muscle shows enhanced TGF-beta signaling, fibro- ated from induced pluripotent stem cells  induction and
sis and induction of fibro/adipogenic progenitor markers. application. World J Stem Cells 9:89–97.
PloS ONE 12:e0177649. 115. Tanaka KK, Hall JK, Troy AA, et al. 2009. Syndecan-4-
96. Uezumi A, Ito T, Morikawa D, et al. 2011. Fibrosis and expressing muscle progenitor cells in the SP engraft as
adipogenesis originate from a common mesenchymal pro- satellite cells during muscle regeneration. Cell Stem Cell
genitor in skeletal muscle. J Cell Sci 124:3654–3664. 4:217–225.

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1260
2 DUNN ET AL.

116. Bachrach E, Li S, Perez AL, et al. 2004. Systemic delivery isolated satellite cells embedded in a photopolymerizable
of human microdystrophin to regenerating mouse dystro- hydrogel. FASEB J 25:2296–2304.
phic muscle by muscle progenitor cells. Proc Natl Acad Sci 136. Pollot BE, Rathbone CR, Wenke JC, et al. 2018. Natural
U S A 101:3581–3586. polymeric hydrogel evaluation for skeletal muscle tissue
117. Doyle MJ, Zhou S, Tanaka KK, et al. 2011. Abcg2 labels engineering. J Biomed Mater Res B Appl Biomater
multiple cell types in skeletal muscle and participates in 106:672–679.
muscle regeneration. J Cell Biol 195:147–163. 137. Huang YC, Dennis RG, Larkin L, et al. 2005. Rapid
118. Penton CM, Thomas-Ahner JM, Johnson EK, et al. 2013. formation of functional muscle in vitro using fibrin gels.
Muscle side population cells from dystrophic or injured J Appl Physiol (1985) 98:706–713.
muscle adopt a fibro-adipogenic fate. PLoS ONE 8:e54553. 138. Page RL, Malcuit C, Vilner L, et al. 2011. Restoration of
119. Li Z. 2013. CD133: a stem cell biomarker and beyond. Exp skeletal muscle defects with adult human cells delivered on
Hematol Oncol 2:17. fibrin microthreads. Tissue Eng Part A 17:2629–2640.
120. Negroni E, Riederer I, Chaouch S, et al. 2009. In vivo 139. Gilbert-Honick J, Iyer SR, Somers SM, et al. 2018.
myogenic potential of human CD133þ muscle-derived stem Engineering functional and histological regeneration of
cells: a quantitative study. Mol Ther 17:1771–1778. vascularized skeletal muscle. Biomaterials 164:70–79.
121. Meng J, Chun S, Asfahani R, et al. 2014. Human skeletal 140. Corona BT, Wu X, Ward CL, et al. 2013. The promotion of
muscle-derived CD133(þ) cells form functional satellite a functional fibrosis in skeletal muscle with volumetric
cells after intramuscular transplantation in immunode- muscle loss injury following the transplantation of muscle-
ficient host mice. Mol Ther 22:1008–1017. ECM. Biomaterials 34:3324–3335.
122. Shi M, Ishikawa M, Kamei N, et al. 2009. Acceleration of 141. Tomblyn S, Pettit Kneller EL, Walker SJ, et al. 2016.
skeletal muscle regeneration in a rat skeletal muscle injury Keratin hydrogel carrier system for simultaneous delivery
model by local injection of human peripheral blood-derived of exogenous growth factors and muscle progenitor cells.
CD133-positive cells. Stem Cells 27:949–960. J Biomed Mater Res B Appl Biomater 104:864–879.
123. Torrente Y, Belicchi M, Marchesi C, et al. 2007. Autologous 142. Baker HB, Passipieri JA, Siriwardane M, et al. 2017. Cell
transplantation of muscle-derived CD133þ stem cells in and growth factor-loaded keratin hydrogels for treatment
Duchenne muscle patients. Cell Transplant 16:563–577. of volumetric muscle loss in a mouse model. Tissue Eng
124. Murua A, Portero A, Orive G, et al. 2008. Cell microencap- Part A 23:572–584.
sulation technology: towards clinical application. J Control 143. Machingal MA, Corona BT, Walters TJ, et al. 2011.
Release 132:76–83. A tissue-engineered muscle repair construct for functional
125. Gilbert PM, Havenstrite KL, Magnusson KEG, et al. 2010. restoration of an irrecoverable muscle injury in a murine
Substrate elasticity regulates skeletal muscle stem cell self- model. Tissue Eng Part A 17:2291–2303.
renewal in culture. Science 329:1078–1081. 144. Passipieri JA, Baker HB, Siriwardane M, et al. 2017.
126. Lacraz G, Rouleau AJ, Couture V, et al. 2015. Increased Keratin hydrogel enhances in vivo skeletal muscle function
stiffness in aged skeletal muscle impairs muscle progenitor in a rat model of volumetric muscle loss. Tissue Eng Part A
cell proliferative activity. PLoS ONE 10:e0136217. 23:556–571.
127. Urciuolo A, Quarta M, Morbidoni V, et al. 2013. Collagen 145. Andrade BM, Baldanza MR, Ribeiro KC, et al. 2015. Bone
VI regulates satellite cell self-renewal and muscle regener- marrow mesenchymal cells improve muscle function in a
ation. Nat Commun 4:1964. skeletal muscle re-injury model. PLoS ONE 10:e0127561.
128. Mase VJ, Jr., Hsu JR, Wolf SE, et al. 2010. Clinical 146. Natsu K, Ochi M, Mochizuki Y, et al. 2004. Allogeneic bone
application of an acellular biologic scaffold for surgical marrow-derived mesenchymal stromal cells promote the
repair of a large, traumatic quadriceps femoris muscle regeneration of injured skeletal muscle without differentia-
defect. Orthopedics 33:511. tion into myofibers. Tissue Eng 10:1093–1112.
129. Gentile NE, Stearns KM, Brown EH, et al. 2014. Targeted 147. Pumberger M, Qazi TH, Ehrentraut MC, et al. 2016.
rehabilitation after extracellular matrix scaffold transplan- Synthetic niche to modulate regenerative potential of
tation for the treatment of volumetric muscle loss. Am J MSCs and enhance skeletal muscle regeneration. Biomate-
Phys Med Rehabil 93:S79–S87. rials 99:95–108.
130. Garg K, Ward CL, Rathbone CR, et al. 2014. Transplanta- 148. Merritt EK, Cannon MV, Hammers DW, et al. 2010. Repair
tion of devitalized muscle scaffolds is insufficient for of traumatic skeletal muscle injury with bone-marrow-
appreciable de novo muscle fiber regeneration after volu- derived mesenchymal stem cells seeded on extracellular
metric muscle loss injury. Cell and Tissue Res matrix. Tissue Eng Part A 16:2871–2881.
358:857–873. 149. Qiu X, Liu S, Zhang H, et al. 2018. Mesenchymal stem cells
131. Aurora A, Roe JL, Corona BT, et al. 2015. An acellular and extracellular matrix scaffold promote muscle regenera-
biologic scaffold does not regenerate appreciable de novo tion by synergistically regulating macrophage polarization
muscle tissue in rat models of volumetric muscle loss toward the M2 phenotype. Stem Cell Res Ther 9:88.
injury. Biomaterials 67:393–407. 150. Hwang JH, Kim IG, Piao S, et al. 2013. Combination
132. Turner NJ, Yates AJ, Jr., Weber DJ, et al. 2010. Xenoge- therapy of human adipose-derived stem cells and basic
neic extracellular matrix as an inductive scaffold for fibroblast growth factor hydrogel in muscle regeneration.
regeneration of a functioning musculotendinous junction. Biomaterials 34:6037–6045.
Tissue Eng Part A 16:3309–3317. 151. Huang H, Liu J, Hao H, et al. 2017. Preferred M2
133. Sicari BM, Agrawal V, Siu BF, et al. 2012. A murine model polarization by ASC-based hydrogel accelerated angiogene-
of volumetric muscle loss and a regenerative medicine sis and myogenesis in volumetric muscle loss rats. Stem
approach for tissue replacement. Tissue Eng Part A Cells Int 2017:2896874.
18:1941–1948. 152. Aurora A, Wrice N, Walters TJ, et al. 2018. A PEGylated
134. Matthias N, Hunt SD, Wu J, et al. 2018. Volumetric muscle platelet free plasma hydrogel based composite scaffold
loss injury repair using in situ fibrin gel cast seeded with enables stable vascularization and targeted cell delivery for
muscle-derived stem cells (MDSCs). Stem Cell Res 27:65–73. volumetric muscle loss. Acta Biomater 65:150–162.
135. Rossi CA, Flaibani M, Blaauw B, et al. 2011. In vivo 153. Corona BT, Garg K, Ward CL, et al. 2013. Autologous
tissue engineering of functional skeletal muscle by freshly minced muscle grafts: a tissue engineering therapy for the

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019
BIOMATERIALS AND STEM CELLS 1261
3

volumetric loss of skeletal muscle. Am J Physiol Cell 172. Vorotnikova E, McIntosh D, Dewilde A, et al. 2010.
Physiol 305:C761–C775. Extracellular matrix-derived products modulate endothelial
154. Hurtgen BJ, Ward CL, Leopold Wager CM, et al. 2017. and progenitor cell migration and proliferation in vitro and
Autologous minced muscle grafts improve endogenous stimulate regenerative healing in vivo. Matrix Biol
fracture healing and muscle strength after musculoskeletal 29:690–700.
trauma. Physiol Rep 5:e13362. 173. Tottey S, Johnson SA, Crapo PM, et al. 2011. The effect of
155. Ward CL, Pollot BE, Goldman SM, et al. 2016. Autologous source animal age upon extracellular matrix scaffold
minced muscle grafts improve muscle strength in a porcine properties. Biomaterials 32:128–136.
model of volumetric muscle loss injury. J Orthop Trauma 174. Tottey S, Corselli M, Jeffries EM, et al. 2011. Extracellular
30:e396–e403. matrix degradation products and low-oxygen conditions
156. Ward CL, Ji L, Corona BT. 2015. An autologous muscle enhance the regenerative potential of perivascular stem
tissue expansion approach for the treatment of volumetric cells. Tissue Eng Part A 17:37–44.
muscle loss. Biores Open Access 4:198–208. 175. Agrawal V, Siu BF, Chao H, et al. 2012. Partial characteri-
157. Goldman SM, Corona BT. 2017. Co-delivery of micronized zation of the Sox2þ cell population in an adult murine
urinary bladder matrix damps regenerative capacity of model of digit amputation. Tissue Eng Part A
minced muscle grafts in the treatment of volumetric muscle 18:1454–1463.
loss injuries. PloS ONE 12:e0186593. 176. Crapo PM, Tottey S, Slivka PF, et al. 2014. Effects of
158. Brown BN, Badylak SF. 2014. Extracellular matrix as an biologic scaffolds on human stem cells and implications for
inductive scaffold for functional tissue reconstruction. CNS tissue engineering. Tissue Eng Part A 20:313–323.
Transl Res 163:268–285. 177. Fishman JM, Lowdell MW, Urbani L, et al. 2013. Immuno-
159. Brown BN, Londono R, Tottey S, et al. 2012. Macrophage modulatory effect of a decellularized skeletal muscle scaf-
phenotype as a predictor of constructive remodeling follow- fold in a discordant xenotransplantation model. Proc Natl
ing the implantation of biologically derived surgical mesh Acad Sci U S A 110:14360–14365.
materials. Acta Biomater 8:978–987. 178. Zhang P, Sun F, Liu S, et al. 2016. Anti-PEG antibodies in
160. Brown BN, Valentin JE, Stewart-Akers AM, et al. 2009. the clinic: current issues and beyond PEGylation. J Control
Macrophage phenotype and remodeling outcomes in re- Release 244:184–193.
sponse to biologic scaffolds with and without a cellular 179. Jiang D, Liang J, Noble PW. 2011. Hyaluronan as an immune
component. Biomaterials 30:1482–1491. regulator in human diseases. Physiol Rev 91:221–264.
161. Sicari BM, Rubin JP, Dearth CL, et al. 2014. An acellular 180. Li R, Liang J, He Y, et al. 2018. Sustained release of
biologic scaffold promotes skeletal muscle formation in immunosuppressant by nanoparticle-anchoring hydrogel
mice and humans with volumetric muscle loss. Sci Transl scaffold improved the survival of transplanted stem cells
Med 6:234ra258. and tissue regeneration. Theranostics 8:878–893.
162. Valentin JE, Turner NJ, Gilbert TW, et al. 2010. Func- 181. Chang CW, Petrie T, Clark A, et al. 2016. Mesenchymal
tional skeletal muscle formation with a biologic scaffold. stem cell seeding of porcine small intestinal submucosal
Biomaterials 31:7475–7484. extracellular matrix for cardiovascular applications. PLoS
163. Goldman SM, Henderson BEP, Walters TJ, et al. 2018. ONE 11:e0153412.
Co-delivery of a laminin-111 supplemented hyaluronic 182. Trounson A, Thakar RG, Lomax G, et al. 2011. Clinical
acid based hydrogel with minced muscle graft in the trials for stem cell therapies. BMC Med 9:52.
treatment of volumetric muscle loss injury. PLoS ONE 13: 183. Fishman JM, Tyraskis A, Maghsoudlou P, et al. 2013.
e0191245. Skeletal muscle tissue engineering: which cell to use?
164. Corona BT, Greising SM. 2016. Challenges to acellular Tissue Eng Part B Rev 19:503–515.
biological scaffold mediated skeletal muscle tissue regener- 184. Kaplan FS, Glaser DL, Hebela N, et al. 2004. Heterotopic
ation. Biomaterials 104:238–246. ossification. JAAOS 12:116–125.
165. Garg K, Corona BT, Walters TJ. 2014. Losartan adminis- 185. Hoyt BW, Pavey GJ, Potter BK, et al. 2018. Heterotopic
tration reduces fibrosis but hinders functional recovery ossification and lessons learned from fifteen years at war: a
after volumetric muscle loss injury. J Appl Physiol (1985) review of therapy, novel research, and future directions for
117:1120–1131. military and civilian orthopaedic trauma. Bone 109:3–11.
166. Garg K, Corona BT, Walters TJ. 2015. Therapeutic strate- 186. Olmsted-Davis E, Gannon FH, Ozen M, et al. 2007.
gies for preventing skeletal muscle fibrosis after injury. Hypoxic adipocytes pattern early heterotopic bone forma-
Front Pharmacol 6:87. tion. Am J Pathol 170:620–632.
167. Elmashhady H, Kraemer BA, Patel KH, et al. 2017. 187. Nelson ER, Wong VW, Krebsbach PH, et al. 2012. Hetero-
Decellularized extracellular matrices for tissue engineering topic ossification following burn injury: the role of stem
applications. Electrospinnning 1:87–99. cells. J Burn Care Res 33:463–470.
168. Gentile NE, Stearns KM, Brown EHP, et al. 2014. Targeted 188. Hashimoto N, Kiyono T, Wada MR, et al. 2008. Osteogenic
rehabilitation after extracellular matrix scaffold transplan- properties of human myogenic progenitor cells. Mech Dev
tation for the treatment of volumetric muscle loss. Am J 125:257–269.
Phys Med Rehabil 93:S79–S87. 189. Lees-Shepard JB, Yamamoto M, Biswas AA, et al. 2018.
169. Machingal MA, Corona BT, Walters TJ, et al. 2011. Activin-dependent signaling in fibro/adipogenic progenitors
A tissue-engineered muscle repair construct for functional causes fibrodysplasia ossificans progressiva. Nat Commu
restoration of an irrecoverable muscle injury in a murine 9:471.
model. Tissue Eng Part A 17:2291–2303. 190. Pollot BE, Goldman SM, Wenke JC, et al. 2016. Decellular-
170. Merritt EK, Hammers DW, Tierney M, et al. 2010. ized extracellular matrix repair of volumetric muscle loss
Functional assessment of skeletal muscle regeneration injury impairs adjacent bone healing in a rat model of
utilizing homologous extracellular matrix as scaffolding. complex musculoskeletal trauma. J Trauma Acute Care
Tissue Eng Part A 16:1395–1405. Surg 81:S184.
171. Reing JE, Zhang L, Myers-Irvin J, et al. 2009. Degradation 191. Huntsman HD, Rendeiro C, Merritt JR, et al. 2018. The
products of extracellular matrix affect cell migration and impact of mechanically stimulated muscle-derived stromal
proliferation. Tissue Eng Part A 15:605–614. cells on aged skeletal muscle. Exp Gerontol 103:35–46.

JOURNAL OF ORTHOPAEDIC RESEARCH1 JUNE


MONTH 2018
2019
1262
2 DUNN ET AL.

192. Huntsman HD, Zachwieja N, Zou K, et al. 2012. Mesenchy- 197. Egusa H, Kobayashi M, Matsumoto T, et al. 2013. Applica-
mal stem cells contribute to vascular growth in skeletal tion of cyclic strain for accelerated skeletal myogenic
muscle in response to eccentric exercise. Am J Physiol differentiation of mouse bone marrow-derived mesenchy-
Heart Circ Physiol 304:H72–H81. mal stromal cells with cell alignment. Tissue Eng Part A
193. Moon DG, Christ G, Stitzel JD, et al. 2008. Cyclic mechani- 19:770–782.
cal preconditioning improves engineered muscle contrac- 198. Yilgor Huri P, Cook CA, Hutton DL, et al. 2013. Biophysi-
tion. Tissue Eng Part A 14:473–482. cal cues enhance myogenesis of human adipose derived
194. Corona BT, Machingal MA, Criswell T, et al. 2012. Further stem/stromal cells. Biochem Biophys Res Commun 438:
development of a tissue engineered muscle repair construct 180–185.
in vitro for enhanced functional recovery following implan- 199. Jo H, Sim M, Kim S, et al. 2017. Electrically conductive
tation in vivo in a murine model of volumetric muscle loss graphene/polyacrylamide hydrogels produced by mild
injury. Tissue Eng Part A 18:1213–1228. chemical reduction for enhanced myoblast growth and
195. Quarta M, Cromie M, Chacon R, et al. 2017. Bioengineered differentiation. Acta Biomater 48:100–109.
constructs combined with exercise enhance stem cell-medi- 200. Nakamura Y, Miyaki S, Ishitobi H, et al. 2015. Mesenchy-
ated treatment of volumetric muscle loss. Nat Commun mal-stem-cell-derived exosomes accelerate skeletal muscle
8:15613. regeneration. FEBS Letters 589:1257–1265.
196. Glaeser JD, Geissler S, Ode A, et al. 2010. Modulation of 201. Choi JS, Yoon HI, Lee KS, et al. 2016. Exosomes from
matrix metalloprotease-2 levels by mechanical loading of differentiating human skeletal muscle cells trigger myo-
three-dimensional mesenchymal stem cell constructs: im- genesis of stem cells and provide biochemical cues for
pact on in vitro tube formation. Tissue Eng Part A 16: skeletal muscle regeneration. J Controlled Release 222:
3139–3148. 107–115.

1 1
ALJOURNAL
OF ORTHOPAEDIC
OF ORTHOPAEDIC
RESEARCH
RESEARCH
JUNE 2019
JUNE 2019

You might also like