G. Engineering
G. Engineering
G. Engineering
Engineering
Page 1 of 57
© Mary Ann Liebert, Inc.
DOI: 10.1089/ten.TEB.2017.0451
1
Stem Cells for Skeletal Muscle Tissue Engineering
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Molly N. Pantelic MS1 and Lisa M. Larkin PhD1,2
1
Molecular and Integrative Physiology, University of Michigan, 2025 BSRB, 109 Zina Pitcher
Place, Ann Arbor, MI, 48109‐2200
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
2
Biomedical Engineering, University of Michigan, 2025 BSRB, 109 Zina Pitcher Place, Ann
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Arbor, MI, 48109‐2200
Tissue Engineering
Corresponding Author: Lisa Larkin, PhD
Associate Professor
Molecular and Integrative Physiology
Biomedical Engineering
University of Michigan
Biomedical Science Research Building (BSRB)
109 Zina Pitcher Place
Room #2025
48109‐2200
Phone: (734) 936‐8181
Fax: (734) 615‐3292
e‐mail: [email protected]
Page 2 of 57
2
Abstract
Volumetric muscle loss (VML) is a debilitating condition wherein muscle loss
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
overwhelms the body’s normal physiological repair mechanism. VML is particularly
common among military service members who have sustained war injuries. Because of the
high social and medical cost associated with VML and suboptimal current surgical
treatments, there is great interest in developing better VML therapies. Skeletal muscle
tissue engineering (SMTE) is a promising alternative to traditional VML surgical treatments
that use autogenic tissue grafts, and rather uses isolated stem cells with myogenic
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
potential to generate de novo skeletal muscle tissues to treat VML. Satellite cells are the
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
native precursors to skeletal muscle tissue, and are thus the most commonly studied
starting source for SMTE. However, satellite cells are difficult to isolate and purify, and it is
presently unknown whether they would be a practical source in clinical SMTE applications.
Alternative myogenic stem cells, including adipose‐derived stem cells (ADSCs), bone
marrow‐derived mesenchymal stem cells (BM‐MSCs), perivascular stem cells (PVSCs),
Tissue Engineering
umbilical cord mesenchymal stem cells (UC‐MSCs), induced pluripotent stem cells (iPSCs)
and embryonic stem cells (ESCs) each have myogenic potential and have been identified as
possible starting sources for SMTE, though they have yet to be studied in detail for this
purpose. These alternative stem cell varieties offer unique advantages and disadvantages
that are worth exploring further to advance the SMTE field towards highly functional, safe,
and practical VML treatments. The following review summarizes the current state of
satellite cell‐based SMTE, details the properties and practical advantages of alternative
myogenic stem cells, and offers guidance to tissue engineers on how alternative myogenic
stem cells can be incorporated into SMTE research.
Page 3 of 57
3
Introduction
Volumetric muscle loss (VML) is skeletal muscle loss due to injury, denervation,
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
disease, or tumor excision that exceeds the body’s regenerative capacity. Thus, VML
cannot be cured via the endogenous repair process, and often requires surgical
intervention. VML is particularly prevalent among military service members who have
sustained war injuries, which is a general category that includes blast injuries, gunshot
wounds, and fragmentation injuries (1,2). Recent military conflicts like Operation Iraqi
Freedom have especially high rates of extremity injury and VML due to the nature of
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
combat (3,4), so VML cases are becoming more prevalent over time.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
VML causes chronic pain, impaired function, and reduced mobility. A study by
Corona et al. (5) found that VML was directly responsible for disability in 8% of all disabled
veterans surveyed, and within a group of veterans with type III open tibia fractures, VML
was an important contributor to 65% of the permanent disability they experienced. Corona
et al. also found that the average lifetime disability cost for surveyed veterans with VML in
Tissue Engineering
the general disability population was $341,300 per person (5). Compounding this problem,
current surgical procedures to treat injury‐induced VML are suboptimal at best, and full
structural and functional recovery is rare (6).
The most common procedure that is currently used to treat VML is muscle flap
surgery (6), which involves transferring autologous tissue with intact blood and nerve
supply from a donor site into the patient's injury site. Typically, this tissue is rotated from
an adjacent donor site directly into the injury site; however, in the case of free functional
muscle transfer, autologous donor muscle is transplanted from a site not immediately
adjacent to the patient's injury, and the arteries, veins, and nerves are cut at the donor site
and sewed into the injury site (6). Muscle flap procedures are severely limited in terms of
the amount of tissue that can be taken and rarely result in full functional recovery (6).
Donor site morbidity, which refers to any complication in the patient's donor site as it
heals, is also a significant concern with these procedures (6).
Composite tissue allografts (CTAs) have been used successfully in recent years for
face, hand, larynx, and abdominal wall transplants (7–12), and they may emerge as a more
Page 4 of 57
4
popular surgical treatment for VML in the coming years. CTA procedures transplant
combinations of skeletal muscle, skin, bone, and tendon from an organ donor that is
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
typically not genetically identical to the patient. More tissue can be implanted using CTA,
but this procedure also carries significant negative side effects. Patients require lifelong
immunosuppression, and tissue rejection, malignancies, and infections are serious
concerns.
Because of the complications and inadequacies associated with current VML
surgeries, there is intense interest in devising better treatment options for VML. In more
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
recent years, skeletal muscle tissue engineering (SMTE) has emerged as a promising
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
alternative to traditional VML surgical procedures. Generally, SMTE protocols start with a
biopsy to isolate a stem cell population with myogenic potential. After the stem cells are
allowed to proliferate, they may be differentiated in vitro. Depending on the specific
protocol used, the cells can be seeded onto a scaffold for structural support (13–15) or
allowed to develop into a scaffoldless skeletal muscle tissue prior to implantation (16). The
Tissue Engineering
engineered tissue is implanted back into the VML injury site to restore structure and
function. Vascularization of engineered skeletal muscle tissues is critical for proper
functioning in vivo, and is thus a burgeoning field of study, with several published methods
to date (17–21). As summarized in a review of engineered skeletal muscle techniques by
Bian et al. (22), innervation methods are also being developed for skeletal muscle tissue
engineering, as innervation is essential for the development of functional, non‐atrophic
skeletal muscle.
As an alternative to the general SMTE process, some labs have experimented with
decellularization techniques that use biologic scaffolds composed of extracellular matrix
(ECM) proteins. When implanted into a VML site, ECM scaffolds encourage some degree of
stem cell recruitment and de novo fiber regeneration without a prior biopsy and in vitro
cell culture (23). These methods rely on ECM's ability to create an environment for wound
healing that minimizes infection, promotes moisture balance, and regulates cell behavior
to promote tissue repair (24). However, Garg et al. (25) found that using an ECM scaffold
alone without a stem cell source was unable to recruit enough host stem cells in vivo to
promote sufficient de novo fiber regeneration for functional recovery, and most SMTE
Page 5 of 57
5
laboratories currently use stem cells in their protocols.
Developing a successful SMTE protocol relies on choosing the proper stem cell
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
starting source, and making this determination requires a detailed understanding of the
properties of myogenic stem cells. Ideally, a stem cell population for SMTE should be easy
to isolate, easy to purify, easy to expand in vitro without sacrificing myogenic potential,
and able to properly fuse with existing skeletal muscle tissue in vivo to contribute to
structural and functional VML recovery.
Because satellite cells are the native stem cell precursor to skeletal muscle, they
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
are the most common starting source for SMTE. However, there are difficulties associated
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
with using satellite cells. Satellite cell isolations are laborious, time‐consuming, and
variable between research labs. There is no consensus as to whether purification is
required, but in protocols that use some degree of purification, the purification process
can be challenging, and currently used methods are inconsistent, wasteful, or impractical.
Looking towards personalized medicine applications, there are concerns as to how an
Tissue Engineering
adequate satellite cell population could be obtained. Acquiring enough satellite cells from
a small skeletal muscle biopsy to produce a sufficiently sized tissue is challenging, yet a
large skeletal muscle biopsy would not be clinically feasible because of the potential
irreversible injury at the biopsy site. Senescence of satellite cells as they are expanded in
vivo causes them to lose proliferative capacity, which reduces their ability to contribute to
myogenesis over time (26,27). More generally, growing skeletal muscle in vitro is also
challenging because there is not a devoted collection of growth factors whose only
purpose is to induce skeletal muscle growth, whereas, for instance, bone and cartilage
tissue engineering methods have had great success using bone morphogenic proteins
(BMPs) (28).
To overcome the problems currently associated with satellite cell‐based SMTE,
tissue engineers should investigate the best stem cell source for SMTE. Alternative
varieties of myogenic stem cells, including adipose‐derived stem cells, bone marrow‐
derived mesenchymal stem cells, perivascular stem cells, and induced pluripotent stem
cells may offer attractive practical advantages over satellite cells, and should thus be
explored in future SMTE studies in addition to satellite cells to advance SMTE towards
Page 6 of 57
6
highly functional, safe, and clinically feasible VML treatments. Before discussing these
alternative stem cells, it is first important to understand satellite cell properties and the
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
current state of satellite cell‐based SMTE.
Satellite Cells
Satellite cells are located in all skeletal muscle tissues beneath the basal lamina and
outside the surrounding myofiber plasma membrane (29,30) and are the primary
contributors to skeletal muscle regeneration. In fact, Lepper et al. (31) showed that
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
satellite cells are absolutely required for normal muscle regeneration in vivo. Normal
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
satellite cell myogenesis (Figure 1) in response to injury begins when satellite cells are
activated by local signals and upregulate the transcription factor myogenic differentiation
1 (MyoD) and myogenic regulatory factor 5 (Myf5), thereby committing to a myoblastic
lineage and becoming myoblasts (32,33) in a process termed induction. Myoblasts
proliferate and differentiate into myocytes, which fuse into multinucleated myotubes after
Tissue Engineering
upregulating the transcription factor myogenin in a process called terminal differentiation.
Alternatively, myoblasts can directly fuse with damaged myofibers in vivo. After terminal
differentiation, myotubes can mature into myofibers.
The essential satellite cell marker is paired box protein‐7 (Pax7), which is a
transcription factor that is required for normal satellite cell function, and is expressed by
both quiescent and proliferating satellite cells (34). Beyond Pax7, there is significant
heterogeneity within the satellite cell population along with differences in satellite cell
marker expression depending on the anatomical location of their niche (35).
The anatomical location of the satellite cell niche also affects satellite cell
differentiation potential. For instance, as demonstrated by Collins et al. (36), satellite cells
from mouse extensor digitorum longus and soleus muscles contribute significantly more to
regeneration in vivo than tibialis anterior (TA) muscle satellite cells. These differences are
caused by local stem cell niche and injury microenvironment (37), and are also affected by
differences in cellular composition of the niche that arise from two distinct satellite cell
populations that Shultz (38) identified: a reserve population and a responsive population.
The responsive satellite cell population comprises roughly 80% of total satellite cells and is
the population that readily divides to contribute to de novo myofiber formation (38). The
Page 7 of 57
7
remaining 20% of satellite cells are part of a reserve population that divides slowly and
only in response to an intensive need for muscle regeneration (38).
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Satellite Cell Isolation
All satellite cell‐based SMTE protocols must create an in vitro approximation of the
in vivo stem cell niche and injury environment to induce proper induction, proliferation,
terminal differentiation, and tissue formation, and each protocol must begin with proper
satellite cell isolation. Satellite cell isolations begin with a skeletal muscle biopsy to obtain
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
skeletal muscle tissue. Skeletal muscle biopsies are painful, and only a small amount of
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
tissue can be taken before a permanent functional deficit is introduced. After biopsy, most
satellite cell isolation methods fall into one of two general categories: single fiber explant
culture or enzymatic digestion.
Explant culture is a general category of cell isolation wherein pieces of tissue are
plated and cells are allowed to egress out of the tissue and onto the cell culture dish.
Tissue Engineering
Bischoff et al. (39) developed the first single fiber explant culture isolation protocol for
satellite cell isolation in rats, and it was later improved upon by Rosenblatt et al. (26). In
their current state, single fiber explant culture methods (Figure 2, top) (40) are highly pure
but low yield due to the nature of isolation (40). In contrast, enzymatic digestion methods
(Figure 2, bottom) (40) are less pure but yield larger satellite cell populations, and are thus
the most common isolation method currently used in SMTE (40). Enzymatic digestion
protocols involve dissecting muscle biopsies, removing any connective tissue by hand,
mincing the muscle, and then performing enzymatic digestion, using enzymes like trypsin,
Pronase, dispase, and collagenase to reduce the tissue into individual cells (40). Enzymatic
digestion is laborious, and efficiency varies widely between labs.
Satellite Cell Purification
After enzymatic digestion, the resulting isolate is typically purified to remove debris
and other abundant cell types, including epithelial cells and fibroblasts. Fluorescent
Activated Cell Sorting (FACS) (Figure 3A) (33) is a purification technique that uses
fluorophore conjugated cell markers to dye cells and applies electrical charges to samples
to sort cells by surface marker (41,42). Determining appropriate positive and negative
Page 8 of 57
8
satellite cell markers can be challenging due to satellite cell heterogeneity. Although
satellite cells have successfully been purified using FACS (43–46), applying electrical
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
charges to live cells can alter gene expression (47) and reduce proliferative capacity (48),
which could limit their SMTE applications.
Magnetic Activated Cell Sorting (MACS) (Figure 3B) (33) is an alternative to FACS
that works by incubating cells with antibodies for surface markers that are conjugated to
magnetic microbeads. A magnetic field is then applied to sort the cells out of the cell
suspension. However, appropriate markers must also be determined for MACS, and the
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
tight antibody‐antigen association can sometimes cause the magnetic microbeads to be
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
endocytosed by the sorted cells (40), rendering the beads impossible to remove prior to
implantation. Although MACS and FACS are powerful experimental techniques, given their
limitations, it is presently unclear whether they will ever be clinically useful purification
techniques for SMTE.
In contrast, the pre‐plate protocol (Figure 3C) (33) developed by Richler and Yaffe
Tissue Engineering
(49) is a simple, fast method to remove fibroblasts and epithelial cells by repeatedly
plating and recollecting the cell suspension obtained from enzymatic dissociation over
time. Lavasani et al. (50) later developed the modified pre‐plate technique, which coats
pre‐plates in collagen I to purify cells by their affinity for collagen I. Pre‐plating is attractive
for its simplicity, but it is variable and results in significant loss of satellite cells on pre‐
plates. Recently, Syverud et al. (51) published a novel, promising purification method that
uses a microfluidic sorting device termed a "Labyrinth" to obtain significantly purer
satellite cell populations using label‐free, inertial separation; the higher‐purity population
was then used to grow skeletal muscle constructs able to produce significantly greater
tetanic forces than unsorted controls.
Satellite Cell Preservation
To date, satellite cell preservation is not well studied, and the field has yet to
comprehensively address the best method for satellite cell cryopreservation or
cryopreservation's effects on satellite cell viability, proliferation, marker expression, and
differentiation potential. Bian et al. (52) found that satellite cells isolated from Luxi cattle
embryo could be successfully cryopreserved after fifteen passages and retained an average
Page 9 of 57
9
viability of 97.90 ± .96% after cryopreservation. Though it is perhaps likely that
cryopreservation would be an effective method for satellite cell preservation, more
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
research must be done to assess the most effective satellite cell preservation techniques
and any negative effects of the cryopreservation process.
Satellite Cell Proliferation and Differentiation
After isolation and purification, most SMTE protocols proliferate satellite cells by
suspending them in a muscle growth medium with high serum and high glucose content
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
(40). A selection of biomolecules are typically added to enhance proliferation and
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
myogenic potential in culture, including fibroblast growth factor (FGF), hepatocyte growth
factor (HGF), platelet‐derived growth factor (PDGF), and insulin‐like growth factor 1 (IGF‐1)
(39,53–55). Weist et al. (56) found that when added to muscle medium, transforming
growth factor beta (TGF‐β1) enhances contractility and extracellular matrix organization in
scaffoldless, tissue‐engineered skeletal muscle tissues. Syverud et al. (57) found that the
Tissue Engineering
glucocorticoid dexamethasone (DEX) is a useful addition to muscle media because it
enhances the myogenic potential of rat satellite cells in culture, thereby improving the
terminal differentiation process by increasing myotube fusion and thickness and enhancing
sarcomeric organization.
After being suspended in appropriate media, satellite cells are often seeded onto
tissue culture plastic that is coated with an adhesion protein. Laminin, fibronectin,
matrigel, and polylysine have all been successfully used in various protocols (53,55,58),
and there is no consensus as to which adhesion protein works best. Alternatively,
hydrogels, which are gelatinous mixtures of proteins and water, are sometimes used in lieu
of protein‐coated, stiff glass dishes. Pollot et al. (59) recently compiled a useful review of
natural polymeric hydrogels for SMTE. It has been hypothesized that hydrogels can
promote growth in a way that is more similar to a stem cell niche by mimicking the
stiffness of the extracellular matrix (ECM) niche in vivo. Indeed, Engler et al. (60) found
that increasing or decreasing hydrogel stiffness outside of physiologically‐normal range for
skeletal muscle significantly reduces myogenic differentiation in mouse myoblasts, which
Page 10 of 57
10
underscores the importance of engineering an in vitro environment that is as similar to the
stem cell niche as possible.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
After allowing the cells to proliferate into myoblasts, SMTE protocols induce
myogenic differentiation via a muscle differentiation medium (MDM). MDMs typically
contain low serum content, because myoblasts interpret serum deprivation in the local
environment as a differentiation signal (61). A wide variety of growth factors, including
IGF‐1, are also known to promote skeletal muscle differentiation and increase skeletal
muscle mass in vivo (62). Kang et al. (63) found that adding recombinant netrin, a secreted
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
axonal guidance protein that associates with the cell membrane and ECM (64,65), to
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
mouse myoblasts in vitro can induce myotube production and increase myotube size via
nuclear factor of activated T cell (NFAT) activation. Arachidonic acid enhances myotube
growth in vitro (66), as do cytokines via the Janus kinase/Signal Transducer and Activator
of Transcription (JAK/STAT) pathway (67).
The presence of ECM also encourages myogenic differentiation due to interactions
Tissue Engineering
between integrins in the ECM and myoblasts that induce signaling events to promote
myogenesis. A study by Stern et al. (68) found that adding ECM extracts from the thigh
muscles of adult rats to myoblasts enhances myoblast differentiation in vitro. Mechanical
stimulation can also be used to encourage myogenic differentiation. In vivo, mechanical
stimulation is required to prevent muscle atrophy, and this has been attributed to the
effects that mechanical stimulation has on the mammalian target of rapamycin (mTOR)
pathway to regulate gene and protein expression (69); it is thought that muscle behaves
similarly in vitro.
Because proper alignment of skeletal muscle fibers is crucial for function, tissue
engineers must also develop strategies to align skeletal muscle myotubes as they fuse in
vitro. Methods to align cells in vitro include topography methods (70,71), surface
patterning methods (72), mechanical stimulation (73–75), and electrical and magnetic
stimulation (76,77). Topography methods use surface features to manipulate cell behavior.
For instance, growing myoblasts on parallel, micropatterned grooves on tissue culture
plates promotes myoblast alignment (78) and myotube fusion in organized, end‐to‐end
configurations (79). Custom bioreactors (80), which contain lined, cylindrical microwells,
can be used to promote cell alignment and myoblast fusion using topographical principles.
Page 11 of 57
11
The same concepts are applied in surface patterning. Surface patterning is an umbrella
term for a variety of techniques that promote cell alignment, including soft lithography
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
(81). ECM proteins, including collagen, fibronectin, and laminin can be printed onto cell
surfaces via soft lithography to promote proper myotube alignment (61). As summarized in
several review articles(82,83), mechanical stimulation can also be used to promote
myotube alignment in vitro by using custom, dynamic bioreactors to add cyclic strain,
which aligns myotubes parallel to the direction of strain (84). As summarized in a useful
review by Ahadian et al. (85), electrical stimulation works by applying an electric field to
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
muscle cells in vitro; because these cells are electroactive, applying an electric field
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
regulates cell behavior and induces muscle tissue alignment parallel to the direction of an
applied electric field (86,87). Similarly, magnetic stimulation applies a magnetic field to
align myoblasts and elongates them as they differentiate along the axis of the applied
magnetic poles (88); however, it must be noted that magnetic stimulation is presently a
clinically infeasible process, as it requires that cells be electroporated with magnetic
Tissue Engineering
nanoparticles.
Satellite Cells in SMTE for VML
Several studies have tested satellite cell‐based SMTE in VML animal models (89,90).
In 2011, Machingal et al. (13) created and tested the first tissue‐engineered muscle repair
(TEMR) construct. The researchers created the TEMR by performing an enzymatic
dissociation isolation of rat muscle to obtain muscle progenitor cells (MPCs), which is a
more general category of Pax7+ cells that includes satellite cells. They cultured the MPCs to
obtain rat myoblasts, and seeded the myoblasts onto a porcine bladder acellular matrix
(BAM) scaffold to produce the TEMR. Afterwards, Machingal et al. used a custom
bioreactor for a week after seeding to provide mechanical stretch, thereby promoting
differentiation and alignment within the TEMR. Fluorescent microscopy and scanning
electron microscopy analysis of the TEMR after bioreactor preconditioning confirmed that
the scaffold contained aligned, multinucleated myotubes. After implanting final TEMRs
into a murine model for VML injury in the lattissimus dorsi (LD) muscle and allowing the
mice to recover for 2 months, they measured isometric tetanic force and observed
Page 12 of 57
12
significant functional recovery of the injured LD in mice treated with TEMRs compared to
no repair, although function was not returned to pre‐injury levels.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
This TEMR protocol was further refined by Corona et al. (91) by experimenting with
three different varieties of TEMR. The first type of TEMR, termed TEMR‐1SP, was seeded
with rat‐derived MPCs and allowed to proliferate for only a short period of time without
bioreactor preconditioning. The second TEMR variety, called TEMR‐1SPD, was given a
longer cellular proliferation and differentiation period and was conditioned with a
bioreactor; this is the TEMR variety that was used in the original Machingal et al. (13)
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
study. The third TEMR variety, named TEMR‐2SPD, was given similar treatment to TEMR‐
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
1SPD but additional MDCs were seeded onto the scaffold during bioreactor
preconditioning to simulate functional recovery during exercise, wherein MPCs are
typically recruited to injured fibers. After implanting these TEMRs into a murine model for
VML injury, they measured in vitro isometric tetanic and twitch force. Results showed that
TEMR‐1SPs and TEMR‐1SPDs conferred similar functional recovery, while TEMR‐2SPDs
Tissue Engineering
induced functional recovery that had twice the magnitude of TEMR‐1SPs and TEMR‐1SPDs.
Corona et al. (14) later used the same protocol used to generate TEMR‐1SPDs in a
Lewis rat model of VML. When VML rats were treated with TEMRs, results were binary; the
rats were either responsive or nonresponsive to the tissue repair. Nonresponsive rats with
TEMR implants had no significant improvements in structural or functional recovery
compared to untreated rats, while responsive rats had significant structural and functional
recovery compared to untreated rats, as assessed by histology and in vivo torque
measurements. The researchers noted that responsive and nonresponsive rats had
different immune responses in their injury site that could have contributed to the
observed variability.
VanDusen et al. (16) published a method for engineering scaffoldless, three‐
dimensional, functional skeletal muscle units (SMUs) and tested them in a rat VML model.
The researchers used enzymatic digestion of rat soleus muscles to isolate satellite cells,
cultivated them using muscle growth medium, and differentiated them in vitro using a
medium with low serum content. After the muscle monolayers rolled up into a 3‐D skeletal
muscle tissue due to passive tension, they were pinned to the plate and engineered
bone/tendon anchors were added to either end, generating the final SMU. In this specific
Page 13 of 57
13
study, the researchers measured isometric tetanic force of their SMUs to ensure
functionality. The SMUs were then implanted into a VML site in the rat's TA muscle. After
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
28 days of recovery, they observed a significant increase in isometric tetanic force for VML
muscles treated with SMUs compared to rats with VML receiving no treatment, and a
force per unit mass for SMU‐treated muscles that was equivalent to native muscle.
Satellite Cell Limitations
Satellite cell‐based SMTE is undoubtedly a promising field with demonstrated
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
potential as a VML treatment, but using satellite cells has significant limitations. A skeletal
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
muscle biopsy is painful and invasive, and only a small biopsy would be clinically feasible.
Enzymatic dissociation is the only isolation method that is able to achieve sufficient
satellite cell yields, yet it is a laborious process and produces impure populations of
satellite cells. With respect to purification, FACS, MACS and pre‐plating all have significant
practical limitations, while microfluidic inertial separation is a promising, yet nascent
Tissue Engineering
method. Satellite cell preservation is not well studied, and satellite cell self‐renewal is not
unlimited, and satellite cell differentiation potential declines rapidly in vivo. To address
these issues, it is important to consider alternative stem cell sources, including adipose‐
derived stem cells, bone marrow‐derived mesenchymal stem cells, umbilical cord‐derived
mesenchymal stem cells, perivascular stem cells, induced pluripotent stem cells, and
embryonic stem cells.
Adipose‐Derived Stem Cells (ADSCs)
Adipose‐derived stem cells (ADSCs) are an abundant, adult, multipotent,
mesenchymal stem cell (MSC) variety with myogenic potential. MSCs are a general
category of non‐hematopoietic stem cells with specific cluster of differentiation (CD)
protein surface markers that reside in many tissue types, including adipose tissue. MSCs
have the capacity to differentiate into tissues of mesodermal lineage, including cartilage,
bone, adipose tissue, and skeletal muscle (92–94). A useful ADSC tissue engineering review
by Zuk (95) contains a comprehensive discussion of human ADSC surface markers.
ADSCs are attractive to tissue engineers not just for their abundance, but because
cultured ADSCs express very low levels of major histocompatibility complex class I (MHC I)
Page 14 of 57
14
and do not express major histocompatibility complex class II (MHC II) (96). Thus, ADSCs
from allogeneic or xenogeneic donors might be able to evade the patient's immune system
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
after implantation, potentially eliminating the need for lifelong immunosuppression in
patients (96). It is important to note that the low levels of MHC I and MHC II observed in
ADSCs are present before differentiation; there has not yet been a study that investigates
MHC I and MHC II levels in ADSC‐derived cells after myogenic differentiation.
ADSC Isolation and Purification
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
ADSC isolation protocols begin with an adipose tissue biopsy (97). As noted in the
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Forcales (98) review of ADSCs for muscle regenerative therapies, adipose tissue is a highly‐
abundant, cosmetically favorable tissue type to biopsy in most individuals, and a simple
liposuction procedure to collect lipoaspirate typically suffices. Moreover, ADSC isolations
are generally efficient, with an average of 404,000 ± 206,000 cells isolated per milliliter of
lipoaspirate (99). Importantly, ADSC yield does not depend on the donor site (100), so fat
Tissue Engineering
can be taken from the most convenient location possible. Thus, ADSCs offer significant
advantages over satellite cells with respect to biopsy.
After biopsy, ADSC isolations typically use enzymatic digestion of the biopsied
adipose tissue or lipoaspirate with collagenase type 1A (98). After digestion, the mixture is
centrifuged to obtain the stromal vascular fraction (SVF) pellet, which contains many cell
types, including preadipocytes, pericytes, fibroblasts, red blood cells, and smooth muscle
cells (98). The SVF is plated in an ADSC media, which can vary by protocol but typically
contains Dulbecco’s Modified Eagle Medium (DMEM) with 10% fetal bovine serum (FBS).
The cells that adhere to the plate are considered to be ADSCs (98). FACS and MACS can be
used to purify ADSC populations, but since ADSCs lack a universal marker, these processes
are complicated and imperfect. Cryopreservation is a suitable method for ADSC
preservation, as summarized in a review by Miyagi‐Shiohira et al. (101).
Several specific ADSC isolation methods may offer advantages over satellite cell
enzymatic dissociation. Like satellite cells, explant culture can be used for ADSC isolations,
and Priya et al. (102) developed an explant culture method that achieved equivalent to
superior human ADSC yields when compared to enzymatic dissociation. Because explant
culture tends to result in purer cell populations than enzymatic dissociation (40), using
Page 15 of 57
15
explant culture could eliminate the need for further purification. There is also a novel
protocol developed by Francis et al. (103) to isolate human ADSCs in just 30 minutes.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Future SMTE studies could consider using one of these protocols to obtain a starting ADSC
population.
ADSC Induction
There are several different methods for inducing ADSCs into myoblasts, including
using an appropriate myogenic media with myogenic biochemical factors, co‐culturing
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
ADSCs with myoblasts, adding biophysical stimuli, and genetically manipulating the ADSCs.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Interestingly, ADSC myogenesis is similar to satellite cell myogenesis in terms of when
Pax7, Myf5, and MyoD are expressed (104).
Di Rocco et al. (105) observed that a small proportion of mouse ADSCs are able to
sporadically convert into a myogenic lineage, which suggests that certain ADSCs do have
inherent myogenic potential. They also experimented with 50‐50 co‐cultures of myoblasts
Tissue Engineering
and ADSCs, and found that ADSCs will fuse into myotubes when cultured with myoblasts,
but only when the ADSCs and myoblasts are in direct contact. Afterwards, they tested
ADSC regenerative capacity in vivo, and found that when uncultured ADSCs were injected
into a mouse with ischemic skeletal muscle, they were able to fuse with muscle fibers to
contribute to structural recovery in vivo. Thus, simply placing ADSCs into the site of injury
can induce some degree of induction and differentiation towards a myogenic lineage,
though it is unknown whether this would contribute to meaningful functional recovery.
Biophysical stimuli can also induce ADSCs to a myogenic lineage, and Huri et al.
(106) found that adding cyclic strain to human ADSCs (hADSCs) treated with muscle
induction media not only encourages proper cell alignment, but also enhances myogenic
differentiation into myotubes. In fact, after adding cyclic strain, the researchers observed a
4.5‐fold increase in nuclei per cell, which suggests enhanced myogenesis. They also
observed significantly higher myogenic protein expression compared to cultures not given
cyclic strain.
Altering gene expression has also been explored as a method to differentiate
ADSCs, and Goudenege et al. (107) developed an ADSC differentiation protocol that uses a
mouse MyoD lentiviral vector to forcibly induce MyoD expression, thereby causing
Page 16 of 57
16
myogenic induction. MyoD is an essential myogenic regulatory factor that induces an open
chromatin structure at myogenic genes (108). The researchers found that using their
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
protocol to induce MyoD expression in hADSCs was a fairly efficient way to achieve
myogenic induction, with 30% of treated hADSCs expressing myogenic markers, as
compared to just 2% of untreated hADSCs. Notably, Goudenege et al. also found that
when injected into TA cryoinjuries in a murine model, the transfected hADSCs were
capable of fusing with regenerating fibers in the injury site to contribute to structural and
functional recovery in vivo. Sung et al. (109) tested a safer method for induction by
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
engineering and applying MyoD protein to hADSC cultures to induce myogenesis in vitro.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
From a clinical perspective, applying MyoD protein is a far more practical and safe method
than altering MyoD gene expression, and would be a more feasible method to try in SMTE.
ADSCs in SMTE for VML
In 2016, Kesireddy (110) published the first comparative study that used rat ADSCs
Tissue Engineering
to create a TEMR construct to treat VML injury in a murine model and compared it to MPC
TEMRs. To make a TEMR, he isolated ADSCs and MPCs from donor rats, cultivated them,
and seeded either ADSCs or MPCs into BAM scaffolds. Afterwards, he implanted them into
a murine VML injury model in the LD muscle. Histological analysis of the injury site 2
months post‐repair revealed that ADSC and MPC TEMRs displayed similar regeneration
potential, and both showed signs of neotissue formation, which is characterized by
scaffold remodeling, fiber formation, and cell fusion in the VML site (110).
Kesireddy also did qualitative observation of lentivirus‐Cherry‐labeled donor cells
to determine the origin of the structural recovery that was observed, and discovered that
ADSCs contributed fairly little to new myofiber formation compared to donor MPCs.
However, ADSCs contributed greatly to vascularization in the injury site, and histological
images revealed greater vascularization after being treated with ADSCs than MPCs (110),
which is a positive sign of healing. Unfortunately, this experiment did not conduct force
testing to assess functional recovery, but the results nevertheless suggest that ADSCs
could be useful for treating VML.
Page 17 of 57
17
Bone Marrow‐Derived Mesenchymal Stem Cells (BM‐MSCs)
Bone marrow‐derived mesenchymal stem cells (BM‐MSCs) are another notable
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
MSC variety with myogenic potential. BM‐MSCs are a multipotent, adult stem cell
population that resides in the bone marrow (BM) on the abluminal side of vessels and can
differentiate into mesodermal tissues like skeletal muscle, bone, and cartilage (111). It is
important to note that BM‐MSCs are distinct from BM hematopoietic stem cells and do
not directly participate in hematopoiesis; rather, BM‐MSCs play an immunomodulatory
role, and provide microenvironment support for hematopoietic stem cells in vivo (112).
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
BM‐MSCs are a heterogeneous stem cell population, and individual subsets have
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
significant differences in markers, as detailed in a review by Boxall et al. (113). Because of
the similarities between BM‐MSCs and ADSCs, which are both MSCs with myogenic
potential that are found in vessel walls in their respective tissues of origin, Bayati et al.
(114) compared rat BM‐MSCs and ADSCs, and discovered that the two cell populations had
differential surface marker expression but did not significantly differ in terms of
Tissue Engineering
proliferative capacity or myogenic potential. Also like ADSCs, BM‐MSCs have poor ability to
stimulate an in vitro allogeneic T cell response due to absent MHC II and low MHC I
expression (115), which makes them well‐suited to transplantation.
BM‐MSC Isolation and Purification
BM‐MSC isolation begins with a BM biopsy, which is a painful and invasive process;
however, it is still advantaged over satellite cell biopsies in that more tissue can be taken
without introducing a functional deficit. After a biopsy, typical BM‐MSC isolations use
enzymatic digestion, just like ADSC and satellite cell protocols (116). BM isolates are
filtered and washed, and enzymes are used to digest the BM isolate to retrieve the BM‐
MSCs. Similarly to ADSCs, explant culture has also more recently been proposed as an
efficient method for BM‐MSC isolation (117). Generally speaking, BM‐MSC isolation from
BM samples is a fairly low‐yield process, because there are less than .01% BM‐MSCs
present in total BM cells (92). FACS and MACS can be used to purify BM‐MSC populations if
that is deemed necessary, and cryopreservation methods for BM‐MSCs have been
developed (118,119).
Page 18 of 57
18
BM‐MSC Induction
BM‐MSC induction methods are similar to what was already discussed for ADSCs
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
and satellite cells; most involve muscle induction media, co‐culture, biophysical stimuli, or
some combination of these methods. Treatment with 5‐azacytidine is a common method
for induction of BM‐MSCs to a myogenic lineage in vitro, though this is not unique to BM‐
MSCs, and can also be used to for induction of ADSCs (120). Although using 5‐azacytidine
for myogenic induction is a useful research technique, it is known that 5‐azacytidine is a
carcinogen (121), and is therefore impractical for SMTE. A study by Stern‐Straeter et al.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
(122) evaluated the efficacy of six different muscle induction media formulations for
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
hADSCs and human BM‐MSCs (hBM‐MSCs) to offer guidance on which induction medias
work best. Beier et al. (123) perfected a rat BM‐MSC/myoblast co‐culture method by using
basic fibroblast growth factor (bFGF) and dexamethasone to maximize myogenic potential.
Egusa et al. (124) were the first to discover that adding cyclic strain to mouse BM‐MSCs
accelerates myogenic induction and promotes proper fiber alignment during the terminal
Tissue Engineering
differentiation process.
An important study by Meligy et al. (125) compared proliferation and myogenic
differentiation capacity between ADSCs, BM‐MSCs, and skeletal muscle tissue
mesenchymal stem cells (MC‐MSCs), which are found in skeletal muscle but have a MSC
phenotype and are distinct from satellite cells. After isolating and cultivating rat MSCs
from their respective tissues of origin, they treated the cells with 5‐azacytidine to promote
induction to a myogenic lineage. ADSCs had the highest observed proliferation rate and
BM‐MSCs had the lowest proliferation rate (125), though all were fairly high. The
researchers measured differentiation potential by myogenin expression, and observed
that BM‐MSCs had higher myogenic differentiation capacity than ADSCs, though both were
inferior to MC‐MSCs (125). From this, Meligy et al. concluded that ADSCs were the most
easily accessible and highly proliferative MSC. Although MC‐MSCs had the highest
observed myogenic differentiation potential, the researchers noted that MC‐MSCs isolated
from a skeletal muscle biopsy are no more accessible than satellite cells, suggesting that
ADSCs and BM‐MSCs could still be a viable starting source for SMTE regardless of their
lower myogenic differentiation potential.
Page 19 of 57
19
BM‐MSCs in SMTE
There has never been a published study that used BM‐MSCs to engineer skeletal
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
muscle tissue specifically for VML. However, there are several studies that have used BM‐
MSC injections to treat skeletal muscle injuries, and they offer clues as to how BM‐MSCs
would behave in SMTE specifically for VML. Natsu et al. (126) isolated BM‐MSCs from
green fluorescent protein (GFP) rats and injected them into lacerated TA muscles of
Sprague‐Dawley (SD) rats after implanting a fibrin scaffold. One month after
transplantation, the rats treated with BM‐MSCs showed significant functional recovery
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
that was almost that of pre‐injury levels. Interestingly, histological analysis did not indicate
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
that the GFP BM‐MSCs actually differentiated or fused into myofibers in the SD rats, which
suggests that BM‐MSCs may play an indirect role in healing, possibly by promoting a
healing immune environment in the injury site.
Merritt et al. (127) repaired skeletal muscle injury in a rat model by implanting an
extracellular matrix (ECM) scaffold into injured lateral gastrocnemius muscles and injecting
Tissue Engineering
BM‐MSCs into a group of rats treated with the ECM scaffold a week after surgery. After 42
days, the injured muscles treated with ECM and BM‐MSCs displayed significant structural
recovery (127) and partial functional recovery that was superior to muscles treated with
ECM alone (127). BM‐MSC‐injected muscles also had more blood vessels and regenerating
fibers. From this, the researchers concluded that BM‐MSCs could be a viable therapy for
skeletal muscle injury in the future. Andrade et al. (128) found that BM‐MSCs administered
via intramuscular injection into a rat model of successive injury significantly increased
maximal skeletal muscle contraction, muscle fiber cross‐sectional area, and the number of
muscle fibers in the injury site 28 days after injection.
Helal et al. (129) found that BM‐MSCs modulate the local immune system
environment in the site of VML to encourage recovery. Specifically, administration of BM‐
MSCs via intramuscular injection into a VML injury in a rat was shown to improve
structural and functional recovery by suppressing inflammatory cytokines, promoting anti‐
inflammatory cytokines, and also by directly contributing to myogenesis and angiogenesis.
BM‐MSCs also downregulate transforming growth factor beta (TGF‐β) in the injury site,
which prevents fibrosis. As this study demonstrates, BM‐MSCs show promise for SMTE
Page 20 of 57
20
applications not just because of their myogenic potential, but also because of the role they
play in immunomodulation to promote proper healing.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Umbilical Cord‐Derived Mesenchymal Stem Cells (UC‐MSCs)
Umbilical cord‐derived mesenchymal stem cells (UC‐MSCs) are derived from
compartments of the umbilical cord, with UC‐MSCs isolated from the cord‐placenta
junction region showing particularly high differentiation potential (130). UC‐MSCs are
more similar to embryonic stem cells than adult stem cells, and they are capable of faster
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
self‐renewal than adult MSCs (131). Collecting UC‐MSCs is also a painless process, and UC‐
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
MSCs lack MHC II, which is useful for transplantation (132). UC‐MSCs can be isolated via
enzymatic digestion (133) or explant culture methods (130,134). A good manufacturing
practice (GMP) protocol for clinical‐grade human UC‐MSC isolation was recently published
(135), and a useful summary of UC‐MSC markers, characteristics, and isolation methods is
included in a review by Nagamura et al. (136).
Tissue Engineering
Several myogenic induction protocols exist for UC‐MSCs. Gang et al. (137)
demonstrated that UC‐MSCs are capable of being induced into a myogenic lineage by
cultivating them in a myogenic media that contained 5% horse serum, 0.1 μM
dexamethasone, and 50 μM hydrocortisone for six weeks. Kocaefe et al. (132) transfected
human UC‐MSCs with MyoD to force induction into a myoblastic lineage. They found that
the resulting cells were capable of undergoing terminal differentiation by fusing with rat
myoblasts to form myotubes, which suggests that UC‐MSCs could be a viable starting
source for SMTE in the future.
To date, UC‐MSCs are best studied in the context of muscular dystrophy therapies.
A study by Zucconi et al. (138) found that undifferentiated UC‐MSCs injected into mouse
and dog muscular dystrophy models did not differentiate into skeletal muscle in vivo, yet
dystrophic animals injected with them displayed significant functional recovery compared
to untreated controls. In contrast, a different study by Nunes et al. (139) was able to
observe some degree of structural recovery, with a small number of mouse UC‐MSCs able
to differentiate into dystrophin‐expressing myotubes in vivo when transplanted into mdx
mice. While the exact reason for these conflicting results is not clear, it was shown in a
study by Koponen et al. (140) that UC‐MSCs are able to promote skeletal muscle
Page 21 of 57
21
regeneration in lacerated mouse skeletal muscle not by directly fusing with myofibers, but
by promoting a healing cytokine environment. While it appears that UC‐MSCs could be a
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
useful source for clinical skeletal muscle treatments in the future, more research must be
done to understand the mechanism and extent of healing in traumatic injury applications.
Perivascular Stem Cells (PVSCs)
Perivascular stem cells (PVSCs) are yet another MSC population that can be
subdivided into pericytes and adventitial cells (141). Adventitial cells are found in the
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
tunica adventitia (Figure 4, right) (142), which is the outermost layer of arteries and veins
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
(143), and they are defined as positive for CD34 and negative for CD31, CD146, and CD45
(144). Although adventitial cells share developmental features and general MSC properties
with pericytes, they are a phenotypically and genotypically distinct population (143). Their
myogenic potential has not yet been studied, whereas pericytes have been studied in
greater detail, discussed below.
Tissue Engineering
Pericytes
Pericytes are a stem cell population found in capillary and microvessel walls (142)
underneath the basal lamina (Figure 4, left) (142) in a variety of tissues, including skeletal
muscle (145) and adipose tissue (141). Pericytes isolated from skeletal muscle, white
adipose tissue, pancreas, and placenta have myogenic potential (146), and adipose tissue
in particular is an attractive biopsy site to target. Dellavale et al. (147) discovered that
mouse pericytes actually participate in normal postnatal skeletal muscle development by
differentiating into muscle fibers and infiltrating the satellite cell niche to generate satellite
cells. Thus, pericyte myogenesis is a natural process that occurs during development and
increases in response to acute injury. Pericytes are defined as positive for CD146 and
negative for CD34 and CD45 (141). Most pericyte isolations involve enzymatic digestion,
though explant culture has also been used (148), and Bryan et al. (149) wrote a useful
review of methods to isolate and culture pericytes. After isolation, pericytes can be
purified using FACS or MACS.
An important study by Dellavalle et al. (148) first discovered the promise of
pericytes for skeletal muscle therapies. Dellavale et al. began by isolating pericytes from
Page 22 of 57
22
human skeletal muscle biopsies with and without Duchenne Muscular Dystrophy (DMD),
and then expanded them out to 20 population doublings, which allowed them to obtain at
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
least 1 x 109 cells from a single biopsy, an amount they considered sufficient for clinical
DMD therapy. After culturing the pericytes, they promoted induction to a myogenic
lineage and differentiation using both a myogenic differentiation medium and co‐culture
with myoblasts, and observed that pericytes formed differentiated myotubes
spontaneously with high efficiency using both differentiation methods (148). In fact, the
differentiation potential they observed for pericytes was strikingly high, and up to an order
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
of magnitude higher than most other myogenic stem cells they compared them to,
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
excluding satellite cells. Dellavale et al. also injected cultured human pericytes into a
severe combined immune deficient–X‐linked, mouse muscular dystrophy (scid–mdx)
mouse model, and found that pericytes could successfully engraft with dystrophic muscle
fibers to contribute to structural recovery in vivo. The qualities of pericytes Dellavale et al.
observed ‐ adequate proliferative capacity, high myogenic potential, ability to contribute
Tissue Engineering
to fiber regeneration in vivo ‐ coupled with the fact that pericytes can be isolated from
other, more convenient tissue types to biopsy, suggests that pericytes could be a highly
useful starting cell for SMTE. Moreover, pericytes do not express MHC II or essential T‐cell
activating costimulatory molecules like CD80 and CD86, which suggests that pericytes are a
promising candidate for allotransplantation (150).
To date, pericytes have never been used in a SMTE study specifically for VML;
however, a study by Fuoco et al. (151) offers guidance to tissue engineers looking to
develop a pericyte SMTE protocol. Fuoco et al. isolated skeletal muscle‐derived pericytes
(MPs) from young and old pigs using enzymatic digestion with collagenase II and induced
myogenesis via a media with low serum content. Myogenic potential of pericytes declines
with age, and the researchers found that old MPs had reduced ability to form myotube
and capillary‐like structures in vitro on their own. However, when the old MPs were
cultured on mixtures of polyethylene glycol, fibrinogen, and water called PF hydrogels,
they displayed enhanced myogenic potential in vitro and in vivo (152). PF hydrogels are
believed to rejuvenate myogenic potential because they better mimic the stem cell niche
in vivo, and they could be a useful scaffold material to try in a pericyte‐based SMTE
protocol in the future.
Page 23 of 57
23
Induced Pluripotent Stem Cells (iPSCs)
Induced pluripotent stem cells (iPSCs) are produced in the lab by culturing adult
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
cells and forcing them to upregulate transcription factors that induce pluripotency.
Because they are pluripotent, they are capable of unlimited self‐renewal in culture, which
is a major advantage over satellite cells. Nishikawa et al. (153) wrote a useful review of the
promises and present limitations of iPSCs.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Making iPSCs
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Technically, any tissue type could be used to make iPSCs; however, in practice,
making iPSCs for use in skeletal muscle therapies (Figure 5) (154) typically begins with a
simple skin biopsy to isolate epithelial cells and fibroblasts (154). These cell types are
chosen for their abundance and convenience; thus, iPSC biopsy offers a clear advantage
over satellite cell biopsy. After biopsy, pluripotency must be induced in the cells. Takahashi
Tissue Engineering
et al.'s (155) pioneering paper determined four essential transcription factors, termed
Yamanaka factors, that must be transduced into starting cells using a viral vector to
reprogram the cells into iPSCs: KLF4, c‐MYC, OCT4, and SOX2. Although fibroblasts and
epithelial cells are abundant, iPSC generation is not efficient, with the percentage of adult
cells successfully reprogramming to pluripotency often somewhere in the range of 0.1‐3%
(155,156). This is partially attributable to the fact that cells retain epigenetic markers from
their tissue of origin that make them resistant to reprogramming (157,158).
In the years since Yamanaka's pioneering study, other scientists have worked to
develop more efficient iPSC protocols. Warren et al. (159) optimized the original iPSC
method using synthetic modified mRNA to transfect cells, causing them to be
reprogrammed to pluripotency at a significantly higher efficiency, while Yulin et al. (160)
and Kunisato et al. (161) have developed more efficient iPSC generation protocols that use
BM‐MSCs as a starting source; given that BM‐MSCs have inherent myogenic differentiation
potential, it is presently unclear whether these would have any relevance to SMTE. Malik
(162) compiled a more comprehensive review of human iPSC generation methods.
Regardless, the development of more efficient iPSC protocols suggests that iPSCs could
Page 24 of 57
24
eventually be a more practical therapeutic option in SMTE applications. After iPSC
generation, cryopreservation is an efficient method for iPSC storage (163).
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
iPSC Induction
iPSCs must be properly induced into a myogenic lineage prior to implantation. A
variety of different protocols have been developed to induce myogenesis in iPSCs, and
most of these methods involve inducing myogenic gene expression. Shoji et al. (164)
developed a reliable myogenic induction protocol for iPSCs that works by delivering a
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
tetracycline‐inducible MyoD piggyBac (PB) vector to human iPSCs, achieving robust
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
differentiation efficiency that is as high as 70‐90% in vitro. Chal et al. (165) developed a
serum‐free method for inducing iPSCs into muscle fibers and satellite‐like cells by
mimicking embryonic signaling events to induce myogenesis in vitro. Maffioletti et al. (166)
demonstrated a complex, thorough method for myogenic induction and differentiation of
human embryonic stem cells (hESC) and iPSCs that used a four‐stage system with different
Tissue Engineering
culture densities, media combinations, and protein coatings on the cell culture dish. First,
they drove ES and iPSCs to a lineage that was similar to mesoangioblasts, and then they
induced MyoD1 expression via a lentivirus vector with MyoD1 cDNA fused to an estrogen
receptor, thereby driving the cells to a myogenic lineage. Darabi et al. (167) developed a
protocol that makes iPSCs from fibroblasts derived from inducible Pax7 mice and then
induces Pax7 expression to induce the iPSCs into myogenic progenitors.
It is important to note that iPSCs are ultimately only useful in SMTE if they are
clinically safe. Reprogramming iPSCs to pluripotency and then to a myogenic lineage can
induce genetic instability and abnormalities that cause cancers, and iPSCs are known to be
tumorigenic (168,169). More emphasis should be placed on developing iPSC myogenic
induction and differentiation methods that do not require genetic modification, and
researchers have already begun this work. Hosoyama et al. (170) derived myogenic
progenitors from human iPSCs by using a medium with fibroblast growth factor‐2 (FGF‐2)
and epidermal growth factor to form free‐floating spherical cultures that contained
myogenic progenitors after 6 weeks. After culturing these progenitors in a muscle
differentiation medium for 2 weeks, the researchers observed multinucleated myotubes.
Xu et al. (171) identified three specific myogenic factors: a glycogen synthase kinase 3β
Page 25 of 57
25
(GSK3β) inhibitor called 6‐bromo‐indirubin‐3'‐oxime (BIO), FGF, and forskolin. By treating
human iPSCs with a combination of these three factors, they were able to produce
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
myogenic progenitors that could engraft into mdx mouse dystrophic muscle fibers in vivo.
Any of these methods could be tried on iPSCs in a SMTE study.
iPSCs in Skeletal Muscle Therapies
To date, iPSCs have never been used in a SMTE study to treat VML, but iPSC
injection therapy for DMD is better studied, and offers clues as to how iPSCs might behave
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
in SMTE. Darabi et al. (172) found that human iPSC‐derived myogenic progenitors restore
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
dystrophin and improve contractility upon transplantation in dystrophic mice, and Xu et al.
(171) found that human iPSC‐derived myogenic precursors can engraft with mdx mouse
muscle fibers in vivo. Quattrocelli et al. (173) found that iPSCs that have been induced to a
mesodermal lineage are able to contribute to functional recovery of cardiac and skeletal
muscle in a mdx mouse and dystrophic canines. Goudenege et al. (174) found that
Tissue Engineering
myoblasts derived from human iPSCs are able to efficiently fuse with existing muscle fibers
following transplantation. These positive results suggest that iPSCs could integrate with
existing tissue in an injury site in SMTE for VML.
Embryonic Stem Cells (ESCs)
Embryonic stem cells (ESCs) are pluripotent stem cells isolated from the inner cell
mass of a blastocyst. Beyond the ethical challenges that using ESCs poses (175), ESCs are
difficult and inefficient to isolate, and the isolation process requires high specialization and
training (176). ESCs are also tumorigenic (177). Partially because of these factors, ESCs
have not been used in muscle therapy studies to any meaningful degree.
Discussion
Although satellite cells remain the best‐studied cell type in SMTE for VML, they can
be difficult to isolate, purify, and biopsy. Alternative stem cells have characteristics that
may provide solutions to these problems, although much is presently unknown about how
alternative stem cell varieties will perform in SMTE studies.
Page 26 of 57
26
Judging on the biopsy alone (Table 1), ADSCs and pericytes are highly attractive
starting sources that offer advantages over satellite cells, because both can be isolated
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
from adipose tissue. Adipose tissue is the most promising location to target for pericyte
isolation not just for convenience, but also because pericytes make up 15% of the SVF that
is obtained during normal ADSC isolation (178), but constitute less than .5% of cells
isolated from bone marrow. Adventitial cells make up 20% of the SVF (144), but their
myogenic potential is currently unknown, and should be explored in future studies. BM‐
MSCs are superior to satellite cells in that a larger biopsy can be taken without introducing
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
a functional deficit. Yet, a BM biopsy is inferior to an adipose tissue biopsy, and BM‐MSCs
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
make up less than .01% of total BM cells biopsied (92). UC‐MSCs can be easily obtained
after birth, but unless allotransplanted UC‐MSCs become viable, UC‐MSCs will not be an
option for many current VML patients. iPSCs can be obtained from a convenient skin
biopsy, but iPSC reprogramming is not highly efficient.
With respect to isolation (Table 1), ADSCs and BM‐MSCs may offer advantages over
Tissue Engineering
satellite cells in SMTE. ADSC isolation in particular is highly efficient (98), and in recent
years, explant culture has been developed as an efficient method for ADSC and BM‐MSC
isolation that results in purer cell populations than enzymatic dissociation. Explant culture
may also decrease the cost, processing time, and labor required for cell isolation (179). The
Francis et al. (103) protocol to isolate ADSCs in 30 minutes is a unique and promising
method that also does not use enzymatic dissociation, and it is far faster and simpler than
satellite cell isolation. Pericytes can also use explant culture; although yields are fairly low,
pericytes can be expanded for at least 20 population doublings in culture (148). Notably, a
clinically safe GMP protocol for UC‐MSC isolation has also been developed (135).
No alternative stem cell is inherently easier to purify than satellite cells, and FACS
and MACS remain the most common purification techniques (Table 2) across cell types.
Alternative stem cell varieties have significant marker heterogeneity, so FACS and MACS
can be particularly challenging, in addition to the inherent limitations of these methods.
However, it must also be noted that purification is more commonly required when
enzymatic dissociation is performed, because the resulting isolate is less pure. If explant
culture methods to isolate ADSCs, BM‐MSCs, or pericytes could be used effectively in
SMTE, it is perhaps less likely that purification would be required, which is an important
Page 27 of 57
27
potential advantage that these cell types offer. Notably, a microfluidic inertial sorting
method was recently developed to obtain purer satellite cell populations (51); this method
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
is advantaged in that it is label‐free, but it has yet to be widely experimented with.
In terms of proliferative capacity and myogenic differentiation potential (Table 2),
it is difficult to say which alternative stem cell is best, though pericytes have particular
promise. As Dellavale et al. (148) noted, pericytes have higher innate myogenic potential
than most other stem cells, and their proliferative capacity is sufficient for clinical therapy.
Pierantozzi et al. (180) found that pericytes have similar proliferative capacity to ADSCs,
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
and are able to differentiate towards myocytes more efficiently than ADSCs. Pericytes'
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
proliferative capacity and myogenic potential compared to UC‐MSCs and BM‐MSCs should
be determined in future studies. UC‐MSCs also have great promise. It is known that UC‐
MSCs have higher proliferative capacity than adult MSCs like ADSCs and BM‐MSCs (181),
but the myogenic differentiation ability of UC‐MSCs compared to ADSCs and BM‐MSCs is
not well understood. Between BM‐MSCs and ADSCs, it is difficult to determine which cell
Tissue Engineering
type is superior. Meligy et al. (125) found that ADSCs were more highly proliferative than
BM‐MSCs, but BM‐MSCs had a higher myogenin expression than ADSCs, which correlates
to myogenic differentiation potential. Conflicting with this finding, Bayati et al. (114)
observed that ADSCs and BM‐MSCs had similar differentiation potentials, though, as Bayati
et al. noted, their protocol differed from Meligy et al.'s in terms of the rat species and
differentiation method used. At this point, pericytes, ADSCs, UC‐MSCs, and BM‐MSCs all
appear worthy of future exploration in SMTE VML studies, and many of them have already
been tested to some extent in muscle therapy studies (Table 3).
iPSCs and ESCs are presently less promising than other cell types. iPSCs can be
generated from an abundant tissue like skin, can be patient‐matched, have unlimited self‐
renewal, and can have high myogenic differentiation capacity in culture. Yet, inducing
pluripotency is not efficient, iPSCs are tumorigenic, and most differentiation methods rely
on gene overexpression, which is not clinically feasible. Moreover, unlike pericytes, ADSCs,
and BM‐MSCs, iPSCs do not have immunological characteristics that are useful for
allotransplantation (96,150). Although iPSCs are useful in experimental settings, and offer
some potential advantages over satellite cells, they have more significant barriers to
overcome than other stem cells before they can be used in SMTE. ESCs have many of the
Page 28 of 57
28
same limitations as iPSCs, but they are also harder to obtain than iPSCs, carry enormous
ethical and legal burdens, and are not patient‐specific.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Although there is no perfect alternative to satellite cells, based on what is currently
known, ADSCs, BM‐MSCs, pericytes, and UC‐MSCs are particularly promising candidates
for SMTE studies. Each offers potential advantages over satellite cells with respect to
biopsy, isolation, and purification while also having high proliferative capacities and
significant myogenic differentiation potentials. Future studies should consider using these
cell types to develop new SMTE protocols and should compare them with current satellite
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
cell‐based engineering methods. In the process, it will be important to optimize isolation
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
and differentiation methods across cell types, to work towards clinical grade SMTE
protocols that do not use gene overexpression, exogenous DNA, or media that contain
animal products. Ensuring proper myogenic differentiation of alternative stem cells either
in vitro or in vivo will be particularly crucial, because, unlike satellite cells, they are capable
of differentiating into multiple tissue types. Despite the challenges ahead, alternative stem
Tissue Engineering
cells may be the impetus that advances SMTE towards highly functional and practical VML
treatments, to give VML patients the quality of life they deserve.
Page 29 of 57
29
References
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
1. Mannion, S.J. and Chaloner, E. Principles of war surgery. BMJ Br Med J 330, 1498,
2005.
2. O’Brien, P.J. and Cox, M.W. Stents in tents: endovascular therapy on the battlefields
of the global war on terror. J Surg Radiol 2, 50, 2011.
3. Owens, B.D., Kragh, J.F., Macaitis, J., Svoboda, S.J., and Wenke, J.C. Characterization
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
of extremity wounds in Operation Iraqi Freedom and Operation Enduring Freedom.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
J Orthop Trauma 21, 254, 2007.
4. Owens, B.D., Kragh, J.F., Wenke, J.C., Macaitis, J., Wade, C.E., and Holcomb, J.B.
Combat wounds in operation Iraqi Freedom and operation Enduring Freedom. J
Trauma 64, 295, 2008.
Tissue Engineering
5. Corona, B.T., Rivera, J.C., Owens, J.G., Wenke, J.C., and Rathbone, C.R. Volumetric
muscle loss leads to permanent disability following extremity trauma. J Rehabil Res
Dev 52, 785, 2015.
6. Mertens, J.P., Sugg, K.B., Lee, J.D., and Larkin, L.M. Engineering muscle constructs
for the creation of functional engineered musculoskeletal tissue. Regen Med 9, 89,
2014.
7. Siemionow, M., Papay, F., Alam, D., Bernard, S., Djohan, R., Gordon, C., Hendrickson,
M., Lohman, R., Eghtesad, B., Coffman, K., Kodish, E., Paradis, C., Avery, R., and
Fung, J. Near‐total human face transplantation for a severely disfigured patient in
the USA. Lancet 374, 203, 2009.
8. Dubernard, J.M., Lengelé, B., Morelon, E., Testelin, S., Badet, L., Moure, C., Beziat,
J.L., Dakpé, S., Kanitakis, J., D’Hauthuille, C., El Jaafari, A., Petruzzo, P., Lefrancois, N.,
Taha, F., Sirigu, A., Di Marco, G., Carmi, E., Bachmann, D., Cremades, S., Giraux, P.,
Burloux, G., Hequet, O., Parquet, N., Francès, C., Michallet, M., Martin, X., and
Devauchelle, B. Outcomes 18 months after the first human partial face
transplantation. N Engl J Med 357, 2451, 2007.
Page 30 of 57
30
9. Jones, J.W., Gruber, S.A., Barker, J.H., and Breidenbach, W.C. Successful Hand
Transplantation — One‐Year Follow‐up. N Engl J Med 343, 468, 2000.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
10. Piza‐Katzer, H., Ninkovic, M., Pechlaner, S., Gabl, M., Ninkovic, M., and Hussl, H.
Double hand transplantation: functional outcome after 18 months. J Hand Surg Br
27, 285, 2002.
11. Strome, M., Stein, J., Esclamado, R., Hicks, D., Lorenz, R.R., Braun, W., Yetman, R.,
Eliachar, I., and Mayes, J. Laryngeal transplantation and 40‐month follow‐up. N Engl
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
J Med 344, 1676, 2001.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
12. Levi, D.M., Tzakis, A.G., Kato, T., Madariaga, J., Mittal, N.K., Nery, J., Nishida, S., and
Ruiz, P. Transplantation of the abdominal wall. Lancet 361, 2173, 2003.
13. Machingal, M.A., Corona, B.T., Walters, T.J., Kesireddy, V., Koval, C.N., Dannahower,
A., Zhao, W., Yoo, J.J., and Christ, G.J. A tissue‐engineered muscle repair construct
Tissue Engineering
for functional restoration of an irrecoverable muscle injury in a murine model.
Tissue Eng Part A 17, 2291, 2011.
14. Corona, B.T., Ward, C.L., Baker, H.B., Walters, T.J., and Christ, G.J. Implantation of in
vitro tissue engineered muscle repair constructs and bladder acellular matrices
partially restore in vivo skeletal muscle function in a rat model of volumetric muscle
loss injury. Tissue Eng Part A 20, 705, 2014.
15. Huang, Y.C., Dennis, R.G., Larkin, L., and Baar, K. Rapid formation of functional
muscle in vitro using fibrin gels. J Appl Physiol 98, 706, 2005.
16. VanDusen, K.W., Syverud, B.C., Williams, M.L., Lee, J.D., and Larkin, L.M. Engineered
skeletal muscle units for repair of volumetric muscle loss in the tibialis anterior
muscle of a rat. Tissue Eng Part A 20, 2920, 2014.
17. Levenberg, S., Rouwkema, J., Macdonald, M., Garfein, E.S., Kohane, D.S., Darland,
D.C., Marini, R., van Blitterswijk, C.A., Mulligan, R.C., D’Amore, P.A., and Langer, R.
Engineering vascularized skeletal muscle tissue. Nat Biotechnol 23, 879, 2005.
18. Messina, A., Bortolotto, S.K., Cassell, O.C., Kelly, J., Abberton, K.M., and Morrison,
Page 31 of 57
31
W.A. Generation of a vascularized organoid using skeletal muscle as the inductive
source. FASEB J 19, 1570, 2005.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
19. Borschel, G.H., Dow, D.E., Dennis, R.G., and Brown, D.L. Tissue‐Engineered Axially
Vascularized Contractile Skeletal Muscle. Plast Reconstr Surg 117, 2235, 2006.
20. Coppi, P, Delo, D., Farrugia, L., Udompanyanan, K., Yoo, J.J., Nomi, M., Atala, A., and
Soker, S. Angiogenic Gene‐Modified Muscle Cells for Enhancement of Tissue
Formation. Tissue Eng 11, 1034, 2005.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
21. Zisch, A.H., Lutolf, M.P., and Hubbell, J.A. Biopolymeric delivery matrices for
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
angiogenic growth factors. Cardiovasc Pathol 12, 295, 2003.
22. Bian, W. and Bursac, N. Tissue engineering of functional skeletal muscle: challenges
and recent advances. IEEE Eng Med Biol Mag 27, 109, 2008.
23. Sicari, B.M., Rubin, J.P., Dearth, C.L., Wolf, M.T., Ambrosio, F., Boninger, M., Turner,
Tissue Engineering
N.J., Weber, D.J., Simpson, T.W., Wyse, A., Brown, E.H., Dziki, J.L., Fisher, L.E.,
Brown, S., and Badylak, S.F. An acellular biologic scaffold promotes skeletal muscle
formation in mice and humans with volumetric muscle loss. Sci Transl Med 6,
234ra58, 2014.
24. Turner, N.J. and Badylak, S.F. The Use of Biologic Scaffolds in the Treatment of
Chronic Nonhealing Wounds. Adv wound care 4, 490, 2015.
25. Garg, K., Ward, C.L., Rathbone, C.R., and Corona, B.T. Transplantation of devitalized
muscle scaffolds is insufficient for appreciable de novo muscle fiber regeneration
after volumetric muscle loss injury. Cell Tissue Res 358, 857, 2014.
26. Rosenblatt, J.D., Lunt, A.I., Parry, D.J., and Partridge, T.A. Culturing satellite cells
from living single muscle fiber explants. In Vitro Cell Dev Biol Anim 31, 773, 1995.
27. Renault, V., Thornell, L.E., Butler‐Browne, G., and Mouly, V. Human skeletal muscle
satellite cells: aging, oxidative stress and the mitotic clock. Exp Gerontol 37, 1229,
2002.
Page 32 of 57
32
28. Bessa, P.C., Casal, M., and Reis, R.L. Bone morphogenetic proteins in tissue
engineering: the road from laboratory to clinic, part II (BMP delivery). J Tissue Eng
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Regen Med 2, 81, 2008.
29. Mauro, A. Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol 9, 493,
1961.
30. Danoviz, M.E. and Yablonka‐Reuveni, Z. Skeletal Muscle Satellite Cells: Background
and Methods for Isolation and Analysis in a Primary Culture System. Methods Mol
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Biol 798, 21, 2012.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
31. Lepper, C., Partridge, T.A., and Fan, C.M. An absolute requirement for Pax7‐positive
satellite cells in acute injury‐induced skeletal muscle regeneration. Development
138, 3639, 2011.
32. Dhawan, J. and Rando, T.A. Stem cells in postnatal myogenesis: Molecular
Tissue Engineering
mechanisms of satellite cell quiescence, activation and replenishment. Trends Cell
Biol 15, 666, 2005.
33. Zammit, P.S., Partridge, T.A., and Yablonka‐Reuveni, Z. The Skeletal Muscle Satellite
Cell: The Stem Cell That Came in From the Cold. J Histochem Cytochem 54, 1177,
2006.
34. von Maltzahn, J., Jones, A.E., Parks, R.J., and Rudnicki, M.A. Pax7 is critical for the
normal function of satellite cells in adult skeletal muscle. Proc Natl Acad Sci U S A
110, 16474, 2013.
35. Yin, H., Price, F., and Rudnicki, M.A. Satellite cells and the muscle stem cell niche.
Physiol Rev 93, 23, 2013.
36. Collins, C.A., Olsen, I., Zammit, P.S., Heslop, L., Petrie, A., Partridge, T.A., and
Morgan, J.E. Stem cell function, self‐renewal, and behavioral heterogeneity of cells
from the adult muscle satellite cell niche. Cell 122, 289, 2005.
37. Ono, Y., Boldrin, L., Knopp, P., Morgan, J.E., and Zammit, P.S. Muscle satellite cells
are a functionally heterogeneous population in both somite‐derived and
Page 33 of 57
33
branchiomeric muscles. Dev Biol 337, 29, 2010.
38. Schultz, E. Satellite Cell Proliferative Compartments in Growing Skeletal Muscles.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Dev Biol 175, 84, 1996.
39. Bischoff, R. Proliferation of muscle satellite cells on intact myofibers in culture. Dev
Biol 115, 129, 1986.
40. Syverud, B.C., Lee, J.D., VanDusen, K.W., and Larkin, L.M. Isolation and Purification
of Satellite Cells for Skeletal Muscle Tissue Engineering. J Regen Med 3, 117, 2015.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
41. Bonner, W.A., Hulett, H.R., Sweet, R.G., and Herzenberg, L.A. Fluorescence activated
cell sorting. Rev Sci Instrum 43, 404, 1972.
42. McClure, C.D. and Southall, T.D. Getting Down to Specifics: Profiling Gene
Expression and Protein‐DNA Interactions in a Cell Type‐Specific Manner. Adv Genet
91, 103, 2015.
Tissue Engineering
43. Asakura, A., Seale, P., Girgis‐Gabardo, A., and Rudnicki, M.A. Myogenic specification
of side population cells in skeletal muscle. J Cell Biol 159, 123, 2002.
44. Pasut, A., Oleynik, P., and Rudnicki, M.A. Isolation of muscle stem cells by
fluorescence activated cell sorting cytometry. Methods Mol Biol 798, 53, 2012.
45. Bosnakovski, D., Xu, Z., Li, W., Thet, S., Cleaver, O., Perlingeiro, R.C., and Kyba, M.
Prospective isolation of skeletal muscle stem cells with a Pax7 reporter. Stem Cells
26, 3194, 2008.
46. Montarras, D., Morgan, J., Collins, C., Relaix, F., Zaffran, S., Cumano, A., Partridge, T.,
and Buckingham, M. Direct Isolation of Satellite Cells for Skeletal Muscle
Regeneration. Science, 309, 2064, 2005.
47. Beliakova‐Bethell, N., Massanella, M., White, C., Lada, S.M., Du, P., Vaida, F., Blanco,
J., Spina, C.A., and Woelk, C.H. The effect of cell subset isolation method on gene
expression in leukocytes. Cytom Part A 85, 94, 2014.
48. Fong, C.Y., Peh, G.S., Gauthaman, K., and Bongso, A. Separation of SSEA‐4 and TRA‐
Page 34 of 57
34
1‐60 labelled undifferentiated human embryonic stem cells from a heterogeneous
cell population using magnetic‐activated cell sorting (MACS) and fluorescence‐
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
activated cell sorting (FACS). Stem Cell Rev Reports 5, 72, 2009.
49. Richler, C. and Yaffe, D. The in vitro cultivation and differentiation capacities of
myogenic cell lines. Dev Biol 23, 1, 1970.
50. Lavasani, M., Lu, A., Thompson, S.D., Robbins, P.D., Huard, J., and Niedernhofer, L.J.
Isolation of muscle‐derived stem/progenitor cells based on adhesion characteristics
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
to collagen‐coated surfaces. Methods Mol Biol 976, 53, 2013.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
51. Syverud, B.C., Lin, E., Nagrath, S., and Larkin, L.M. Label‐Free, High‐Throughput
Purification of Satellite Cells Using Microfluidic Inertial Separation. Tissue Eng Part C
Methods 24, 32, 2018.
52. Bian, Y., He, X., Mu, R., Li, X., Ma, Y., Li, Y., and Guan, W. [Isolation, culture and
Tissue Engineering
differentiation of skeletal muscle satellite cells of Luxi cattle embryo]. Xi Bao Yu Fen
Zi Mian Yi Xue Za Zhi 29, 1196, 2013.
53. Doumit, M.E. and Merkel, R.A. Conditions for isolation and culture of porcine
myogenic satellite cells. Tissue Cell 24, 253, 1992.
54. Blau, H.M. and Webster, C. Isolation and characterization of human muscle cells.
Proc Natl Acad Sci U S A 78, 5623, 1981.
55. McFarland, D.C., Doumit, M.E., and Minshall, R.D. The turkey myogenic satellite cell:
Optimization of in vitro proliferation and differentiation. Tissue Cell 20, 899, 1988.
56. Weist, M.R., Wellington, M.S., Bermudez, J.E., Kostrominova, T.Y., Mendias, C.L.,
Arruda, E.M., and Larkin, L.M. TGF‐β1 enhances contractility in engineered skeletal
muscle. J Tissue Eng Regen Med 7, 562, 2013.
57. Syverud, B.C., VanDusen, K.W., and Larkin, L.M. Effects of Dexamethasone on
Satellite Cells and Tissue Engineered Skeletal Muscle Units. Tissue Eng Part A 22,
480, 2016.
Page 35 of 57
35
58. Allen, R.E., Temm‐Grove, C.J., Sheehan, S.M., and Rice, G. Chapter 8 Skeletal Muscle
Satellite Cell Cultures. Methods Cell Biol 52, 155, 1997.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
59. Pollot, B.E., Rathbone, C.R., Wenke, J.C., and Guda, T. Natural polymeric hydrogel
evaluation for skeletal muscle tissue engineering. J Biomed Mater Res B Appl
Biomater 2017.
60. Engler, A.J., Griffin, M.A., Sen, S., Bönnemann, C.G., Sweeney, H.L., and Discher, D.E.
Myotubes differentiate optimally on substrates with tissue‐like stiffness:
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
pathological implications for soft or stiff microenvironments. J Cell Biol 166, 877,
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
2004.
61. Ostrovidov, S., Hosseini, V., Ahadian, S., Fujie, T., Parthiban, S.P., Ramalingam, M.,
Bae, H., Kaji, H., and Khademhosseini, A. Skeletal muscle tissue engineering:
methods to form skeletal myotubes and their applications. Tissue Eng Part B Rev 20,
403, 2014.
Tissue Engineering
62. Matheny, R.W. and Nindl, B.C. Loss of IGF‐IEa or IGF‐IEb Impairs Myogenic
Differentiation. Endocrinology 152, 1923, 2011.
63. Kang, J.S., Yi, M.J., Zhang, W., Feinleib, J.L., Cole, F., and Krauss, R.S. Netrins and
neogenin promote myotube formation. J Cell Biol 167, 493, 2004.
64. Manitt, C., Colicos, M.A., Thompson, K.M., Rousselle, E., Peterson, A.C., and
Kennedy, T.E. Widespread expression of netrin‐1 by neurons and oligodendrocytes
in the adult mammalian spinal cord. J Neurosci 21, 3911, 2001.
65. Serafini, T., Kennedy, T.E., Galko, M.J., Mirzayan, C., Jessell, T.M., and Tessier‐
Lavigne, M. The netrins define a family of axon outgrowth‐promoting proteins
homologous to C. elegans UNC‐6. Cell 78, 409, 1994.
66. Markworth, J.F. and Cameron‐Smith, D. Arachidonic acid supplementation enhances
in vitro skeletal muscle cell growth via a COX‐2‐dependent pathway. Am J Physiol
Cell Physiol 304, C56, 2013.
67. Jang, Y.N. and Baik, E.J. JAK‐STAT pathway and myogenic differentiation. JAKSTAT 2,
Page 36 of 57
36
e23282, 2013.
68. Stern, M.M., Myers, R.L., Hammam, N., Stern, K.A., Eberli, D., Kritchevsky, S.B.,
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Soker, S., and Van Dyke, M. The influence of extracellular matrix derived from
skeletal muscle tissue on the proliferation and differentiation of myogenic
progenitor cells ex vivo. Biomaterials 30, 2393, 2009.
69. Zanchi, N.E. and Lancha, A.H. Mechanical stimuli of skeletal muscle: implications on
mTOR/p70s6k and protein synthesis. Eur J Appl Physiol 102, 253, 2007.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
70. Nikkhah, M., Edalat, F., Manoucheri, S., and Khademhosseini, A. Engineering
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
microscale topographies to control the cell–substrate interface. Biomaterials 33,
5230, 2012.
71. Curtis, A. and Wilkinson, C. Topographical control of cells. Biomaterials 18, 1573,
1997.
Tissue Engineering
72. McDonald, J.C., Duffy, D.C., Anderson, J.R., Chiu, D.T., Wu, H., Schueller, O.J., and
Whitesides, G.M. Fabrication of microfluidic systems in poly(dimethylsiloxane).
Electrophoresis 21, 27, 2000.
73. Pennisi, C.P., Olesen, C.G., de Zee, M., Rasmussen, J., and Zachar, V. Uniaxial cyclic
strain drives assembly and differentiation of skeletal myocytes. Tissue Eng Part A 17,
2543, 2011.
74. Vandenburgh, H.H. and Karlisch, P. Longitudinal growth of skeletal myotubes in vitro
in a new horizontal mechanical cell stimulator. In Vitro Cell Dev Biol 25, 607, 1989.
75. Segurola, R.J., Mills, I., and Sumpio, B.E. Strain‐induced dual alignment of L6 rat
skeletal muscle cells. In Vitro Cell Dev Biol Anim 34, 609, 1998.
76. Matsue, T., Matsumoto, N., and Uchida, I. Rapid micropatterning of living cells by
repulsive dielectrophoretic force. Electrochim Acta 42, 3251, 1997.
77. Suzuki, M., Yasukawa, T., Shiku, H., and Matsue, T. Negative dielectrophoretic
patterning with different cell types. Biosens Bioelectron 24, 1043, 2008.
Page 37 of 57
37
78. Evans, D.J., Britland, S., and Wigmore, P.M. Differential response of fetal and
neonatal myoblasts to topographical guidance cues in vitro. Dev Genes Evol 209,
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
438, 1999.
79. Clark, P., Dunn, G.A., Knibbs, A., and Peckham, M. Alignment of myoblasts on
ultrafine gratings inhibits fusion in vitro. Int J Biochem Cell Biol 34, 816, 2002.
80. Neal, D., Sakar, M.S., Ong, L.S., and Asada, H. Formation of elongated fascicle‐
inspired 3D tissues consisting of high‐density, aligned cells using sacrificial outer
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
molding. Lab Chip 14, 1907, 2014.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
81. Xia, Y. and Whitesides, G.M. Soft Lithography. Angew Chemie Int Ed 37, 550, 1998.
82. Bach, A.D., Stem‐Straeter, J., Beier, J.P., Bannasch, H., and Stark, G.B. Engineering of
muscle tissue. Clin Plast Surg 30, 589, 2003.
83. Dennis, R.G., Smith, B., Philp, A., Donnelly, K., and Baar, K. Bioreactors for guiding
Tissue Engineering
muscle tissue growth and development. Adv Biochem Eng Biotechnol 112, 39, 2009.
84. Akiyama, Y., Terada, R., Hashimoto, M., Hoshino, T., Furukawa, Y., and Morishima, K.
Rod‐shaped Tissue Engineered Skeletal Muscle with Artificial Anchors to Utilize as a
Bio‐Actuator. J Biomech Sci Eng 5, 236, 2010.
85. Ahadian, S., Ostrovidov, S., Hosseini, V., Kaji, H., Ramalingam, M., Bae, H., and
Khademhosseini, A. Electrical stimulation as a biomimicry tool for regulating muscle
cell behavior. Organogenesis 9, 87, 2013.
86. Ahadian, S., Ramón‐Azcón, J., Ostrovidov, S., Camci‐Unal, G., Hosseini, V., Kaji, H.,
Ino, K., Shiku, H., Khademhosseini, A., and Matsue, T. Interdigitated array of Pt
electrodes for electrical stimulation and engineering of aligned muscle tissue. Lab
Chip 12, 3491, 2012.
87. Hosseini, V., Ahadian, S., Ostrovidov, S., Camci‐Unal, G., Chen, S., Kaji, H.,
Ramalingam, M., and Khademhosseini, A. Engineered contractile skeletal muscle
tissue on a microgrooved methacrylated gelatin substrate. Tissue Eng Part A 18,
2453, 2012.
Page 38 of 57
38
88. Yuge, L. and Kataoka, K. Differentiation Of Myoblasts Is Accelerated In Culture In A
Magnetic Field. Vitr Cell Dev Biol Anim 36, 383, 2000.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
89. Sicari, B.M., Agrawal, V., Siu, B.F., Medberry, C.J., Dearth, C.L., Turner, N.J., and
Badylak, S.F. A murine model of volumetric muscle loss and a regenerative medicine
approach for tissue replacement. Tissue Eng Part A 18, 1941, 2012.
90. Wu, X., Corona, B.T., Chen, X., and Walters, T.J. A standardized rat model of
volumetric muscle loss injury for the development of tissue engineering therapies.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Biores Open Access 1, 280, 2012.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
91. Corona, B.T., Machingal, M.A., Criswell, T., Vadhavkar, M., Dannahower, A.C.,
Bergman, C., Zhao, W., and Christ, G.J. Further development of a tissue engineered
muscle repair construct in vitro for enhanced functional recovery following
implantation in vivo in a murine model of volumetric muscle loss injury. Tissue Eng
Part A 18, 1213, 2012.
Tissue Engineering
92. Pittenger, M.F. Multilineage Potential of Adult Human Mesenchymal Stem Cells.
Science 284, 143, 1999.
93. Zuk, P.A., Zhu, M., Ashjian, P., De Ugarte, D.A., Huang, J.I., Mizuno, H., Alfonso, Z.C.,
Fraser, J.K., Benhaim, P., and Hedrick, M.H. Human adipose tissue is a source of
multipotent stem cells. Mol Biol Cell 13, 4279, 2002.
94. Dominici, M., Le Blanc, K., Mueller, I., Slaper‐Cortenbach, I., Marini, F., Krause, D.S.,
Deans, R.J., Keating, A., Prockop, D.J., and Horwitz, E.M. Minimal criteria for defining
multipotent mesenchymal stromal cells. The International Society for Cellular
Therapy position statement. Cytotherapy 8, 315, 2006.
95. Zuk, P. Adipose‐Derived Stem Cells in Tissue Regeneration: A Review. ISRN Stem
Cells 2013, 1, 2013.
96. Lin, C.S., Lin, G., and Lue, T.F. Allogeneic and Xenogeneic Transplantation of
Adipose‐Derived Stem Cells in Immunocompetent Recipients Without
Immunosuppressants. Stem Cells Dev 21, 2770, 2012.
Page 39 of 57
39
97. Naderi, N., Combellack, E.J., Griffin, M., Sedaghati, T., Javed, M., Findlay, M.W.,
Wallace, C.G., Mosahebi, A., Butler, P.E., Seifalian, A.M., and Whitaker. I.S. The
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
regenerative role of adipose‐derived stem cells (ADSC) in plastic and reconstructive
surgery. Int Wound J 14, 112, 2017.
98. Forcales S.V. Potential of adipose‐derived stem cells in muscular regenerative
therapies. Front Aging Neurosci 7, 123, 2015.
99. Aust, L., Devlin, B., Foster, S.J., Halvorsen, Y.D., Hicok, K., du Laney, T., Sen, A.,
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Willingmyre, G.D., and Gimble, J.M. Yield of human adipose‐derived adult stem cells
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
from liposuction aspirates. Cytotherapy 6, 7, 2004.
100. Oedayrajsingh‐Varma, M.J., van Ham, S.M., Knippenberg, M., Helder, M.N., Klein‐
Nulend, J., Schouten, T.E., Ritt, M.J., and van Milligen, F.J. Adipose tissue‐derived
mesenchymal stem cell yield and growth characteristics are affected by the tissue‐
harvesting procedure. Cytotherapy 8, 166, 2006.
Tissue Engineering
101. Miyagi‐Shiohira, C., Kurima, K., Kobayashi, N., Saitoh, I., Watanabe, M., Noguchi, Y.,
Matsushita, M., Noguchi, H. Cryopreservation of Adipose‐Derived Mesenchymal
Stem Cells. Cell Med 8, 3, 2015.
102. Priya, N., Sarcar, S., Majumdar, A.S., and SundarRaj, S. Explant culture: a simple,
reproducible, efficient and economic technique for isolation of mesenchymal
stromal cells from human adipose tissue and lipoaspirate. J Tissue Eng Regen Med 8,
706, 2014.
103. Francis, M.P., Sachs, P.C., Elmore, L.W., and Holt, S.E. Isolating adipose‐derived
mesenchymal stem cells from lipoaspirate blood and saline fraction. Organogenesis
6, 11, 2010.
104. Huri, P.Y., Wang, A., Spector, A.A., S, and Grayson, W.L. Multistage Adipose‐Derived
Stem Cell Myogenesis: An Experimental and Modeling Study. Cell Mol Bioeng 7, 497,
2014.
105. Di Rocco, G., Iachininoto, M.G., Tritarelli, A., Straino, S., Zacheo, A., Germani, A.,
Page 40 of 57
40
Crea, F., and Capogrossi, M.C. Myogenic potential of adipose‐tissue‐derived cells. J
Cell Sci 119, 2945, 2006.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
106. Yilgor Huri, P., Cook, C.A., Hutton, D.L., Goh, B.C., Gimble, J.M., DiGirolamo. D.J., and
Grayson, W.L. Biophysical cues enhance myogenesis of human adipose derived
stem/stromal cells. Biochem Biophys Res Commun 438, 180, 2013.
107. Goudenege, S., Pisani, D.F., Wdziekonski, B., Di Santo, J.P., Bagnis, C., Dani, C., and
Dechesne, C.A. Enhancement of myogenic and muscle repair capacities of human
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
adipose‐derived stem cells with forced expression of MyoD. Mol Ther 17, 1064,
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
2009.
108. Singh, K. and Dilworth, F.J. Differential modulation of cell cycle progression
distinguishes members of the myogenic regulatory factor family of transcription
factors. FEBS J 280, 3991, 2013.
Tissue Engineering
109. Sung, M.S., Mun, J.Y., Kwon, O., Kwon, K.S., and Oh, D.B. Efficient myogenic
differentiation of human adipose‐derived stem cells by the transduction of
engineered MyoD protein. Biochem Biophys Res Commun 437, 156, 2013.
110. Kesireddy, V. Evaluation of adipose‐derived stem cells for tissue‐engineered muscle
repair construct‐mediated repair of a murine model of volumetric muscle loss
injury. Int J Nanomedicine 11, 1461, 2016.
111. Owen, M., Friedenstein, A.J. Stromal stem cells: marrow‐derived osteogenic
precursors. Ciba Found Symp 136, 42, 1988.
112. Pontikoglou, C., Deschaseaux, F., Sensebé, L., Papadaki, H.A. Bone Marrow
Mesenchymal Stem Cells: Biological Properties and Their Role in Hematopoiesis and
Hematopoietic Stem Cell Transplantation. Stem Cell Rev Reports 7, 569, 2011.
113. Boxall, S.A. and Jones, E. Markers for characterization of bone marrow
multipotential stromal cells. Stem Cells Int 2012, 975871, 2012.
114. Bayati, V., Hashemitabar, M., Gazor, R., Nejatbakhsh, R., and Bijannejad, D.
Expression of surface markers and myogenic potential of rat bone marrow and
Page 41 of 57
41
adipose‐derived stem cells: a comparative study. Anat Cell Biol 46, 113, 2013.
115. Jacobs, S.A., Roobrouck, V.D., Verfaillie, C.M., and Van Gool, S.W. Immunological
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
characteristics of human mesenchymal stem cells and multipotent adult progenitor
cells. Immunol Cell Biol 91, 32, 2013.
116. Bara, J.J., Richards, R.G., Alini, M., and Stoddart, M.J. Concise Review: Bone Marrow‐
Derived Mesenchymal Stem Cells Change Phenotype Following In Vitro Culture:
Implications for Basic Research and the Clinic. Stem Cells 32, 1713, 2014.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
117. Xing, W., Pang, A.M., Yao, J.F., Li, Y., Shi, H., Sheng, M.Y., Zhou, Y., Zhao, Y.X., Xu,
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
M.J., and Yang, F.C. [Efficient isolation of mesenchymal stem cells from human bone
marrow by direct plating method combined with modified primary explant culture].
Zhongguo shi yan xue ye xue za zhi 21, 451, 2013.
118. Ginis, I., Grinblat, B., and Shirvan, M.H. Evaluation of bone marrow‐derived
Tissue Engineering
mesenchymal stem cells after cryopreservation and hypothermic storage in clinically
safe medium. Tissue Eng Part C Methods 18, 453, 2012.
119. Shen, J., Huang, Y., Xu, S., Zheng, P., Yin, W., Cen, J., and Gong, L. Effectiveness of
human mesenchymal stem cells derived from bone marrow cryopreserved for 23–
25 years. Cryobiology 64, 167, 2012.
120. Wakitani, S., Saito, T., and Caplan, A.I. Myogenic cells derived from rat bone marrow
mesenchymal stem cells exposed to 5‐azacytidine. Muscle Nerve 18, 1417, 1995.
121. Carr, B.I., Reilly, J.G., Smith, S.S., Winberg, C., and Riggs, A. The tumorigenicity of 5‐
azacytidine in the male Fischer rat. Carcinogenesis 5, 1583, 1984.
122. Stern‐Straeter, J., Bonaterra, G.A., Juritz, S., Birk, R., Goessler, U.R., Bieback, K.,
Bugert, P., Schultz, J., Hörmann, K., Kinscherf, R., and Faber, A. Evaluation of the
effects of different culture media on the myogenic differentiation potential of
adipose tissue‐ or bone marrow‐derived human mesenchymal stem cells. Int J Mol
Med 33, 160, 2013.
123. Beier, J.P., Bitto, F.F., Lange, C., Klumpp, D., Arkudas, A., Bleiziffer, O., Boos, A.M.,
Page 42 of 57
42
Horch, R.E., and Kneser, U. Myogenic differentiation of mesenchymal stem cells co‐
cultured with primary myoblasts. Cell Biol Int 35, 397, 2011.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
124. Egusa, H,. Kobayashi, M., Matsumoto, T., Sasaki, J.I., Uraguchi, S., and Yatani, H.
Application of cyclic strain for accelerated skeletal myogenic differentiation of
mouse bone marrow‐derived mesenchymal stromal cells with cell alignment. Tissue
Eng Part A 19, 770, 2013.
125. Meligy, F.Y., Shigemura, K., Behnsawy, H.M., Fujisawa, M., Kawabata, M., and
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Shirakawa, T. The efficiency of in vitro isolation and myogenic differentiation of
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
MSCs derived from adipose connective tissue, bone marrow, and skeletal muscle
tissue. Vitr Cell Dev Biol Anim 48, 203, 2012.
126. Natsu, K., Ochi, M., Mochizuki, Y., Hachisuka, H., Yanada, S., and Yasunaga, Y.
Allogeneic Bone Marrow‐Derived Mesenchymal Stromal Cells Promote the
Regeneration of Injured Skeletal Muscle without Differentiation into Myofibers.
Tissue Engineering
Tissue Eng 10, 1093, 2004.
127. Merritt, E.K., Cannon, M.V., Hammers, D.W., Le, L.N., Gokhale, R., Sarathy, A., Song,
T.J., Tierney, M.T., Suggs, L.J., Walters, T.J., and Farrar, R.P. Repair of traumatic
skeletal muscle injury with bone‐marrow‐derived mesenchymal stem cells seeded
on extracellular matrix. Tissue Eng Part A 16, 2871, 2010.
128. Andrade, B.M., Baldanza, M.R., Ribeiro, K.C., Porto, A., Peçanha, R., Fortes, F.S.,
Zapata‐Sudo, G., Campos‐de‐Carvalho, A.C., Goldenberg, R.C., and Werneck‐de‐
Castro, J.P. Bone Marrow Mesenchymal Cells Improve Muscle Function in a Skeletal
Muscle Re‐Injury Model. PLoS One 10, e0127561, 2015.
129. Helal, M.A., Shaheen, N.E., and Abu Zahra, F.A. Immunomodulatory capacity of the
local mesenchymal stem cells transplantation after severe skeletal muscle injury in
female rats. Immunopharmacol Immunotoxicol 38, 1, 2016.
130. Beeravolu, N., McKee, C., Alamri, A., Mikhael, S., Brown, C., Perez‐Cruet, M., and
Chaudhry, G.R. Isolation and Characterization of Mesenchymal Stromal Cells from
Human Umbilical Cord and Fetal Placenta. J Vis Exp 3, 2017.
Page 43 of 57
43
131. Hsieh, J.Y., Fu, Y.S., Chang, S.J., Tsuang, Y.H., and Wang, H.W. Functional Module
Analysis Reveals Differential Osteogenic and Stemness Potentials in Human
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Mesenchymal Stem Cells from Bone Marrow and Wharton’s Jelly of Umbilical Cord.
Stem Cells Dev 19, 1895, 2010.
132. Kocaefe, C., Balci, D., Hayta, B.B., and Can, A. Reprogramming of human umbilical
cord stromal mesenchymal stem cells for myogenic differentiation and muscle
repair. Stem Cell Rev 6, 512, 2010.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
133. Lu, L., Liu, Y., Yang, S., Zhao, Q., Wang, X., Gong, W., Han, Z., Xu, Z., Lu, Y., Liu, D.,
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Chen, Z., and Han, Z. Isolation and characterization of human umbilical cord
mesenchymal stem cells with hematopoiesis‐supportive function and other
potentials. Haematologica 91, 1017, 2006.
134. Hendijani, F., Sadeghi‐Aliabadi, H., and Haghjooy Javanmard, S. Comparison of
human mesenchymal stem cells isolated by explant culture method from entire
Tissue Engineering
umbilical cord and Wharton’s jelly matrix. Cell Tissue Bank 15, 555, 2014.
135. Van Pham, P., Truong, N.C., Le, P.T., Tran, T.D., Vu, N.B., Bui, K.H., and Phan, N.K.
Isolation and proliferation of umbilical cord tissue derived mesenchymal stem cells
for clinical applications. Cell Tissue Bank 17, 289, 2016.
136. Nagamura‐Inoue, T. and He, H. Umbilical cord‐derived mesenchymal stem cells:
Their advantages and potential clinical utility. World J Stem Cells 6, 195, 2014.
137. Gang, E.J., Jeong, J.A., Hong, S.H., Hwang, S.H., Kim, S.W., Yang, I.H., Ahn, C., Han, H.,
and Kim, H. Skeletal Myogenic Differentiation of Mesenchymal Stem Cells Isolated
from Human Umbilical Cord Blood. Stem Cells 22, 617, 2004.
138. Zucconi, E., Vieira, N.M., Bueno, C.R., Secco, M., Jazedje, T., Costa Valadares, M.,
Fussae Suzuki, M., Bartolini, P., Vainzof, M., and Zatz, M. Preclinical studies with
umbilical cord mesenchymal stromal cells in different animal models for muscular
dystrophy. J Biomed Biotechnol 2011, 715251, 2011.
139. Nunes, V.A., Cavaçana, N., Canovas, M., Strauss, B.E., and Zatz, M. Stem cells from
Page 44 of 57
44
umbilical cord blood differentiate into myotubes and express dystrophin in vitro
only after exposure to in vivo muscle environment. Biol Cell 99, 185, 2007.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
140. Koponen, J.K., Kekarainen, T., Heinonen, S.E., Laitinen, A., Nystedt, J., Laine, J., and
Ylä‐Herttuala, S. Umbilical Cord Blood–derived Progenitor Cells Enhance Muscle
Regeneration in Mouse Hindlimb Ischemia Model. Mol Ther 15, 2172, 2007.
141. James, A.W., Zara, J.N., Zhang, X., Askarinam, A., Goyal, R., Chiang, M., Yuan, W.,
Chang, L., Corselli, M., Shen, J., Pang, S., Stoker, D., Wu, B., Ting, K., Péault, B., and
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Soo, C. Perivascular stem cells: a prospectively purified mesenchymal stem cell
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
population for bone tissue engineering. Stem Cells Transl Med 1, 510, 2012.
142. Bautch, V.L. Stem cells and the vasculature. Nat Med 17, 1437, 2011.
143. Corselli, M., Chen, C.W., Sun, B., Yap, S., Rubin, J.P., and Péault, B. The tunica
adventitia of human arteries and veins as a source of mesenchymal stem cells. Stem
Tissue Engineering
Cells Dev 21, 1299, 2012.
144. Corselli, M., Chen, C.W., Sun, B., Yap, S., Rubin, J.P., and Péault, B. The tunica
adventitia of human arteries and veins as a source of mesenchymal stem cells. Stem
Cells Dev 21, 1299, 2012.
145. Fuoco, C., Salvatori, M.L., Biondo, A., Shapira‐Schweitzer, K., Santoleri, S., Antonini,
S., Bernardini, S., Tedesco, F.S., Cannata, S., Seliktar, D., Cossu, G., and Gargioli, C.
Injectable polyethylene glycol‐fibrinogen hydrogel adjuvant improves survival and
differentiation of transplanted mesoangioblasts in acute and chronic skeletal‐
muscle degeneration. Skelet Muscle 2, 24, 2012.
146. Crisan, M., Corselli, M., Chen, W.C., Péault, B., Moldovan, N. Perivascular cells for
regenerative medicine. J Cell Mol Med 16, 2851, 2012.
147. Dellavalle, A., Maroli, G., Covarello, D., Azzoni, E., Innocenzi, A., Perani, L., Antonini,
S., Sambasivan, R., Brunelli, S., Tajbakhsh, S., and Cossu, G. Pericytes resident in
postnatal skeletal muscle differentiate into muscle fibres and generate satellite
cells. Nat Commun 2, 499, 2011.
Page 45 of 57
45
148. Dellavalle, A., Sampaolesi, M., Tonlorenzi, R., Tagliafico, E., Sacchetti, B., Perani, L.,
Innocenzi, A., Galvez, B.G., Messina, G., Morosetti, R., Li, S., Belicchi, M., Peretti, G.,
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Chamberlain, J.S., Wright, W.E., Torrente, Y., Ferrari, S., Bianco, P., and Cossu, G.
Pericytes of human skeletal muscle are myogenic precursors distinct from satellite
cells. Nat Cell Biol 9, 255, 2007.
149. Bryan, B.A. and D’Amore, P.A. Pericyte Isolation and Use in Endothelial/Pericyte
Coculture Models. Methods in enzymology 443, 315, 2008.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
150. Domev, H., Milkov, I., Itskovitz‐Eldor, J., and Dar, A. Immunoevasive Pericytes From
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Human Pluripotent Stem Cells Preferentially Modulate Induction of Allogeneic
Regulatory T Cells. Stem Cells Transl Med 3, 1169, 2014.
151. Fuoco, C., Sangalli, E., Vono, R., Testa, S., Sacchetti, B., Latronico, M.V., Bernardini,
S., Madeddu, P., Cesareni, G., Seliktar, D., Rizzi, R., Bearzi, C., Cannata, S.M., Spinetti,
G., and Gargioli, C. 3D hydrogel environment rejuvenates aged pericytes for skeletal
Tissue Engineering
muscle tissue engineering. Front Physiol 5, 203, 2014.
152. Fuoco, C., Sangalli, E., Vono, R., Testa, S., Sacchetti, B., Latronico, M.V., Bernardini,
S., Madeddu, P., Cesareni, G., Seliktar, D., Rizzi, R., Bearzi, C., Cannata, S.M., Spinetti,
G., and Gargioli, C. 3D hydrogel environment rejuvenates aged pericytes for skeletal
muscle tissue engineering. Front Physiol 5, 203, 2014.
153. Nishikawa, S., Goldstein, R.A., and Nierras, C.R. The promise of human induced
pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol 9, 725, 2008.
154. McCullagh, K.J. and Perlingeiro, R.C. Coaxing stem cells for skeletal muscle repair.
Adv Drug Deliv Rev 84, 198, 2015.
155. Takahashi, K and Yamanaka, S. Induction of Pluripotent Stem Cells from Mouse
Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 126, 663, 2006.
156. Apostolou, E. and Hochedlinger, K. Chromatin dynamics during cellular
reprogramming. Nature 502, 462, 2013.
157. Djuric, U. and Ellis, J. Epigenetics of induced pluripotency, the seven‐headed dragon.
Page 46 of 57
46
Stem Cell Res Ther 1, 3, 2010.
158. Gładych, M., Andrzejewska, A., Oleksiewicz, U., and Estécio, M.R. Epigenetic
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
mechanisms of induced pluripotency. Contemp Oncol (Pozn) 19, A30, 2015.
159. Warren, L., Manos, P.D., Ahfeldt, T., Loh, Y.H., Li, H., Lau, F., Ebina, W., Mandal, P.K.,
Smith, Z.D., Meissner, A., Daley, G.Q., Brack, A.S., Collins, J.J., Cowan, C., Schlaeger,
T.M., and Rossi, D.J. Highly efficient reprogramming to pluripotency and directed
differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7, 618,
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
2010.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
160. Yulin, X., Lizhen, L., Lifei, Z., Shan, F., Ru, L., Kaimin, H., and Huang, H. Efficient
generation of induced pluripotent stem cells from human bone marrow
mesenchymal stem cells. Folia Biol (Praha) 58, 221, 2012.
161. Kunisato, A., Wakatsuki, M., Kodama, Y., Shinba, H., Ishida, I., and Nagao, K.
Tissue Engineering
Generation of Induced Pluripotent Stem Cells by Efficient Reprogramming of Adult
Bone Marrow Cells. Stem Cells Dev 19, 229, 2010.
162. Malik, N. and Rao, M.S. A review of the methods for human iPSC derivation.
Methods Mol Biol 997, 23, 2013.
163. Liu, W. and Chen, G. Cryopreservation of human pluripotent stem cells in defined
medium. Curr Protoc Stem Cell Biol 31, 1C.17.1, 2014.
164. Shoji, E., Woltjen, K., and Sakurai, H. Directed Myogenic Differentiation of Human
Induced Pluripotent Stem Cells. Methods Mol Biol 1353, 89, 2015.
165. Chal, J., Al Tanoury, Z., Hestin, M., Gobert, B., Aivio, S., Hick, A., Cherrier, T.,
Nesmith, A.P., Parker, K.K., and Pourquié, O. Generation of human muscle fibers and
satellite‐like cells from human pluripotent stem cells in vitro. Nat Protoc 11, 1833,
2016.
166. Maffioletti, S.M., Gerli, M.F., Ragazzi, M., Dastidar, S., Benedetti, S., Loperfido, M.,
VandenDriessche, T., Chuah, M.K., and Tedesco, F.S. Efficient derivation and
inducible differentiation of expandable skeletal myogenic cells from human ES and
Page 47 of 57
47
patient‐specific iPS cells. Nat Protoc 10, 941, 2015.
167. Darabi, R., Pan, W., Bosnakovski, D., Baik, J., Kyba, M., and Perlingeiro, R.C.
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Functional myogenic engraftment from mouse iPS cells. Stem Cell Rev 7, 948, 2011.
168. Roca, I., Requena, J., Edel, M.J., and Alvarez‐Palomo, A.B. Myogenic Precursors from
iPS Cells for Skeletal Muscle Cell Replacement Therapy. J Clin Med 4, 243, 2015.
169. Ben‐David, U. and Benvenisty, N. The tumorigenicity of human embryonic and
induced pluripotent stem cells. Nat Rev Cancer 11, 268, 2011.
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
170. Hosoyama, T., McGivern, J.V., Van Dyke, J.M., Ebert, A.D., and Suzuki M. Derivation
of myogenic progenitors directly from human pluripotent stem cells using a sphere‐
based culture. Stem Cells Transl Med 3, 564, 2014.
171. Xu, C., Tabebordbar, M., Iovino, S., Ciarlo, C., Liu, J., Castiglioni, A., Price, E., Liu, M.,
Barton, E.R., Kahn, C.R., Wagers, A.J., and Zon. L.I. A zebrafish embryo culture
Tissue Engineering
system defines factors that promote vertebrate myogenesis across species. Cell 155,
909, 2013.
172. Darabi, R. and Perlingeiro, R.C. Derivation of Skeletal Myogenic Precursors from
Human Pluripotent Stem Cells Using Conditional Expression of PAX7. In: Methods
Mol Bio 1357, 423, 2014.
173. Quattrocelli, M., Swinnen, M., Giacomazzi, G., Camps, J., Barthélemy, I., Ceccarelli,
G., Caluwé, E., Grosemans, H., Thorrez, L., Pelizzo, G., Muijtjens, M., Verfaillie, C.M.,
Blot, S., Janssens, S., and Sampaolesi, M. Mesodermal iPSC–derived progenitor cells
functionally regenerate cardiac and skeletal muscle. J Clin Invest 125, 4463, 2015.
174. Goudenege, S., Lebel, C., Huot, N.B., Dufour, C., Fujii, I., Gekas, J., Rousseau, J., and
Tremblay, J.P. Myoblasts derived from normal hESCs and dystrophic hiPSCs
efficiently fuse with existing muscle fibers following transplantation. Mol Ther 20,
2153, 2012.
175. Lo, B. and Parham, L. Ethical issues in stem cell research. Endocr Rev 30, 204, 2009.
Page 48 of 57
48
176. Tě Zslav Bryja, V., Bonilla, S., Š Č Nek, L.A., Parish, C.L., Schwartz, C.M., Luo, Y., Rao,
M.S., and Arenas, E. An Efficient Method for the Derivation of Mouse Embryonic
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Stem Cells. Stem Cells 24, 844, 2005.
177. Blum, B. and Benvenisty, N. The Tumorigenicity of Human Embryonic Stem Cells.
Advances in cancer research 100, 133, 2008:133–158.
178. Crisan, M., Yap, S., Casteilla, L., Chen, C.W., Corselli, M., Park, T.S., Andriolo, G., Sun,
B., Zheng, B., Zhang, L., Norotte, C., Teng, P.N., Traas, J., Schugar, R., Deasy, B.M.,
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Badylak, S., Bűhring, H.J., Giacobino, J.P., Lazzari, L., Huard, J., and Péault, B. A
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Perivascular Origin for Mesenchymal Stem Cells in Multiple Human Organs. Cell
Stem Cell 3, 301, 2008.
179. Hendijani, F. Explant culture: An advantageous method for isolation of
mesenchymal stem cells from human tissues. Cell Prolif 50, e12334, 2017.
Tissue Engineering
180. Pierantozzi, E., Badin, M., Vezzani, B., Curina, C., Randazzo, D., Petraglia, F., Rossi,
D., and Sorrentino, V. Human pericytes isolated from adipose tissue have better
differentiation abilities than their mesenchymal stem cell counterparts. Cell Tissue
Res 361, 769, 2015.
181. Christodoulou, I., Kolisis, F.N., Papaevangeliou, D., and Zoumpourlis, V. Comparative
Evaluation of Human Mesenchymal Stem Cells of Fetal (Wharton’s Jelly) and Adult
(Adipose Tissue) Origin during Prolonged In Vitro Expansion: Considerations for
Cytotherapy. Stem Cells Int 2013, 246134, 2013.
Page 49 of 57
49
Table 1. Biopsy, Isolation Summary
Characteristics
Satellite Cells Skeletal muscle Painful, invasive, lower Enzymatic dissociation is
(29,30) volume tissue yield higher yield but less pure,
while single fiber explant
culture is highly pure but low
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
yield (40).
ADSCs White adipose Less painful, less Enzymatic dissociation is
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Potential
Satellite Cells Have been expanded to at least 50 Usually highest, but senescence
population doublings in culture (26) as cells are expanded contributes
to decline in myogenesis in vivo
(26, 27).
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
(125). Unknown whether BM‐MSCs
Higher than MC‐MSCs (125) have higher myogenic
differentiation potential.
BM‐MSCs Less than ADSCs (125). Lower than satellite cells and
Tissue Engineering
MC‐MSCs (125). Unknown
whether ADSCs have higher
myogenic differentiation
potential (114, 125).
UC‐MSCs Higher than adult MSCs (181). Relative myogenic potential
unknown.
Pericytes Similar to ADSCs (180). Determined to be an order of
magnitude higher than most
alternative myogenic stem cells,
excluding satellite cells (134).
iPSCs, ESCs Unlimited Can be high when gene
overexpression is used (164‐
167), but these methods are not
clinically feasible. Using medium,
relative myogenic potential
compared to other types is
unknown.
Page 52 of 57
52
Table 5. Muscle Therapy Summary
Cell Type Muscle Therapy Applications To Date
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
Satellite Cells Used in SMTE studies specifically for VML.
Both scaffold (13‐15) and scaffoldless (16) constructs are able to
partially restore structure and function in VML rats and mice.
ADSCs Used in one SMTE study specifically for VML (110).
Undifferentiated ADSC scaffold construct was able to partially
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
restore structure in a VML rat (110). Functional recovery is
presently unknown.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
BM‐MSCs Administered undifferentiated via injection to treat skeletal muscle
injuries in several SMTE studies (126‐128) that are not strictly
confined to VML. Able to partially restore structure and function,
and may also play an immunomodulatory role in healing (129).
Tissue Engineering
UC‐MSCs UC‐MSC injections promoted functional recovery in DMD mice and
dogs (138). UC‐MSC injections promoted both structural and
functional recovery (139) in a separate study. UC‐MSC injections
were able to promote skeletal muscle regeneration in lacerated
mouse muscle by promoting a healing cytokine environment, rather
than directly fusing with fibers (140).
Pericytes After systemic injection, pericytes can fuse with dystrophic mouse
fibers to contribute to structural recovery in a DMD mouse model
(148).
iPSCs After systemic injection, human iPSCs can fuse with dystrophic
fibers to contribute to structural and functional recovery in
dystrophic mice and canines (171‐174).
ESCs Not well‐studied due to technical and ethical challenges (175‐177)
as well as their tumorigenicity (178).
Page 53 of 57
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
53
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Figure 1. Pathway of normal satellite cell myogenesis. Satellite cells express express
paired box protein‐7 (Pax7) and can commit to myoblasts in a process called induction or
can self‐renew. Myoblasts express Pax7, transcription factor myogenic differentiation 1
(MyoD), myogenic regulatory factor 5 (Myf5), and the myogenic protein desmin, and can
Tissue Engineering
differentiate into myocytes, which express MyoD, desmin, and the transcription factor
myogenin. In a process termed terminal differentiation, myocytes fuse into
multinucleated, differentiated myotubes that express the myogenic proteins alpha actinin,
desmin, and myosin heavy chain. Afterwards, myotubes can further mature into
myofibers. Figure adapted from (33).
Page 54 of 57
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
54
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Tissue Engineering
Figure 2. Methods for satellite cell isolation. Single fiber isolation (top) typically involves
digesting a whole muscle and dissecting out individual fibers. Fibers have attached satellite
cells that are exposed during the digestion process, and the fibers are plated, allowing the
satellite cells to egress onto tissue culture plates. Depending on the protocol, fibers may
later be removed from the plates to obtain a more pure satellite cell population. Enzymatic
dissociation (bottom) involves mincing a muscle biopsy, performing an enzymatic digestion
to obtain a heterogeneous population of isolated cells that is typically less pure than the
product from single fiber isolation, and then typically purifying the population to obtain an
isolated satellite cell product. Figure adapted from (40).
Page 55 of 57
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
55
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Tissue Engineering
Figure 3. Methods for satellite cell purification. FACS (A), MACS (B), and pre‐plating (C)
methods to purify satellite cells. FACS uses fluorophore‐conjugated antibody dyes to sort
cells into groups by electrical charge using a laser, which causes the cells to deflect to
specific regions depending on their charge. MACS uses magnetic microbeads coated in
antibody to sort cells by using a magnet. First, a magnet is applied. Cells that aren't
attracted to the magnet are negative for antibody markers and will thus be collected first.
Then, the magnet is removed to elute cells that are attracted to the magnet. Pre‐plating is
done by serially plating a satellite cell suspension over time to purify satellite cells, and
works because other cell types like fibroblasts and epithelial cells tend to adhere to the
plate first. Figure adapted from (42).
Page 56 of 57
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
56
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Tissue Engineering
Figure 4. Visual summary of the relative locations and basic properties of the two varieties
of perivascular stem cells: pericytes and adventitial cells. Pericytes are located in capillary
and microvessel walls and are defined as positive for CD146 and negative for CD34 and
CD45. Adventitial cells are located in the walls of large vessels (veins and arteries) and are
positive for CD34 and negative for CD31, CD145, and CD45. Figure adapted from (142).
Page 57 of 57
This paper has been peer‐reviewed and accepted for publication, but has yet to undergo copyediting and proof correction. The final published version may differ from this proof.
57
Downloaded by Tufts University package NERL from www.liebertpub.com at 03/19/18. For personal use only.
Stem Cells for Skeletal Muscle Tissue Engineering (DOI: 10.1089/ten.TEB.2017.0451)
Tissue Engineering
Figure 5. Visual summary of induced pluripotent stem cell (iPSC) generation for muscle
therapy applications with a comparison to satellite cell methods. Making iPSCs starts with
taking a skin biopsy to isolate fibroblasts. The four Yamanaka transcription factors (Oct4,
Sox2, KLF4, and c‐Myc) are added to induce pluripotency. iPSC cells can become muscle
precursor cells by inducing muscle‐specific genes like Pax7 and Pax3. Then, the myogenic
precursors can be transplanted into a diseased patient. Unlike iPSCs, satellite cells are
isolated from skeletal muscle biopsy and must be expanded in culture, but do not have the
capacity for unlimited self‐renewal. After expansion, satellite cells can be transplanted
back into a patient. Figure from (154).