Sunteți pe pagina 1din 18

J Appl Physiol

91: 534551, 2001.

invited review
Myogenic satellite cells: physiology
to molecular biology
THOMAS J. HAWKE1 AND DANIEL J. GARRY1,2
1
Department of Internal Medicine and 2Department of Molecular Biology,
University of Texas Southwestern Medical Center, Dallas, Texas 75390
Hawke, Thomas J., and Daniel J. Garry. Myogenic satellite cells:
physiology to molecular biology. J Appl Physiol 91: 534551,
2001.Adult skeletal muscle has a remarkable ability to regenerate
following myotrauma. Because adult myofibers are terminally differentiated, the regeneration of skeletal muscle is largely dependent on a
small population of resident cells termed satellite cells. Although this
population of cells was identified 40 years ago, little is known regarding
the molecular phenotype or regulation of the satellite cell. The use of cell
culture techniques and transgenic animal models has improved our
understanding of this unique cell population; however, the capacity and
potential of these cells remain ill-defined. This review will highlight the
origin and unique markers of the satellite cell population, the regulation
by growth factors, and the response to physiological and pathological
stimuli. We conclude by highlighting the potential therapeutic uses of
satellite cells and identifying future research goals for the study of
satellite cell biology.
skeletal muscle; stem cells; regeneration; aging; transgenic models

mammalian species exhibit a


remarkable capacity to adapt to physiological demands
such as growth, training, and injury. The processes by
which these adaptations occur are largely attributed to
a small population of cells that are resident in adult
skeletal muscle and are referred to as satellite cells.
After their initial identification in 1961 (98), Mauro
(117) described a cell closely associated with the periphery of the frog myofiber and termed it a satellite
cell based on its location. Quiescent satellite cells are
physically distinct from the adult myofiber as they
reside in indentations between the sarcolemma and
the basal lamina (130). Adult skeletal muscle fibers are
terminally differentiated such that muscle growth and
regeneration are accomplished by satellite cells. In the
unperturbed state, these cells remain in a nonproliferative, quiescent state. However, in response to stimuli such as myotrauma, satellite cells become activated, proliferate, and express myogenic markers
SKELETAL MUSCLES OF ADULT

Address for reprint requests and other correspondence: D. J.


Garry, UT Southwestern Medical Center at Dallas, 5323 Harry
Hines Blvd., NB11.200, Dallas, TX 75390-8573 (E-mail:
daniel.garry@utsouthwestern.edu).
534

(satellite cells expressing myogenic markers are also


termed myoblasts). Ultimately, these cells fuse to existing muscle fibers or fuse together to form new myofibers during regeneration of damaged skeletal muscle
(12, 167).
Since the original description of the myogenic satellite cell, considerable interest and research efforts
have focused on myogenic satellite cell biology.
These research efforts have enhanced our understanding of muscle growth, remodeling, and regeneration. In addition, new paradigms have been proposed regarding the regenerative capacity and the
plasticity of the myogenic satellite cell population
(108, 119, 139, 168, 169). These paradigms suggest
that the satellite cell population not only has a
remarkable capacity for muscle regeneration but
may also contribute to alternative muscle and nonmuscle lineages and may have clinical applications
in the treatment of devastating and deadly diseases
such as muscular dystrophy.
The current review attempts to integrate the anatomic, physiological, biochemical, and molecular properties that regulate the myogenic satellite cell population. We begin with a brief overview of vertebrate

8750-7587/01 $5.00 Copyright 2001 the American Physiological Society

http://www.jap.org

535

INVITED REVIEW

myogenesis, highlighting the populations of myoblast


precursor cells that contribute to muscle development,
and outline a well-described genetic hierarchy that is
important in muscle specification. We describe the limited gene expression profile and the distinguishing
morphological characteristics of the satellite cell population. We describe the inductive signals that regulate the satellite cell in vitro and in well-described
physiological models. Finally, we will review the stem
cell-like features of the satellite cells with emphasis on
the novel strategies that may be pursued in the future
for the treatment of debilitating myopathies.
Importantly, myogenic satellite cell biology remains an emerging field of scientific inquiry, such
that satisfying and complete answers to the most
fundamental questions are currently unavailable. In
this review, we will provide a summary of the current knowledge in this area and highlight fertile
areas for future research.

BUILDING OF MUSCLE WITH WAVES OF PRECURSOR


CELLS

Anatomic and molecular mechanisms during muscle


regeneration have been postulated to recapitulate
muscle development. Although current evidence suggests the regenerative process may be more complex,
an understanding of muscle development is important
to appreciate the anatomic and molecular network
associated with muscle regeneration.
During embryogenesis, the head, trunk, and limb
skeletal muscles develop as separate lineages. Of the
three germ layers in the early embryo, the paraxial
mesoderm gives rise to the somite (Fig. 1A). The somite
is subdivided into the dorsomedial (epaxial) domain,
which generates the muscles of the back, and the
ventrolateral (hypaxial) domain, which gives rise to the
abdominal, intercostal, and limb musculature (see
Refs. 123 and 140 for review).

Fig. 1. Derivation of muscle precursor


cells during mouse embryogenesis. A:
precursor cells from the epaxial region
of the somite migrate to form the back
musculature. Precursor cells from the
hypaxial region of the somite migrate
to the newly formed limb buds. Note
the migrating limb precursor cells form
dorsal and ventral masses, which will
later become the extensor and flexor
muscle groups of the limb. Surrounding structures such as the neural tube,
notochord, dorsal aorta, and overlying
ectoderm potentially provide signals
regulating precursor cell movements
and fate. B: members of the MyoD family play an integral role in skeletal
muscle myogenesis. MyoD and my f 5
expression is involved in determination
of precursor cells to a myogenic fate,
whereas myogenin and MRF4 expression is associated with terminal
differentiation.

J Appl Physiol VOL

91 AUGUST 2001

www.jap.org

536

INVITED REVIEW

During somitogenesis, cross talk involving growth


factors (Wnt proteins, Sonic hedge hog, bone morphogenic proteins, and so forth) and transcription factors
(myf 5, MyoD, Pax-3, and so forth) occurs between the
developing somite and the anatomically adjacent structures, including the overlying ectoderm, the ventromedial neurotube and notochord, and the vascular structures including the aorta (38, 113, 123, 131, 145). The
constellation of these positional cues (i.e., growth and
transcription factors) results in the specification of
muscle through the regulation of a distinct molecular
(hierarchical) cascade.
Since the discovery of MyoD in 1987, the role of the
myogenic basic helix-loop-helix (bHLH) transcription factors in skeletal myogenesis has been defined
in elegant detail by several groups (19, 126, 137, 147,
148, 192, 200, 207). This subset of the bHLH family
includes MyoD, my f 5, myogenin, and MRF4. Each of
these myogenic bHLH proteins forms heterodimeric
DNA binding complexes that include other bHLH
proteins of the E2 gene family (E12 and E47) and
bind a canonical DNA sequence, CANNTG (E-box),
within enhancer elements of genes that encode terminal differentiation markers of the skeletal muscle
lineage (41, 103). MyoD family members share the
ability to activate skeletal muscle differentiation
when expressed ectopically in nonmuscle cells (115,
193). The essential role played by bHLH proteins in
skeletal myogenesis has been demonstrated unambiguously by gene disruption experiments (78, 138,
142, 147, 148, 157, 185). The results of these gene
knockout experiments support a role for my f 5 and
MyoD in the determination of the myogenic cell fate
and the formation of myoblasts during embryogenesis (Fig. 1B). Myogenin and MRF4 appear to function
in activation of muscle differentiation (149, 175).
Although a number of biochemical and transgenic
studies support an integral role for the myogenic
bHLH proteins during development, it is also clear
that the MyoD family members interact in a combinatorial fashion with known transcription factors
such as members of the MADS box family (i.e., myocyte enhancer factor 2 or MEF-2; Refs. 41, 103), cell
cycle regulatory proteins, and currently undefined
factors to regulate myogenesis (see Refs. 126 and 175
for review). Although the role of MyoD family members during embryogenesis has been defined in great
detail, the functional role of these family members in
established postnatal skeletal muscle remains unclear. Studies support the localization of MyoD and
myogenin in fast-twitch and slow-twitch adult myofibers, respectively, suggesting a function for these
myogenic regulatory factors in fiber-specific contractile protein gene expression (84, 85, 171). Future
studies utilizing transgenic technologies such as
conditional or tissue-restricted knockout strategies
of these myogenic regulatory factors will be useful
in the definition of their role in adult skeletal
muscle.
J Appl Physiol VOL

MUSCLE PRECURSOR CELLS AND LIMB


DEVELOPMENT

Whereas the cells associated with the epaxial region


of the somite contribute to the primary myocytes of the
myotome, the hypaxial region of the somite (at the level
of the limb) contributes to the migratory limb muscle
precursor cells (141). Specification, migration, and differentiation of the myogenic precursor cells to their
distal targets are complex processes involving intrinsic
and extrinsic cues of which little is known. Recent
studies have shown that these myogenic precursor
cells express the paired domain transcription factor
Pax-3 (181), the tyrosine kinase receptor c-Met (205),
the homeodomain transcriptional repressor msx1 (11,
189), and the homeodomain transcription factor Lbx 1
(17, 71) but lack expression of the myogenic regulatory
factors of the MyoD family. After migration to the
developing limb, these precursor cells coalesce into the
dorsal and ventral premuscle masses, which will be the
future flexor and extensor compartments of the forelimb and now express members of the MyoD family (19,
137, 141).
Targeted gene disruption studies (i.e., knockout
mouse models) have begun to provide insight into the
molecular regulation of limb development. In mice
lacking either Pax-3 (16, 60) or c-Met (50), limb precursor cells fail to migrate into the limb, resulting in
the complete loss of limb muscles. Combinatorial
knockout experiments crossing the Pax-3 mutant
mouse (Splotch) into the myf 5 null background result
in a further perturbation of myogenesis and an absence
of both limb and body wall muscle (181). Additional
studies support a genetic hierarchy where Pax-3 mediates the activation of other myogenic regulatory factors
(myf 5 and MyoD) and functions as a key regulator of
somitic myogenesis (113, 181). In addition, Pax-3 functions in the specification of the limb precursor cells and
is upstream of both c-Met and Lbx 1 (17, 50, 60, 181).
Inactivation of the Lbx 1 locus by homologous recombination results in an extensive loss of limb muscles,
although residual muscle groups are still present. This
finding suggests that Lbx 1 is not required for the
specification of limb muscles but may function in the
determination of which migratory highway the precursors should pursue (17, 71).
Proliferating limb myoblasts coalesce into the ventral and dorsal premuscle masses, withdraw from the
cell cycle, and form multinucleated primary myofibers
at approximately embryonic day 13 postcoitum (E13)
in the mouse embryo. In a process that is less clearly
defined, secondary myofibers form parallel to the primary fibers and constitute the predominant multinucleated myofibers during the latter stages of embryogenesis (E15E16 in the mouse) and in postnatal
skeletal muscle (56, 123). Studies suggest that two
distinct lineages generate primary myofibers (i.e., embryonic myoblasts) and the secondary myofibers (i.e.,
fetal myoblasts). Furthermore, primary myofibers differ from secondary myofibers in their temporal devel-

91 AUGUST 2001

www.jap.org

537

INVITED REVIEW

opment, size, number, and expression of myosin heavychain isoforms (123, 175).
The discovery of satellite cells in the early 1960s
demonstrated the existence of yet an additional population of proliferative cells that contributed to postnatal growth, the maintenance of adult skeletal muscle,
and the repair of damaged myofibers. Myogenic satellite cells are present in the limbs of midgestational
mouse embryos after E15 (35). After birth, the satellite
cell population accounts for 30% of sublaminar muscle nuclei in neonatal hindlimb skeletal muscle (12).
These neonatal satellite cells fuse to growing myofibers
to contribute additional nuclei during postnatal growth
of skeletal muscle.
SOMITIC VS. NONSOMITIC ORIGIN OF SATELLITE
CELLS

Previous studies support the hypothesis that muscle


precursor cells, including the myogenic satellite cell
population, originate from the multipotential mesodermal cells of the somite (58, 141, 167). The support for
this hypothesis is primarily derived from chimeric or
interspecies grafting experiments that have been performed in avian models. These fate-mapping studies
involved the transplantation (or exchange) of embryonic somites from donor quail embryos into host chick
embryos (27, 105). The transplanted quail cells have
distinguishing morphological characteristics and were
observed to migrate from the somite and contribute to
both the limb muscles and the satellite cell population
in postnatal chick skeletal muscle. Satellite cells have
been isolated from the fetal skeletal muscle from an
E15 or older mouse embryo, suggesting that satellite
cells populate the developing limb during the latter
stages of embryogenesis (34, 35, 36). Whether the satellite cells migrate from the somite as a distinct lineage
or whether they originate from a preexisting lineage
(i.e., embryonic or fetal myoblasts) in the developing
limb is unclear. Nevertheless, the underlying concept
was that each of the myoblast precursor cells (i.e.,
embryonic myoblasts, fetal myoblasts, and satellite
cells) was a derivative of the somite.
This paradigm has recently been challenged, as studies have suggested that multipotential cells of nonsomitic origin may be the precursors of the satellite cell
(45, 139). De Angelis et al. (45) reported that cells
isolated from the embryonic dorsal aorta had a similar
morphological appearance and a similar profile of gene
expression to that of satellite cells. Furthermore,
transplantation of the aorta-derived myogenic cells
into newborn mice revealed that this cell population
participated in postnatal muscle growth, regeneration,
and fusion with resident satellite cells. The authors
proposed that satellite cells may be derived from endothelial cells or a precursor common to both the satellite
cell and the endothelial cell.
Derivation of the satellite cell from the somite or a
nonsomitic source need not be mutually exclusive. Conceivably, both lineages may contribute under physiological or pathological states to the satellite cell popuJ Appl Physiol VOL

lation. Additional fate-mapping strategies will be


needed to further define and dissect the lineage derivation of all the satellite cells that reside in adult
skeletal muscle. These studies will be important in
defining whether there is a common lineage source for
the entire satellite cell population and a common lineage source for cells that have regenerative capacities
in both muscle and nonmuscle tissues.
SATELLITE CELL IDENTIFICATION

Anatomic identification. Resident within adult skeletal muscle is a pool of undifferentiated mononuclear
cells termed satellite cells because of their anatomic
location at the periphery of the mature, multinucleated
myotube. The defining characteristic of the satellite
cell is that the basal lamina that surrounds the satellite cell and the associated myofiber is continuous
(167). As shown in Fig. 2, the identification of this cell
population has historically utilized ultrastructural
techniques (66, 161). Other distinguishing morphological features of the satellite cell population include a
relatively high nuclear-to-cytoplasmic ratio with few
organelles, a smaller nuclear size compared with the
adjacent nucleus of the myotube, and an increase in
the amount of nuclear heterochromatin compared with
that of the myonucleus (167). These morphological
features are consistent with the finding that satellite
cells are relatively quiescent and transcriptionally less
active. These distinguishing features are absent following activation or proliferation of the satellite cells in
response to growth, remodeling, or muscle injury. After
activation, the satellite cells are more easily identified
as they appear as a swelling on the myofiber with
cytoplasmic processes that extend from one or both
poles of the cell (Fig. 2; Ref. 167). Associated with the
increase in mitotic activity, there is a reduction in
heterochromatin, an increase in cytoplasmic-to-nuclear ratio, and an increase in the number of intracellular organelles (167).
Satellite cell markers. The profile of gene expression
of the quiescent satellite cell as well as their activated
and proliferating progeny is largely unknown. The
quiescent satellite cells do not express myogenic regulatory factors of the MyoD or MEF2 families or other
known markers of terminal differentiation (33, 119,
201). Through the identification of satellite cell markers, biologists will be able to address issues related to
the developmental origin of the satellite cell, the cell
cycle control, and the molecular regulation of this
unique cell population during growth and regeneration. Several satellite cell markers have been identified
and are restricted to either the quiescent, activated, or
proliferative state or are expressed more broadly (Table 1).
We have previously determined that myocyte nuclear factor (MNF or Foxk1), a member of the winged
helix transcription factor family, is localized to the
quiescent satellite cell in adult skeletal muscle (64).
We have identified two alternatively spliced isoforms
for MNF and termed them MNF- and MNF- (64,

91 AUGUST 2001

www.jap.org

538

INVITED REVIEW

Fig. 2. Satellite cells occupy a sublaminar position in adult skeletal muscle.


In the uninjured muscle fiber, the satellite cell is quiescent and rests in an
indentation in the adult muscle fiber.
The satellite cells can be distinguished
from the myonuclei by a surrounding
basal lamina and more abundant heterochromatin. When the fiber becomes
injured, the satellite cells become activated and increase their cytoplasmic
content. The cytoplasmic processes allow for chemotaxis of the satellite cell
along the myofiber. Bar 1 m.

203, 204). These two alternatively spliced isoforms are


reciprocally expressed during myogenesis and during
muscle regeneration, suggesting that the two isoforms
of MNF may exert opposing effects on target genes at
discrete steps during muscle regeneration or in renewal of the satellite cell population (63). Using a
RT-PCR assay, we have shown that MNF- is the
principal form expressed in quiescent satellite cells,
whereas MNF- predominates in proliferating satellite
cells following muscle injury. Disruption of the MNF
locus, mutating both isoforms, resulted in a severe
Table 1. Expression patterns of satellite cell markers
in adult skeletal muscle
Molecular Marker

MNF
Pax7
c-Met
M-cadherin
NCAM
VCAM-1
Desmin
myf5
MyoD
BrdU
PCNA
[3H]thymidine

Expression Observed in the Adult

Quiescent, activated, and proliferating


satellite cells
Quiescent, activated, and proliferating
satellite cells
Quiescent, activated, and proliferating
satellite cells
Quiescent, activated, and proliferating
satellite cells
Quiescent, activated, and proliferating
satellite cells; synaptic junctions in
adult myofibers
Quiescent, activated, and proliferating
satellite cells
Activated and proliferating satellite
cells
Activated and proliferating satellite
cells
Activated and proliferating satellite
cells
Proliferating cells
Proliferating cells
Proliferating cells

Reference

64
169
33
33
39
89
15
33
33, 124
163
90
128

Expression of selected molecular markers used to identify the


satellite cell population in adult skeletal muscle is outlined. MNF,
myocyte nuclear factor; NCAM and VCAM, neural cell and vascular
adhesion molecule; BrdU, bromodeoxyuridine; PCNA, proliferating
cell nuclear antigen.
J Appl Physiol VOL

growth deficit, a marked impairment in muscle regeneration (63), and a decreased number of satellite cells
in adult MNF mutant skeletal muscle (Hawke and
Garry, unpublished observations). Additional winged
helix family members have also been identified in stem
cells or regenerating cells including Genesis (Foxd3;
Ref. 83), which is expressed selectively in embryonic
stem cells, and a protein related to hepatocyte nuclear
factor-3 (HNF3/forkhead homolog 11), which has been
identified in regenerating hepatocytes (206).
Utilizing single cell RT-PCR analysis, Cornelison
and Wold (33) characterized the satellite cells as a
heterogeneous population based on their profile of
gene expression. Additionally, they identified c-Met,
the receptor for hepatocyte growth factor (HGF), as a
marker of quiescent satellite cells. HGF is a potent
mitogen for satellite cells and has been shown to be
important in the migration of the myogenic precursor cells from the somite to the developing limb (5,
14). Moreover, c-Met deficient embryos fail to form
limb skeletal muscle due to a lack of myogenic precursor cells (14, 112).
Irintchev and colleagues (87) identified M-cadherin,
a calcium-dependent cell adhesion molecule, as a
unique marker of the satellite cell pool. M-cadherin is
only expressed in a subpopulation of the quiescent cell
pool; however, its expression is increased when the
satellite cells become activated in response to a stimulus (9, 33). Recent studies suggest that other cell
adhesion molecules, neural cell adhesion molecule
(NCAM) and vascular adhesion molecule-1 (VCAM-1),
are also potential markers of quiescent satellite cells
(39, 89). The role of these adhesion molecules is unclear
but collectively (NCAM, VCAM-1, and M-cadherin)
may function in the adhesion of the satellite cell to the
basal lamina of the myofiber and may participate in
the migratory capacity of this cell population in response to stimuli. NCAM is expressed in both myofibers and satellite cells, whereas VCAM-1 is broadly

91 AUGUST 2001

www.jap.org

539

INVITED REVIEW

expressed during embryogenesis but limited to satellite cells in adult muscle (39). Furthermore, VCAM-1
has been shown to mediate satellite cell interaction
with leukocytes following injury (89).
Recent work from the Rudnicki laboratory (169)
identified the paired box transcription factor, Pax7,
expressed selectively in quiescent and proliferating
satellite cells. Analysis of the Pax7 mutant skeletal
muscle revealed a complete absence of satellite cells.
This novel finding supports the hypothesis that Pax7 is
essential for the specification of the satellite cell population. Future studies will be important in the definition of Pax7 downstream target genes in the satellite
cell population and may provide insight into the regulation of this cellular pool.
Identification of other satellite cell markers is the
focus of current research efforts. Emerging candidates
include the cell surface antigen Sca-1 (stem cell antigen-1; Refs. 88, 177), the glycoprotein Leu-19 (94, 162),
the anti-apoptotic factor Bcl-2 (106, 123), CD34 (9), and
interferon regulatory factor-2, which is a transcription
factor that mediates VCAM-1 expression in skeletal
muscle (89).
SATELLITE CELL QUANTITATION AND DISTRIBUTION

Quantitation of the satellite cell population in adult


skeletal muscle has been possible primarily through
the use of ultrastructural techniques. More recently,

immunohistochemical techniques have been utilized


for the identification of the satellite cell pool. Although
a limited number of markers for the quiescent satellite
cell population exist, proliferating satellite cells, as
measured by [3H]thymidine or bromodeoxyuridine
(BrdU) incorporation, can be identified immunohistochemically for the coexpression of the MyoD family
members or the intermediate filament protein, desmin
(15, 104, 123). MyoD expression occurs early during
the activation of the satellite cell population (within 6 h
following muscle injury) (63, 74, 100, 119, 134). In
addition, several nonselective markers of cellular proliferation have been used to characterize the proliferating satellite cell pool. These markers of cellular proliferation include proliferating cell nuclear antigen
(90), BrdU (163), and [3H]thymidine (128).
Satellite cell number is dependent on the species,
age, and muscle fiber type (Table 2; see Ref. 167 for
review). Satellite cells constitute 30% of the muscle
nuclei in the neonate and decrease with age to 4% in
the adult and 2% in the senile (2930 mo) mouse
(176). The decrease in the percentage of satellite cells
with aging (i.e., senile rodent) is the result of an increase in myonuclei (oxidative and glycolytic myofibers) and a decrease in total number of satellite cells
(glycolytic myofibers) (12, 66, 167).
The satellite cell distribution between muscle groups
is a result of the heterogeneity in satellite cell content

Table 2. Satellite cell content in skeletal muscle


Model

Rat
Rat
Rat

Rat
Rat
Mouse
Mouse
Quail

Muscle

TA
Soleus
Diaphragm
LD
Soleus
Soleus
Soleus
EDL
EDL
EDL
EDL
EDL
EDL
Levator ani
Levator ani
Soleus
Soleus
Gastroc
Gastroc
LD
LD

Human
Human
Human
Pig
Lizard

Trapezius
Trapezius
Biceps brachii
Biceps brachii
Sartorius
PL
Tail
Tail

Age and Protocol

%SC

79 wk
79 wk
79 wk
adult
1 mo
1 yr
2 yr
1 mo
1 yr
2 yr
4 mo
Hypothyroid
Hypothyroid; chronic stimulation
4 mo
32 mo
8 mo
30 mo
710 days
Pax 7/
6 wk
Stretched
Control patients
DMD patients
Control (38 yr)
Resistance trained
30 mo
Werdnig-Hoffman infants
64 wk
64 wk
Lygosoma species
Anolis species

4
11
8
4.5
9.6
6.6
4.7
7.0
2.9
1.9
3.8
3.8
7.913.8
1.9
1.2
4.6
2.4
25
0
15.6
16.7
15
25
3.7
5.4
8.4
14.4
1.1
4.3
7.5
4.8

SC#/muscle

5.2 105
7.3 105
5.4 105
3.1 105
2.1 105
1.3 105

Reference

161
161
161
2
66
66
66
66
66
66
146
146
146
135
135
176
176
169
169
198
198
190
190
94
94
158
158
21
21
95
95

Satellite cell (SC) content in various species under varying physiological and pathological conditions. TA, tibialis anterior; LD, latissimus
dorsi; Gastroc, gastrocnemius; EDL, extensor digitorum longus; PL, peroneus longus; DMD, Duchenne muscular dystrophy.
J Appl Physiol VOL

91 AUGUST 2001

www.jap.org

540

INVITED REVIEW

between muscle fiber types. An increase in satellite cell


density has been demonstrated in association with the
proximity of capillaries (161), myonuclei (18, 161), and
motoneuron junctions (199). The proximity of satellite
cells to these anatomic structures suggests a permissive role in the regulation of the cellular pool. In support of this hypothesis, oxidative fibers (characterized
by increased capillary and motoneuron density compared with glycolytic myofibers) demonstrate a five to
six times greater satellite cell content (65, 161).
GROWTH FACTORS AS REGULATORS OF THE
SATELLITE CELL POPULATION

Growth Factor

Chemotactic
Ability
Proliferation Differentiation Reference

1
1
1

FGF
NE
1
1

HGF

IGF-I

The process of muscle regeneration requires the influence of growth factors and a sequence of cellular
events, which results in the regulation of the satellite
cell population (Table 3, Fig. 3; Refs. 73, 168). Many of
the studies that have examined the effect of growth
factors on satellite cell biology have utilized satellite
cell cultures. These studies have defined the effect of
growth factors alone or in combination and have provided valuable insight into the regulation of the satellite cell. Admittedly, in vitro studies are limited due to
the lack of permissive and repressive factors that are
present in vivo and may influence cellular activity.
Currently, most satellite cell cultures are derived from
neonatal skeletal muscle due to the abundance of satellite cells in these tissues compared with older animals (30% in young animals vs. 5% in older animals; Ref. 143). The population and age of the satellite
cell is an important consideration in cell culture preparations as the response of aged, quiescent satellite
cells to growth factor stimulation differs compared
with young, proliferating satellite cells (182). This section will provide a brief introduction of growth factors
that are important in the regulation of satellite cell
proliferation, differentiation, and motility (for review,
see Table 3).
Insulin-like growth factors. Skeletal muscle secretes
insulin-like growth factors I and II (IGF-I and IGF-II),
which are known to be important in the regulation of
insulin metabolism (3, 109, 186). In addition, these
growth factors are important in the regulation of skeletal muscle regeneration. IGF-I and IGF-II increase
satellite cell proliferation and differentiation in vitro
(Table 3). The importance of these growth factors was
demonstrated with the intramuscular administration
of IGF-I into older, injured animals. In this study,
IGF-I administration (using an osmotic minipump) resulted in enhanced satellite cell proliferation and increased muscle mass (26). Moreover, skeletal muscle
overload or eccentric exercise results in elevated IGF-I
levels, increased DNA content (suggesting an increase
in satellite cell proliferation), and a compensatory hypertrophy of skeletal muscle (1, 202).
IGF-I appears to utilize multiple signaling pathways
in the regulation of the satellite cell pool. The calcineurin/NFAT, mitogen-activated protein (MAP) kinase, and phosphatidylinositol-3-OH kinase (PI-3K)
pathways have all been implicated in satellite cell
J Appl Physiol VOL

Table 3. Factors affecting satellite cell activity

IGF-II
TGF-

1
1

Macrophages
NE
Crushed muscle/
plateletderived
extract
LIF
IL-6
PDGFAA/AB
PDGFBB
EGF

1
1
NE
1
NE
NE

2
1
1

1
1

79
3
54
13
152
13
4
124
182
79
3
54
26(t)
79
54
79
152
3
13
208
152
121
13
86(t)

79

1
1
NE
1
NE
NE
1

13
7
152
7
152
54
152
13
13
*54

1
1
1
1
1
1
1
1
1
1
1
2

1*

2
1
1

2
2

A number of factors affect satellite cell proliferation, differentiation, and chemotaxis. All studies were performed using cell culture
techniques except those denoted with a (t), which were performed
using skeletal muscle tissue. NE, no effect. FBF, HGF, IGF, TGF,
PDGF, and EGF, fibroblast, hepatocyte, insulin-like, transforming,
platelet-derived, and endothelial growth factor, respectively. LIF,
leukemia-inhibitory factor. * Stimulation of satellite cell proliferation in the presence of serum but no effect in serum-free medium. 1
and 2, Increase or decrease in effect, respectively.

proliferation (25, 32, 170). IGF-I-stimulated satellite


cell differentiation appears to be mediated through the
PI-3K pathway (32). Additional studies, utilizing genetic mouse models (i.e., transgenic overexpression or
knockout models), may further define the regulation
and signaling pathways of the IGFs and satellite cell
biology (8, 25, 132).
Hepatocyte growth factor. Hepatocyte growth factor
(HGF) is a multifunctional cytokine initially described
as a mitogen in mature hepatocytes (122). Recently,
HGF and its receptor c-Met have been localized to
satellite cells and adjacent myofibers but are absent in
the adjacent fibroblasts. In addition, HGF expression is
proportional to the degree of muscle injury (33, 174,

91 AUGUST 2001

www.jap.org

541

INVITED REVIEW

Fig. 3. Factors modulate satellite cell activity.


Growth factors and hormones are released from
a number of tissues and modulate satellite cell
activity (chemotaxis, proliferation, and differentiation). These factors utilize signaling pathways in the regulation of the satellite cell. LIF,
leukemia inhibitory factor; IL-6, interleukin-6.
EGF, FGF, HGF, IGF, PDGF, and TGF, endothelial-derived, fibroblast, hepatocyte, insulinlike, platelet-derived, and transforming growth
factors, respectively.

182). Multiple roles for HGF have been proposed for


the regulation of the satellite cell, including a role as a
potent chemotactic factor, an activator of the satellite
cell, and an inhibitor of myoblast differentiation (Table
3). HGF is capable of activating and selectively promoting satellite cell proliferation (4). Furthermore, HGF
administration attenuates satellite cell differentiation
through the transcriptional inhibition of the myogenic
regulatory factors (i.e., MyoD and myogenin) (62).
Fibroblast growth factors. Fibroblast growth factor
(FGF) has nine different isoforms (FGF-1 to FGF-9).
Although many of the FGF isoforms are broadly expressed, FGF-6 is restricted to skeletal muscle (59).
Sheehan and Allen (173) investigated in detail the role
of the FGF family on satellite cell proliferation in
culture. In these studies, it was demonstrated that
FGF-1, -2, -4, -6, and -9 stimulated cellular proliferation, whereas FGF-5, -7, and -8 had no mitogenic activity. The investigators further observed that addition
of HGF to either FGF-2, -4, -6, or -9 resulted in a
synergistic increase in satellite cell proliferation. In
addition to an increase in satellite cell proliferation,
the FGF family has also been observed to attenuate
satellite cell differentiation to myofibers (30, 91, 173,
178).
Floss et al. (59) reported that mice deficient for
FGF-6 (i.e., knockout mice at the FGF-6 locus) have
impaired satellite cell proliferation and a subsequent
defect in muscle regeneration in response to a crush
J Appl Physiol VOL

injury. In contrast, Fiore et al. (57) pursued a similar


targeting strategy to mutate the FGF-6 locus and observed an absence of defects in response to either a
crush injury or chemically induced injury (notexin).
Consequently, the functional role(s) of FGF-6 during
muscle regeneration remains unclear. Nevertheless,
these studies underscore the ability of redundant family members to compensate for one another and result
in preserved function under pathological conditions
(i.e., mouse knockout models).
The release of FGF-2 from the damaged myofibers,
like HGF, is proportional to the degree of injury (29).
FGF levels are coordinated with FGF receptor expression. When receptor expression is increased, satellite
cells propagated in culture demonstrate an increased
proliferation and decreased differentiation (160). Conversely, when receptor expression is diminished, proliferation is decreased and there is a concomitant increase in satellite cell differentiation. Interestingly,
during the period of satellite cell activation and proliferation (048 h after injury), FGF receptor (FGF-R1)
mRNA is increased fivefold, and this increase is further
enhanced in the presence of HGF (173).
The signaling pathway(s) that transduces the FGF
signal has recently been investigated with the use of
both transgenic techniques and pharmacological inhibitors (92). These studies revealed that the MAP kinase
pathway is important in transducing the FGF-induced
increase in satellite cell proliferation; however, the

91 AUGUST 2001

www.jap.org

542

INVITED REVIEW

MAP kinase signaling pathway did not mediate the


FGF-mediated repression of satellite cell differentiation.
Transforming growth factors. Transforming growth
factor- (TGF-) is the prototypical family member of
cytokines that includes bone morphogenic protein and
growth-differentiation factors. The TGF- family of
cytokines transduces their signal through the SMAD
family of proteins (see Ref. 196 for review). Generally,
the TGF- family members function to inhibit muscle
proliferation and differentiation (3, 70, 97, 99, 208) by
silencing the transcriptional activation of the MyoD
family members (115). This inhibition of differentiation by TGF-, like FGF, persists even in nonmuscle
cell lines engineered to ectopically express members of
the MyoD family (97, 115, 184). The combination of
IGF-I or FGF with TGF- was unable to alter the
TGF--induced attenuation of satellite cell differentiation; however, TGF- action had little effect on IGF-Ior FGF-mediated increases in proliferation (70). During muscle regeneration, TGF- receptor levels (TGF-
RII) and TGF ligand are reciprocally expressed, resulting in the initial promotion of cellular proliferation
followed by enhanced muscle differentiation (159).
Interleukin-6 cytokines. Leukemia inhibitory factor
(LIF) and interleukin-6 (IL-6) are members of the IL-6
family of cytokines produced by many different cells,
including myoblasts and macrophages. These cytokines share a common receptor component, and their
actions are mediated through the same signaling pathways (81, 144). Skeletal muscle regeneration after injury in LIF mutant mice is attenuated, whereas exogenous administration of LIF increased the regenerative
process and produced enlarged myofibers (101). The permissive effect of LIF was associated only with the muscle
lineage and had no effect on nonmuscle cells in skeletal
muscle (101). IL-6 promotes the degradation of necrotic
tissue, synchronizes the cell cycle of satellite cells, and
induces apoptosis of macrophages following muscle injury (22). Unlike LIF, however, IL-6 expression in injured
muscle does not increase satellite cell proliferation (96).
Collectively, this family of growth factors appears to play
an integral role in skeletal muscle regeneration.
Many other factors may be involved in the regulation
of the satellite cell in adult skeletal muscle. Nitric
oxide, platelet-derived growth factor, endothelial-derived growth factor, and testosterone have been shown
to mediate satellite cell activity (6, 93, 133). Obviously,
the regulation of satellite cells is orchestrated by numerous factors in a temporal and concentration-dependent fashion during regeneration.
Few animal studies have examined the effect of
growth factors in vivo. Chakravarthy et al. (26) observed that local IGF-I administration to atrophied
muscle increased satellite cell proliferation and muscle
mass within 2 wk. Unlike the observations with IGF-I,
the intramuscular injection of HGF, at specified intervals following skeletal muscle injury, increased satellite cell proliferation and either had no effect or impaired the rate of regeneration (124). Similarly,
administration of FGF at timed intervals and selected
J Appl Physiol VOL

dosages did not appreciably affect muscle regeneration


(125). Future studies that combine cell culture methodologies and overexpression or loss of function models
using molecular technologies will be helpful in the
definition of the role of growth factors in satellite cell
biology.
FUNCTIONAL RESPONSES OF SATELLITE CELLS
TO PHYSIOLOGICAL STIMULI

Hypertrophic stimuli. Load-induced hypertrophy


(chronic stretch, agonist ablation, tenotomy) and resistance training are physiological challenges that promote a hypertrophic response in both human and animal models (154156, 197). Hypertrophic growth of
skeletal muscle is stimulated by short bursts of muscle
activity against high resistance. Resistance training
induces muscle hypertrophy through a process of satellite cell activation, proliferation, chemotaxis, and fusion to existing myofibers to contribute to muscle
growth (Fig. 4; Ref. 167). The migratory capacity (chemotaxis) of satellite cells is dependent on the integrity
of the basal lamina. After the rupture or interruption
of the basal lamina in response to myotrauma, satellite
cells may migrate to adjacent myofibers utilizing tissue
bridges (164, 191). In response to limited myotrauma,
where no rupture of the basal lamina occurs, satellite
cells migrate from the proximal intact portion of the
myofiber, under the basal lamina, to the site of injury
to participate in the repair process (165, 167).
Exercise-induced myotrauma initiates an immune
response, resulting in the influx of macrophages into
the damaged region. After the acute insult, macrophage infiltration peaks within 48 h (186). Initially, the
role of these blood-borne macrophages was believed to
be limited to phagocytosis and the digestion of myonecrotic fibers. However, additional roles for macrophages during the early stages of muscle repair are
emerging. Macrophages are essential in the orchestration of the repair process as they secrete a collection of
cytokine factors that regulate the satellite cell pool
(133). Importantly, in the absence of a macrophage
response, muscle regeneration is absent; in the presence of an enhanced macrophage response, there is an
increase in satellite cell proliferation and differentiation (110).
In response to resistance training, myotrauma results in the release of growth factors that will, in part,
regulate the satellite cell population during regeneration (Fig. 3 and Table 3). For example, IGF-I is upregulated in response to hypertrophic signals in skeletal
muscle and promotes proliferation and fusion of the
satellite cell pool (1, 26, 186). As outlined in the previous section, additional growth factors and/or cytokines
including LIF and members of the TGF- family may
play a role in the signaling or commissioning of the
satellite cells to participate in the hypertrophic remodeling response. Although a number of questions remain
regarding the role of the satellite cell in muscle remodeling, the primary physiological consequence of the

91 AUGUST 2001

www.jap.org

543

INVITED REVIEW

Fig. 4. Satellite cell response to myotrauma.


*Skeletal muscle trauma or injury may be minor
(e.g., resistance training) or may be more extensive (e.g., toxin injection, Duchenne muscular
dystrophy). In response to an injury, satellite
cells become activated and proliferate. Some of
the satellite cells will reestablish a quiescent
satellite cell pool through a process of self-renewal. Satellite cells will migrate to the damaged
region and, depending on the severity of the
injury, fuse to the existing myofiber or align and
fuse to produce a new myofiber. In the regenerated myofiber, the newly fused satellite cell nuclei will initially be centralized but will later
migrate to assume a more peripheral location.

hypertrophic response is to produce a muscle with a


greater capacity for peak force generation.
Atrophic stimuli. Atrophy of skeletal muscle results
in a reduction in myonuclei number and can be induced
by numerous factors including denervation, hindlimb
suspension, and malnutrition (73). Atrophy and remodeling that result from muscle disuse can be produced in laboratory rodents physiologically by tail/
hindlimb suspension or immobilization of specific
muscle groups in plaster casts or pathologically
through denervation. The response of the satellite cells
appears to be pleiotropic and dependent on the atrophic stimulus.
In adolescent rats, hindlimb suspension results in an
irreversible remodeling process, including decreased
satellite cell content and an impaired proliferative capacity within 3 days of unloading in both the oxidative
slow-twitch soleus and the fast-twitch extensor digitorum longus (EDL) muscles (44, 129). Thus the atrophic
stimulus in the adolescent animal may irreversibly
alter the developmental program for myofibers to accrue nuclei even with the resumption of weight bearing. A similar reduction in the satellite cell population
was observed in adult rat hindlimbs using an immobilization model as an alternative atrophic stimulus
(188). In contrast to adolescent animals, remobilization
of the hindlimb was accompanied by increased myofiber regeneration, supporting the hypothesis that, following completion of the developmental program, adult
satellite cells are capable of activation and proliferation to repopulate atrophied muscle (129, 188).
Unlike the other forms of atrophy, denervation is a
pathological rather than physiological stimulus. Denervation produces a form of disuse atrophy that includes myofiber degeneration and is accompanied by
distinctive changes in the myonuclei and quiescent
satellite cells (102, 111, 153). In the unperturbed conJ Appl Physiol VOL

dition, there are an increased number of satellite cells


associated with the neuromuscular junction (199). It is
conceivable that neurotrophic factors are important in
satellite cell homeostasis. A number of laboratories
have reported that the percentage of satellite cells
increase (from 3 to 9%) during the initial period following denervation (118, 187). However, a prolonged period of denervation results in a significant decrease in
the percentage of satellite cells (3 to 1% following an
18-mo period of denervation). Viguie et al. (187) hypothesize that the progressive decline in the satellite
cell pool may be the result of satellite cell apoptosis.
Alternatively, denervation may result in a lack of neurotrophic input (including growth factors) that negatively impacts satellite cell function and content.
Long-term denervation has considerable clinical implications. Denervation for periods of 618 mo results
in the inability of skeletal muscle to reestablish a
preinjury functional capacity even if neuronal sprouting and regeneration occurs (180). The mechanisms for
this phenomenon are unclear, but considerable data
support the conclusion that the intact neuromuscular
junction and the denervation model mediate positive
and repressive influences, respectively, on the satellite
cell pool (187).
Aging. Recent advances have allowed biologists to
interrogate the proliferative history of a cellular population. With each cell replication, there is 100 bp lost
from the ends of eukaryotic chromosomes (47, 48, 151,
172). The ends of the chromosomes contain TTAGGG
repeats termed telomeres. The length of these telomeres reflects the number of replications of a particular cell and its proliferative capacity. Using this technology, investigators now have the ability to analyze
the proliferative history and future capacity of the
satellite cell, providing valuable insight into the effects
of aging and disease in this cell population.

91 AUGUST 2001

www.jap.org

544

INVITED REVIEW

As age progresses, there is an impairment of skeletal


muscle regeneration following injury (see Ref. 72 for
review). A decrease in satellite cell number and/or
proliferative capacity has been used to explain this
phenomena. Support for this hypothesis is observed in
rodent models, as increasing age is associated with a
decrease in satellite cell number and a reduced proliferative capacity (52, 66, 166). In contrast to the rodent
model, a decrease in human satellite cell population
and proliferative ability is observed only during the
childhood years. For example, neonatal (5-day-old) and
infant (5-mo-old) satellite cells are capable of 60 and
45 replications, respectively, whereas 9-yr-old and
60-yr-old humans are both capable of 2030 replications (151). However, as aging progresses, satellite
cells (60-yr-old skeletal muscle) fuse to form thinner,
more fragile myotubes (151). Thus, despite a normal
ability to proliferate, the satellite cells of older humans
have a reduced capacity to repopulate the myofiber
population.
The impaired regenerative response that is observed
with aging in humans thus appears to be much more
complex than satellite cell senescence alone. Results
from cross-transplantation experiments suggest that
the host environment is a critical factor in the ability of
older skeletal muscle to regenerate (23, 24). Cross
transplantation of EDL muscles between 4-mo-old and
24-mo-old rats demonstrated that mature skeletal
muscle was as capable as young skeletal muscle in
recovering from transplant and from toxin-induced injury. However, young (4-mo-old) nerve-muscle autografts functioned significantly better than old (24mo-old) nerve-muscle autografts, as determined by the
measurement of mass and maximum isometric force,
suggesting that the ability for neuronal regeneration
may be a critical factor in activating the satellite cell
response and, ultimately, regenerating the damaged
muscle (23, 24).
Other factors within the host environment affect the
efficiency of skeletal muscle regeneration as aging
progresses. A thickening of the basal lamina (176),
increased fibrosis within skeletal muscle (114), and
reduced capillary density (31) may also contribute to
impaired regeneration. Inflammatory factors (macrophages and associated cytokines) are essential for the
normal satellite cell response to injury. Aging negatively impacts the immune response, resulting in a
decrease of the inflammatory factors and macrophages
(43). In association with an impaired immune response, there are reduced serum levels of growth factors including IGF-I in aged rats and humans (150,
183). After multiple cycles of atrophy, there is no restoration of muscle mass or satellite cell proliferation in
aged rats even after 9 wk of recovery. However, local
IGF-I administration to the atrophied muscle resulted
in significant increases in mass and satellite cell proliferation within 2 wk (26).
An unresolved question with regard to satellite cells
and the aging process is whether repeated exhaustive
and/or resistance training exercise programs have a
negative impact on the long-term satellite cell content?
J Appl Physiol VOL

This question is applicable to the young population as


well. If satellite cells have a limited proliferative capacity (60 doublings), does a lifetime of intense exercise have a negative influence on their ability to regenerate skeletal muscle as aging progresses?
FUNCTIONAL RESPONSES TO DISEASE STATES

Most myopathies have a molecular mutation that


affects the structural or cytoskeletal proteins in skeletal muscle. Duchenne muscular dystrophy (DMD) is
the most common and the most devastating of the
muscular dystrophies (20, 55, 76, 82, 127). Disease
progression and death are ultimately due to a failure of
the myogenic satellite cells to maintain muscle regeneration (36, 80). DMD is a recessive X-linked disease
that results in a null mutation at the dystrophin locus
(20, 82, 127). The absence of this cytoskeletal protein
renders the muscle fiber extremely fragile. In response
to mechanical stress associated with repeated contraction, there is widespread degeneration. The satellite
cells respond to the injury by repopulating the injured
skeletal muscle with defective myofibers lacking dystrophin. This process results in continuous degeneration-regeneration cycles and ultimately exhausts the
satellite cell pool (36, 80).
Clinical symptoms are apparent by 45 yr of age in
boys with DMD (10, 20, 127). DMD patients (45 yr of
age) have been shown to undergo more skeletal muscle
regeneration than that measured in a total of six normal patients over 60 yr of age (46). These results were
confirmed by Renault et al. (151), who demonstrated
that the proliferative life span of satellite cells derived
from a 9-yr-old DMD patient was approximately onethird of an age-matched control. Proliferative fatigue
or senescence of the satellite cell population and the
milieu of the DMD skeletal muscle may collectively
impair the proliferative or regenerative capacity of this
cell population. In the DMD patient, increased levels of
IGF binding proteins (IGFBP) are released by fibroblasts. The elevated IGFBP sequesters IGF-I, limiting
its bioavailability for satellite cells and ultimately resulting in increased skeletal muscle fibrosis (120). The
evidence to date demonstrates the tremendous strain
and ultimate failure of the satellite cell population to
adequately compensate for the persistent degeneration-regeneration process that is occurring in the DMD
skeletal muscle.
GENETIC MOUSE MODELS OF MYOPATHY

A number of gene knockout mouse models and experimental methods are available for studies of muscle
regeneration and satellite cell biology. Although the
mdx mouse, which lacks dystrophin, has provided important insights into the pathophysiology of DMD, the
myopathy in these animals does not represent the
myopathic process of DMD in humans (75). The mdx
mice have a normal life span, a temporally restricted
myofiber degeneration, and adapt to muscle degeneration with an expansion of the satellite cell pool and
muscle hypertrophy, thereby avoiding the compro-

91 AUGUST 2001

www.jap.org

545

INVITED REVIEW

mised muscle function that afflicts humans who lack


dystrophin (51, 69, 116, 179). In addition to the spontaneous dystrophin mutation in the mdx mouse, myopathic mouse models include genetically engineered
mouse strains with knockouts of utrophin (49, 69),
MyoD (119), or MNF (63) crossed into the mdx background. Each of these double mutant mouse models
exhibit features that more closely resemble DMD in
humans, including a severe myopathy, an impaired
regenerative capacity, and a decreased life span.
MUSCLE REGENERATION MODELS

An alternative strategy that has been successfully


used for the study of satellite cell activation, proliferation, regeneration, and self-renewal is to experimentally produce a controlled skeletal muscle injury. Strategies including crush (101), freeze (40), or chemically
induced injury (42, 63) have all been successfully used

to study satellite cell biology. Perhaps the most extensive and reproducible muscle injury is the delivery of
cardiotoxin (purified from the venom of the Naja nigricollis snake) into the hindlimb skeletal muscle of the
mouse (53, 63, 134). The intramuscular injection of 100
l of 10 M cardiotoxin into the gastrocnemius muscle
results in 8090% muscle degeneration (Fig. 5). After
cardiotoxin-induced injury, satellite cells become activated within 6 h of injury (Garry, unpublished observations). In response to locally released growth factors
from injured myotubes and macrophages, the satellite
cells proliferate extensively within 23 days of injury
(63, 64). Approximately 5 days after injury, the satellite cells withdraw from the cell cycle and either selfrenew or form differentiated myotubes that contain a
central nucleus (63, 64). With the use of this cardiotoxin-induced injury protocol, the architecture of the
injured muscle is largely restored within 10 days after

Fig. 5. Skeletal muscle response to toxin-induced injury. A: transverse section


of the adult gastrocnemius muscle
stained with hematoxylin and eosin at
defined intervals following cardiotoxininduced injury. After cardiotoxin delivery, there is evidence of extensive myonecrosis and edema of the myofibers
(12 h; denoted by a). A hypercellular
response (proliferating satellite cells
and inflammatory cells) is observed
within 2 days of injury. Muscle regeneration is evident within 5 days of injury. At this time, newly regenerated
myofibers are evident as small, basophilic, central-nucleated myofibers (denoted by b). The architecture of the
muscle is largely restored within 10
days following injury. The newly regenerated myofiber displays numerous
centrally aligned nuclei, demonstrating the fusion of many satellite cells to
form a single myofiber (hyperplasia) as
denoted by the myofiber designated c.
B: schematic diagram emphasizing the
temporal pattern of satellite cell proliferation and muscle differentiation following a chemically induced injury of
adult mouse skeletal muscle. Bar
100 m.

J Appl Physiol VOL

91 AUGUST 2001

www.jap.org

546

INVITED REVIEW

injury (Fig. 5). The complete profile of intrinsic and


extrinsic cues that regulate the satellite cell population
during muscle regeneration remains unclear. Therefore, the use of reproducible experimental injuries such
as cardiotoxin-induced injury will be important to evaluate and define the regulation of the satellite cell
population in molecular and physiological myopathic
models. Additional myonecrotic agents such as notexin
have been used with similar success (107), resulting in
a well-characterized regenerative response in skeletal
muscle.
SATELLITE CELLS AS MUSCLE PRECURSOR CELLS OR
STEM CELLS

Myogenic satellite cells have a tremendous proliferative capacity and are capable of self-renewal. These
tissue-specific progenitor cells or satellite cells are important in the maintenance and regeneration of skeletal muscle. Progenitor cells that are resident in other
adult tissues have stem cell characteristics, as they are
capable of self-renewal and multilineage differentiation along a specified molecular pathway (61, 194, 195).
For example, Clarke et al. (28) revealed that neural
stem cells isolated from the adult mouse brain had a
very broad developmental capacity and could contribute to virtually all tissues when delivered into the
developing embryo. These results suggest that adult
stem cells or progenitor cells are capable of dedetermination and/or transdifferentiation to adopt alternative
lineages in a permissive environment. Unlike the neural stem cell population, the potential of the satellite
cell pool in adult skeletal muscle remains incompletely
defined. Further definition of the gene expression profile of the satellite cell population and an enhanced
understanding of the molecular events responsible for
satellite cell development, activation, proliferation,
and self-renewal will define the stem cell characteristics of the satellite cell.
SIDE POPULATION CELLS

Recent studies have identified a population of pluripotent stem cells from adult skeletal muscle termed
side population (SP) cells. Skeletal muscle- and bone
marrow-derived SP cells are isolated using a DNAbinding dye (Hoechst 33342) and dual-wavelength flow
cytometric analysis (67, 68, 77). This method, which
relies on the differential ability of the SP cells to efflux
the Hoechst dye, defines a small and homogeneous
population of cells that can adopt alternative fates in
permissive environments (77, 88). For example, Gussoni et al. (77) demonstrated that SP cells isolated from
adult bone marrow were able to reconstitute the irradiated mdx mouse bone marrow, and later these cells
were recruited from the bone marrow to participate in
muscle repair. Furthermore, Jackson et al. (88) utilized
the same protocol to isolate muscle SP cells from adult
mice and observed that as few as 100 SP cells could
reconstitute the entire bone marrow of a lethally irradiated mouse. These results demonstrate that adult
skeletal muscle contains a stem cell population that
J Appl Physiol VOL

can be purified based on the exclusion of Hoechst dye


and can adopt alternative fates. Further studies are
necessary to determine whether the SP cells are satellite cell progenitors, a subpopulation of the satellite
cells, or an independent progenitor cell population that
are resident in skeletal muscle.
Alternative sources of proliferating myoblasts have
recently been described. Odelberg et al. (136) recently
reported that terminally differentiated myotubes are
capable of dedifferentiation to form myoblasts when
exposed to the homeobox-containing transcriptional repressor, msx1. Furthermore, in response to appropriate cues (i.e., conditioned media), these dedifferentiated myoblasts have the capacity to adopt alternative
phenotypes. Collectively, these studies are challenging
the established developmental paradigms regarding
satellite cell biology and the plasticity or the potential
of the terminally differentiated myotube for dedifferentiation. Further studies are necessary to define the
molecular regulation of this dedifferentiation process
but clearly have applications for the treatment of myopathies including DMD.
FUTURE PROSPECTS

Since the initial description of the satellite cell approximately 40 years ago, a number of anatomic and
physiological studies have established the importance
of this cellular pool in the growth, remodeling, and
regeneration of adult skeletal muscle. The profile of
gene expression that regulates the cell cycle progression of the satellite cell from a quiescent to an activated
and proliferating state remains ill-defined. Despite its
complexity, future challenges for this field will include
the definition of the development, maintenance, selfrenewal, and potentiality of the satellite cell population. These challenges will require an integration of
each of the biological disciplines, including the use of
increasingly powerful molecular biological tools.
The authors recognize the research efforts of those included and
those omitted (due only to space considerations) who have contributed to this field of study. Special thanks to Drs. M. I. Lindinger, P.
Mammen, and R. S. Williams for critical review of this manuscript.
The authors work is supported by grants from the Muscular
Dystrophy Association, March of Dimes, Doris Duke Charitable
Foundation, Texas Advanced Research (Technology) Program, and
the Donald W. Reynolds Foundation.
REFERENCES
1. Adams GR, Haddad F, and Baldwin KM. Time course of
changes in markers of myogenesis in overloaded rat skeletal
muscles. J Appl Physiol 87: 17051712, 1999.
2. Allam Ali M. Satellite cells in the latissimus dorsi of the rat.
Acta Anat (Basel) 110: 287290, 1981.
3. Allen RE and Boxhorn LK. Regulation of skeletal muscle
satellite cell proliferation and differentiation by transforming
growth factor-beta, insulin-like growth factor I, and fibroblast
growth factor. J Cell Physiol 138: 311315, 1989.
4. Allen RE, Sheehan SM, Taylor RG, Kendall TL, and Rice
GM. Hepatocyte growth factor activates quiescent skeletal
muscle satellite cells in vitro. J Cell Physiol 165: 307312, 1995.
5. Andermarcher E, Surani MA, and Gherardi E. Co-expression of the HGF/SF and c-met genes during early mouse embryogenesis precedes reciprocal expression in adjacent tissues
during organogenesis. Dev Genet 18: 254266, 1996.

91 AUGUST 2001

www.jap.org

547

INVITED REVIEW
6. Anderson JE. A role for nitric oxide in muscle repair: nitric
oxide-mediated activation of muscle satellite cells. Mol Biol Cell
11: 18591874, 2000.
7. Austin L and Burgess AW. Stimulation of myoblast proliferation in culture by leukaemia inhibitory factor and other cytokines. J Neurol Sci 101: 193197, 1991.
8. Barton-Davis ER, Shoturma DI, Musaro A, Rosenthal N,
and Sweeney HL. Viral mediated expression of insulin-like
growth factor I blocks the aging-related loss of skeletal muscle
function. Proc Natl Acad Sci USA 95: 1560315607, 1998.
9. Beauchamp JR, Heslop L, Yu DS, Tajbakhsh S, Kelly RG,
Wernig A, Buckingham ME, Partridge TA, and Zammit
PS. Expression of CD34 and myf5 defines the majority of
quiescent adult skeletal muscle satellite cells. J Cell Biol 151:
12211234, 2000.
10. Bell CD and Conen PE. Histopathological changes in Duchenne muscular dystrophy. J Neurol Sci 7: 529544, 1968.
11. Bendall AJ, Ding J, Hu G, Shen MM, and Abate-Shen C.
Msx1 antagonizes the myogenic activity of Pax3 in migrating
limb muscle precursors. Development 126: 49654976, 1999.
12. Bischoff R. The satellite cell and muscle regeneration. In:
Myology, edited by Engel AG and Frazini-Armstrong C. New
York: McGraw-Hill, 1994, p. 97118.
13. Bischoff R. Chemotaxis of skeletal muscle satellite cells. Dev
Dyn 208: 505515, 1997.
14. Bladt F, Riethmacher D, Isenmann S, Aguzzi A, and
Birchmeier C. Essential role for the c-met receptor in the
migration of myogenic precursor cells into the limb bud. Nature
376: 768771, 1995.
15. Bockhold KJ, Rosenblatt JD, and Partridge TA. Aging
normal and dystrophic mouse muscle: analysis of myogenicity
in cultures of living single fibers. Muscle Nerve 21: 173183,
1998.
16. Borycki AG, Li J, Jin F, Emerson CP, and Epstein JA.
Pax3 functions in cell survival and in pax7 regulation. Development 126: 16651674, 1999.
17. Brohmann H, Jagla K, and Birchmeier C. The role of Lbx1
in migration of muscle precursor cells. Development 127: 437
445, 2000.
18. Brown SC and Stickland NC. Satellite cell content in muscles of large and small mice. J Anat 183: 9196, 1993.
19. Buckingham M. Making muscle in mammals. Trends Genet 8:
144148, 1992.
20. Burghes AH, Logan C, Hu X, Belfall B, Worton RG, and
Ray PN. A cDNA clone from the Duchenne/Becker muscular
dystrophy gene. Nature 328: 434437, 1987.
21. Campion DR, Richardson RL, Reagan JO, and Kraeling
RR. Changes in the satellite cell population during postnatal
growth of pig skeletal muscle. J Anim Sci 52: 10141018, 1981.
22. Cantini M and Carraro F. Control of cell proliferation by
macrophage-myoblast interactions. Basic Appl Myol 6: 485
489, 1996.
23. Carlson BM and Faulkner JA. Muscle transplantation between young and old rats: age of host determines recovery.
Am J Physiol Cell Physiol 256: C1262C1266, 1989.
24. Carlson BM and Faulkner JA. The regeneration of noninnervated muscle grafts and marcaine-treated muscles in young
and old rats. J Gerontol A Biol Sci Med Sci 51: B4349, 1996.
25. Chakravarthy MV, Abraha TW, Schwartz RJ, Fiorotto
ML, and Booth FW. Insulin-like growth factor-I extends in
vitro replicative life span of skeletal muscle satellite cells by
enhancing G1/S cell cycle progression via the activation of
phosphatidylinositol 3-kinase/Akt signaling pathway. J Biol
Chem 275: 3594235952, 2000.
26. Chakravarthy MV, Davis BS, and Booth FW. IGF-I restores
satellite cell proliferative potential in immobilized old skeletal
muscle. J Appl Physiol 89: 13651379, 2000.
27. Christ B, Jacob HJ, and Jacob M. Origin of wing musculature. Experimental studies on quail and chick embryos. Experientia 30: 14461449, 1974.
28. Clarke DL, Johansson CB, Wilbertz J, Veress B, Nilsson
E, Karlstrom H, Lendahl U, and Frisen J. Generalized
potential of adult neural stem cells. Science 288: 16601663,
2000.
J Appl Physiol VOL

29. Clarke MS, Khakee R, and McNeil PL. Loss of cytoplasmic


basic fibroblast growth factor from physiologically wounded
myofibers of normal and dystrophic muscle. J Cell Sci 106:
121133, 1993.
30. Clegg CH, Linkhart TA, Olwin BB, and Hauschka SD.
Growth factor control of skeletal muscle differentiation: commitment to terminal differentiation occurs in G1 phase and is
repressed by fibroblast growth factor. J Cell Biol 105: 949956,
1987.
31. Coggan AR, Spina RJ, King DS, Rogers MA, Brown M,
Nemeth PM, and Holloszy JO. Histochemical and enzymatic
comparison of the gastrocnemius muscle of young and elderly
men and women. J Gerontol B Psychol Sci Soc Sci 47: B71B76,
1992.
32. Coolican SA, Samuel DS, Ewton DZ, McWade FJ, and
Florini JR. The mitogenic and myogenic actions of insulin-like
growth factors utilize distinct signaling pathways. J Biol Chem
272: 66536662, 1997.
33. Cornelison DD and Wold BJ. Single-cell analysis of regulatory gene expression in quiescent and activated mouse skeletal
muscle satellite cells. Dev Biol 191: 270283, 1997.
34. Cossu G. Unorthodox myogenesis: possible developmental significance and implications for tissue histogenesis and regeneration. Histol Histopathol 12: 755760, 1997.
35. Cossu G, Eusebi F, Grassi F, and Wanke E. Acetylcholine
receptor channels are present in undifferentiated satellite cells
but not in embryonic myoblasts in culture. Dev Biol 123: 4350,
1987.
36. Cossu G and Mavilio F. Myogenic stem cells for the therapy
of primary myopathies: wishful thinking or therapeutic perspective? J Clin Invest 105: 16691674, 2000.
37. Cossu G and Molinaro M. Cell heterogeneity in the myogenic
lineage. Curr Top Dev Biol 23: 185208, 1987.
38. Cossu G, Tajbakhsh S, and Buckingham M. How is myogenesis initiated in the embryo? Trends Genet 12: 218223,
1996.
39. Covault J and Sanes JR. Distribution of N-CAM in synaptic
and extrasynaptic portions of developing and adult skeletal
muscle. J Cell Biol 102: 716730, 1986.
40. Creuzet S, Lescaudron L, Li Z, and Fontaine-Perus J.
MyoD, myogenin, and desmin-nls-lacZ transgene emphasize
the distinct patterns of satellite cell activation in growth and
regeneration. Exp Cell Res 243: 241253, 1998.
41. Cserjesi P and Olson EN. Myogenin induces the myocytespecific enhancer binding factor MEF-2 independently of other
muscle-specific gene products. Mol Cell Biol 11: 48544862,
1991.
42. DAlbis A, Couteaux R, Janmot C, Roulet A, and Mira JC.
Regeneration after cardiotoxin injury of innervated and denervated slow and fast muscles of mammals. Myosin isoform analysis. Eur J Biochem 174: 103110, 1988.
43. Danon D, Kowatch MA, and Roth GS. Promotion of wound
repair in old mice by local injection of macrophages. Proc Natl
Acad Sci USA 86: 20182020, 1989.
44. Darr KC and Schultz E. Hindlimb suspension suppresses
muscle growth and satellite cell proliferation. J Appl Physiol
67: 18271834, 1989.
45. De Angelis L, Berghella L, Coletta M, Lattanzi L, Zanchi
M, Cusella-De Angelis MG, Ponzetto C, and Cossu G.
Skeletal myogenic progenitors originating from embryonic dorsal aorta coexpress endothelial and myogenic markers and
contribute to postnatal muscle growth and regeneration. J Cell
Biol 147: 869878, 1999.
46. Decary S, Hamida CB, Mouly V, Barbet JP, Hentati F,
and Butler-Browne GS. Shorter telomeres in dystrophic muscle consistent with extensive regeneration in young children.
Neuromuscul Disord 10: 113120, 2000.
47. Decary S, Mouly V, and Butler-Browne GS. Telomere
length as a tool to monitor satellite cell amplification for cellmediated gene therapy. Hum Gene Ther 7: 13471350, 1996.
48. Decary S, Mouly V, Hamida CB, Sautet A, Barbet JP, and
Butler-Browne GS. Replicative potential and telomere length
in human skeletal muscle: implications for satellite cell-mediated gene therapy. Hum Gene Ther 8: 14291438, 1997.

91 AUGUST 2001

www.jap.org

548

INVITED REVIEW

49. Deconinck AE, Rafael JA, Skinner JA, Brown SC, Potter
AC, Metzinger L, Watt DJ, Dickson JG, Tinsley JM, and
Davies KE. Utrophin-dystrophin-deficient mice as a model for
Duchenne muscular dystrophy. Cell 90: 717727, 1997.
50. Dietrich S, Schubert FR, Gruss P, and Lumsden A. The
role of the notochord for epaxial myotome formation in the
mouse. Cell Mol Biol (Oxf) 45: 601616, 1999.
51. DiMario JX, Uzman A, and Strohman RC. Fiber regeneration is not persistent in dystrophic (MDX) mouse skeletal muscle. Dev Biol 148: 314321, 1991.
52. Dodson MV and Allen RE. Interaction of multiplication stimulating activity/rat insulin-like growth factor II with skeletal
muscle satellite cells during aging. Mech Ageing Dev 39: 121
128, 1987.
53. Doroshow JH, Tallent C, and Schechter JE. Ultrastructural features of adriamycin-induced skeletal and cardiac muscle toxicity. Am J Pathol 118: 288297, 1985.
54. Doumit ME, Cook DR, and Merkel RA. Fibroblast growth
factor, epidermal growth factor, insulin-like growth factors, and
platelet-derived growth factor-BB stimulate proliferation of
clonally derived porcine myogenic satellite cells. J Cell Physiol
157: 326332, 1993.
55. Engel AG, Yamamoto M, and Fischbeck KH. Comprehensive description of the clinical and laboratory features and
therapy for polymyositis, dermamyositis and inclusion body
myositis. In: Myology, edited by Engel AG and Frazini-Armstrong C. New York: McGraw-Hill, 1994, p. 11301188.
56. Feldman JL and Stockdale FE. Temporal appearance of
satellite cells during myogenesis. Dev Biol 153: 217226, 1992.
57. Fiore F, Sebille A, and Birnbaum D. Skeletal muscle regeneration is not impaired in Fgf6 / mutant mice. Biochem
Biophys Res Commun 272: 138143, 2000.
58. Fischel A. Zur entwicklung der ventralen rumpf- und extremitatenmuskulatur der Vogel und Saugetiere. Morphol Jb 23:
544561, 1895.
59. Floss T, Arnold HH, and Braun T. A role for FGF-6 in
skeletal muscle regeneration. Genes Dev 11: 20402051, 1997.
60. Franz T, Kothary R, Surani MA, Halata Z, and Grim M.
The Splotch mutation interferes with muscle development in
the limbs. Anat Embryol (Berl) 187: 153160, 1993.
61. Fuchs E and Segre JA. Stem cells: a new lease on life. Cell
100: 143155, 2000.
62. Gal-Levi R, Leshem Y, Aoki S, Nakamura T, and Halevy
O. Hepatocyte growth factor plays a dual role in regulating
skeletal muscle satellite cell proliferation and differentiation.
Biochim Biophys Acta 1402: 3951, 1998.
63. Garry DJ, Meeson A, Elterman J, Zhao Y, Yang P, BasselDuby R, and Williams RS. Myogenic stem cell function is
impaired in mice lacking the forkhead/winged helix protein
MNF. Proc Natl Acad Sci USA 97: 54165421, 2000.
64. Garry DJ, Yang Q, Bassel-Duby R, and Sanders Williams
R. Persistent expression of MNF identifies myogenic stem cells
in postnatal muscles. Dev Biol 188: 280294, 1997.
65. Gibson MC and Schultz E. The distribution of satellite cells
and their relationship to specific fiber types in soleus and
extensor digitorum longus muscles. Anat Rec 202: 329337,
1982.
66. Gibson MC and Schultz E. Age-related differences in absolute numbers of skeletal muscle satellite cells. Muscle Nerve 6:
574580, 1983.
67. Goodell MA, Brose K, Paradis G, Conner AS, and Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med 183:
17971806, 1996.
68. Goodell MA, Rosenzweig M, Kim H, Marks DF, DeMaria
M, Paradis G, Grupp SA, Sieff CA, Mulligan RC, and
Johnson RP. Dye efflux studies suggest that hematopoietic
stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species. Nat Med 3: 13371345, 1997.
69. Grady RM, Merlie JP, and Sanes JR. Subtle neuromuscular
defects in utrophin-deficient mice. J Cell Biol 136: 871882,
1997.
70. Greene EA and Allen RE. Growth factor regulation of bovine
satellite cell growth in vitro. J Anim Sci 69: 146152, 1991.
J Appl Physiol VOL

71. Gross MK, Moran-Rivard L, Velasquez T, Nakatsu MN,


Jagla K, and Goulding M. Lbx1 is required for muscle precursor migration along a lateral pathway into the limb. Development 127: 413424, 2000.
72. Grounds MD. Age-associated changes in the response of skeletal muscle cells to exercise and regeneration. Ann NY Acad Sci
854: 7891, 1998.
73. Grounds MD. Muscle regeneration: molecular aspects and
therapeutic implications. Curr Opin Neurol 12: 535543, 1999.
74. Grounds MD, Garrett KL, Lai MC, Wright WE, and Beilharz MW. Identification of skeletal muscle precursor cells in
vivo by use of MyoD1 and myogenin probes. Cell Tissue Res 267:
99104, 1992.
75. Grounds MD and McGeachie JK. Skeletal muscle regeneration after crush injury in dystrophic mdx mice: an autoradiographic study. Muscle Nerve 15: 580586, 1992.
76. Gussoni E, Blau HM, and Kunkel LM. The fate of individual
myoblasts after transplantation into muscles of DMD patients.
Nat Med 3: 970977, 1997.
77. Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan
MK, Flint AF, Kunkel LM, and Mulligan RC. Dystrophin
expression in the mdx mouse restored by stem cell transplantation. Nature 401: 390394, 1999.
78. Hasty P, Bradley A, Morris JH, Edmondson DG, Venuti
JM, Olson EN, and Klein WH. Muscle deficiency and neonatal death in mice with a targeted mutation in the myogenin
gene. Nature 364: 501506, 1993.
79. Haugk KL, Roeder RA, Garber MJ, and Schelling GT.
Regulation of muscle cell proliferation by extracts from crushed
muscle. J Anim Sci 73: 19721981, 1995.
80. Heslop L, Morgan JE, and Partridge TA. Evidence for a
myogenic stem cell that is exhausted in dystrophic muscle.
J Cell Sci 113: 22992308, 2000.
81. Hibi M, Nakajima K, and Hirano T. IL-6 cytokine family and
signal transduction: a model of the cytokine system. J Mol Med
74: 112, 1996.
82. Hoffman EP, Monaco AP, Feener CC, and Kunkel LM.
Conservation of the Duchenne muscular dystrophy gene in mice
and humans. Science 238: 347350, 1987.
83. Hromas R, Ye H, Spinella M, Dmitrovsky E, Xu D, and
Costa RH. Genesis, a Winged Helix transcriptional repressor,
has embryonic expression limited to the neural crest, and stimulates proliferation in vitro in a neural development model. Cell
Tissue Res 297: 371382, 1999.
84. Hughes SM, Koishi K, Rudnicki M, and Maggs AM. MyoD
protein is differentially accumulated in fast and slow skeletal
muscle fibres and required for normal fibre type balance in
rodents. Mech Dev 61: 151163, 1997.
85. Hughes SM, Taylor JM, Tapscott SJ, Gurley CM, Carter
WJ, and Peterson CA. Selective accumulation of MyoD and
myogenin mRNAs in fast and slow adult skeletal muscle is
controlled by innervation and hormones. Development 118:
11371147, 1993.
86. Hurme T and Kalimo H. Activation of myogenic precursor
cells after muscle injury. Med Sci Sports Exerc 24: 197205,
1992.
87. Irintchev A, Zeschnigk M, Starzinski-Powitz A, and
Wernig A. Expression pattern of M-cadherin in normal, denervated, and regenerating mouse muscles. Dev Dyn 199: 326337,
1994.
88. Jackson KA, Mi T, and Goodell MA. Hematopoietic potential
of stem cells isolated from murine skeletal muscle. Proc Natl
Acad Sci USA 96: 1448214486, 1999.
89. Jesse TL, LaChance R, Iademarco MF, and Dean DC.
Interferon regulatory factor-2 is a transcriptional activator in
muscle where It regulates expression of vascular cell adhesion
molecule-1. J Cell Biol 140: 12651276, 1998.
90. Johnson SE and Allen RE. Proliferating cell nuclear antigen
(PCNA) is expressed in activated rat skeletal muscle satellite
cells. J Cell Physiol 154: 3943, 1993.
91. Johnson SE and Allen RE. Activation of skeletal muscle
satellite cells and the role of fibroblast growth factor receptors.
Exp Cell Res 219: 449453, 1995.

91 AUGUST 2001

www.jap.org

549

INVITED REVIEW
92. Jones NC, Fedorov YV, Rosenthal RS, and Olwin BB.
ERK1/2 is required for myoblast proliferation but is dispensable for muscle gene expression and cell fusion. J Cell Physiol
186: 104115, 2001.
93. Joubert Y and Tobin C. Testosterone treatment results in
quiescent satellite cells being activated and recruited into cell
cycle in rat levator ani muscle. Dev Biol 169: 286294, 1995.
94. Kadi F and Thornell LE. Concomitant increases in myonuclear and satellite cell content in female trapezius muscle
following strength training. Histochem Cell Biol 113: 99103,
2000.
95. Kahn EB and Simpson SB. Satellite cells in mature, uninjured skeletal muscle of the lizard tail. Dev Biol 37: 219223,
1974.
96. Kami K and Senba E. Localization of leukemia inhibitory
factor and interleukin-6 messenger ribonucleic acids in regenerating rat skeletal muscle. Muscle Nerve 21: 819822, 1998.
97. Katagiri T, Akiyama S, Namiki M, Komaki M, Yamaguchi
A, Rosen V, Wozney JM, Fujisawa-Sehara A, and Suda T.
Bone morphogenetic protein-2 inhibits terminal differentiation
of myogenic cells by suppressing the transcriptional activity of
MyoD and myogenin. Exp Cell Res 230: 342351, 1997.
98. Katz FRS. The termination of the afferent nerve fiber in the
muscle spindle of the frog. Philos Trans R Soc Lond B Biol Sci
243: 221225, 1961.
99. Kocamis H, McFarland DC, and Killefer J. Temporal expression of growth factor genes during myogenesis of satellite
cells derived from the biceps femoris and pectoralis major
muscles of the chicken. J Cell Physiol 186: 146152, 2001.
100. Koishi K, Zhang M, McLennan IS, and Harris AJ. MyoD
protein accumulates in satellite cells and is neurally regulated
in regenerating myotubes and skeletal muscle fibers. Dev Dyn
202: 244254, 1995.
101. Kurek JB, Bower JJ, Romanella M, Koentgen F, Murphy
M, and Austin L. The role of leukemia inhibitory factor in
skeletal muscle regeneration. Muscle Nerve 20: 815822, 1997.
102. Kuschel R, Yablonka-Reuveni Z, and Bornemann A. Satellite cells on isolated myofibers from normal and denervated
adult rat muscle. J Histochem Cytochem 47: 13751384, 1999.
103. Lassar AB, Davis RL, Wright WE, Kadesch T, Murre C,
Voronova A, Baltimore D, and Weintraub H. Functional
activity of myogenic HLH proteins requires hetero-oligomerization with E12/E47-like proteins in vivo. Cell 66: 305315, 1991.
104. Lawson-Smith MJ and McGeachie JK. The identification of
myogenic cells in skeletal muscle, with emphasis on the use of
tritiated thymidine autoradiography and desmin antibodies. J
Anat 192: 161171, 1998.
105. Le Douarin N and Barq G. Use of Japanese quail cells as
biological markers in experimental embryology. CR Acad Sci
Hebd Seances Acad Sci D 269: 15431546, 1969.
106. Lee JY, Qu-Petersen Z, Cao B, Kimura S, Jankowski R,
Cummins J, Usas A, Gates C, Robbins P, Wernig A, and
Huard J. Clonal isolation of muscle-derived cells capable of
enhancing muscle regeneration and bone healing. J Cell Biol
150: 10851100, 2000.
107. Lefaucheur JP and Sebille A. The cellular events of injured
muscle regeneration depend on the nature of the injury. Neuromuscul Disord 5: 501509, 1995.
108. Lemischka I. The power of stem cells reconsidered? Proc Natl
Acad Sci USA 96: 1419314195, 1999.
109. LeRoith D, McGuinness M, Shemer J, Stannard B, Lanau
F, Faria TN, Kato H, Werner H, Adamo M, and Roberts
CT. Insulin-like growth factors. Biol Signals 1: 173181, 1992.
110. Lescaudron L, Peltekian E, Fontaine-Perus J, Paulin D,
Zampieri M, Garcia L, and Parrish E. Blood borne macrophages are essential for the triggering of muscle regeneration
following muscle transplant. Neuromuscul Disord 9: 7280,
1999.
111. Lu DX, Huang SK, and Carlson BM. Electron microscopic
study of long-term denervated rat skeletal muscle. Anat Rec
248: 355365, 1997.
112. Maina F, Casagranda F, Audero E, Simeone A, Comoglio
PM, Klein R, and Ponzetto C. Uncoupling of Grb2 from the
J Appl Physiol VOL

113.

114.

115.

116.

117.
118.
119.
120.

121.

122.
123.
124.

125.

126.
127.

128.
129.
130.
131.

132.

133.
134.

Met receptor in vivo reveals complex roles in muscle development. Cell 87: 531542, 1996.
Maroto M, Reshef R, Munsterberg AE, Koester S, Goulding M, and Lassar AB. Ectopic Pax-3 activates MyoD and
Myf-5 expression in embryonic mesoderm and neural tissue.
Cell 89: 139148, 1997.
Marshall PA, Williams PE, and Goldspink G. Accumulation of collagen and altered fiber-type ratios as indicators of
abnormal muscle gene expression in the mdx dystrophic mouse.
Muscle Nerve 12: 528537, 1989.
Martin JF, Li L, and Olson EN. Repression of myogenin
function by TGF-1 is targeted at the basic helix-loop-helix
motif and is independent of E2A products. J Biol Chem 267:
1095610960, 1992.
Matsuda R, Nishikawa A, and Tanaka H. Visualization of
dystrophic muscle fibers in mdx mouse by vital staining with
Evans blue: evidence of apoptosis in dystrophin-deficient muscle. J Biochem (Tokyo) 118: 959964, 1995.
Mauro A. Satellite cell of skeletal muscle fibers. J Biophys
Biochem Cytol 9: 493498, 1961.
McGeachie JK. Sustained cell proliferation in denervated
skeletal muscle of mice. Cell Tissue Res 257: 455457, 1989.
Megeney LA, Kablar B, Garrett K, Anderson JE, and
Rudnicki MA. MyoD is required for myogenic stem cell function in adult skeletal muscle. Genes Dev 10: 11731183, 1996.
Melone MA, Peluso G, Galderisi U, Petillo O, and Cotrufo
R. Increased expression of IGF-binding protein-5 in Duchenne
muscular dystrophy (DMD) fibroblasts correlates with the fibroblast-induced downregulation of DMD myoblast growth: an
in vitro analysis. J Cell Physiol 185: 143153, 2000.
Merly F, Lescaudron L, Rouaud T, Crossin F, and Gardahaut MF. Macrophages enhance muscle satellite cell proliferation and delay their differentiation. Muscle Nerve 22: 724
732, 1999.
Michalopoulos GK and Zarnegav R. Hepatocyte growth
factor. Hepatology 15: 149155, 1992.
Miller JB, Schaefer L, and Dominov JA. Seeking muscle
stem cells. Curr Top Dev Biol 43: 191219, 1999.
Miller KJ, Thaloor D, Matteson S, and Pavlath GK. Hepatocyte growth factor affects satellite cell activation and differentiation in regenerating skeletal muscle. Am J Physiol Cell
Physiol 278: C174C181, 2000.
Mitchell CA, McGeachie JK, and Grounds MD. The exogenous administration of basic fibroblast growth factor to regenerating skeletal muscle in mice does not enhance the process of
regeneration. Growth Factors 13: 3755, 1996.
Molkentin JD and Olson EN. Defining the regulatory networks for muscle development. Curr Opin Genet Dev 6: 445
453, 1996.
Monaco AP, Neve RL, Colletti-Feener C, Bertelson CJ,
Kurnit DM, and Kunkel LM. Isolation of candidate cDNAs
for portions of the Duchenne muscular dystrophy gene. Nature
323: 646650, 1986.
Moss FP and Leblond CP. Satellite cells as the source of
nuclei in muscles of growing rats. Anat Rec 170: 421435, 1971.
Mozdziak PE, Pulvermacher PM, and Schultz E. Unloading of juvenile muscle results in a reduced muscle size 9 wk
after reloading. J Appl Physiol 88: 158164, 2000.
Muir AR, Kanji AH, and Allbrook D. The structure of the
satellite cells in skeletal muscle. J Anat 99: 435444, 1965.
Munsterberg AE and Lassar AB. Combinatorial signals
from the neural tube, floor plate and notochord induce myogenic bHLH gene expression in the somite. Development 121:
651660, 1995.
Musaro A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, Barton ER, Sweeney HL, and
Rosenthal N. Localized Igf-1 transgene expression sustains
hypertrophy and regeneration in senescent skeletal muscle.
Nat Genet 27: 195200, 2001.
Nathan CF. Secretory products of macrophages. J Clin Invest
79: 319326, 1987.
Nicolas N, Gallien CL, and Chanoine C. Analysis of MyoD,
myogenin, and muscle-specific gene mRNAs in regenerating
Xenopus skeletal muscle. Dev Dyn 207: 6068, 1996.

91 AUGUST 2001

www.jap.org

550

INVITED REVIEW

135. Nnodim JO. Satellite cell numbers in senile rat levator ani
muscle. Mech Ageing Dev 112: 99111, 2000.
136. Odelberg SJ, Kollhoff A, and Keating MT. Dedifferentiation of mammalian myotubes induced by msx1. Cell 103: 1099
1109, 2000.
137. Olson EN. MyoD family: a paradigm for development? Genes
Dev 4: 14541461, 1990.
138. Olson EN, Brown D, Burgess R, and Cserjesi P. A new
subclass of helix-loop-helix transcription factors expressed in
paraxial mesoderm and chondrogenic cell lineages. Ann NY
Acad Sci 785: 108118, 1996.
139. Ordahl CP. Myogenic shape-shifters. J Cell Biol 147: 695698,
1999.
140. Ordahl CP and Le Douarin NM. Two myogenic lineages
within the developing somite. Development 114: 339353, 1992.
141. Ordahl CP, Williams BA, and Denetclaw W. Determination
and morphogenesis in myogenic progenitor cells: an experimental embryological approach. Curr Top Dev Biol 48: 319367,
2000.
142. Patapoutian A, Yoon JK, Miner JH, Wang S, Stark K, and
Wold B. Disruption of the mouse MRF4 gene identifies multiple waves of myogenesis in the myotome. Development 121:
33473358, 1995.
143. Pavlath G. Isolation, purification and growth of skeletal muscle cells. In: Methods of Molecular Medicine: Human Cell Culture Protocols, edited by Jones GE. Totawa, NJ: Humana, 1996,
p. 307318.
144. Pennica D, Shaw KJ, Swanson TA, Moore MW, Shelton
DL, Zioncheck KA, Rosenthal A, Taga T, Paoni NF, and
Wood WI. Cardiotrophin-1. Biological activities and binding to
the leukemia inhibitory factor receptor/gp130 signaling complex. J Biol Chem 270: 1091510922, 1995.
145. Pirskanen A, Kiefer JC, and Hauschka SD. IGFs, insulin,
Shh, bFGF, and TGF-1 interact synergistically to promote
somite myogenesis in vitro. Dev Biol 224: 189203, 2000.
146. Putman CT, Dusterhoft S, and Pette D. Changes in satellite
cell content and myosin isoforms in low frequency-stimulated
fast muscle of hypothyroid rat. J Appl Physiol 86: 4051, 1999.
147. Rawls A, Morris JH, Rudnicki M, Braun T, Arnold HH,
Klein WH, and Olson EN. Myogenins functions do not overlap with those of MyoD or Myf-5 during mouse embryogenesis.
Dev Biol 172: 3750, 1995.
148. Rawls A, Valdez MR, Zhang W, Richardson J, Klein WH,
and Olson EN. Overlapping functions of the myogenic bHLH
genes MRF4 and MyoD revealed in double mutant mice. Development 125: 23492358, 1998.
149. Rawls A, Wilson-Rawls J, and Olson EN. Genetic regulation
of somite formation. Curr Top Dev Biol 47: 131154, 2000.
150. Reeves I, Abribat T, Laramee P, Jasmin G, and Brazeau
P. Age-related serum levels of insulin-like growth factor-I, -II
and IGF- binding protein-3 following myocardial infarction.
Growth Horm IGF Res 10: 7884, 2000.
151. Renault V, Piron-Hamelin G, Forestier C, DiDonna S,
Decary S, Hentati F, Saillant G, Butler-Browne GS, and
Mouly V. Skeletal muscle regeneration and the mitotic clock.
Exp Gerontol 35: 711719, 2000.
152. Robertson TA, Maley MA, Grounds MD, and Papadimitriou JM. The role of macrophages in skeletal muscle regeneration with particular reference to chemotaxis. Exp Cell Res
207: 321331, 1993.
153. Rodrigues A and Schmalbruch H. Satellite cells and myonuclei in long-term denervated rat muscles. Anat Rec 243:
430437, 1995.
154. Rosenblatt J and Parry DJ. Gamma irradiation prevents
compensatory hypertrophy of overload mouse extensor digitorum longus muscle. J Appl Physiol 73: 25382543, 1992.
155. Rosenblatt JD and Parry DJ. Adaptation of rat extensor
digitorum longus muscle to gamma irradiation and overload.
Pflugers Arch 423: 255264, 1993.
156. Rosenblatt JD, Yong D, and Parry DJ. Satellite cell activity
is required for hypertrophy of overloaded adult rat muscle.
Muscle Nerve 17: 608613, 1994.
J Appl Physiol VOL

157. Rudnicki MA, Schnegelsberg PN, Stead RH, Braun T,


Arnold HH, and Jaenisch R. MyoD or Myf-5 is required for
the formation of skeletal muscle. Cell 75: 13511359, 1993.
158. Saito Y. Muscle fibre type differentiation and satellite cell
population in Werdnig-Hoffmann disease. J Neurol Sci 68:
7587, 1985.
159. Sakuma K, Watanabe K, Sano M, Kitajima S, Sakamoto
K, Uramoto I, and Totsuka T. The adaptive response of
transforming growth factor-2 and -RII in the overloaded,
regenerating and denervated muscles of rats. Acta Neuropathol
(Berl) 99: 177185, 2000.
160. Scata KA, Bernard DW, Fox J, and Swain JL. FGF receptor
availability regulates skeletal myogenesis. Exp Cell Res 250:
1021, 1999.
161. Schmalbruch H and Hellhammer U. The number of nuclei
in adult rat muscles with special reference to satellite cells.
Anat Rec 189: 169175, 1977.
162. Schubert W, Zimmermann K, Cramer M, and StarzinskiPowitz A. Lymphocyte antigen Leu-19 as a molecular marker
of regeneration in human skeletal muscle. Proc Natl Acad Sci
USA 86: 307311, 1989.
163. Schultz E. Satellite cell proliferative compartments in growing
skeletal muscles. Dev Biol 175: 8494, 1996.
164. Schultz E, Jaryszak DL, Gibson MC, and Albright DJ.
Absence of exogenous satellite cell contribution to regeneration
of frozen skeletal muscle. J Muscle Res Cell Motil 7: 361367,
1986.
165. Schultz E, Jaryszak DL, and Valliere CR. Response of
satellite cells to focal skeletal muscle injury. Muscle Nerve 8:
217222, 1985.
166. Schultz E and Lipton BH. Skeletal muscle satellite cells:
changes in proliferation potential as a function of age. Mech
Ageing Dev 20: 377383, 1982.
167. Schultz E and McCormick KM. Skeletal muscle satellite
cells. Rev Physiol Biochem Pharmacol 123: 213257, 1994.
168. Seale P and Rudnicki MA. A new look at the origin, function,
and stem-cell status of muscle satellite cells. Dev Biol 218:
115124, 2000.
169. Seale P, Sabourin LA, Girgis-Gabardo A, Mansouri A,
Gruss P, and Rudnicki MA. Pax7 is required for the specification of myogenic satellite cells. Cell 102: 777786, 2000.
170. Semsarian C, Wu MJ, Ju YK, Marciniec T, Yeoh T, Allen
DG, Harvey RP, and Graham RM. Skeletal muscle hypertrophy is mediated by a Ca2-dependent calcineurin signalling
pathway. Nature 400: 576581, 1999.
171. Seward DJ, Haney JC, Rudnicki MA, and Swoap SJ.
bHLH transcription factor MyoD affects myosin heavy chain
expression pattern in a muscle-specific fashion. Am J Physiol
Cell Physiol 280: C408C413, 2001.
172. Shay JW and Wright WE. Telomeres and telomerase: implications for cancer and aging. Radiat Res 155: 188193, 2001.
173. Sheehan SM and Allen RE. Skeletal muscle satellite cell
proliferation in response to members of the fibroblast growth
factor family and hepatocyte growth factor. J Cell Physiol 181:
499506, 1999.
174. Sheehan SM, Tatsumi R, Temm-Grove CJ, and Allen RE.
HGF is an autocrine growth factor for skeletal muscle satellite
cells in vitro. Muscle Nerve 23: 239245, 2000.
175. Smith TH, Block NE, Rhodes SJ, Konieczny SF, and
Miller JB. A unique pattern of expression of the four muscle
regulatory factor proteins distinguishes somitic from embryonic, fetal and newborn mouse myogenic cells. Development
117: 11251133, 1993.
176. Snow MH. The effects of aging on satellite cells in skeletal
muscles of mice and rats. Cell Tissue Res 185: 399408, 1977.
177. Spangrude GJ, Heimfeld S, and Weissman IL. Purification
and characterization of mouse hematopoietic stem cells. Science
241: 5862, 1988.
178. Spizz G, Roman D, Strauss A, and Olson EN. Serum and
fibroblast growth factor inhibit myogenic differentiation
through a mechanism dependent on protein synthesis and
independent of cell proliferation. J Biol Chem 261: 94839488,
1986.

91 AUGUST 2001

www.jap.org

551

INVITED REVIEW
179. Straub V, Rafael JA, Chamberlain JS, and Campbell KP.
Animal models for muscular dystrophy show different patterns
of sarcolemmal disruption. J Cell Biol 139: 375385, 1997.
180. Sunderland S. Traumatized nerves, roots and ganglia: musculoskeletal factors and neuropathological consequences. In:
The Neurobiologic Mechanisms in Manipulative Therapy, edited by Korr IM. New York: Plenum, 1978, p. 137166.
181. Tajbakhsh S, Rocancourt D, Cossu G, and Buckingham
M. Redefining the genetic hierarchies controlling skeletal myogenesis: Pax-3 and Myf-5 act upstream of MyoD. Cell 89: 127
138, 1997.
182. Tatsumi R, Anderson JE, Nevoret CJ, Halevy O, and
Allen RE. HGF/SF is present in normal adult skeletal muscle
and is capable of activating satellite cells. Dev Biol 194: 114
128, 1998.
183. Ullman M, Ullman A, Sommerland H, Skottner A, and
Oldfors A. Effects of growth hormone on muscle regeneration
and IGF-I concentration in old rats. Acta Physiol Scand 140:
521525, 1990.
184. Vaidya TB, Rhodes SJ, Taparowsky EJ, and Konieczny
SF. Fibroblast growth factor and transforming growth factor
beta repress transcription of the myogenic regulatory gene
MyoD1. Mol Cell Biol 9: 35763579, 1989.
185. Valdez MR, Richardson JA, Klein WH, and Olson EN.
Failure of Myf5 to support myogenic differentiation without
myogenin, MyoD, and MRF4. Dev Biol 219: 287298, 2000.
186. Vierck J, OReilly B, Hossner K, Antonio J, Byrne K,
Bucci L, and Dodson M. Satellite cell regulation following
myotrauma caused by resistance exercise. Cell Biol Int 24:
263272, 2000.
187. Viguie CA, Lu DX, Huang SK, Rengen H, and Carlson BM.
Quantitative study of the effects of long-term denervation on
the extensor digitorum longus muscle of the rat. Anat Rec 248:
346354, 1997.
188. Wanek LJ and Snow MH. Activity-induced fiber regeneration
in rat soleus muscle. Anat Rec 258: 176185, 2000.
189. Wang Y and Sassoon D. Ectoderm-mesenchyme and mesenchyme-mesenchyme interactions regulate Msx- 1 expression
and cellular differentiation in the murine limb bud. Dev Biol
168: 374382, 1995.
190. Watkins SC and Cullen MJ. A quantitative study of myonuclear and satellite cell nuclear size in Duchennes muscular
dystrophy, polymyositis and normal human skeletal muscle.
Anat Rec 222: 611, 1988.
191. Watt DJ, Morgan JE, Clifford MA, and Partridge TA. The
movement of muscle precursor cells between adjacent regenerating muscles in the mouse. Anat Embryol (Berl) 175: 527536,
1987.
192. Weintraub H. The MyoD family and myogenesis: redundancy,
networks, and thresholds. Cell 75: 12411244, 1993.
193. Weintraub H, Tapscott SJ, Davis RL, Thayer MJ, Adam
MA, Lassar AB, and Miller AD. Activation of muscle-specific
genes in pigment, nerve, fat, liver, and fibroblast cell lines by

J Appl Physiol VOL

194.
195.
196.
197.

198.
199.

200.
201.

202.
203.
204.

205.

206.

207.
208.

forced expression of MyoD. Proc Natl Acad Sci USA 86: 5434
5438, 1989.
Weissman IL. Stem cells: units of development, units of regeneration, and units in evolution. Cell 100: 157168, 2000.
Weissman IL. Translating stem and progenitor cell biology to
the clinic: barriers and opportunities. Science 287: 14421446,
2000.
Whitman M. Smads and early developmental signaling by the
TGF superfamily. Genes Dev 12: 24452462, 1998.
Williams RS and Neufer PD. Regulation of gene expression
in skeletal muscle by contractile activity. In: Handbook of
Physiology. Exercise: Regulation and Integration of Multiple
Systems. Bethesda, MD: Am. Physiol. Soc., 1995, sect. 12,
chapt. 27, p. 11241150.
Winchester PK and Gonyea WJ. A quantitative study of
satellite cells and myonuclei in stretched avian slow tonic
muscle. Anat Rec 232: 369377, 1992.
Wokke JH, Van den Oord CJ, Leppink GJ, and Jennekens FG. Perisynaptic satellite cells in human external intercostal muscle: a quantitative and qualitative study. Anat Rec
223: 174180, 1989.
Wright WE, Sassoon DA, and Lin VK. Myogenin, a factor
regulating myogenesis, has a domain homologous to MyoD. Cell
56: 607617, 1989.
Yablonka-Reuveni Z and Rivera AJ. Temporal expression
of regulatory and structural muscle proteins during myogenesis
of satellite cells on isolated adult rat fibers. Dev Biol 164:
588603, 1994.
Yan Z, Biggs RB, and Booth FW. Insulin-like growth factor
immunoreactivity increases in muscle after acute eccentric
contractions. J Appl Physiol 74: 410414, 1993.
Yang Q, Bassel-Duby R, and Williams RS. Transient expression of a winged-helix protein, MNF-, during myogenesis.
Mol Cell Biol 17: 52365243, 1997.
Yang Q, Kong Y, Rothermel B, Garry DJ, Bassel-Duby R,
and Williams RS. The winged-helix/forkhead protein myocyte
nuclear factor beta (MNF-) forms a co-repressor complex with
mammalian sin3B. Biochem J 345: 335343, 2000.
Yang XM, Vogan K, Gros P, and Park M. Expression of the
met receptor tyrosine kinase in muscle progenitor cells in
somites and limbs is absent in Splotch mice. Development 122:
21632171, 1996.
Ye H, Kelly TF, Samadani U, Lim L, Rubio S, Overdier
DG, Roebuck KA, and Costa RH. Hepatocyte nuclear factor
3/fork head homolog 11 is expressed in proliferating epithelial
and mesenchymal cells of embryonic and adult tissues. Mol Cell
Biol 17: 16261641, 1997.
Yun K and Wold B. Skeletal muscle determination and differentiation: story of a core regulatory network and its context.
Curr Opin Cell Biol 8: 877889, 1996.
Zentella A and Massague J. Transforming growth factor beta
induces myoblast differentiation in the presence of mitogens.
Proc Natl Acad Sci USA 89: 51765180, 1992.

91 AUGUST 2001

www.jap.org

S-ar putea să vă placă și