You are on page 1of 18

J Appl Physiol

91: 534–551, 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: 534–551,
2001.—Adult skeletal muscle has a remarkable ability to regenerate
following myotrauma. Because adult myofibers are terminally differen-
tiated, 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

SKELETAL MUSCLES OF ADULT mammalian species exhibit a (satellite cells expressing myogenic markers are also
remarkable capacity to adapt to physiological demands termed myoblasts). Ultimately, these cells fuse to ex-
such as growth, training, and injury. The processes by isting muscle fibers or fuse together to form new myo-
which these adaptations occur are largely attributed to fibers during regeneration of damaged skeletal muscle
a small population of cells that are resident in adult (12, 167).
skeletal muscle and are referred to as satellite cells. Since the original description of the myogenic sat-
After their initial identification in 1961 (98), Mauro ellite cell, considerable interest and research efforts
(117) described a cell closely associated with the pe- have focused on myogenic satellite cell biology.
riphery of the frog myofiber and termed it a satellite These research efforts have enhanced our under-
cell based on its location. Quiescent satellite cells are standing of muscle growth, remodeling, and regen-
physically distinct from the adult myofiber as they eration. In addition, new paradigms have been pro-
reside in indentations between the sarcolemma and posed regarding the regenerative capacity and the
the basal lamina (130). Adult skeletal muscle fibers are plasticity of the myogenic satellite cell population
terminally differentiated such that muscle growth and (108, 119, 139, 168, 169). These paradigms suggest
regeneration are accomplished by satellite cells. In the that the satellite cell population not only has a
unperturbed state, these cells remain in a nonprolif- remarkable capacity for muscle regeneration but
erative, quiescent state. However, in response to stim- may also contribute to alternative muscle and non-
uli such as myotrauma, satellite cells become acti- muscle lineages and may have clinical applications
vated, proliferate, and express myogenic markers in the treatment of devastating and deadly diseases
such as muscular dystrophy.
The current review attempts to integrate the ana-
Address for reprint requests and other correspondence: D. J.
Garry, UT Southwestern Medical Center at Dallas, 5323 Harry tomic, physiological, biochemical, and molecular prop-
Hines Blvd., NB11.200, Dallas, TX 75390-8573 (E-mail: erties that regulate the myogenic satellite cell popula-
daniel.garry@utsouthwestern.edu). tion. We begin with a brief overview of vertebrate
534 8750-7587/01 $5.00 Copyright © 2001 the American Physiological Society http://www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 535

myogenesis, highlighting the populations of myoblast BUILDING OF MUSCLE WITH WAVES OF PRECURSOR
precursor cells that contribute to muscle development, CELLS
and outline a well-described genetic hierarchy that is
important in muscle specification. We describe the lim- Anatomic and molecular mechanisms during muscle
ited gene expression profile and the distinguishing regeneration have been postulated to recapitulate
morphological characteristics of the satellite cell pop- muscle development. Although current evidence sug-
ulation. We describe the inductive signals that regu- gests the regenerative process may be more complex,
late the satellite cell in vitro and in well-described an understanding of muscle development is important
physiological models. Finally, we will review the stem to appreciate the anatomic and molecular network
cell-like features of the satellite cells with emphasis on associated with muscle regeneration.
the novel strategies that may be pursued in the future During embryogenesis, the head, trunk, and limb
for the treatment of debilitating myopathies. skeletal muscles develop as separate lineages. Of the
Importantly, myogenic satellite cell biology re- three germ layers in the early embryo, the paraxial
mains an emerging field of scientific inquiry, such mesoderm gives rise to the somite (Fig. 1A). The somite
that satisfying and complete answers to the most is subdivided into the dorsomedial (epaxial) domain,
fundamental questions are currently unavailable. In which generates the muscles of the back, and the
this review, we will provide a summary of the cur- ventrolateral (hypaxial) domain, which gives rise to the
rent knowledge in this area and highlight fertile abdominal, intercostal, and limb musculature (see
areas for future research. 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. Surround-
ing 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 fam-
ily 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 expres-
sion is associated with terminal
differentiation.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


536 INVITED REVIEW

During somitogenesis, cross talk involving growth MUSCLE PRECURSOR CELLS AND LIMB
factors (Wnt proteins, Sonic hedge hog, bone morpho- DEVELOPMENT
genic proteins, and so forth) and transcription factors
Whereas the cells associated with the epaxial region
(myf 5, MyoD, Pax-3, and so forth) occurs between the
of the somite contribute to the primary myocytes of the
developing somite and the anatomically adjacent struc-
myotome, the hypaxial region of the somite (at the level
tures, including the overlying ectoderm, the ventrome-
of the limb) contributes to the migratory limb muscle
dial neurotube and notochord, and the vascular struc- precursor cells (141). Specification, migration, and dif-
tures including the aorta (38, 113, 123, 131, 145). The ferentiation of the myogenic precursor cells to their
constellation of these positional cues (i.e., growth and distal targets are complex processes involving intrinsic
transcription factors) results in the specification of and extrinsic cues of which little is known. Recent
muscle through the regulation of a distinct molecular studies have shown that these myogenic precursor
(hierarchical) cascade. cells express the paired domain transcription factor
Since the discovery of MyoD in 1987, the role of the Pax-3 (181), the tyrosine kinase receptor c-Met (205),
myogenic basic helix-loop-helix (bHLH) transcrip- the homeodomain transcriptional repressor msx1 (11,
tion factors in skeletal myogenesis has been defined 189), and the homeodomain transcription factor Lbx 1
in elegant detail by several groups (19, 126, 137, 147, (17, 71) but lack expression of the myogenic regulatory
148, 192, 200, 207). This subset of the bHLH family factors of the MyoD family. After migration to the
includes MyoD, my f 5, myogenin, and MRF4. Each of developing limb, these precursor cells coalesce into the
these myogenic bHLH proteins forms heterodimeric dorsal and ventral premuscle masses, which will be the
DNA binding complexes that include other bHLH future flexor and extensor compartments of the fore-
proteins of the E2 gene family (E12 and E47) and limb and now express members of the MyoD family (19,
bind a canonical DNA sequence, CANNTG (E-box), 137, 141).
within enhancer elements of genes that encode ter- Targeted gene disruption studies (i.e., knockout
minal differentiation markers of the skeletal muscle mouse models) have begun to provide insight into the
lineage (41, 103). MyoD family members share the molecular regulation of limb development. In mice
ability to activate skeletal muscle differentiation lacking either Pax-3 (16, 60) or c-Met (50), limb pre-
when expressed ectopically in nonmuscle cells (115, cursor cells fail to migrate into the limb, resulting in
193). The essential role played by bHLH proteins in the complete loss of limb muscles. Combinatorial
skeletal myogenesis has been demonstrated unam- knockout experiments crossing the Pax-3 mutant
biguously by gene disruption experiments (78, 138, mouse (Splotch) into the myf 5 null background result
142, 147, 148, 157, 185). The results of these gene in a further perturbation of myogenesis and an absence
knockout experiments support a role for my f 5 and of both limb and body wall muscle (181). Additional
MyoD in the determination of the myogenic cell fate studies support a genetic hierarchy where Pax-3 medi-
and the formation of myoblasts during embryogene- ates the activation of other myogenic regulatory factors
sis (Fig. 1B). Myogenin and MRF4 appear to function (myf 5 and MyoD) and functions as a key regulator of
in activation of muscle differentiation (149, 175). somitic myogenesis (113, 181). In addition, Pax-3 func-
Although a number of biochemical and transgenic tions in the specification of the limb precursor cells and
studies support an integral role for the myogenic is upstream of both c-Met and Lbx 1 (17, 50, 60, 181).
bHLH proteins during development, it is also clear Inactivation of the Lbx 1 locus by homologous recom-
that the MyoD family members interact in a combi- bination results in an extensive loss of limb muscles,
natorial fashion with known transcription factors although residual muscle groups are still present. This
such as members of the MADS box family (i.e., myo- finding suggests that Lbx 1 is not required for the
cyte enhancer factor 2 or MEF-2; Refs. 41, 103), cell specification of limb muscles but may function in the
cycle regulatory proteins, and currently undefined determination of which migratory highway the precur-
factors to regulate myogenesis (see Refs. 126 and 175 sors should pursue (17, 71).
for review). Although the role of MyoD family mem- Proliferating limb myoblasts coalesce into the ven-
bers during embryogenesis has been defined in great tral and dorsal premuscle masses, withdraw from the
detail, the functional role of these family members in cell cycle, and form multinucleated primary myofibers
established postnatal skeletal muscle remains un- at approximately embryonic day 13 postcoitum (E13)
clear. Studies support the localization of MyoD and in the mouse embryo. In a process that is less clearly
myogenin in fast-twitch and slow-twitch adult myo- defined, secondary myofibers form parallel to the pri-
fibers, respectively, suggesting a function for these mary fibers and constitute the predominant multinu-
myogenic regulatory factors in fiber-specific contrac- cleated myofibers during the latter stages of embryo-
tile protein gene expression (84, 85, 171). Future genesis (E15–E16 in the mouse) and in postnatal
studies utilizing transgenic technologies such as skeletal muscle (56, 123). Studies suggest that two
conditional or tissue-restricted knockout strategies distinct lineages generate primary myofibers (i.e., em-
of these myogenic regulatory factors will be useful bryonic myoblasts) and the secondary myofibers (i.e.,
in the definition of their role in adult skeletal fetal myoblasts). Furthermore, primary myofibers dif-
muscle. fer from secondary myofibers in their temporal devel-
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 537

opment, size, number, and expression of myosin heavy- lation. Additional fate-mapping strategies will be
chain isoforms (123, 175). needed to further define and dissect the lineage deri-
The discovery of satellite cells in the early 1960s vation of all the satellite cells that reside in adult
demonstrated the existence of yet an additional popu- skeletal muscle. These studies will be important in
lation of proliferative cells that contributed to postna- defining whether there is a common lineage source for
tal growth, the maintenance of adult skeletal muscle, the entire satellite cell population and a common lin-
and the repair of damaged myofibers. Myogenic satel- eage source for cells that have regenerative capacities
lite cells are present in the limbs of midgestational in both muscle and nonmuscle tissues.
mouse embryos after E15 (35). After birth, the satellite
cell population accounts for ⬃30% of sublaminar mus- SATELLITE CELL IDENTIFICATION
cle nuclei in neonatal hindlimb skeletal muscle (12).
These neonatal satellite cells fuse to growing myofibers Anatomic identification. Resident within adult skel-
to contribute additional nuclei during postnatal growth etal muscle is a pool of undifferentiated mononuclear
of skeletal muscle. cells termed satellite cells because of their anatomic
location at the periphery of the mature, multinucleated
SOMITIC VS. NONSOMITIC ORIGIN OF SATELLITE
myotube. The defining characteristic of the satellite
CELLS
cell is that the basal lamina that surrounds the satel-
lite cell and the associated myofiber is continuous
Previous studies support the hypothesis that muscle (167). As shown in Fig. 2, the identification of this cell
precursor cells, including the myogenic satellite cell population has historically utilized ultrastructural
population, originate from the multipotential mesoder- techniques (66, 161). Other distinguishing morpholog-
mal cells of the somite (58, 141, 167). The support for ical features of the satellite cell population include a
this hypothesis is primarily derived from chimeric or relatively high nuclear-to-cytoplasmic ratio with few
interspecies grafting experiments that have been per- organelles, a smaller nuclear size compared with the
formed in avian models. These fate-mapping studies adjacent nucleus of the myotube, and an increase in
involved the transplantation (or exchange) of embry- the amount of nuclear heterochromatin compared with
onic somites from donor quail embryos into host chick that of the myonucleus (167). These morphological
embryos (27, 105). The transplanted quail cells have features are consistent with the finding that satellite
distinguishing morphological characteristics and were cells are relatively quiescent and transcriptionally less
observed to migrate from the somite and contribute to active. These distinguishing features are absent follow-
both the limb muscles and the satellite cell population ing activation or proliferation of the satellite cells in
in postnatal chick skeletal muscle. Satellite cells have response to growth, remodeling, or muscle injury. After
been isolated from the fetal skeletal muscle from an activation, the satellite cells are more easily identified
E15 or older mouse embryo, suggesting that satellite as they appear as a swelling on the myofiber with
cells populate the developing limb during the latter cytoplasmic processes that extend from one or both
stages of embryogenesis (34, 35, 36). Whether the sat- poles of the cell (Fig. 2; Ref. 167). Associated with the
ellite cells migrate from the somite as a distinct lineage increase in mitotic activity, there is a reduction in
or whether they originate from a preexisting lineage heterochromatin, an increase in cytoplasmic-to-nu-
(i.e., embryonic or fetal myoblasts) in the developing clear ratio, and an increase in the number of intracel-
limb is unclear. Nevertheless, the underlying concept lular organelles (167).
was that each of the myoblast precursor cells (i.e., Satellite cell markers. The profile of gene expression
embryonic myoblasts, fetal myoblasts, and satellite of the quiescent satellite cell as well as their activated
cells) was a derivative of the somite. and proliferating progeny is largely unknown. The
This paradigm has recently been challenged, as stud- quiescent satellite cells do not express myogenic regu-
ies have suggested that multipotential cells of non- latory factors of the MyoD or MEF2 families or other
somitic origin may be the precursors of the satellite cell known markers of terminal differentiation (33, 119,
(45, 139). De Angelis et al. (45) reported that cells 201). Through the identification of satellite cell mark-
isolated from the embryonic dorsal aorta had a similar ers, biologists will be able to address issues related to
morphological appearance and a similar profile of gene the developmental origin of the satellite cell, the cell
expression to that of satellite cells. Furthermore, cycle control, and the molecular regulation of this
transplantation of the aorta-derived myogenic cells unique cell population during growth and regenera-
into newborn mice revealed that this cell population tion. Several satellite cell markers have been identified
participated in postnatal muscle growth, regeneration, and are restricted to either the quiescent, activated, or
and fusion with resident satellite cells. The authors proliferative state or are expressed more broadly (Ta-
proposed that satellite cells may be derived from endo- ble 1).
thelial cells or a precursor common to both the satellite We have previously determined that myocyte nu-
cell and the endothelial cell. clear factor (MNF or Foxk1), a member of the winged
Derivation of the satellite cell from the somite or a helix transcription factor family, is localized to the
nonsomitic source need not be mutually exclusive. Con- quiescent satellite cell in adult skeletal muscle (64).
ceivably, both lineages may contribute under physio- We have identified two alternatively spliced isoforms
logical or pathological states to the satellite cell popu- for MNF and termed them MNF-␣ and MNF-␤ (64,
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


538 INVITED REVIEW

Fig. 2. Satellite cells occupy a sublami-


nar position in adult skeletal muscle.
In the uninjured muscle fiber, the sat-
ellite 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 het-
erochromatin. When the fiber becomes
injured, the satellite cells become acti-
vated and increase their cytoplasmic
content. The cytoplasmic processes al-
low for chemotaxis of the satellite cell
along the myofiber. Bar ⫽ 1 ␮m.

203, 204). These two alternatively spliced isoforms are growth deficit, a marked impairment in muscle regen-
reciprocally expressed during myogenesis and during eration (63), and a decreased number of satellite cells
muscle regeneration, suggesting that the two isoforms in adult MNF mutant skeletal muscle (Hawke and
of MNF may exert opposing effects on target genes at Garry, unpublished observations). Additional winged
discrete steps during muscle regeneration or in re- helix family members have also been identified in stem
newal of the satellite cell population (63). Using a cells or regenerating cells including Genesis (Foxd3;
RT-PCR assay, we have shown that MNF-␤ is the Ref. 83), which is expressed selectively in embryonic
principal form expressed in quiescent satellite cells, stem cells, and a protein related to hepatocyte nuclear
whereas MNF-␣ predominates in proliferating satellite factor-3 (HNF3/forkhead homolog 11), which has been
cells following muscle injury. Disruption of the MNF identified in regenerating hepatocytes (206).
locus, mutating both isoforms, resulted in a severe Utilizing single cell RT-PCR analysis, Cornelison
and Wold (33) characterized the satellite cells as a
Table 1. Expression patterns of satellite cell markers heterogeneous population based on their profile of
in adult skeletal muscle gene expression. Additionally, they identified c-Met,
the receptor for hepatocyte growth factor (HGF), as a
Molecular Marker Expression Observed in the Adult Reference marker of quiescent satellite cells. HGF is a potent
MNF Quiescent, activated, and proliferating 64 mitogen for satellite cells and has been shown to be
satellite cells important in the migration of the myogenic precur-
Pax7 Quiescent, activated, and proliferating 169 sor cells from the somite to the developing limb (5,
satellite cells 14). Moreover, c-Met deficient embryos fail to form
c-Met Quiescent, activated, and proliferating 33
satellite cells limb skeletal muscle due to a lack of myogenic pre-
M-cadherin Quiescent, activated, and proliferating 33 cursor cells (14, 112).
satellite cells Irintchev and colleagues (87) identified M-cadherin,
NCAM Quiescent, activated, and proliferating 39 a calcium-dependent cell adhesion molecule, as a
satellite cells; synaptic junctions in
adult myofibers
unique marker of the satellite cell pool. M-cadherin is
VCAM-1 Quiescent, activated, and proliferating 89 only expressed in a subpopulation of the quiescent cell
satellite cells pool; however, its expression is increased when the
Desmin Activated and proliferating satellite 15 satellite cells become activated in response to a stim-
cells ulus (9, 33). Recent studies suggest that other cell
myf5 Activated and proliferating satellite 33
cells adhesion molecules, neural cell adhesion molecule
MyoD Activated and proliferating satellite 33, 124 (NCAM) and vascular adhesion molecule-1 (VCAM-1),
cells are also potential markers of quiescent satellite cells
BrdU Proliferating cells 163 (39, 89). The role of these adhesion molecules is unclear
PCNA Proliferating cells 90
[3H]thymidine Proliferating cells 128 but collectively (NCAM, VCAM-1, and M-cadherin)
may function in the adhesion of the satellite cell to the
Expression of selected molecular markers used to identify the basal lamina of the myofiber and may participate in
satellite cell population in adult skeletal muscle is outlined. MNF,
myocyte nuclear factor; NCAM and VCAM, neural cell and vascular
the migratory capacity of this cell population in re-
adhesion molecule; BrdU, bromodeoxyuridine; PCNA, proliferating sponse to stimuli. NCAM is expressed in both myofi-
cell nuclear antigen. bers and satellite cells, whereas VCAM-1 is broadly
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 539

expressed during embryogenesis but limited to satel- immunohistochemical techniques have been utilized
lite cells in adult muscle (39). Furthermore, VCAM-1 for the identification of the satellite cell pool. Although
has been shown to mediate satellite cell interaction a limited number of markers for the quiescent satellite
with leukocytes following injury (89). cell population exist, proliferating satellite cells, as
Recent work from the Rudnicki laboratory (169) measured by [3H]thymidine or bromodeoxyuridine
identified the paired box transcription factor, Pax7, (BrdU) incorporation, can be identified immunohisto-
expressed selectively in quiescent and proliferating chemically for the coexpression of the MyoD family
satellite cells. Analysis of the Pax7 mutant skeletal members or the intermediate filament protein, desmin
muscle revealed a complete absence of satellite cells. (15, 104, 123). MyoD expression occurs early during
This novel finding supports the hypothesis that Pax7 is the activation of the satellite cell population (within 6 h
essential for the specification of the satellite cell pop- following muscle injury) (63, 74, 100, 119, 134). In
ulation. Future studies will be important in the defini- addition, several nonselective markers of cellular pro-
tion of Pax7 downstream target genes in the satellite liferation have been used to characterize the prolifer-
cell population and may provide insight into the regu- ating satellite cell pool. These markers of cellular pro-
lation of this cellular pool. liferation include proliferating cell nuclear antigen
Identification of other satellite cell markers is the (90), BrdU (163), and [3H]thymidine (128).
focus of current research efforts. Emerging candidates Satellite cell number is dependent on the species,
include the cell surface antigen Sca-1 (stem cell anti- age, and muscle fiber type (Table 2; see Ref. 167 for
gen-1; Refs. 88, 177), the glycoprotein Leu-19 (94, 162), review). Satellite cells constitute ⬃30% of the muscle
the anti-apoptotic factor Bcl-2 (106, 123), CD34 (9), and nuclei in the neonate and decrease with age to ⬃4% in
interferon regulatory factor-2, which is a transcription the adult and ⬃2% in the senile (29–30 mo) mouse
factor that mediates VCAM-1 expression in skeletal (176). The decrease in the percentage of satellite cells
muscle (89). with aging (i.e., senile rodent) is the result of an in-
SATELLITE CELL QUANTITATION AND DISTRIBUTION
crease in myonuclei (oxidative and glycolytic myofi-
bers) and a decrease in total number of satellite cells
Quantitation of the satellite cell population in adult (glycolytic myofibers) (12, 66, 167).
skeletal muscle has been possible primarily through The satellite cell distribution between muscle groups
the use of ultrastructural techniques. More recently, is a result of the heterogeneity in satellite cell content

Table 2. Satellite cell content in skeletal muscle


Model Muscle Age and Protocol %SC SC#/muscle Reference

Rat TA 7–9 wk 4 161


Soleus 7–9 wk 11 161
Diaphragm 7–9 wk 8 161
Rat LD adult 4.5 2
Rat Soleus 1 mo 9.6 5.2 ⫻ 105 66
Soleus 1 yr 6.6 7.3 ⫻ 105 66
Soleus 2 yr 4.7 5.4 ⫻ 105 66
EDL 1 mo 7.0 3.1 ⫻ 105 66
EDL 1 yr 2.9 2.1 ⫻ 105 66
EDL 2 yr 1.9 1.3 ⫻ 105 66
Rat EDL 4 mo 3.8 146
EDL Hypothyroid 3.8 146
EDL Hypothyroid; chronic stimulation 7.9–13.8 146
Rat Levator ani 4 mo 1.9 135
Levator ani 32 mo 1.2 135
Mouse Soleus 8 mo 4.6 176
Soleus 30 mo 2.4 176
Mouse Gastroc 7–10 days 25 169
Gastroc Pax 7⫺/⫺ 0 169
Quail LD 6 wk 15.6 198
LD Stretched 16.7 198
Human Control patients 15 190
DMD patients 25 190
Human Trapezius Control (38 yr) 3.7 94
Trapezius Resistance trained 5.4 94
Human Biceps brachii ⬍30 mo 8.4 158
Biceps brachii Werdnig-Hoffman infants 14.4 158
Pig Sartorius 64 wk 1.1 21
PL 64 wk 4.3 21
Lizard Tail Lygosoma species 7.5 95
Tail Anolis species 4.8 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

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


540 INVITED REVIEW

between muscle fiber types. An increase in satellite cell Table 3. Factors affecting satellite cell activity
density has been demonstrated in association with the
Chemotactic
proximity of capillaries (161), myonuclei (18, 161), and Growth Factor Ability Proliferation Differentiation Reference
motoneuron junctions (199). The proximity of satellite
cells to these anatomic structures suggests a permis- FGF 1 79
1 2 3
sive role in the regulation of the cellular pool. In sup- 1 54
port of this hypothesis, oxidative fibers (characterized NE 13
by increased capillary and motoneuron density com- 1 152
pared with glycolytic myofibers) demonstrate a five to HGF 1 13
six times greater satellite cell content (65, 161). 1 4
1 2 124
1 182
GROWTH FACTORS AS REGULATORS OF THE
IGF-I 1 79
SATELLITE CELL POPULATION 1 1 3
1 54
The process of muscle regeneration requires the in- 1 1 26(t)
fluence of growth factors and a sequence of cellular IGF-II 1 79
events, which results in the regulation of the satellite 1 54
cell population (Table 3, Fig. 3; Refs. 73, 168). Many of TGF-␤ 1 79
the studies that have examined the effect of growth 1 1 2 152
factors on satellite cell biology have utilized satellite 2 2 3
cell cultures. These studies have defined the effect of 1 13
2 1 208
growth factors alone or in combination and have pro-
vided valuable insight into the regulation of the satel- Macrophages 1 1 152
1 121
lite cell. Admittedly, in vitro studies are limited due to NE 13
the lack of permissive and repressive factors that are 1 1 86(t)
present in vivo and may influence cellular activity. Crushed muscle/
Currently, most satellite cell cultures are derived from platelet- 1 2 79
neonatal skeletal muscle due to the abundance of sat- derived
ellite cells in these tissues compared with older ani- extract 1 1 2 13
mals (⬎30% in young animals vs. ⬃5% in older ani- LIF 1 7
1 NE 152
mals; Ref. 143). The population and age of the satellite
IL-6 1 7
cell is an important consideration in cell culture prep-
arations as the response of aged, quiescent satellite PDGFAA/AB NE NE 152
NE 54
cells to growth factor stimulation differs compared
PDGFBB 1 1 152
with young, proliferating satellite cells (182). This sec- NE 13
tion will provide a brief introduction of growth factors
EGF NE 13
that are important in the regulation of satellite cell 1* *54
proliferation, differentiation, and motility (for review,
see Table 3). A number of factors affect satellite cell proliferation, differentia-
tion, and chemotaxis. All studies were performed using cell culture
Insulin-like growth factors. Skeletal muscle secretes techniques except those denoted with a (t), which were performed
insulin-like growth factors I and II (IGF-I and IGF-II), using skeletal muscle tissue. NE, no effect. FBF, HGF, IGF, TGF,
which are known to be important in the regulation of PDGF, and EGF, fibroblast, hepatocyte, insulin-like, transforming,
insulin metabolism (3, 109, 186). In addition, these platelet-derived, and endothelial growth factor, respectively. LIF,
growth factors are important in the regulation of skel- leukemia-inhibitory factor. * Stimulation of satellite cell prolifera-
tion in the presence of serum but no effect in serum-free medium. 1
etal muscle regeneration. IGF-I and IGF-II increase and 2, Increase or decrease in effect, respectively.
satellite cell proliferation and differentiation in vitro
(Table 3). The importance of these growth factors was
demonstrated with the intramuscular administration proliferation (25, 32, 170). IGF-I-stimulated satellite
of IGF-I into older, injured animals. In this study, cell differentiation appears to be mediated through the
IGF-I administration (using an osmotic minipump) re- PI-3K pathway (32). Additional studies, utilizing ge-
sulted in enhanced satellite cell proliferation and in- netic mouse models (i.e., transgenic overexpression or
creased muscle mass (26). Moreover, skeletal muscle knockout models), may further define the regulation
overload or eccentric exercise results in elevated IGF-I and signaling pathways of the IGFs and satellite cell
levels, increased DNA content (suggesting an increase biology (8, 25, 132).
in satellite cell proliferation), and a compensatory hy- Hepatocyte growth factor. Hepatocyte growth factor
pertrophy of skeletal muscle (1, 202). (HGF) is a multifunctional cytokine initially described
IGF-I appears to utilize multiple signaling pathways as a mitogen in mature hepatocytes (122). Recently,
in the regulation of the satellite cell pool. The cal- HGF and its receptor c-Met have been localized to
cineurin/NFAT, mitogen-activated protein (MAP) ki- satellite cells and adjacent myofibers but are absent in
nase, and phosphatidylinositol-3-OH kinase (PI-3K) the adjacent fibroblasts. In addition, HGF expression is
pathways have all been implicated in satellite cell proportional to the degree of muscle injury (33, 174,
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 541

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 differen-
tiation). These factors utilize signaling path-
ways in the regulation of the satellite cell. LIF,
leukemia inhibitory factor; IL-6, interleukin-6.
EGF, FGF, HGF, IGF, PDGF, and TGF, endo-
thelial-derived, fibroblast, hepatocyte, insulin-
like, platelet-derived, and transforming growth
factors, respectively.

182). Multiple roles for HGF have been proposed for injury. In contrast, Fiore et al. (57) pursued a similar
the regulation of the satellite cell, including a role as a targeting strategy to mutate the FGF-6 locus and ob-
potent chemotactic factor, an activator of the satellite served an absence of defects in response to either a
cell, and an inhibitor of myoblast differentiation (Table crush injury or chemically induced injury (notexin).
3). HGF is capable of activating and selectively promot- Consequently, the functional role(s) of FGF-6 during
ing satellite cell proliferation (4). Furthermore, HGF muscle regeneration remains unclear. Nevertheless,
administration attenuates satellite cell differentiation these studies underscore the ability of redundant fam-
through the transcriptional inhibition of the myogenic ily members to compensate for one another and result
regulatory factors (i.e., MyoD and myogenin) (62). in preserved function under pathological conditions
Fibroblast growth factors. Fibroblast growth factor (i.e., mouse knockout models).
(FGF) has nine different isoforms (FGF-1 to FGF-9). The release of FGF-2 from the damaged myofibers,
Although many of the FGF isoforms are broadly ex- like HGF, is proportional to the degree of injury (29).
pressed, FGF-6 is restricted to skeletal muscle (59). FGF levels are coordinated with FGF receptor expres-
Sheehan and Allen (173) investigated in detail the role sion. When receptor expression is increased, satellite
of the FGF family on satellite cell proliferation in cells propagated in culture demonstrate an increased
culture. In these studies, it was demonstrated that proliferation and decreased differentiation (160). Con-
FGF-1, -2, -4, -6, and -9 stimulated cellular prolifera- versely, when receptor expression is diminished, pro-
tion, whereas FGF-5, -7, and -8 had no mitogenic ac- liferation is decreased and there is a concomitant in-
tivity. The investigators further observed that addition crease in satellite cell differentiation. Interestingly,
of HGF to either FGF-2, -4, -6, or -9 resulted in a during the period of satellite cell activation and prolif-
synergistic increase in satellite cell proliferation. In eration (0–48 h after injury), FGF receptor (FGF-R1)
addition to an increase in satellite cell proliferation, mRNA is increased fivefold, and this increase is further
the FGF family has also been observed to attenuate enhanced in the presence of HGF (173).
satellite cell differentiation to myofibers (30, 91, 173, The signaling pathway(s) that transduces the FGF
178). signal has recently been investigated with the use of
Floss et al. (59) reported that mice deficient for both transgenic techniques and pharmacological inhib-
FGF-6 (i.e., knockout mice at the FGF-6 locus) have itors (92). These studies revealed that the MAP kinase
impaired satellite cell proliferation and a subsequent pathway is important in transducing the FGF-induced
defect in muscle regeneration in response to a crush increase in satellite cell proliferation; however, the
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


542 INVITED REVIEW

MAP kinase signaling pathway did not mediate the dosages did not appreciably affect muscle regeneration
FGF-mediated repression of satellite cell differentia- (125). Future studies that combine cell culture meth-
tion. odologies and overexpression or loss of function models
Transforming growth factors. Transforming growth using molecular technologies will be helpful in the
factor-␤ (TGF-␤) is the prototypical family member of definition of the role of growth factors in satellite cell
cytokines that includes bone morphogenic protein and biology.
growth-differentiation factors. The TGF-␤ family of
cytokines transduces their signal through the SMAD FUNCTIONAL RESPONSES OF SATELLITE CELLS
family of proteins (see Ref. 196 for review). Generally, TO PHYSIOLOGICAL STIMULI
the TGF-␤ family members function to inhibit muscle
proliferation and differentiation (3, 70, 97, 99, 208) by Hypertrophic stimuli. Load-induced hypertrophy
silencing the transcriptional activation of the MyoD (chronic stretch, agonist ablation, tenotomy) and resis-
family members (115). This inhibition of differentia- tance training are physiological challenges that pro-
tion by TGF-␤, like FGF, persists even in nonmuscle mote a hypertrophic response in both human and ani-
cell lines engineered to ectopically express members of mal models (154–156, 197). Hypertrophic growth of
the MyoD family (97, 115, 184). The combination of skeletal muscle is stimulated by short bursts of muscle
IGF-I or FGF with TGF-␤ was unable to alter the activity against high resistance. Resistance training
TGF-␤-induced attenuation of satellite cell differentia- induces muscle hypertrophy through a process of sat-
tion; however, TGF-␤ action had little effect on IGF-I- ellite cell activation, proliferation, chemotaxis, and fu-
or FGF-mediated increases in proliferation (70). Dur- sion to existing myofibers to contribute to muscle
ing muscle regeneration, TGF-␤ receptor levels (TGF-␤ growth (Fig. 4; Ref. 167). The migratory capacity (che-
RII) and TGF ligand are reciprocally expressed, result- motaxis) of satellite cells is dependent on the integrity
ing in the initial promotion of cellular proliferation of the basal lamina. After the rupture or interruption
followed by enhanced muscle differentiation (159). of the basal lamina in response to myotrauma, satellite
Interleukin-6 cytokines. Leukemia inhibitory factor cells may migrate to adjacent myofibers utilizing tissue
(LIF) and interleukin-6 (IL-6) are members of the IL-6 bridges (164, 191). In response to limited myotrauma,
family of cytokines produced by many different cells, where no rupture of the basal lamina occurs, satellite
including myoblasts and macrophages. These cyto- cells migrate from the proximal intact portion of the
kines share a common receptor component, and their myofiber, under the basal lamina, to the site of injury
actions are mediated through the same signaling path- to participate in the repair process (165, 167).
ways (81, 144). Skeletal muscle regeneration after in- Exercise-induced myotrauma initiates an immune
jury in LIF mutant mice is attenuated, whereas exog- response, resulting in the influx of macrophages into
enous administration of LIF increased the regenerative the damaged region. After the acute insult, macro-
process and produced enlarged myofibers (101). The per-
phage infiltration peaks within 48 h (186). Initially, the
missive effect of LIF was associated only with the muscle
role of these blood-borne macrophages was believed to
lineage and had no effect on nonmuscle cells in skeletal
be limited to phagocytosis and the digestion of myone-
muscle (101). IL-6 promotes the degradation of necrotic
crotic fibers. However, additional roles for macro-
tissue, synchronizes the cell cycle of satellite cells, and
phages during the early stages of muscle repair are
induces apoptosis of macrophages following muscle in-
jury (22). Unlike LIF, however, IL-6 expression in injured emerging. Macrophages are essential in the orchestra-
muscle does not increase satellite cell proliferation (96). tion of the repair process as they secrete a collection of
Collectively, this family of growth factors appears to play cytokine factors that regulate the satellite cell pool
an integral role in skeletal muscle regeneration. (133). Importantly, in the absence of a macrophage
Many other factors may be involved in the regulation response, muscle regeneration is absent; in the pres-
of the satellite cell in adult skeletal muscle. Nitric ence of an enhanced macrophage response, there is an
oxide, platelet-derived growth factor, endothelial-de- increase in satellite cell proliferation and differentia-
rived growth factor, and testosterone have been shown tion (110).
to mediate satellite cell activity (6, 93, 133). Obviously, In response to resistance training, myotrauma re-
the regulation of satellite cells is orchestrated by nu- sults in the release of growth factors that will, in part,
merous factors in a temporal and concentration-depen- regulate the satellite cell population during regenera-
dent fashion during regeneration. tion (Fig. 3 and Table 3). For example, IGF-I is upregu-
Few animal studies have examined the effect of lated in response to hypertrophic signals in skeletal
growth factors in vivo. Chakravarthy et al. (26) ob- muscle and promotes proliferation and fusion of the
served that local IGF-I administration to atrophied satellite cell pool (1, 26, 186). As outlined in the previ-
muscle increased satellite cell proliferation and muscle ous section, additional growth factors and/or cytokines
mass within 2 wk. Unlike the observations with IGF-I, including LIF and members of the TGF-␤ family may
the intramuscular injection of HGF, at specified inter- play a role in the signaling or commissioning of the
vals following skeletal muscle injury, increased satel- satellite cells to participate in the hypertrophic remod-
lite cell proliferation and either had no effect or im- eling response. Although a number of questions remain
paired the rate of regeneration (124). Similarly, regarding the role of the satellite cell in muscle remod-
administration of FGF at timed intervals and selected eling, the primary physiological consequence of the
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 543

Fig. 4. Satellite cell response to myotrauma.


*Skeletal muscle trauma or injury may be minor
(e.g., resistance training) or may be more exten-
sive (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-re-
newal. 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 regener-
ated myofiber, the newly fused satellite cell nu-
clei will initially be centralized but will later
migrate to assume a more peripheral location.

hypertrophic response is to produce a muscle with a dition, there are an increased number of satellite cells
greater capacity for peak force generation. associated with the neuromuscular junction (199). It is
Atrophic stimuli. Atrophy of skeletal muscle results conceivable that neurotrophic factors are important in
in a reduction in myonuclei number and can be induced satellite cell homeostasis. A number of laboratories
by numerous factors including denervation, hindlimb have reported that the percentage of satellite cells
suspension, and malnutrition (73). Atrophy and re- increase (from 3 to 9%) during the initial period follow-
modeling that result from muscle disuse can be pro- ing denervation (118, 187). However, a prolonged pe-
duced in laboratory rodents physiologically by tail/ riod of denervation results in a significant decrease in
hindlimb suspension or immobilization of specific the percentage of satellite cells (3 to 1% following an
muscle groups in plaster casts or pathologically 18-mo period of denervation). Viguie et al. (187) hy-
through denervation. The response of the satellite cells pothesize that the progressive decline in the satellite
appears to be pleiotropic and dependent on the atro- cell pool may be the result of satellite cell apoptosis.
phic stimulus. Alternatively, denervation may result in a lack of neu-
In adolescent rats, hindlimb suspension results in an rotrophic input (including growth factors) that nega-
irreversible remodeling process, including decreased tively impacts satellite cell function and content.
satellite cell content and an impaired proliferative ca- Long-term denervation has considerable clinical im-
pacity within 3 days of unloading in both the oxidative plications. Denervation for periods of 6–18 mo results
slow-twitch soleus and the fast-twitch extensor digito- in the inability of skeletal muscle to reestablish a
rum longus (EDL) muscles (44, 129). Thus the atrophic preinjury functional capacity even if neuronal sprout-
stimulus in the adolescent animal may irreversibly ing and regeneration occurs (180). The mechanisms for
alter the developmental program for myofibers to ac- this phenomenon are unclear, but considerable data
crue nuclei even with the resumption of weight bear- support the conclusion that the intact neuromuscular
ing. A similar reduction in the satellite cell population junction and the denervation model mediate positive
was observed in adult rat hindlimbs using an immobi- and repressive influences, respectively, on the satellite
lization model as an alternative atrophic stimulus cell pool (187).
(188). In contrast to adolescent animals, remobilization Aging. Recent advances have allowed biologists to
of the hindlimb was accompanied by increased myofi- interrogate the proliferative history of a cellular popu-
ber regeneration, supporting the hypothesis that, fol- lation. With each cell replication, there is ⬃100 bp lost
lowing completion of the developmental program, adult from the ends of eukaryotic chromosomes (47, 48, 151,
satellite cells are capable of activation and prolifera- 172). The ends of the chromosomes contain TTAGGG
tion to repopulate atrophied muscle (129, 188). repeats termed telomeres. The length of these telo-
Unlike the other forms of atrophy, denervation is a meres reflects the number of replications of a particu-
pathological rather than physiological stimulus. De- lar cell and its proliferative capacity. Using this tech-
nervation produces a form of disuse atrophy that in- nology, investigators now have the ability to analyze
cludes myofiber degeneration and is accompanied by the proliferative history and future capacity of the
distinctive changes in the myonuclei and quiescent satellite cell, providing valuable insight into the effects
satellite cells (102, 111, 153). In the unperturbed con- of aging and disease in this cell population.
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


544 INVITED REVIEW

As age progresses, there is an impairment of skeletal This question is applicable to the young population as
muscle regeneration following injury (see Ref. 72 for well. If satellite cells have a limited proliferative ca-
review). A decrease in satellite cell number and/or pacity (⬃60 doublings), does a lifetime of intense exer-
proliferative capacity has been used to explain this cise have a negative influence on their ability to regen-
phenomena. Support for this hypothesis is observed in erate skeletal muscle as aging progresses?
rodent models, as increasing age is associated with a
decrease in satellite cell number and a reduced prolif- FUNCTIONAL RESPONSES TO DISEASE STATES
erative capacity (52, 66, 166). In contrast to the rodent
Most myopathies have a molecular mutation that
model, a decrease in human satellite cell population
affects the structural or cytoskeletal proteins in skele-
and proliferative ability is observed only during the
tal muscle. Duchenne muscular dystrophy (DMD) is
childhood years. For example, neonatal (5-day-old) and
the most common and the most devastating of the
infant (5-mo-old) satellite cells are capable of ⬃60 and
muscular dystrophies (20, 55, 76, 82, 127). Disease
45 replications, respectively, whereas 9-yr-old and
progression and death are ultimately due to a failure of
ⱖ60-yr-old humans are both capable of 20–30 replica-
the myogenic satellite cells to maintain muscle regen-
tions (151). However, as aging progresses, satellite
eration (36, 80). DMD is a recessive X-linked disease
cells (ⱖ60-yr-old skeletal muscle) fuse to form thinner,
that results in a null mutation at the dystrophin locus
more fragile myotubes (151). Thus, despite a normal
(20, 82, 127). The absence of this cytoskeletal protein
ability to proliferate, the satellite cells of older humans
renders the muscle fiber extremely fragile. In response
have a reduced capacity to repopulate the myofiber
to mechanical stress associated with repeated contrac-
population.
tion, there is widespread degeneration. The satellite
The impaired regenerative response that is observed
cells respond to the injury by repopulating the injured
with aging in humans thus appears to be much more
skeletal muscle with defective myofibers lacking dys-
complex than satellite cell senescence alone. Results
trophin. This process results in continuous degenera-
from cross-transplantation experiments suggest that
tion-regeneration cycles and ultimately exhausts the
the host environment is a critical factor in the ability of
satellite cell pool (36, 80).
older skeletal muscle to regenerate (23, 24). Cross
Clinical symptoms are apparent by 4–5 yr of age in
transplantation of EDL muscles between 4-mo-old and
boys with DMD (10, 20, 127). DMD patients (4–5 yr of
24-mo-old rats demonstrated that mature skeletal
age) have been shown to undergo more skeletal muscle
muscle was as capable as young skeletal muscle in
regeneration than that measured in a total of six nor-
recovering from transplant and from toxin-induced in-
mal patients over 60 yr of age (46). These results were
jury. However, young (4-mo-old) nerve-muscle au-
confirmed by Renault et al. (151), who demonstrated
tografts functioned significantly better than old (24-
that the proliferative life span of satellite cells derived
mo-old) nerve-muscle autografts, as determined by the
from a 9-yr-old DMD patient was approximately one-
measurement of mass and maximum isometric force,
third of an age-matched control. Proliferative fatigue
suggesting that the ability for neuronal regeneration
or senescence of the satellite cell population and the
may be a critical factor in activating the satellite cell
milieu of the DMD skeletal muscle may collectively
response and, ultimately, regenerating the damaged
impair the proliferative or regenerative capacity of this
muscle (23, 24).
cell population. In the DMD patient, increased levels of
Other factors within the host environment affect the
IGF binding proteins (IGFBP) are released by fibro-
efficiency of skeletal muscle regeneration as aging
blasts. The elevated IGFBP sequesters IGF-I, limiting
progresses. A thickening of the basal lamina (176),
its bioavailability for satellite cells and ultimately re-
increased fibrosis within skeletal muscle (114), and
sulting in increased skeletal muscle fibrosis (120). The
reduced capillary density (31) may also contribute to
evidence to date demonstrates the tremendous strain
impaired regeneration. Inflammatory factors (macro-
and ultimate failure of the satellite cell population to
phages and associated cytokines) are essential for the
adequately compensate for the persistent degenera-
normal satellite cell response to injury. Aging nega-
tion-regeneration process that is occurring in the DMD
tively impacts the immune response, resulting in a
skeletal muscle.
decrease of the inflammatory factors and macrophages
(43). In association with an impaired immune re- GENETIC MOUSE MODELS OF MYOPATHY
sponse, there are reduced serum levels of growth fac-
tors including IGF-I in aged rats and humans (150, A number of gene knockout mouse models and ex-
183). After multiple cycles of atrophy, there is no res- perimental methods are available for studies of muscle
toration of muscle mass or satellite cell proliferation in regeneration and satellite cell biology. Although the
aged rats even after 9 wk of recovery. However, local mdx mouse, which lacks dystrophin, has provided im-
IGF-I administration to the atrophied muscle resulted portant insights into the pathophysiology of DMD, the
in significant increases in mass and satellite cell pro- myopathy in these animals does not represent the
liferation within 2 wk (26). myopathic process of DMD in humans (75). The mdx
An unresolved question with regard to satellite cells mice have a normal life span, a temporally restricted
and the aging process is whether repeated exhaustive myofiber degeneration, and adapt to muscle degenera-
and/or resistance training exercise programs have a tion with an expansion of the satellite cell pool and
negative impact on the long-term satellite cell content? muscle hypertrophy, thereby avoiding the compro-
J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 545

mised muscle function that afflicts humans who lack to study satellite cell biology. Perhaps the most exten-
dystrophin (51, 69, 116, 179). In addition to the spon- sive and reproducible muscle injury is the delivery of
taneous dystrophin mutation in the mdx mouse, myo- cardiotoxin (purified from the venom of the Naja ni-
pathic mouse models include genetically engineered gricollis snake) into the hindlimb skeletal muscle of the
mouse strains with knockouts of utrophin (49, 69), mouse (53, 63, 134). The intramuscular injection of 100
MyoD (119), or MNF (63) crossed into the mdx back- ␮l of 10 ␮M cardiotoxin into the gastrocnemius muscle
ground. Each of these double mutant mouse models results in 80–90% muscle degeneration (Fig. 5). After
exhibit features that more closely resemble DMD in cardiotoxin-induced injury, satellite cells become acti-
humans, including a severe myopathy, an impaired vated within 6 h of injury (Garry, unpublished obser-
regenerative capacity, and a decreased life span. vations). In response to locally released growth factors
from injured myotubes and macrophages, the satellite
MUSCLE REGENERATION MODELS
cells proliferate extensively within 2–3 days of injury
An alternative strategy that has been successfully (63, 64). Approximately 5 days after injury, the satel-
used for the study of satellite cell activation, prolifer- lite cells withdraw from the cell cycle and either self-
ation, regeneration, and self-renewal is to experimen- renew or form differentiated myotubes that contain a
tally produce a controlled skeletal muscle injury. Strat- central nucleus (63, 64). With the use of this cardio-
egies including crush (101), freeze (40), or chemically toxin-induced injury protocol, the architecture of the
induced injury (42, 63) have all been successfully used injured muscle is largely restored within 10 days after

Fig. 5. Skeletal muscle response to tox-


in-induced injury. A: transverse section
of the adult gastrocnemius muscle
stained with hematoxylin and eosin at
defined intervals following cardiotoxin-
induced injury. After cardiotoxin deliv-
ery, there is evidence of extensive myo-
necrosis 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 regen-
eration is evident within 5 days of in-
jury. At this time, newly regenerated
myofibers are evident as small, baso-
philic, central-nucleated myofibers (de-
noted by b). The architecture of the
muscle is largely restored within ⬃10
days following injury. The newly regen-
erated myofiber displays numerous
centrally aligned nuclei, demonstrat-
ing 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 prolif-
eration and muscle differentiation fol-
lowing a chemically induced injury of
adult mouse skeletal muscle. Bar ⫽
100 ␮m.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


546 INVITED REVIEW

injury (Fig. 5). The complete profile of intrinsic and can be purified based on the exclusion of Hoechst dye
extrinsic cues that regulate the satellite cell population and can adopt alternative fates. Further studies are
during muscle regeneration remains unclear. There- necessary to determine whether the SP cells are satel-
fore, the use of reproducible experimental injuries such lite cell progenitors, a subpopulation of the satellite
as cardiotoxin-induced injury will be important to eval- cells, or an independent progenitor cell population that
uate and define the regulation of the satellite cell are resident in skeletal muscle.
population in molecular and physiological myopathic Alternative sources of proliferating myoblasts have
models. Additional myonecrotic agents such as notexin recently been described. Odelberg et al. (136) recently
have been used with similar success (107), resulting in reported that terminally differentiated myotubes are
a well-characterized regenerative response in skeletal capable of dedifferentiation to form myoblasts when
muscle. exposed to the homeobox-containing transcriptional re-
pressor, msx1. Furthermore, in response to appropri-
SATELLITE CELLS AS MUSCLE PRECURSOR CELLS OR ate cues (i.e., conditioned media), these dedifferenti-
STEM CELLS ated myoblasts have the capacity to adopt alternative
phenotypes. Collectively, these studies are challenging
Myogenic satellite cells have a tremendous prolifer-
the established developmental paradigms regarding
ative capacity and are capable of self-renewal. These
satellite cell biology and the plasticity or the potential
tissue-specific progenitor cells or satellite cells are im-
of the terminally differentiated myotube for dediffer-
portant in the maintenance and regeneration of skele-
entiation. Further studies are necessary to define the
tal muscle. Progenitor cells that are resident in other
molecular regulation of this dedifferentiation process
adult tissues have stem cell characteristics, as they are
but clearly have applications for the treatment of my-
capable of self-renewal and multilineage differentia-
opathies including DMD.
tion along a specified molecular pathway (61, 194, 195).
For example, Clarke et al. (28) revealed that neural FUTURE PROSPECTS
stem cells isolated from the adult mouse brain had a
very broad developmental capacity and could contrib- Since the initial description of the satellite cell ap-
ute to virtually all tissues when delivered into the proximately 40 years ago, a number of anatomic and
developing embryo. These results suggest that adult physiological studies have established the importance
stem cells or progenitor cells are capable of dedetermi- of this cellular pool in the growth, remodeling, and
nation and/or transdifferentiation to adopt alternative regeneration of adult skeletal muscle. The profile of
lineages in a permissive environment. Unlike the neu- gene expression that regulates the cell cycle progres-
ral stem cell population, the potential of the satellite sion of the satellite cell from a quiescent to an activated
cell pool in adult skeletal muscle remains incompletely and proliferating state remains ill-defined. Despite its
defined. Further definition of the gene expression pro- complexity, future challenges for this field will include
file of the satellite cell population and an enhanced the definition of the development, maintenance, self-
understanding of the molecular events responsible for renewal, and potentiality of the satellite cell popula-
satellite cell development, activation, proliferation, tion. These challenges will require an integration of
and self-renewal will define the stem cell characteris- each of the biological disciplines, including the use of
tics of the satellite cell. increasingly powerful molecular biological tools.
The authors recognize the research efforts of those included and
SIDE POPULATION CELLS
those omitted (due only to space considerations) who have contrib-
uted to this field of study. Special thanks to Drs. M. I. Lindinger, P.
Recent studies have identified a population of pluri- Mammen, and R. S. Williams for critical review of this manuscript.
potent stem cells from adult skeletal muscle termed The authors’ work is supported by grants from the Muscular
side population (SP) cells. Skeletal muscle- and bone Dystrophy Association, March of Dimes, Doris Duke Charitable
marrow-derived SP cells are isolated using a DNA- Foundation, Texas Advanced Research (Technology) Program, and
binding dye (Hoechst 33342) and dual-wavelength flow the Donald W. Reynolds Foundation.
cytometric analysis (67, 68, 77). This method, which REFERENCES
relies on the differential ability of the SP cells to efflux
the Hoechst dye, defines a small and homogeneous 1. Adams GR, Haddad F, and Baldwin KM. Time course of
changes in markers of myogenesis in overloaded rat skeletal
population of cells that can adopt alternative fates in muscles. J Appl Physiol 87: 1705–1712, 1999.
permissive environments (77, 88). For example, Gus- 2. Allam Ali M. Satellite cells in the latissimus dorsi of the rat.
soni et al. (77) demonstrated that SP cells isolated from Acta Anat (Basel) 110: 287–290, 1981.
adult bone marrow were able to reconstitute the irra- 3. Allen RE and Boxhorn LK. Regulation of skeletal muscle
diated mdx mouse bone marrow, and later these cells satellite cell proliferation and differentiation by transforming
growth factor-beta, insulin-like growth factor I, and fibroblast
were recruited from the bone marrow to participate in growth factor. J Cell Physiol 138: 311–315, 1989.
muscle repair. Furthermore, Jackson et al. (88) utilized 4. Allen RE, Sheehan SM, Taylor RG, Kendall TL, and Rice
the same protocol to isolate muscle SP cells from adult GM. Hepatocyte growth factor activates quiescent skeletal
mice and observed that as few as 100 SP cells could muscle satellite cells in vitro. J Cell Physiol 165: 307–312, 1995.
5. Andermarcher E, Surani MA, and Gherardi E. Co-expres-
reconstitute the entire bone marrow of a lethally irra- sion of the HGF/SF and c-met genes during early mouse em-
diated mouse. These results demonstrate that adult bryogenesis precedes reciprocal expression in adjacent tissues
skeletal muscle contains a stem cell population that during organogenesis. Dev Genet 18: 254–266, 1996.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 547

6. Anderson JE. A role for nitric oxide in muscle repair: nitric 29. Clarke MS, Khakee R, and McNeil PL. Loss of cytoplasmic
oxide-mediated activation of muscle satellite cells. Mol Biol Cell basic fibroblast growth factor from physiologically wounded
11: 1859–1874, 2000. myofibers of normal and dystrophic muscle. J Cell Sci 106:
7. Austin L and Burgess AW. Stimulation of myoblast prolifer- 121–133, 1993.
ation in culture by leukaemia inhibitory factor and other cyto- 30. Clegg CH, Linkhart TA, Olwin BB, and Hauschka SD.
kines. J Neurol Sci 101: 193–197, 1991. Growth factor control of skeletal muscle differentiation: com-
8. Barton-Davis ER, Shoturma DI, Musaro A, Rosenthal N, mitment to terminal differentiation occurs in G1 phase and is
and Sweeney HL. Viral mediated expression of insulin-like repressed by fibroblast growth factor. J Cell Biol 105: 949–956,
growth factor I blocks the aging-related loss of skeletal muscle 1987.
function. Proc Natl Acad Sci USA 95: 15603–15607, 1998. 31. Coggan AR, Spina RJ, King DS, Rogers MA, Brown M,
9. Beauchamp JR, Heslop L, Yu DS, Tajbakhsh S, Kelly RG, Nemeth PM, and Holloszy JO. Histochemical and enzymatic
Wernig A, Buckingham ME, Partridge TA, and Zammit comparison of the gastrocnemius muscle of young and elderly
PS. Expression of CD34 and myf5 defines the majority of men and women. J Gerontol B Psychol Sci Soc Sci 47: B71–B76,
quiescent adult skeletal muscle satellite cells. J Cell Biol 151: 1992.
1221–1234, 2000. 32. Coolican SA, Samuel DS, Ewton DZ, McWade FJ, and
10. Bell CD and Conen PE. Histopathological changes in Du- Florini JR. The mitogenic and myogenic actions of insulin-like
chenne muscular dystrophy. J Neurol Sci 7: 529–544, 1968. growth factors utilize distinct signaling pathways. J Biol Chem
11. Bendall AJ, Ding J, Hu G, Shen MM, and Abate-Shen C. 272: 6653–6662, 1997.
Msx1 antagonizes the myogenic activity of Pax3 in migrating 33. Cornelison DD and Wold BJ. Single-cell analysis of regula-
limb muscle precursors. Development 126: 4965–4976, 1999. tory gene expression in quiescent and activated mouse skeletal
12. Bischoff R. The satellite cell and muscle regeneration. In: muscle satellite cells. Dev Biol 191: 270–283, 1997.
Myology, edited by Engel AG and Frazini-Armstrong C. New 34. Cossu G. Unorthodox myogenesis: possible developmental sig-
York: McGraw-Hill, 1994, p. 97–118. nificance and implications for tissue histogenesis and regener-
13. Bischoff R. Chemotaxis of skeletal muscle satellite cells. Dev ation. Histol Histopathol 12: 755–760, 1997.
Dyn 208: 505–515, 1997. 35. Cossu G, Eusebi F, Grassi F, and Wanke E. Acetylcholine
14. Bladt F, Riethmacher D, Isenmann S, Aguzzi A, and receptor channels are present in undifferentiated satellite cells
Birchmeier C. Essential role for the c-met receptor in the but not in embryonic myoblasts in culture. Dev Biol 123: 43–50,
migration of myogenic precursor cells into the limb bud. Nature 1987.
376: 768–771, 1995. 36. Cossu G and Mavilio F. Myogenic stem cells for the therapy
15. Bockhold KJ, Rosenblatt JD, and Partridge TA. Aging of primary myopathies: wishful thinking or therapeutic per-
normal and dystrophic mouse muscle: analysis of myogenicity spective? J Clin Invest 105: 1669–1674, 2000.
in cultures of living single fibers. Muscle Nerve 21: 173–183, 37. Cossu G and Molinaro M. Cell heterogeneity in the myogenic
1998. lineage. Curr Top Dev Biol 23: 185–208, 1987.
16. Borycki AG, Li J, Jin F, Emerson CP, and Epstein JA. 38. Cossu G, Tajbakhsh S, and Buckingham M. How is myo-
Pax3 functions in cell survival and in pax7 regulation. Devel- genesis initiated in the embryo? Trends Genet 12: 218–223,
opment 126: 1665–1674, 1999. 1996.
17. Brohmann H, Jagla K, and Birchmeier C. The role of Lbx1 39. Covault J and Sanes JR. Distribution of N-CAM in synaptic
in migration of muscle precursor cells. Development 127: 437– and extrasynaptic portions of developing and adult skeletal
445, 2000. muscle. J Cell Biol 102: 716–730, 1986.
18. Brown SC and Stickland NC. Satellite cell content in mus- 40. Creuzet S, Lescaudron L, Li Z, and Fontaine-Perus J.
cles of large and small mice. J Anat 183: 91–96, 1993. MyoD, myogenin, and desmin-nls-lacZ transgene emphasize
19. Buckingham M. Making muscle in mammals. Trends Genet 8: the distinct patterns of satellite cell activation in growth and
144–148, 1992. regeneration. Exp Cell Res 243: 241–253, 1998.
20. Burghes AH, Logan C, Hu X, Belfall B, Worton RG, and 41. Cserjesi P and Olson EN. Myogenin induces the myocyte-
Ray PN. A cDNA clone from the Duchenne/Becker muscular specific enhancer binding factor MEF-2 independently of other
dystrophy gene. Nature 328: 434–437, 1987. muscle-specific gene products. Mol Cell Biol 11: 4854–4862,
21. Campion DR, Richardson RL, Reagan JO, and Kraeling 1991.
RR. Changes in the satellite cell population during postnatal 42. D’Albis A, Couteaux R, Janmot C, Roulet A, and Mira JC.
growth of pig skeletal muscle. J Anim Sci 52: 1014–1018, 1981. Regeneration after cardiotoxin injury of innervated and dener-
22. Cantini M and Carraro F. Control of cell proliferation by vated slow and fast muscles of mammals. Myosin isoform anal-
macrophage-myoblast interactions. Basic Appl Myol 6: 485– ysis. Eur J Biochem 174: 103–110, 1988.
489, 1996. 43. Danon D, Kowatch MA, and Roth GS. Promotion of wound
23. Carlson BM and Faulkner JA. Muscle transplantation be- repair in old mice by local injection of macrophages. Proc Natl
tween young and old rats: age of host determines recovery. Acad Sci USA 86: 2018–2020, 1989.
Am J Physiol Cell Physiol 256: C1262–C1266, 1989. 44. Darr KC and Schultz E. Hindlimb suspension suppresses
24. Carlson BM and Faulkner JA. The regeneration of nonin- muscle growth and satellite cell proliferation. J Appl Physiol
nervated muscle grafts and marcaine-treated muscles in young 67: 1827–1834, 1989.
and old rats. J Gerontol A Biol Sci Med Sci 51: B43–49, 1996. 45. De Angelis L, Berghella L, Coletta M, Lattanzi L, Zanchi
25. Chakravarthy MV, Abraha TW, Schwartz RJ, Fiorotto M, Cusella-De Angelis MG, Ponzetto C, and Cossu G.
ML, and Booth FW. Insulin-like growth factor-I extends in Skeletal myogenic progenitors originating from embryonic dor-
vitro replicative life span of skeletal muscle satellite cells by sal aorta coexpress endothelial and myogenic markers and
enhancing G1/S cell cycle progression via the activation of contribute to postnatal muscle growth and regeneration. J Cell
phosphatidylinositol 3⬘-kinase/Akt signaling pathway. J Biol Biol 147: 869–878, 1999.
Chem 275: 35942–35952, 2000. 46. Decary S, Hamida CB, Mouly V, Barbet JP, Hentati F,
26. Chakravarthy MV, Davis BS, and Booth FW. IGF-I restores and Butler-Browne GS. Shorter telomeres in dystrophic mus-
satellite cell proliferative potential in immobilized old skeletal cle consistent with extensive regeneration in young children.
muscle. J Appl Physiol 89: 1365–1379, 2000. Neuromuscul Disord 10: 113–120, 2000.
27. Christ B, Jacob HJ, and Jacob M. Origin of wing muscula- 47. Decary S, Mouly V, and Butler-Browne GS. Telomere
ture. Experimental studies on quail and chick embryos. Expe- length as a tool to monitor satellite cell amplification for cell-
rientia 30: 1446–1449, 1974. mediated gene therapy. Hum Gene Ther 7: 1347–1350, 1996.
28. Clarke DL, Johansson CB, Wilbertz J, Veress B, Nilsson 48. Decary S, Mouly V, Hamida CB, Sautet A, Barbet JP, and
E, Karlstrom H, Lendahl U, and Frisen J. Generalized Butler-Browne GS. Replicative potential and telomere length
potential of adult neural stem cells. Science 288: 1660–1663, in human skeletal muscle: implications for satellite cell-medi-
2000. ated gene therapy. Hum Gene Ther 8: 1429–1438, 1997.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


548 INVITED REVIEW

49. Deconinck AE, Rafael JA, Skinner JA, Brown SC, Potter 71. Gross MK, Moran-Rivard L, Velasquez T, Nakatsu MN,
AC, Metzinger L, Watt DJ, Dickson JG, Tinsley JM, and Jagla K, and Goulding M. Lbx1 is required for muscle pre-
Davies KE. Utrophin-dystrophin-deficient mice as a model for cursor migration along a lateral pathway into the limb. Devel-
Duchenne muscular dystrophy. Cell 90: 717–727, 1997. opment 127: 413–424, 2000.
50. Dietrich S, Schubert FR, Gruss P, and Lumsden A. The 72. Grounds MD. Age-associated changes in the response of skel-
role of the notochord for epaxial myotome formation in the etal muscle cells to exercise and regeneration. Ann NY Acad Sci
mouse. Cell Mol Biol (Oxf) 45: 601–616, 1999. 854: 78–91, 1998.
51. DiMario JX, Uzman A, and Strohman RC. Fiber regenera- 73. Grounds MD. Muscle regeneration: molecular aspects and
tion is not persistent in dystrophic (MDX) mouse skeletal mus- therapeutic implications. Curr Opin Neurol 12: 535–543, 1999.
cle. Dev Biol 148: 314–321, 1991. 74. Grounds MD, Garrett KL, Lai MC, Wright WE, and Beil-
52. Dodson MV and Allen RE. Interaction of multiplication stim- harz MW. Identification of skeletal muscle precursor cells in
ulating activity/rat insulin-like growth factor II with skeletal vivo by use of MyoD1 and myogenin probes. Cell Tissue Res 267:
muscle satellite cells during aging. Mech Ageing Dev 39: 121– 99–104, 1992.
128, 1987. 75. Grounds MD and McGeachie JK. Skeletal muscle regener-
53. Doroshow JH, Tallent C, and Schechter JE. Ultrastruc- ation after crush injury in dystrophic mdx mice: an autoradio-
tural features of adriamycin-induced skeletal and cardiac mus- graphic study. Muscle Nerve 15: 580–586, 1992.
cle toxicity. Am J Pathol 118: 288–297, 1985. 76. Gussoni E, Blau HM, and Kunkel LM. The fate of individual
54. Doumit ME, Cook DR, and Merkel RA. Fibroblast growth myoblasts after transplantation into muscles of DMD patients.
factor, epidermal growth factor, insulin-like growth factors, and Nat Med 3: 970–977, 1997.
platelet-derived growth factor-BB stimulate proliferation of 77. Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan
clonally derived porcine myogenic satellite cells. J Cell Physiol MK, Flint AF, Kunkel LM, and Mulligan RC. Dystrophin
157: 326–332, 1993. expression in the mdx mouse restored by stem cell transplan-
55. Engel AG, Yamamoto M, and Fischbeck KH. Comprehen- tation. Nature 401: 390–394, 1999.
sive description of the clinical and laboratory features and 78. Hasty P, Bradley A, Morris JH, Edmondson DG, Venuti
therapy for polymyositis, dermamyositis and inclusion body JM, Olson EN, and Klein WH. Muscle deficiency and neona-
myositis. In: Myology, edited by Engel AG and Frazini-Arm- tal death in mice with a targeted mutation in the myogenin
strong C. New York: McGraw-Hill, 1994, p. 1130–1188. gene. Nature 364: 501–506, 1993.
56. Feldman JL and Stockdale FE. Temporal appearance of 79. Haugk KL, Roeder RA, Garber MJ, and Schelling GT.
satellite cells during myogenesis. Dev Biol 153: 217–226, 1992. Regulation of muscle cell proliferation by extracts from crushed
57. Fiore F, Sebille A, and Birnbaum D. Skeletal muscle regen- muscle. J Anim Sci 73: 1972–1981, 1995.
eration is not impaired in Fgf6 ⫺/⫺ mutant mice. Biochem 80. Heslop L, Morgan JE, and Partridge TA. Evidence for a
Biophys Res Commun 272: 138–143, 2000. myogenic stem cell that is exhausted in dystrophic muscle.
58. Fischel A. Zur entwicklung der ventralen rumpf- und extremi-
J Cell Sci 113: 2299–2308, 2000.
tatenmuskulatur der Vogel und Saugetiere. Morphol Jb 23:
81. Hibi M, Nakajima K, and Hirano T. IL-6 cytokine family and
544–561, 1895.
signal transduction: a model of the cytokine system. J Mol Med
59. Floss T, Arnold HH, and Braun T. A role for FGF-6 in
74: 1–12, 1996.
skeletal muscle regeneration. Genes Dev 11: 2040–2051, 1997.
82. Hoffman EP, Monaco AP, Feener CC, and Kunkel LM.
60. Franz T, Kothary R, Surani MA, Halata Z, and Grim M.
Conservation of the Duchenne muscular dystrophy gene in mice
The Splotch mutation interferes with muscle development in
and humans. Science 238: 347–350, 1987.
the limbs. Anat Embryol (Berl) 187: 153–160, 1993.
83. Hromas R, Ye H, Spinella M, Dmitrovsky E, Xu D, and
61. Fuchs E and Segre JA. Stem cells: a new lease on life. Cell
Costa RH. Genesis, a Winged Helix transcriptional repressor,
100: 143–155, 2000.
62. Gal-Levi R, Leshem Y, Aoki S, Nakamura T, and Halevy has embryonic expression limited to the neural crest, and stim-
O. Hepatocyte growth factor plays a dual role in regulating ulates proliferation in vitro in a neural development model. Cell
skeletal muscle satellite cell proliferation and differentiation. Tissue Res 297: 371–382, 1999.
Biochim Biophys Acta 1402: 39–51, 1998. 84. Hughes SM, Koishi K, Rudnicki M, and Maggs AM. MyoD
63. Garry DJ, Meeson A, Elterman J, Zhao Y, Yang P, Bassel- protein is differentially accumulated in fast and slow skeletal
Duby R, and Williams RS. Myogenic stem cell function is muscle fibres and required for normal fibre type balance in
impaired in mice lacking the forkhead/winged helix protein rodents. Mech Dev 61: 151–163, 1997.
MNF. Proc Natl Acad Sci USA 97: 5416–5421, 2000. 85. Hughes SM, Taylor JM, Tapscott SJ, Gurley CM, Carter
64. Garry DJ, Yang Q, Bassel-Duby R, and Sanders Williams WJ, and Peterson CA. Selective accumulation of MyoD and
R. Persistent expression of MNF identifies myogenic stem cells myogenin mRNAs in fast and slow adult skeletal muscle is
in postnatal muscles. Dev Biol 188: 280–294, 1997. controlled by innervation and hormones. Development 118:
65. Gibson MC and Schultz E. The distribution of satellite cells 1137–1147, 1993.
and their relationship to specific fiber types in soleus and 86. Hurme T and Kalimo H. Activation of myogenic precursor
extensor digitorum longus muscles. Anat Rec 202: 329–337, cells after muscle injury. Med Sci Sports Exerc 24: 197–205,
1982. 1992.
66. Gibson MC and Schultz E. Age-related differences in abso- 87. Irintchev A, Zeschnigk M, Starzinski-Powitz A, and
lute numbers of skeletal muscle satellite cells. Muscle Nerve 6: Wernig A. Expression pattern of M-cadherin in normal, dener-
574–580, 1983. vated, and regenerating mouse muscles. Dev Dyn 199: 326–337,
67. Goodell MA, Brose K, Paradis G, Conner AS, and Mulli- 1994.
gan RC. Isolation and functional properties of murine hema- 88. Jackson KA, Mi T, and Goodell MA. Hematopoietic potential
topoietic stem cells that are replicating in vivo. J Exp Med 183: of stem cells isolated from murine skeletal muscle. Proc Natl
1797–1806, 1996. Acad Sci USA 96: 14482–14486, 1999.
68. Goodell MA, Rosenzweig M, Kim H, Marks DF, DeMaria 89. Jesse TL, LaChance R, Iademarco MF, and Dean DC.
M, Paradis G, Grupp SA, Sieff CA, Mulligan RC, and Interferon regulatory factor-2 is a transcriptional activator in
Johnson RP. Dye efflux studies suggest that hematopoietic muscle where It regulates expression of vascular cell adhesion
stem cells expressing low or undetectable levels of CD34 anti- molecule-1. J Cell Biol 140: 1265–1276, 1998.
gen exist in multiple species. Nat Med 3: 1337–1345, 1997. 90. Johnson SE and Allen RE. Proliferating cell nuclear antigen
69. Grady RM, Merlie JP, and Sanes JR. Subtle neuromuscular (PCNA) is expressed in activated rat skeletal muscle satellite
defects in utrophin-deficient mice. J Cell Biol 136: 871–882, cells. J Cell Physiol 154: 39–43, 1993.
1997. 91. Johnson SE and Allen RE. Activation of skeletal muscle
70. Greene EA and Allen RE. Growth factor regulation of bovine satellite cells and the role of fibroblast growth factor receptors.
satellite cell growth in vitro. J Anim Sci 69: 146–152, 1991. Exp Cell Res 219: 449–453, 1995.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 549

92. Jones NC, Fedorov YV, Rosenthal RS, and Olwin BB. Met receptor in vivo reveals complex roles in muscle develop-
ERK1/2 is required for myoblast proliferation but is dispens- ment. Cell 87: 531–542, 1996.
able for muscle gene expression and cell fusion. J Cell Physiol 113. Maroto M, Reshef R, Munsterberg AE, Koester S, Gould-
186: 104–115, 2001. ing M, and Lassar AB. Ectopic Pax-3 activates MyoD and
93. Joubert Y and Tobin C. Testosterone treatment results in Myf-5 expression in embryonic mesoderm and neural tissue.
quiescent satellite cells being activated and recruited into cell Cell 89: 139–148, 1997.
cycle in rat levator ani muscle. Dev Biol 169: 286–294, 1995. 114. Marshall PA, Williams PE, and Goldspink G. Accumula-
94. Kadi F and Thornell LE. Concomitant increases in myo- tion of collagen and altered fiber-type ratios as indicators of
nuclear and satellite cell content in female trapezius muscle abnormal muscle gene expression in the mdx dystrophic mouse.
following strength training. Histochem Cell Biol 113: 99–103, Muscle Nerve 12: 528–537, 1989.
2000. 115. Martin JF, Li L, and Olson EN. Repression of myogenin
95. Kahn EB and Simpson SB. Satellite cells in mature, unin- function by TGF-␤1 is targeted at the basic helix-loop-helix
jured skeletal muscle of the lizard tail. Dev Biol 37: 219–223, motif and is independent of E2A products. J Biol Chem 267:
1974. 10956–10960, 1992.
96. Kami K and Senba E. Localization of leukemia inhibitory 116. Matsuda R, Nishikawa A, and Tanaka H. Visualization of
factor and interleukin-6 messenger ribonucleic acids in regen- dystrophic muscle fibers in mdx mouse by vital staining with
erating rat skeletal muscle. Muscle Nerve 21: 819–822, 1998. Evans blue: evidence of apoptosis in dystrophin-deficient mus-
97. Katagiri T, Akiyama S, Namiki M, Komaki M, Yamaguchi cle. J Biochem (Tokyo) 118: 959–964, 1995.
A, Rosen V, Wozney JM, Fujisawa-Sehara A, and Suda T. 117. Mauro A. Satellite cell of skeletal muscle fibers. J Biophys
Bone morphogenetic protein-2 inhibits terminal differentiation Biochem Cytol 9: 493–498, 1961.
of myogenic cells by suppressing the transcriptional activity of 118. McGeachie JK. Sustained cell proliferation in denervated
MyoD and myogenin. Exp Cell Res 230: 342–351, 1997. skeletal muscle of mice. Cell Tissue Res 257: 455–457, 1989.
98. Katz FRS. The termination of the afferent nerve fiber in the 119. Megeney LA, Kablar B, Garrett K, Anderson JE, and
muscle spindle of the frog. Philos Trans R Soc Lond B Biol Sci Rudnicki MA. MyoD is required for myogenic stem cell func-
243: 221–225, 1961. tion in adult skeletal muscle. Genes Dev 10: 1173–1183, 1996.
99. Kocamis H, McFarland DC, and Killefer J. Temporal ex- 120. Melone MA, Peluso G, Galderisi U, Petillo O, and Cotrufo
pression of growth factor genes during myogenesis of satellite R. Increased expression of IGF-binding protein-5 in Duchenne
cells derived from the biceps femoris and pectoralis major muscular dystrophy (DMD) fibroblasts correlates with the fi-
muscles of the chicken. J Cell Physiol 186: 146–152, 2001. broblast-induced downregulation of DMD myoblast growth: an
100. Koishi K, Zhang M, McLennan IS, and Harris AJ. MyoD in vitro analysis. J Cell Physiol 185: 143–153, 2000.
protein accumulates in satellite cells and is neurally regulated 121. Merly F, Lescaudron L, Rouaud T, Crossin F, and Gar-
in regenerating myotubes and skeletal muscle fibers. Dev Dyn dahaut MF. Macrophages enhance muscle satellite cell prolif-
eration and delay their differentiation. Muscle Nerve 22: 724–
202: 244–254, 1995.
732, 1999.
101. Kurek JB, Bower JJ, Romanella M, Koentgen F, Murphy
122. Michalopoulos GK and Zarnegav R. Hepatocyte growth
M, and Austin L. The role of leukemia inhibitory factor in
factor. Hepatology 15: 149–155, 1992.
skeletal muscle regeneration. Muscle Nerve 20: 815–822, 1997.
123. Miller JB, Schaefer L, and Dominov JA. Seeking muscle
102. Kuschel R, Yablonka-Reuveni Z, and Bornemann A. Sat-
stem cells. Curr Top Dev Biol 43: 191–219, 1999.
ellite cells on isolated myofibers from normal and denervated
124. Miller KJ, Thaloor D, Matteson S, and Pavlath GK. Hepa-
adult rat muscle. J Histochem Cytochem 47: 1375–1384, 1999.
tocyte growth factor affects satellite cell activation and differ-
103. Lassar AB, Davis RL, Wright WE, Kadesch T, Murre C,
entiation in regenerating skeletal muscle. Am J Physiol Cell
Voronova A, Baltimore D, and Weintraub H. Functional Physiol 278: C174–C181, 2000.
activity of myogenic HLH proteins requires hetero-oligomeriza- 125. Mitchell CA, McGeachie JK, and Grounds MD. The exog-
tion with E12/E47-like proteins in vivo. Cell 66: 305–315, 1991. enous administration of basic fibroblast growth factor to regen-
104. Lawson-Smith MJ and McGeachie JK. The identification of erating skeletal muscle in mice does not enhance the process of
myogenic cells in skeletal muscle, with emphasis on the use of regeneration. Growth Factors 13: 37–55, 1996.
tritiated thymidine autoradiography and desmin antibodies. J 126. Molkentin JD and Olson EN. Defining the regulatory net-
Anat 192: 161–171, 1998. works for muscle development. Curr Opin Genet Dev 6: 445–
105. Le Douarin N and Barq G. Use of Japanese quail cells as 453, 1996.
“biological markers” in experimental embryology. CR Acad Sci 127. Monaco AP, Neve RL, Colletti-Feener C, Bertelson CJ,
Hebd Seances Acad Sci D 269: 1543–1546, 1969. Kurnit DM, and Kunkel LM. Isolation of candidate cDNAs
106. Lee JY, Qu-Petersen Z, Cao B, Kimura S, Jankowski R, for portions of the Duchenne muscular dystrophy gene. Nature
Cummins J, Usas A, Gates C, Robbins P, Wernig A, and 323: 646–650, 1986.
Huard J. Clonal isolation of muscle-derived cells capable of 128. Moss FP and Leblond CP. Satellite cells as the source of
enhancing muscle regeneration and bone healing. J Cell Biol nuclei in muscles of growing rats. Anat Rec 170: 421–435, 1971.
150: 1085–1100, 2000. 129. Mozdziak PE, Pulvermacher PM, and Schultz E. Unload-
107. Lefaucheur JP and Sebille A. The cellular events of injured ing of juvenile muscle results in a reduced muscle size 9 wk
muscle regeneration depend on the nature of the injury. Neu- after reloading. J Appl Physiol 88: 158–164, 2000.
romuscul Disord 5: 501–509, 1995. 130. Muir AR, Kanji AH, and Allbrook D. The structure of the
108. Lemischka I. The power of stem cells reconsidered? Proc Natl satellite cells in skeletal muscle. J Anat 99: 435–444, 1965.
Acad Sci USA 96: 14193–14195, 1999. 131. Munsterberg AE and Lassar AB. Combinatorial signals
109. LeRoith D, McGuinness M, Shemer J, Stannard B, Lanau from the neural tube, floor plate and notochord induce myo-
F, Faria TN, Kato H, Werner H, Adamo M, and Roberts genic bHLH gene expression in the somite. Development 121:
CT. Insulin-like growth factors. Biol Signals 1: 173–181, 1992. 651–660, 1995.
110. Lescaudron L, Peltekian E, Fontaine-Perus J, Paulin D, 132. Musaro A, McCullagh K, Paul A, Houghton L, Do-
Zampieri M, Garcia L, and Parrish E. Blood borne macro- browolny G, Molinaro M, Barton ER, Sweeney HL, and
phages are essential for the triggering of muscle regeneration Rosenthal N. Localized Igf-1 transgene expression sustains
following muscle transplant. Neuromuscul Disord 9: 72–80, hypertrophy and regeneration in senescent skeletal muscle.
1999. Nat Genet 27: 195–200, 2001.
111. Lu DX, Huang SK, and Carlson BM. Electron microscopic 133. Nathan CF. Secretory products of macrophages. J Clin Invest
study of long-term denervated rat skeletal muscle. Anat Rec 79: 319–326, 1987.
248: 355–365, 1997. 134. Nicolas N, Gallien CL, and Chanoine C. Analysis of MyoD,
112. Maina F, Casagranda F, Audero E, Simeone A, Comoglio myogenin, and muscle-specific gene mRNAs in regenerating
PM, Klein R, and Ponzetto C. Uncoupling of Grb2 from the Xenopus skeletal muscle. Dev Dyn 207: 60–68, 1996.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


550 INVITED REVIEW

135. Nnodim JO. Satellite cell numbers in senile rat levator ani 157. Rudnicki MA, Schnegelsberg PN, Stead RH, Braun T,
muscle. Mech Ageing Dev 112: 99–111, 2000. Arnold HH, and Jaenisch R. MyoD or Myf-5 is required for
136. Odelberg SJ, Kollhoff A, and Keating MT. Dedifferentia- the formation of skeletal muscle. Cell 75: 1351–1359, 1993.
tion of mammalian myotubes induced by msx1. Cell 103: 1099– 158. Saito Y. Muscle fibre type differentiation and satellite cell
1109, 2000. population in Werdnig-Hoffmann disease. J Neurol Sci 68:
137. Olson EN. MyoD family: a paradigm for development? Genes 75–87, 1985.
Dev 4: 1454–1461, 1990. 159. Sakuma K, Watanabe K, Sano M, Kitajima S, Sakamoto
138. Olson EN, Brown D, Burgess R, and Cserjesi P. A new K, Uramoto I, and Totsuka T. The adaptive response of
subclass of helix-loop-helix transcription factors expressed in transforming growth factor-␤2 and -␤RII in the overloaded,
paraxial mesoderm and chondrogenic cell lineages. Ann NY regenerating and denervated muscles of rats. Acta Neuropathol
Acad Sci 785: 108–118, 1996. (Berl) 99: 177–185, 2000.
139. Ordahl CP. Myogenic shape-shifters. J Cell Biol 147: 695–698, 160. Scata KA, Bernard DW, Fox J, and Swain JL. FGF receptor
1999. availability regulates skeletal myogenesis. Exp Cell Res 250:
140. Ordahl CP and Le Douarin NM. Two myogenic lineages 10–21, 1999.
within the developing somite. Development 114: 339–353, 1992. 161. Schmalbruch H and Hellhammer U. The number of nuclei
141. Ordahl CP, Williams BA, and Denetclaw W. Determination in adult rat muscles with special reference to satellite cells.
and morphogenesis in myogenic progenitor cells: an experimen- Anat Rec 189: 169–175, 1977.
tal embryological approach. Curr Top Dev Biol 48: 319–367, 162. Schubert W, Zimmermann K, Cramer M, and Starzinski-
2000. Powitz A. Lymphocyte antigen Leu-19 as a molecular marker
142. Patapoutian A, Yoon JK, Miner JH, Wang S, Stark K, and of regeneration in human skeletal muscle. Proc Natl Acad Sci
Wold B. Disruption of the mouse MRF4 gene identifies multi- USA 86: 307–311, 1989.
ple waves of myogenesis in the myotome. Development 121: 163. Schultz E. Satellite cell proliferative compartments in growing
3347–3358, 1995. skeletal muscles. Dev Biol 175: 84–94, 1996.
143. Pavlath G. Isolation, purification and growth of skeletal mus- 164. Schultz E, Jaryszak DL, Gibson MC, and Albright DJ.
cle cells. In: Methods of Molecular Medicine: Human Cell Cul- Absence of exogenous satellite cell contribution to regeneration
ture Protocols, edited by Jones GE. Totawa, NJ: Humana, 1996, of frozen skeletal muscle. J Muscle Res Cell Motil 7: 361–367,
p. 307–318. 1986.
144. Pennica D, Shaw KJ, Swanson TA, Moore MW, Shelton 165. Schultz E, Jaryszak DL, and Valliere CR. Response of
DL, Zioncheck KA, Rosenthal A, Taga T, Paoni NF, and satellite cells to focal skeletal muscle injury. Muscle Nerve 8:
Wood WI. Cardiotrophin-1. Biological activities and binding to 217–222, 1985.
the leukemia inhibitory factor receptor/gp130 signaling com- 166. Schultz E and Lipton BH. Skeletal muscle satellite cells:
plex. J Biol Chem 270: 10915–10922, 1995. changes in proliferation potential as a function of age. Mech
Ageing Dev 20: 377–383, 1982.
145. Pirskanen A, Kiefer JC, and Hauschka SD. IGFs, insulin,
167. Schultz E and McCormick KM. Skeletal muscle satellite
Shh, bFGF, and TGF-␤1 interact synergistically to promote
cells. Rev Physiol Biochem Pharmacol 123: 213–257, 1994.
somite myogenesis in vitro. Dev Biol 224: 189–203, 2000.
168. Seale P and Rudnicki MA. A new look at the origin, function,
146. Putman CT, Dusterhoft S, and Pette D. Changes in satellite
and “stem-cell” status of muscle satellite cells. Dev Biol 218:
cell content and myosin isoforms in low frequency-stimulated
115–124, 2000.
fast muscle of hypothyroid rat. J Appl Physiol 86: 40–51, 1999.
169. Seale P, Sabourin LA, Girgis-Gabardo A, Mansouri A,
147. Rawls A, Morris JH, Rudnicki M, Braun T, Arnold HH,
Gruss P, and Rudnicki MA. Pax7 is required for the specifi-
Klein WH, and Olson EN. Myogenin’s functions do not over-
cation of myogenic satellite cells. Cell 102: 777–786, 2000.
lap with those of MyoD or Myf-5 during mouse embryogenesis.
170. Semsarian C, Wu MJ, Ju YK, Marciniec T, Yeoh T, Allen
Dev Biol 172: 37–50, 1995. DG, Harvey RP, and Graham RM. Skeletal muscle hyper-
148. Rawls A, Valdez MR, Zhang W, Richardson J, Klein WH, trophy is mediated by a Ca2⫹-dependent calcineurin signalling
and Olson EN. Overlapping functions of the myogenic bHLH pathway. Nature 400: 576–581, 1999.
genes MRF4 and MyoD revealed in double mutant mice. Devel- 171. Seward DJ, Haney JC, Rudnicki MA, and Swoap SJ.
opment 125: 2349–2358, 1998. bHLH transcription factor MyoD affects myosin heavy chain
149. Rawls A, Wilson-Rawls J, and Olson EN. Genetic regulation expression pattern in a muscle-specific fashion. Am J Physiol
of somite formation. Curr Top Dev Biol 47: 131–154, 2000. Cell Physiol 280: C408–C413, 2001.
150. Reeves I, Abribat T, Laramee P, Jasmin G, and Brazeau 172. Shay JW and Wright WE. Telomeres and telomerase: impli-
P. Age-related serum levels of insulin-like growth factor-I, -II cations for cancer and aging. Radiat Res 155: 188–193, 2001.
and IGF- binding protein-3 following myocardial infarction. 173. Sheehan SM and Allen RE. Skeletal muscle satellite cell
Growth Horm IGF Res 10: 78–84, 2000. proliferation in response to members of the fibroblast growth
151. Renault V, Piron-Hamelin G, Forestier C, DiDonna S, factor family and hepatocyte growth factor. J Cell Physiol 181:
Decary S, Hentati F, Saillant G, Butler-Browne GS, and 499–506, 1999.
Mouly V. Skeletal muscle regeneration and the mitotic clock. 174. Sheehan SM, Tatsumi R, Temm-Grove CJ, and Allen RE.
Exp Gerontol 35: 711–719, 2000. HGF is an autocrine growth factor for skeletal muscle satellite
152. Robertson TA, Maley MA, Grounds MD, and Papadimi- cells in vitro. Muscle Nerve 23: 239–245, 2000.
triou JM. The role of macrophages in skeletal muscle regen- 175. Smith TH, Block NE, Rhodes SJ, Konieczny SF, and
eration with particular reference to chemotaxis. Exp Cell Res Miller JB. A unique pattern of expression of the four muscle
207: 321–331, 1993. regulatory factor proteins distinguishes somitic from embry-
153. Rodrigues A and Schmalbruch H. Satellite cells and myo- onic, fetal and newborn mouse myogenic cells. Development
nuclei in long-term denervated rat muscles. Anat Rec 243: 117: 1125–1133, 1993.
430–437, 1995. 176. Snow MH. The effects of aging on satellite cells in skeletal
154. Rosenblatt J and Parry DJ. Gamma irradiation prevents muscles of mice and rats. Cell Tissue Res 185: 399–408, 1977.
compensatory hypertrophy of overload mouse extensor digito- 177. Spangrude GJ, Heimfeld S, and Weissman IL. Purification
rum longus muscle. J Appl Physiol 73: 2538–2543, 1992. and characterization of mouse hematopoietic stem cells. Science
155. Rosenblatt JD and Parry DJ. Adaptation of rat extensor 241: 58–62, 1988.
digitorum longus muscle to gamma irradiation and overload. 178. Spizz G, Roman D, Strauss A, and Olson EN. Serum and
Pflügers Arch 423: 255–264, 1993. fibroblast growth factor inhibit myogenic differentiation
156. Rosenblatt JD, Yong D, and Parry DJ. Satellite cell activity through a mechanism dependent on protein synthesis and
is required for hypertrophy of overloaded adult rat muscle. independent of cell proliferation. J Biol Chem 261: 9483–9488,
Muscle Nerve 17: 608–613, 1994. 1986.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.


INVITED REVIEW 551

179. Straub V, Rafael JA, Chamberlain JS, and Campbell KP. forced expression of MyoD. Proc Natl Acad Sci USA 86: 5434–
Animal models for muscular dystrophy show different patterns 5438, 1989.
of sarcolemmal disruption. J Cell Biol 139: 375–385, 1997. 194. Weissman IL. Stem cells: units of development, units of re-
180. Sunderland S. Traumatized nerves, roots and ganglia: mus- generation, and units in evolution. Cell 100: 157–168, 2000.
culoskeletal factors and neuropathological consequences. In: 195. Weissman IL. Translating stem and progenitor cell biology to
The Neurobiologic Mechanisms in Manipulative Therapy, ed- the clinic: barriers and opportunities. Science 287: 1442–1446,
ited by Korr IM. New York: Plenum, 1978, p. 137–166. 2000.
181. Tajbakhsh S, Rocancourt D, Cossu G, and Buckingham 196. Whitman M. Smads and early developmental signaling by the
M. Redefining the genetic hierarchies controlling skeletal myo- TGF␤ superfamily. Genes Dev 12: 2445–2462, 1998.
genesis: Pax-3 and Myf-5 act upstream of MyoD. Cell 89: 127– 197. Williams RS and Neufer PD. Regulation of gene expression
138, 1997. in skeletal muscle by contractile activity. In: Handbook of
182. Tatsumi R, Anderson JE, Nevoret CJ, Halevy O, and Physiology. Exercise: Regulation and Integration of Multiple
Allen RE. HGF/SF is present in normal adult skeletal muscle Systems. Bethesda, MD: Am. Physiol. Soc., 1995, sect. 12,
and is capable of activating satellite cells. Dev Biol 194: 114– chapt. 27, p. 1124–1150.
128, 1998. 198. Winchester PK and Gonyea WJ. A quantitative study of
183. Ullman M, Ullman A, Sommerland H, Skottner A, and satellite cells and myonuclei in stretched avian slow tonic
Oldfors A. Effects of growth hormone on muscle regeneration muscle. Anat Rec 232: 369–377, 1992.
and IGF-I concentration in old rats. Acta Physiol Scand 140: 199. Wokke JH, Van den Oord CJ, Leppink GJ, and Jenne-
521–525, 1990. kens FG. Perisynaptic satellite cells in human external inter-
184. Vaidya TB, Rhodes SJ, Taparowsky EJ, and Konieczny costal muscle: a quantitative and qualitative study. Anat Rec
SF. Fibroblast growth factor and transforming growth factor 223: 174–180, 1989.
beta repress transcription of the myogenic regulatory gene 200. Wright WE, Sassoon DA, and Lin VK. Myogenin, a factor
MyoD1. Mol Cell Biol 9: 3576–3579, 1989. regulating myogenesis, has a domain homologous to MyoD. Cell
185. Valdez MR, Richardson JA, Klein WH, and Olson EN. 56: 607–617, 1989.
Failure of Myf5 to support myogenic differentiation without 201. Yablonka-Reuveni Z and Rivera AJ. Temporal expression
myogenin, MyoD, and MRF4. Dev Biol 219: 287–298, 2000. of regulatory and structural muscle proteins during myogenesis
186. Vierck J, O’Reilly B, Hossner K, Antonio J, Byrne K, of satellite cells on isolated adult rat fibers. Dev Biol 164:
Bucci L, and Dodson M. Satellite cell regulation following 588–603, 1994.
myotrauma caused by resistance exercise. Cell Biol Int 24: 202. Yan Z, Biggs RB, and Booth FW. Insulin-like growth factor
263–272, 2000. immunoreactivity increases in muscle after acute eccentric
187. Viguie CA, Lu DX, Huang SK, Rengen H, and Carlson BM. contractions. J Appl Physiol 74: 410–414, 1993.
Quantitative study of the effects of long-term denervation on 203. Yang Q, Bassel-Duby R, and Williams RS. Transient ex-
the extensor digitorum longus muscle of the rat. Anat Rec 248: pression of a winged-helix protein, MNF-␤, during myogenesis.
346–354, 1997. Mol Cell Biol 17: 5236–5243, 1997.
188. Wanek LJ and Snow MH. Activity-induced fiber regeneration 204. Yang Q, Kong Y, Rothermel B, Garry DJ, Bassel-Duby R,
in rat soleus muscle. Anat Rec 258: 176–185, 2000. and Williams RS. The winged-helix/forkhead protein myocyte
189. Wang Y and Sassoon D. Ectoderm-mesenchyme and mesen- nuclear factor beta (MNF-␤) forms a co-repressor complex with
chyme-mesenchyme interactions regulate Msx- 1 expression mammalian sin3B. Biochem J 345: 335–343, 2000.
and cellular differentiation in the murine limb bud. Dev Biol 205. Yang XM, Vogan K, Gros P, and Park M. Expression of the
168: 374–382, 1995. met receptor tyrosine kinase in muscle progenitor cells in
190. Watkins SC and Cullen MJ. A quantitative study of myo- somites and limbs is absent in Splotch mice. Development 122:
nuclear and satellite cell nuclear size in Duchenne’s muscular 2163–2171, 1996.
dystrophy, polymyositis and normal human skeletal muscle. 206. Ye H, Kelly TF, Samadani U, Lim L, Rubio S, Overdier
Anat Rec 222: 6–11, 1988. DG, Roebuck KA, and Costa RH. Hepatocyte nuclear factor
191. Watt DJ, Morgan JE, Clifford MA, and Partridge TA. The 3/fork head homolog 11 is expressed in proliferating epithelial
movement of muscle precursor cells between adjacent regener- and mesenchymal cells of embryonic and adult tissues. Mol Cell
ating muscles in the mouse. Anat Embryol (Berl) 175: 527–536, Biol 17: 1626–1641, 1997.
1987. 207. Yun K and Wold B. Skeletal muscle determination and dif-
192. Weintraub H. The MyoD family and myogenesis: redundancy, ferentiation: story of a core regulatory network and its context.
networks, and thresholds. Cell 75: 1241–1244, 1993. Curr Opin Cell Biol 8: 877–889, 1996.
193. Weintraub H, Tapscott SJ, Davis RL, Thayer MJ, Adam 208. Zentella A and Massague J. Transforming growth factor beta
MA, Lassar AB, and Miller AD. Activation of muscle-specific induces myoblast differentiation in the presence of mitogens.
genes in pigment, nerve, fat, liver, and fibroblast cell lines by Proc Natl Acad Sci USA 89: 5176–5180, 1992.

J Appl Physiol • VOL 91 • AUGUST 2001 • www.jap.org

Downloaded from www.physiology.org/journal/jappl (189.120.075.241) on July 11, 2019.

You might also like