Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Asymmetric Self-Renewal and Commitment

of Satellite Stem Cells in Muscle


Shihuan Kuang,1 Kazuki Kuroda,1 Fabien Le Grand,1 and Michael A. Rudnicki1,2,*
1
The Sprott Center for Stem Cell Research, Ottawa Health Research Institute, Molecular Medicine Program, 501 Smyth Road,
Ottawa, ON K1H 8L6, Canada
2
University of Ottawa, Department of Medicine, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
*Correspondence: mrudnicki@ohri.ca
DOI 10.1016/j.cell.2007.03.044

SUMMARY lating satellite cell self-renewal and differentiation remain


poorly understood (McKinnell et al., 2005).
Satellite cells play a central role in mediating the Early experiments using quail-chick chimeras sug-
growth and regeneration of skeletal muscle. gested that satellite cells were derived from the somite
However, whether satellite cells are stem cells, (Armand et al., 1983). Recent experiments support
committed progenitors, or dedifferentiated this work and indicate that a significant proportion of
myoblasts has remained unclear. Using Myf5- the progenitors of satellite cells originate in embryonic
somites as Pax3/Pax7-expressing cells (Ben-Yair and
Cre and ROSA26-YFP Cre-reporter alleles,
Kalcheim, 2005; Gros et al., 2005; Kassar-Duchossoy
we observed that in vivo 10% of sublaminar
et al., 2005; Relaix et al., 2005; Schienda et al., 2006). In
Pax7-expressing satellite cells have never ex- addition, satellite cells may also be derived from cells
pressed Myf5. Moreover, we found that Pax7+/ associated with the embryonic vasculature including
Myf5 satellite cells gave rise to Pax7+/Myf5+ the dorsal aorta (De Angelis et al., 1999) and from other
satellite cells through apical-basal oriented adult stem cells during regeneration (Asakura et al.,
divisions that asymmetrically generated a basal 2002; LaBarge and Blau, 2002; Polesskaya et al., 2003).
Pax7+/Myf5 and an apical Pax7+/Myf5+ cells. However, whether satellite cells are stem cells, committed
Prospective isolation and transplantation into progenitors, or dedifferentiated myoblasts remains
muscle revealed that whereas Pax7+/Myf5+ unresolved.
cells exhibited precocious differentiation, Pax7+/ Satellite cells are widely considered to be a homoge-
neous population of committed muscle progenitors (Bis-
Myf5 cells extensively contributed to the satel-
choff, 1994). However, several studies have raised the
lite cell reservoir throughout the injected mus-
possibility that the satellite cell compartment may be a het-
cle. Therefore, we conclude that satellite cells erogeneous population. Radioisotope labeling of growing
are a heterogeneous population composed of rat muscle revealed that satellite cells are a mixture of
stem cells and committed progenitors. These 80% fast-cycling cells and 20% slow-cycling ‘‘reserve
results provide critical insights into satellite cells’’ (Schultz, 1996). Examination of the expression of
cell biology and open new avenues for thera- satellite cell markers CD34, M-cadherin, and Myf5-nLacZ
peutic treatment of neuromuscular diseases. in freshly prepared myofibers suggested that a subpopula-
tion of satellite cells exhibits heterogeneous expression of
these markers (Beauchamp et al., 2000). However, the
INTRODUCTION molecular identity of any potential subpopulations has
not been defined, and the prospective isolation and char-
Satellite cells reside in a niche beneath the basal lamina acterization of these cells has not been achieved.
closely juxtaposed to the muscle fiber, and are responsi- Transplantation of the cultured primary myoblasts into
ble for the growth, maintenance, and repair of postnatal regenerating muscle typically results in extensive loss of
skeletal muscle. Satellite cells are normally mitotically qui- the transplanted cells, terminal differentiation of the
escent but are activated (i.e., enter the cell cycle) in re- surviving cells, and virtually no contribution to the satellite
sponse to stress induced by weight bearing or by trauma cell compartment (Beauchamp et al., 1999; El Fahime
such as injury. The descendants of activated satellite cells, et al., 2003; Fan et al., 1996; Hodgetts et al., 2000; Qu
called myogenic precursor cells, undergo multiple rounds et al., 1998; Rando and Blau, 1994). By contrast, experi-
of division prior to terminal differentiation and fusion to ments involving transplant of intact fibers carrying satellite
form multinucleated myofibers. Activated satellite cells cells (Collins et al., 2005) or fresh isolated satellite cells
also generate progeny that restore the pool of quiescent (Montarras et al., 2005) have suggested that at least
satellite cells. However, the molecular mechanisms regu- some portion of satellite cells have the capacity to

Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc. 999


repopulate the satellite cell compartment as well as exten-
sively contribute to regenerating muscle.
In the present study, we report the existence of hierar-
chical subpopulations of satellite cells that are distinct in
phenotype and function. We document that sublaminar
satellite cells expressing Pax7 are heterogeneous based
on Myf5 expression. Satellite cells that express Pax7 but
not Myf5 give rise to Myf5-expressing cells through subla-
minar cell divisions in a basal-apical orientation. Finally,
we observe that Pax7+/Myf5 satellite cells are capable
of efficiently contributing to the satellite cell reservoir
following prospective isolation and transplantation into
Pax7 / muscle. Together, these experiments demon-
strate that satellite cells are a heterogeneous population
composed of stem cells and committed myogenic
progenitors.

RESULTS

Satellite Cells Are Heterogeneous


by Myf5 Expression
Satellite cells uniformly express Pax7 (Seale et al., 2000)
and have been suggested to express the Myf5-nLacZ
knockin allele in the quiescent sublaminar state (Beau-
champ et al., 2000). We hypothesized that Myf5 transcrip-
tion occurs in satellite cells that had undergone commit-
ment to the myogenic lineage. We reasoned that if we
could detect satellite cells that had not expressed Myf5,
these would represent a candidate stem cell of the satellite
cell compartment.
We first examined whether all satellite cells expressing
Pax7 also express Myf5 in single myofiber preparations
fixed immediately following isolation from extensor digito-
rum longus (EDL) muscles of Myf5-nLacZ mice. Immuno-
histological analysis revealed that the majority of satellite
cells contained readily detectable levels of Pax7 and
b-Gal proteins. Notably, 13 ± 4% (n = 3 mice, > 100 cells/
mouse) of Pax7+ satellite cells did not contain detectable
levels of b-Gal, indicating that Myf5 was not uniformly
expressed (Figure 1A).
To investigate whether the Pax7+/b-Gal satellite cells
reflect a transient downregulation of Myf5 or represent
a distinct population that never expressed Myf5, we em-
ployed the Cre-LoxP system to genetically mark cells
that have expressed Myf5. We generated heterozygous Figure 1. Satellite Cells Are a Heterogeneous Population
mice carrying a Myf5-Cre allele (Tallquist et al., 2000) Based on Myf5 Expression
and a Cre-dependent YFP reporter knocked into the ubiq- (A) 87% of Pax7+ satellite cells expressed b-Gal (left column), and 13%
uitously expressed ROSA26 locus (Srinivas et al., 2001; of Pax7+ cells were b-Gal (right column, n = 3 mice) on single fibers
isolated from Myf5-nLacZ mice.
Figure S1A). In the Myf5-Cre/ROSA-YFP mice, any satel-
(B) 90% of Pax7+ cells expressed YFP (left column), and 10% of Pax7+
lite cells that have once expressed Myf5-Cre should cells were YFP (right column, n = 18 mice) on single fibers isolated
express YFP irreversibly. Conversely, any cells that are from Myf5-Cre/ROSA-YFP mice.
YFP should have never expressed Myf5-Cre in the past. (C and D) Total number of Pax7+ satellite cells per EDL myofiber and
Analysis of myofibers isolated from Myf5-Cre/ROSA- percentage of Pax7+/YFP cells at different ages (n = 3, 4, 6, and 4
YFP EDL muscle demonstrated that 90 ± 2% of satellite mice for 0.5-, 1-, 2-, and 6-month-old mice, respectively). Error bars
indicate standard error of mean (SEM).
cells expressing Pax7 also expressed YFP. Importantly,
(E) Sublaminar localization of Pax7+/YFP (left) and Pax7+/YFP+ (right)
10 ± 2% (n = 18 mice, >100 cells/mouse) of Pax7+ satellite cells.
cells did not contain detectable levels of YFP, confirming Scale bars are 10 mm in (A) and (B) and 25 mm in (E).
that these cells never expressed Myf5 (Figure 1B). The

1000 Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc.


relative proportion of Pax7+/YFP satellite cells persisted selection marker and further narrowed the gating based
in muscle in mice up to 6 months old (Figures 1C and 1D). on the side-scatter (SSC) range of satellite cells.
Similar proportions of b-Gal-positive and -negative Next, we isolated the YFP+/a7b1-int+/Lin and the
satellite cells were observed when Myf5-Cre/ROSA26R3 YFP /a7b1-int+/Lin (termed YFP+ and YFP hereafter)
reporter mice that express b-Gal only in the presence of cells by FACS (Figure 2A). Immunostaining of freshly
Myf5-Cre were examined (Figure S1B). By contrast, all sorted cells revealed that 90 ± 1% (n = 3) of the YFP+ cells
Pax7+ satellite cells were also YFP+ in Pax3-Cre/ROSA- expressed detectable levels of Pax7, whereas 67 ± 9%
YFP mice (n = 4 mice, data not shown), confirming the (n = 3) of YFP cells expressed Pax7 (Figure 2B). There-
efficacy of the Cre-LoxP system and the notion that fore, we established an approach for the prospective iso-
satellite cells are derived from embryonic Pax3+/Pax7+ lation of the different satellite cell subpopulations with
progenitors. a high degree of purity.
To ask whether the Pax7+/Myf5 represent newborn RT-PCR analysis of YFP+ cells confirmed expression of
satellite cells that have never undergone activation, we Pax7, Myf5, Cre, and YFP mRNAs. By contrast, YFP cells
treated muscles with cardiotoxin (CTX) to induce the acti- expressed similar levels of Pax7 but did not express Myf5,
vation of satellite cells and muscle regeneration. We ob- Cre, or YFP (Figure 2C). Importantly, Myf5 protein was
served substantial numbers of both Pax7+/YFP and readily detectable in the FACS-purified YFP+ but not in
Pax7+/b-Gal cells in regenerating Myf5-Cre/ROSA-YFP the YFP cells (Figure 2D). Together, these data directly
and Myf5-nLacZ muscles, respectively (Figure S2). Many confirmed that the YFP cells did not express Myf5 at
of the Pax7+/b-Gal cells had incorporated BrdU, indicat- either mRNA or protein level.
ing they were undergoing DNA synthesis and hence were
progressing through the cell cycle (Figure S2B). Therefore,
the lack of Myf5 expression in satellite cells does not Pax7+/Myf5 Cells Give Rise to Pax7+/Myf5+ Cells
equate to quiescence. To investigate the developmental origin and relationship
To confirm the satellite cell identity of the Pax7+/YFP between the Pax7+/Myf5 and Pax7+/Myf5+ cells, we ex-
cells, we triple labeled Myf5-Cre/ROSA-YFP muscle sec- amined the satellite cell progenitors in the somites and
tions with antibodies to Pax7, YFP, and laminin. All Pax7+/ limb buds of E11.5 embryos. Interestingly, about 10% of
YFP cells were located beneath the basal lamina in a sat- Pax7-expressing cells in the dermamyotome and migrat-
ellite cell position (Figure 1E). Furthermore, Pax7+/YFP ing progenitors near the limb bud were YFP among the
and Pax7+/b-Gal cells were found to express typical sat- large majority of YFP+ progenitors (Figure S5A and S5B).
ellite cell markers including CD34, M-Cad, Syn4, and To analyze the clonal growth of satellite cells on isolated
NCAM (Figure S3). These data confirm that sublaminar myofibers in vitro, individual myofibers were isolated from
satellite cells are a heterogeneous population on the basis Myf5-Cre/ROSA-YFP EDL muscle and cultured in sus-
of Myf5 expression. pension under growth conditions for 1–3 days. Typically,
satellite cells undergo their first cell division at around
24 hr and form 4–8 cell aggregates within 3 days. Live-
Prospective Isolation of Pax7+/YFP and Pax7+/YFP+ imaging analysis confirmed that these aggregates were
Satellite Cells of clonal origin (not shown). After 3 days of culture almost
To further characterize Pax7+/YFP and Pax7+/YFP+ sat- all clones uniformly expressed YFP, but the levels of Pax7
ellite cells, we developed a method to prospectively iso- within each clone were variable (Figure 3A). Importantly,
late the different satellite cell subpopulations based on nearly 10% of the clones contained both Pax7+/YFP
the previously reported approach using fluorescence- and Pax7+/YFP+ cells at day 3 (Figure 3B) and day 2
activated cell sorting (FACS) with antibody reactive with (Figure 3C; 6 out of 70 clones from 4 experiments). Since a
the cell-surface protein a7-integrin (int; Blanco-Bose colony of mixed YFP+ and YFP cells can only be derived
et al., 2001). Immunostaining of isolated myofibers con- from a common YFP progenitor, these results indicate
firmed that a7- and its dimerization partner b1-int were that Pax7+/Myf5 cells can form additional Pax7+/Myf5
expressed on both YFP and YFP+ satellite cells (Figures satellite cells as well as give rise to Pax7+/Myf5+ satellite
S4A and S4B). cells.
Initially, we employed positive selection for a7-int, to- To confirm that Pax7+/Myf5 cells give rise to Pax7+/
gether with negative selection (Lin , for Sca1, CD31, Myf5+ cells, we conducted time-lapse imaging analysis
CD45). FACS analysis of mononuclear cells from Myf5- of satellite cell division on cultured Myf5-Cre/ROSA-YFP
Cre/ROSA-YFP muscle revealed that the a7-int+ fraction myofibers. We observed that single YFP satellite cells di-
contained both YFP+ and YFP cells (Figure S4B, right rectly gave rise to one YFP and one YFP+ cell (Figure 4;
column). RT-PCR analysis indicated that mRNA for Movie S1). Furthermore, we examined expression of
Pax7, Myf5, and Cre were only present in the a7-int+ frac- MyoD, an established marker for myogenic commitment
tion (Figure S4C), suggesting that the a7-int+ fraction con- and differentiation. Notably, Pax7+/YFP cells did not ex-
tained all the Pax7+ satellite cells. However, only 20 ± 5% press MyoD (Figure S5C), consistent with the hypothesis
(n = 4) FACS-isolated a7-int+Lin YFP cells were Pax7+; that Pax7+/YFP cells have yet to undergo myogenic
therefore, we introduced b1-int as a second positive- commitment.

Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc. 1001


Figure 2. Isolation of Pax7+/YFP and Pax7+/YFP+ Cells from Myf5-Cre/ROSA-YFP Muscle
(A) Top panels: FACS profile of cells from control mice with no staining. Bottom panels: FACS profile of Myf5-Cre/ROSA-YFP cells stained with PE
conjugated to Sca-1, CD31, CD45 (PE-Lin), a7-integrin, and b1-integrin. The gates used to prospectively isolate a7-int+/b1-int+/Lin /YFP and
a7-int+/b1-int+/Lin /YFP+ cells are shown by boxed or circled areas. The bottom right panel shows the a7-int and YFP profile after gating.
(B) Pax7 expression in FACS-isolated YFP+ (upper panels) and YFP (lower panels) cells.
(C) RT-PCR analysis of sorted cells showing the absence of Myf5, Cre, and YFP expression in the YFP cells. Myoblasts indicates cultured primary
myoblasts isolated from Myf5-Cre/ROSA-YFP mice. NTC indicates no template control.
(D) Absence of Myf5 protein in FACS-isolated YFP cells.
Scale bars are 25 mm.

1002 Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc.


Figure 3. Clonal Analysis Implies a Devel-
opmental Relationship between Pax7+/
Myf5 and Pax7+/Myf5+ Satellite Cells
(A) Satellite-cell-derived clonal clusters from
Myf5-Cre/ROSA-YFP reporter mice after 3
days in suspended culture. Shown is a clonal
cluster of cells that uniformly express YFP.
(B) Shown is a clonal cluster containing both
Pax7+/YFP+ (arrow) and Pax7+/YFP (arrow-
head) cells.
(C) A couplet of sister cells with one Pax7+/
YFP cell (arrow head) and one Pax7+/YFP+
cell (arrow) after 2 days in culture.
(D) Extensive in vivo proliferation of satellite
cells on a regenerating EDL myofiber 4 days
post CTX injection. The majority of satellite
cells have undergone mitosis as indicated by
the presence of two adjacent Pax7+ nuclei
(double arrows) within a single satellite cell
niche.
(E) Couplet of identical sister cells that were
both Pax7+/YFP+.
(F) Couplet of sister cells with one Pax7+/YFP
cell (arrowhead) and one Pax7+/YFP+ cell (ar-
row).
(G) Satellite cell couplets expressed the meta-
phase mitosis marker phosphorylated his-
tone-H3 (H3-P).
(H) Satellite cell couplets expressed the prolif-
eration marker Ki67. b-Gal labeling denotes
Myf5-nLacZ expression.
Scale bars are 20 mm in (A), (B), and (D) and
10 mm in (C), (E), (F), (G), and (H).

To examine satellite cell divisions within their natural Pax7+/Myf5 and Pax7+/Myf5+ cells and therefore repre-
niche, we developed an approach to analyze satellite sent a reservoir that maintains a hierarchical composition
cell proliferation in vivo. Typically, induction of muscle of the satellite cell compartment.
regeneration with CTX results in death of myofibers and
massive immunoinfiltration, making it technically difficult Apical-Basal Location Determines Self-Renewal
to isolate viable myofibers. Therefore, we instead isolated versus Differentiation
fibers from the adjacent EDL muscle 4–5 days following We next investigated whether the fate of daughter cells is
CTX injection into the tibialis anterior (TA) muscle. We determined by their relative orientation within the satellite
found that this approach allowed us to isolate viable myo- cell compartment. Theoretically, sister cells can either be
fibers containing numerous satellite cells that were under- in a planar orientation where both cells remain in direct
going cell division in vivo (Figure 3D). contact with the basal lamina and host myofiber (Fig-
Numerous doublets of sister cells (double arrows in ure 5A) or in an apical-basal orientation where one daugh-
Figure 3D) were observed beneath the basal lamina on vi- ter cell is pushed toward the basal lamina and the other
able regenerating myofibers, which contained long arrays cell apically toward the host myofiber (Figure 5B).
of centrally located nuclei (Figure 3D). The close physical Examination of myofibers isolated from regenerating
proximity of the cells within each doublet, the expression EDL muscle revealed doublets of sister satellite cells be-
of proliferation markers in both cells (Ki67 and phospho- neath the basal lamina in both planar and apical-basal ori-
H3; Figures 3G and 3H), and their sublaminar localization entations (Figures 5C and 5D). Doublets of cells within the
within the same satellite cell niche (Refer to Figures 5C and satellite cell niche were predominantly planar in orienta-
5D) evidence the clonal origin of these doublet sister cells. tion (91%, n = 248 pairs). Planar divisions were observed
The vast majority of the doublets in Myf5-Cre/ROSA-YFP to give rise almost exclusively (92%, n = 89 pairs) to iden-
mice displayed identical expression of Pax7 and YFP tical daughter cells that were either both Pax7+/Myf5 or
(Figure 3E). Nonetheless, we identified doublets of prolif- both Pax7+/Myf5+, as reported by Myf5-nLacZ and
erating cells with one Pax7+/YFP cell and one Pax7+/ ROSA-YFP reporter mice (Figures 5A, 5E, and 5G).
YFP+ cell (Figure 3F), presumably derived from a common Strikingly, we observed apical-basal oriented doublets
Pax7+/YFP progenitor. where the sister cells assumed asymmetric cell fates
Altogether, our in vivo and in vitro analyses support (82%, n = 38 pairs; Figure 5B). The majority of apical-basal
the notion that Pax7+/Myf5 cells give rise to both oriented doublets contained a Pax7+/Myf5 cell against

Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc. 1003


Figure 4. Pax7+/Myf5 Satellite Cells Give Rise to Pax7+/
Myf5+ Satellite Cells
Time-lapse image analysis showing a YFP satellite cell (arrowhead at
T = 0) give rise to YFP+ (arrows) and YFP (arrowheads) daughter cells.
EDL myofibers isolated from Myf5-Cre/ROSA-YFP reporter mouse
were plated on Matrigel, and satellite cell growth was monitored with
a live imaging system. Cell division occurred at approximately 0 min
(around 36 hr in real time), and YFP expression became apparent in
the lower daughter cell at 240 min. Scale bar is 25 mm.

the basal surface and a Pax7+/Myf5+ cell located on the


apical side against the muscle fiber (Figures 5F, 5H, and
S6–S7). In addition, we observed rare events where the
basal cell expressed Pax7 and Myf5 and the apical cell
had downregulated Pax7 and was presumably undergo-
ing terminal differentiation (Figure S7).
Taken together, these results demonstrate that satellite
cell niche plays an important role in the maintenance of
stem cell identity of newly divided daughter cells. The Figure 5. Orientation of Cell Division within the Satellite Cell
Niche Determines Cell Fate
daughter cell attached to the basal lamina remains
Regenerating EDL myofibers were isolated from Myf5-Cre/ROSA-YFP
Myf5 , whereas the daughter that loses contact with the (C–F) or Myf5-nLacZ (G–H) reporter mice and fixed immediately for
basal lamina upregulates Myf5 and becomes a committed labeling of Pax7 and YFP or b-Gal. Myofibers were horizontally ori-
myogenic cell. ented in all panels.
(A and B) Schematics of planar (A) and apical-basal (B) oriented sister
cells and the corresponding frequency of symmetric (Sym) versus
asymmetric (Asym) expression of Myf5 (based on YFP or b-Gal). Sat-
Notch Regulation of Asymmetric Self-Renewal
ellite cells (red and green cells) are located beneath the basal lamina
To further explore the signaling mechanism underlying the (green) and are adjacent to the myofiber (brown).
apical-basal asymmetric self-renewal of satellite stem (C and D) Newly formed sister cells (double arrows) were both located
cells, we conducted gene-expression analysis of FACS- under the basal lamina (Lam) but were separated from myofiber by
purified YFP+ and YFP satellite cells. Importantly, YFP+ M-cadherin (M-Cad) regardless of their relative orientation (C, planar;
cells expressed the Notch ligand Delta-1 whereas YFP D, apical-basal).
(E and F) Examples of planar (double arrows) and apical-basal (arrow
cells did not (Figure 6A). Moreover, YFP cells but not
and arrowhead)-oriented Pax7+ sister cells that displayed symmetric
YFP+ cells expressed high levels of Notch-3 (Figure 6A). and asymmetric YFP expression, respectively.
By contrast, Notch-1 and Notch-2 receptors and the (G and H) Examples of planar (double arrows in G) and apical-basal
Notch signaling inhibitor Numb were equally expressed (arrow and arrowhead in H)-oriented Pax7+ sister cells that displayed
in both cell types. symmetric and asymmetric b-Gal expression, respectively. Centrally
Immunostaining of isolated myofibers revealed that located myonuclei indicated the regenerating status of the myofibers.
Scale bar is 12.5 mm.
Delta-1 was expressed at high levels in Pax7+/Myf5+ sat-
ellite cells but low levels in Pax7+/Myf5 cells (Figure 6B).
In cultured satellite cells, Delta-1 expression was upregu-
lated in Myf5+ but not in Myf5 cells (Figure 6C). In newly

1004 Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc.


Figure 6. A Role for Notch Signaling in
Self-Renewal of Pax7+/Myf5 Satellite
Cells
(A) RT-PCR analysis of FACS-purified YFP
and YFP+ satellite cells from Myf5-Cre/ROSA-
YFP muscles.
(B) Fresh isolated Myf5-nLacZ myofiber show-
ing Pax7+/b-Gal (arrowhead) and Pax7+/
b-Gal+ (arrow) satellite cells (B1) and their cor-
responding expressions of Delta-1 (B2).
(C) Satellite-cell-derived cultures showing dif-
ferential expression of Delta-1 in Pax7+/Myf5
(arrowhead) and Pax7+/Myf5+ (arrows) cells.
(D) High levels of Delta-1 were observed in
Pax7+/Myf5+ cells and low levels of Delta-1 in
Pax7+/Myf5 cells in newly formed sister dou-
blets.
(E) Abrogation of satellite cell self-renewal by
the g-secretase inhibitor DAPT. (E1–E2) Ex-
pression of the self-renewing marker Pax7
and myogenic differentiation marker MyoD in
colonies of satellite cell progenies in control
(E1) and DAPT (E2)-treated (3 day) myofiber
cultures. Notice the lack of Pax7 expression
in DAPT-treated cells. (E3–E5) Quantification
of cell number (E3) and the relative proportion
of Pax7+ (E4) and MyoD+ (E5) cells. Numbers
in the graphs indicate n. Error bars indicate
SEM.
Scale bar is 15 mm.

divided cells, high-level Delta-1 expression was observed fore, the transplanted cells can readily colonize the unoc-
specifically in Myf5-expressing cells (Figures 6C–6D). cupied satellite cell niche.
To directly test the role of Notch signaling in satellite cell Three weeks after transplantation (900–10,000 cells/TA
self-renewal, we treated proliferating satellite cells on muscle), we observed donor-derived cells as indicated by
freshly isolated myofibers with DAPT, an inhibitor of the expression of YFP or Pax7 (n = 6 animals in each
g-secretase that is necessary for Notch signaling. DAPT group). Both YFP+ and YFP donor cells were capable
treatment significantly reduced the total number of cells of undergoing terminal differentiation as revealed by the
after 3 days of culture. Importantly, Pax7+/MyoD satellite presence YFP+ myofibers (Figures 7A and 7B). Transplan-
cells were markedly reduced in number following DAPT tation of YFP+ cells typically gave rise to clusters of 20–60
treatment. By contrast, the percentage of Pax7 /MyoD+ YFP+ myofibers at the injection site, whereas transplanted
cells was significantly increased (Figure 6E). Together, YFP cells formed 0–22 YFP+ fibers. Furthermore, we ob-
these results strongly suggest that Notch signaling plays served in half of the transplanted muscles (n = 6) a massive
a critical role in the maintenance of Pax7+/YFP satellite accumulation of cells (DAPI nuclei in Figures 7A and 7E)
cells in an undifferentiated stem cell state. surrounding the newly formed YFP+ myofibers, suggest-
ing a host immune reaction against donor cells. Little or
Transplanted Pax7+/Myf5 Cells Extensively no immune reaction was observed in muscle transplanted
Contribute to the Satellite Cell Compartment with YFP cells (Figures 7B, 7D, and 7F). These data indi-
Our experiments provided compelling evidence that satel- cate that YFP+ donor cells readily differentiated following
lite cells are a hierarchal population composed of Pax7+/ transplantation.
Myf5 and Pax7+/Myf5+ satellite cells. To examine the Transplanted YFP cells efficiently gave rise to both
biological function of these two subpopulations of satellite Pax7+/YFP and Pax7+/YFP+ satellite cells (94 ± 32 cells/
cells, we transplanted FACS-purified YFP+ and YFP sat- section/10000 Pax7+ cells injected or 13 ± 3 cells/cross-
ellite cells from Myf5-Cre/ROSA-YFP mice into Pax7 / section, n = 6 mice). Many YFP satellite cells were
muscle and assessed: (1) their ability to terminally dif- observed to have migrated large distances into the host
ferentiate and (2) their contribution to the satellite cell muscle tissue, and were located in sublaminar positions
compartment. Notably, Pax7 / mice lack functional sat- (Figures 7D and 7F). By contrast, transplanted YFP+ cells
ellite cells (Kuang et al., 2006; Seale et al., 2000). There- only occasionally gave rise to Pax7+/YFP+ satellite cells

Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc. 1005


Finally, we assessed the relative repopulating efficiency
of YFP and YFP+ cells after engrafting myofibers isolated
from Myf5-Cre/ROSA-YFP mice into Pax7 / recipients.
We hypothesized that the relative ratio of reconstituted
YFP and YFP+ satellite cells should remain unchanged
or decline if both populations repopulate equally well
(given that some YFP can turn into YFP+ cells). Ten
freshly isolated EDL myofibers containing 7% YFP and
93% YFP+ satellite cells were grafted into the TA muscle
of Pax7 / mice. Four weeks later, the satellite cells de-
rived from the grafted fibers differentiated into bundles
of 20–100 YFP+ myofibers and also gave rise to numerous
Pax7+ satellite cells (14.7 ± 2.5 cells/cross-section, three
mice; Figure S8). Importantly, 24 ± 8% of engrafted
Pax7+ satellite cells remained YFP . Most of these YFP
satellite cells had migrated away from the injection site
and infiltrated the surrounding muscle tissue to repopulate
the host muscle. By contrast, almost all of the YFP+ satel-
lite cells remained at the injection site (Figure S8). Con-
sistent with the notion that Pax7+/Myf5 satellite cells
represent a stem cell population within the satellite cell
compartment, YFP satellite cells rapidly expanded in
number as they repopulated the vacant satellite cell niche
in Pax7 / muscle.
Together, these data indicate that transplanted Pax7+/
Myf5+ satellite cells preferentially undergo terminal differ-
entiation, whereas transplanted Pax7+/Myf5 satellite
cells extensively contribute to the satellite compartment
Figure 7. Transplanted Pax7+/Myf5 Cells Extensively Con- and give rise to both classes of satellite cells. Therefore,
tribute to the Satellite Cell Compartment
we conclude that the Pax7+/Myf5 satellite cells represent
TA muscles of Pax7 / mice were engrafted with freshly sorted YFP+
and YFP cells from Myf5-Cre/ROSA-YFP mice. Three weeks later,
a novel population of stem cells that actively maintain the
transplanted muscles were examined for the expression of YFP and homeostatic composition of stem cells and committed
Pax7. progenitors within the satellite cell niche.
(A and B) Transplanted YFP+ (A) and YFP (B) satellite cells both differ-
entiated into YFP+ myofibers; however, YFP satellite cells also gave
rise to several Pax7+/YFP cells associated with host (YFP ) myo-
DISCUSSION
fibers.
(C) Transplanted YFP+ cells gave rise to a satellite cell (arrowhead) that
was associated with a YFP+ fiber. Previously, our laboratory found that satellite cells uni-
(D) Transplanted YFP satellite cells gave rise to several Pax7+/YFP formly express the transcription factor Pax7 (Seale et al.,
(arrows) satellite cells that migrated onto host fibers (YFP ) and lo- 2000). Using a Myf5-Cre knockin allele and a ROSA-YFP
cated beneath the basal lamina (white staining). Notice that one Cre reporter, we observed that in vivo about 10% of satel-
Pax7+/YFP+ (arrowhead) cell closely associated with a YFP+ myofiber. lite cells express Pax7 but have never expressed Myf5.
(E) TA muscle transplanted with YFP+ satellite cells showing numerous
Moreover, we found that Pax7+/Myf5 satellite cells give
nuclei surrounding a YFP+ myofiber and two interstitial Pax7+/YFP+
cells (arrowheads) indicative of host immune rejection. rise to Pax7+/Myf5+ satellite cells through basal-apical
(F) TA muscle transplanted with YFP satellite cells showing several oriented divisions within the satellite cell niche. Prospec-
Pax7+/YFP cells (arrows) and two YFP+ cells (arrowheads) associ- tive isolation followed by transplantation confirmed that
ated with host (YFP ) myofibers and minimal immune rejection. Pax7+/Myf5+ satellite cells preferentially differentiate,
(G and H) Number of donor-derived satellite cells/TA cross-section af- whereas Pax7+/Myf5 satellite cells extensively contrib-
ter transplantation of YFP+ (G) and YFP (H) satellite cells (n = 6 mice).
ute to the satellite cell compartment. Therefore, anatomi-
Error bars indicate SEM.
Scale bars are 25 mm in (A) and (B) and 12.5 mm in (C)–(F).
cal and functional experiments provide compelling evi-
dence that the satellite cell population is composed of
hierarchal subpopulations of stem cells (Pax7+/Myf5 )
(12 ± 7 cells/section/10000 Pax7+ cells injected or 4 ± 2 and committed myogenic progenitors (Pax7+/Myf5+).
cells/cross-section, n = 6 mice), and these were virtually This discovery provides a new paradigm that redefines
all associated with the new YFP+ myofibers formed at satellite cell biology and opens new doors for therapeutic
the injection site (Figure 7C). Rare mononuclear YFP+ cells intervention for the treatment of devastating neuromus-
were also observed in interstitial spaces between fibers cular diseases such as Duchenne muscular dystrophy
(Figure 7E). (DMD).

1006 Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc.


Transplantation experiments have raised the possibility 2003; Fuchs et al., 2004; Knoblich, 2001; Knoblich et al.,
that only a portion of satellite cells exhibit repopulating ac- 1995; Moore and Lemischka, 2006; Rhyu et al., 1994).
tivity. Unlike cultured primary myoblasts (Collins et al., We observed that sublaminar satellite stem cells dis-
2005; Cousins et al., 2004; Heslop et al., 2001), some por- played elevated expression of Notch-3, whereas com-
tion of freshly isolated satellite cells or single myofibers mitted progenitors exhibited upregulation of Delta-1. We
carrying satellite cells give rise to satellite cells after trans- further demonstrated that in newly divided primary myo-
plantation (Collins et al., 2005; Montarras et al., 2005). blasts the level of Delta-1 is high in Myf5+ and low in
These results suggest that satellite cells in vivo contain Myf5 sister cells. Moreover, we observed that inhibition
a subpopulation of repopulating cells that are lost in of Notch signaling resulted in the loss of the Pax7+/
culture. Our findings are consistent with this body of Myf5 population. Therefore, activation of the Notch sig-
work and demonstrate that repopulating cells represent naling pathway, probably stimulated by Delta-1 ligand
a pool of premyogenic cell as evidenced by lack of Myf5 and Notch-3 receptor interactions, plays a positive role
expression. in the self-renewal of satellite stem cells.
Recent advances have provided important insights into Several lines of evidence implicate Notch signaling in
the role played by the microenvironment in regulating the regulation of satellite cell function. Delta-1 inhibits
stem cell identity and the asymmetric generation of com- myogenic differentiation in cell culture and during embry-
mitted daughter cells (Fuchs et al., 2004; Knoblich, 2001; onic myogenesis (Delfini et al., 2000; Kuroda et al., 1999).
Moore and Lemischka, 2006). In several other systems, Cosegregation of template DNA strand and asymmetric
stem cell polarity and spindle orientation relative to the distribution of Numb to one daughter cell were observed
basal lamina determines the fate of daughter cells. Stem during satellite cell division (Conboy et al., 2005; Shinin
cell divisions in a planar orientation are symmetric and et al., 2006). Furthermore, asymmetric distribution of
generate identical daughter stem cells. By contrast, Numb to the basal side of dividing cells occurs in the der-
stem cell divisions in an apical-basal orientation are asym- mamyotome during embryonic myogenesis (Holowacz
metric and give rise to a stem cell at the basal surface and et al., 2006; Venters and Ordahl, 2005). Finally, Inscute-
a committed daughter cell destined for differentiation on able and Numb are segregated to the apical and basal sib-
the apical surface (Fuchs et al., 2004; Knoblich, 2001; ling cells, respectively, during muscle differentiation in
Lechler and Fuchs, 2005). Therefore, the discovery of Drosophila, and perturbation of this segregation pattern
the stem cell niche provides an anatomical means to es- leads to the failure of muscle development (Carmena
tablish the identity of a candidate stem cell population et al., 1998). One of the challenges for future study is to
within a particular tissue. pinpoint the intrinsic and extrinsic molecules and signaling
Muscle satellite cells occupy a niche featuring a struc- pathways that control the asymmetric distribution of Delta
tural foundation for asymmetric self-renewal. The basal and Numb in satellite cells and how this asymmetry leads
side of satellite cells is covered by the basal lamina en- to cell fate segregation.
sheathing the muscle fiber, while the apical side of satellite BrdU-labeling experiments in freshly isolated satellite
cells is adjacent to the myofiber. The laminin receptor cells have suggested that template DNA cosegregates
a7b1 integrin is specifically expressed in satellite cells with Numb in label-retaining cells that express the self-re-
on the basal surface, whereas the cell-adhesion molecule newal marker Pax7 (Shinin et al., 2006). The relationship
M-cadherin is specifically expressed on the apical surface between Pax7+/Myf5 satellite cells and label-retaining
toward the muscle fiber (Bornemann and Schmalbruch, cells thus represents an interesting question. In prelimi-
1994; Burkin and Kaufman, 1999; Collo et al., 1993; nary short-term label-retention experiments, we applied
Irintchev et al., 1994). Our data document the asymmetric a short pulse of BrdU to label the newly synthesized
generation of Pax7+/Myf5 stem cells and Pax7+/Myf5+ DNA strand in proliferating satellite cells, which was
committed cells upon apical-basal oriented cell divisions followed by a 3 day chase. From cross-sections, we ob-
as well as symmetric generation of daughters upon planar served that 14% (n = 71) of Pax7+/Myf5 cells were
cell divisions. Notably, we observed that Pax7+/Myf5 BrdU+, whereas 41% (n = 309) of Pax7+/Myf5+ cells
cells remained at the basal surface, whereas Pax7+/Myf5+ were BrdU+. From single myofibers, we observed cose-
cells were localized to the apical surface against the mus- gregation of template DNA to Pax7+/Myf5 cells in 38%
cle fiber. Presumably the apical cell became committed (5 out of 13) of newly divided sister cells. By contrast,
due to loss of contact with the basal lamina and the extra- only 9% (2 out of 23) of newly divided Pax7+/Myf5+ cells
cellular matrix. These results suggest that the satellite cell displayed cosegregation of template DNA to one sister
niche plays an important role in regulating both the main- cell. These results are consistent with the observations
tenance of stem cell identity and the generation of com- of Shinin and coworkers (Shinin et al., 2006).
mitted daughter cells through asymmetric cell divisions. In studies of mdx mice, a mouse model of human DMD,
Within the stem cell niche, signaling pathways such as it has been noted that a radiation-resistant subpopulation
Wnt, Notch, BMP/TGFb, and STAT and proteins such as of satellite cells is depleted relative to wild-type muscle
Numb, PAR, PKCz, LGL, and NUMA have been implicated (Heslop et al., 2000). Moreover, satellite-cell-derived myo-
in the regulation of asymmetric cell division in model sys- blasts display accelerated differentiation kinetics when
tems (Betschinger and Knoblich, 2004; Betschinger et al., isolated from mdx mice (Yablonka-Reuveni and Anderson,

Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc. 1007


2006). These data suggest that under pathological condi- listed in Table S1. Secondary antibodies used were Alexa488,
tions, the equilibrium between stem cells and committed Alexa568, and Alexa647 conjugated to specific IgG types (Invitrogen
Molecular Probes) that matched the primary antibodies (Invitrogen
progenitors within the satellite cell compartment is per-
Molecular Probes). Zenon tricolor anti-Rabbit IgG labeling kit (Invitro-
turbed. It is interesting to speculate that this phenomenon gen Molecular Probes) and Avidin-conjugated fluorophores were
contributes to the progression and severity of DMD. also used where applicable. Nuclei were counterstained with DAPI.
It is worth mentioning that symmetric expansion of sat-
ellite stem cells and myogenic cells may represent a pri-
Cell Sorting, PCR Analysis, and Transplantation
mary means of cell division during CTX-induced muscle Cells were isolated from hindlimb muscle of 6- to 8-week-old mice as
regeneration, as manifested by the focal enrichment of previously described (Megeney et al., 1996). Erythrocytes were re-
Pax7+/YFP cells (Figure S2). We suspect that symmetric moved with the Red Blood Cell Lysing Buffer Hybri-Max (Sigma).
expansion of stem cells represents the primary mecha- Mononuclear cells were blocked with goat serum for 10 min and incu-
nism of self-renewal under certain devastating pathologi- bated with primary antibodies in DMEM with 2% FBS at 1–3 3 107 cell/ml
for 15 min at 4 C. Cells were briefly washed and incubated with
cal conditions, whereas asymmetric self-renewal occurs
appropriate secondary antibodies (1: 1000) at 4 C for 15 min. After
as the primary mechanism for the maintenance of the ho- staining, cells were washed, passed through 30 mm filters (Miltenyi
meostasis of satellite cells under normal physiological Biotec) and suspended at a concentration of 1 3 107 cells/ml. Cells
conditions. were separated on a MoFlo cytometer (DakoCytomation) equipped
The identification of a satellite stem cell represents an with three lasers. Sorting gates were strictly defined based on single
important advance in our understanding of satellite cell bi- antibody-stained control cells as well as the forward and SSC patterns
of satellite cells based on preliminary tests. Total RNA was prepared
ology and opens new avenues to explore for the treatment
from 2 3 104 of sorted cell populations by TRIzol (GIBCO) or MicroRNA
of degenerative neuromuscular diseases. Satellite stem kit (Qiagen). cDNA synthesis and PCR were performed as previously
cells could be used for direct transplantation into diseased described (Minoguchi et al., 1997). Primers are listed in Table S2.
muscle. Molecular characterization of satellite stem cells Equal numbers of FACS-purified YFP and YFP+ cells were injected
should lead to identification of markers for their prospec- with 0.5 cc insulin syringes into the left and right TA muscles of the
tive isolation from human muscle tissue. Alternatively, un- same Pax7 / mouse. The number of cells used for each injection
varied from 900–10,000 based on availability of FACS-purified cells.
derstanding the molecular regulation of satellite stem cell
Injected TA muscles were harvested 3 weeks later for analysis. For
symmetric versus asymmetric cell division will lead to
myofiber graft, 10 EDL myofibers were grafted together with 10 ml
identification of biologics or small drugs that specifically DMEM into the TA muscles of Pax7 / mice with custom-made glass
target the relevant pathway leading to satellite stem cell needles controlled by a microsyringe. Grafted muscles were collected
expansion. Future experiments will investigate both the 4 weeks later for analysis.
utility of satellite stem cells for cell transplantation and
the molecular control of cell fate determination. Microscopy and Live Imaging Analysis
Samples were visualized with a Zeiss Axioplan 2 microscope, and im-
EXPERIMENTAL PROCEDURES ages were acquired using a Zeiss AxioCam camera and the Axioview
3.1 software as previously described (Kuang et al., 2006). For clonal
Mice and Animal Care tracing of satellite cells, single myofiber cultures were monitored using
ROSA26-LacZ (ROSA26R3) reporter mice, mdx mice carrying homol- a Zeiss Axiovert 200 microscope equipped with an incubator (XL-3,
ogous mutated dystrophin gene, and Pax3-Cre mice strains were Zeiss), a CO2 controller, and a heating unit that cooperate to maintain
purchased from JaxMice (Jackson Laboratories, MI). Heterozygous 37 C and 5% CO2 within the incubator. Fluorescent and phase-
Myf5-nLacZ (Tajbakhsh et al., 1996) mice were directly used, whereas contrast images were recorded at one frame every 5 min using a
Myf5-Cre (Tallquist et al., 2000) heterozygous mice were bred with 203 objective (LD Plan-Neofluar N.A. 0.4) with a Zeiss AxioCam and
ROSA26-YFP (Srinivas et al., 2001), also referred to as ‘‘ROSA-YFP’’ the AxioView 4.5 software.
in the text, or ROSA26R3 reporter mice. Pax7 mutant mice carrying
the lacZ knockin mutation were maintained in an inbred 129SV/J back-
Supplemental Data
ground (Kuang et al., 2006). All mice are maintained inside a barrier
Supplemental Data include eight figures, two tables, and one movie
facility, and experiments were performed in accordance with the
and can be found with this article online at http://www.cell.com/cgi/
University of Ottawa regulations for animal care and handling.
content/full/129/5/999/DC1/.

Myofiber Isolation and Immunohistochemistry


Single myofibers were isolated from the EDL muscles as previously de- ACKNOWLEDGMENTS
scribed (Rosenblatt et al., 1995). To induce activation of satellite cells,
6- to 8-week-old Myf5-nLacz or Myf5-Cre/ROSA-YFP mice were We thank Caroline Vergette and Vanessa Seale for expert technical
injected with 25 ml CTX (10 mM, Latoxan, France) into the TA muscles assistance and Iain McKinnell and Mark Gillespie for careful reading
and examined 4 days later. For analysis of satellite cell proliferation of the manuscript. We thank Drs. P. Soriano for the Myf5-Cre mice,
in vitro, single myofibers were either plated on Matrigel-coated cham- F. Costantini for the ROSA-YFP mice, and S. Tajbakhsh for the
ber slides or cultured in suspension in 60 mm Petri dishes coated with Myf5-nLacZ mice. M.A.R. holds the Canada Research Chair in Molec-
horse serum to prevent fiber attachment (Kuang et al., 2006). Primary ular Genetics and is an International Research Scholar of the Howard
myoblasts were isolated from hindlimb muscles and cultured as previ- Hughes Medical Institute. This work was supported by grants to
ously reported (Megeney et al., 1996). M.A.R. from the Canadian Institutes of Health Research, the National
Histological processing and immunochemical labeling of cryo- Institutes of Health, Muscular Dystrophy Association, the Howard
sections, single myofibers, and cells were performed as previously Hughes Medical Institute, and the Canada Research Chair Program.
reported (Kuang et al., 2006). M.O.M. blocking kit (BMK-2202, Vector S.K. was supported by an NSERC Postdoctoral Fellowship.
Lab) was used in staining of muscle sections. Primary antibodies are The authors declare no conflict of interests.

1008 Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc.


Received: June 14, 2006 Delfini, M., Hirsinger, E., Pourquie, O., and Duprez, D. (2000). Delta
Revised: February 7, 2007 1-activated notch inhibits muscle differentiation without affecting
Accepted: March 7, 2007 Myf5 and Pax3 expression in chick limb myogenesis. Development
Published: May 31, 2007 127, 5213–5224.
El Fahime, E., Bouchentouf, M., Benabdallah, B.F., Skuk, D., Lafre-
REFERENCES niere, J.F., Chang, Y.T., and Tremblay, J.P. (2003). Tubulyzine, a novel
tri-substituted triazine, prevents the early cell death of transplanted
Armand, O., Boutineau, A.M., Mauger, A., Pautou, M.P., and Kieny, M. myogenic cells and improves transplantation success. Biochem. Cell
(1983). Origin of satellite cells in avian skeletal muscles. Arch. Anat. Biol. 81, 81–90.
Microsc. Morphol. Exp. 72, 163–181. Fan, Y., Maley, M., Beilharz, M., and Grounds, M. (1996). Rapid death
Asakura, A., Seale, P., Girgis-Gabardo, A., and Rudnicki, M.A. (2002). of injected myoblasts in myoblast transfer therapy. Muscle Nerve 19,
Myogenic specification of side population cells in skeletal muscle. 853–860.
J. Cell Biol. 159, 123–134. Fuchs, E., Tumbar, T., and Guasch, G. (2004). Socializing with the
Beauchamp, J.R., Morgan, J.E., Pagel, C.N., and Partridge, T.A. neighbors: stem cells and their niche. Cell 116, 769–778.
(1999). Dynamics of myoblast transplantation reveal a discrete minor- Gros, J., Manceau, M., Thome, V., and Marcelle, C. (2005). A common
ity of precursors with stem cell-like properties as the myogenic source. somitic origin for embryonic muscle progenitors and satellite cells. Na-
J. Cell Biol. 144, 1113–1122. ture 435, 954–958.
Beauchamp, J.R., Heslop, L., Yu, D.S., Tajbakhsh, S., Kelly, R.G., Wer-
Heslop, L., Morgan, J.E., and Partridge, T.A. (2000). Evidence for
nig, A., Buckingham, M.E., Partridge, T.A., and Zammit, P.S. (2000).
a myogenic stem cell that is exhausted in dystrophic muscle. J. Cell
Expression of CD34 and myf5 defines the majority of quiescent adult
Sci. 113, 2299–2308.
skeletal muscle satellite cells. J. Cell Biol. 151, 1221–1234.
Heslop, L., Beauchamp, J.R., Tajbakhsh, S., Buckingham, M.E., Par-
Ben-Yair, R., and Kalcheim, C. (2005). Lineage analysis of the avian
tridge, T.A., and Zammit, P.S. (2001). Transplanted primary neonatal
dermomyotome sheet reveals the existence of single cells with both
myoblasts can give rise to functional satellite cells as identified using
dermal and muscle progenitor fates. Development 132, 689–701.
the Myf5nlacZl+ mouse. Gene Ther. 8, 778–783.
Betschinger, J., Mechtler, K., and Knoblich, J.A. (2003). The Par com-
Hodgetts, S.I., Beilharz, M.W., Scalzo, A.A., and Grounds, M.D. (2000).
plex directs asymmetric cell division by phosphorylating the cytoskel-
Why do cultured transplanted myoblasts die in vivo? DNA quantifica-
etal protein Lgl. Nature 422, 326–330.
tion shows enhanced survival of donor male myoblasts in host mice
Betschinger, J., and Knoblich, J.A. (2004). Dare to be different: asym- depleted of CD4+ and CD8+ cells or Nk1.1+ cells. Cell Transplant. 9,
metric cell division in Drosophila, C. elegans and vertebrates. Curr. 489–502.
Biol. 14, R674–R685.
Holowacz, T., Zeng, L., and Lassar, A.B. (2006). Asymmetric localiza-
Bischoff, R. (1994). The satellite cell and muscle regeneration. In tion of numb in the chick somite and the influence of myogenic signals.
Myology, A.G. Engel and C. Franzini-Armstrong, eds. (New York: Dev. Dyn. 235, 633–645.
McGraw-Hill), pp. 97–118.
Irintchev, A., Zeschnigk, M., Starzinski-Powitz, A., and Wernig, A.
Blanco-Bose, W.E., Yao, C.C., Kramer, R.H., and Blau, H.M. (2001). (1994). Expression pattern of M-cadherin in normal, denervated, and
Purification of mouse primary myoblasts based on alpha 7 integrin ex- regenerating mouse muscles. Dev. Dyn. 199, 326–337.
pression. Exp. Cell Res. 265, 212–220.
Kassar-Duchossoy, L., Giacone, E., Gayraud-Morel, B., Jory, A.,
Bornemann, A., and Schmalbruch, H. (1994). Immunocytochemistry of Gomes, D., and Tajbakhsh, S. (2005). Pax3/Pax7 mark a novel popu-
M-cadherin in mature and regenerating rat muscle. Anat. Rec. 239, lation of primitive myogenic cells during development. Genes Dev.
119–125. 19, 1426–1431.
Burkin, D.J., and Kaufman, S.J. (1999). The alpha7beta1 integrin in
Knoblich, J.A. (2001). The Drosophila nervous system as a model for
muscle development and disease. Cell Tissue Res. 296, 183–190.
asymmetric cell division. Symp. Soc. Exp. Biol. 53, 75–89.
Carmena, A., Murugasu-Oei, B., Menon, D., Jimenez, F., and Chia, W.
Knoblich, J.A., Jan, L.Y., and Jan, Y.N. (1995). Asymmetric segrega-
(1998). Inscuteable and numb mediate asymmetric muscle progenitor
tion of Numb and Prospero during cell division. Nature 377, 624–627.
cell divisions during Drosophila myogenesis. Genes Dev. 12, 304–315.
Kuang, S., Charge, S.B., Seale, P., Huh, M., and Rudnicki, M.A. (2006).
Collins, C.A., Olsen, I., Zammit, P.S., Heslop, L., Petrie, A., Partridge,
Distinct roles for Pax7 and Pax3 in adult regenerative myogenesis.
T.A., and Morgan, J.E. (2005). Stem cell function, self-renewal, and be-
J. Cell Biol. 172, 103–113.
havioral heterogeneity of cells from the adult muscle satellite cell niche.
Cell 122, 289–301. Kuroda, K., Tani, S., Tamura, K., Minoguchi, S., Kurooka, H., and
Honjo, T. (1999). Delta-induced Notch signaling mediated by RBP-J
Collo, G., Starr, L., and Quaranta, V. (1993). A new isoform of the lam-
inhibits MyoD expression and myogenesis. J. Biol. Chem. 274,
inin receptor integrin alpha 7 beta 1 is developmentally regulated in
7238–7244.
skeletal muscle. J. Biol. Chem. 268, 19019–19024.
LaBarge, M.A., and Blau, H.M. (2002). Biological progression from
Conboy, I.M., Conboy, M.J., Wagers, A.J., Girma, E.R., Weissman,
adult bone marrow to mononucleate muscle stem cell to multinucleate
I.L., and Rando, T.A. (2005). Rejuvenation of aged progenitor cells by
muscle fiber in response to injury. Cell 111, 589–601.
exposure to a young systemic environment. Nature 433, 760–764.
Lechler, T., and Fuchs, E. (2005). Asymmetric cell divisions promote
Cousins, J.C., Woodward, K.J., Gross, J.G., Partridge, T.A., and Mor-
stratification and differentiation of mammalian skin. Nature 437, 275–
gan, J.E. (2004). Regeneration of skeletal muscle from transplanted
280.
immortalised myoblasts is oligoclonal. J. Cell Sci. 117, 3259–3269.
De Angelis, L., Berghella, L., Coletta, M., Lattanzi, L., Zanchi, M., McKinnell, I.W., Parise, G., and Rudnicki, M.A. (2005). Muscle stem
Cusella-De Angelis, M.G., Ponzetto, C., and Cossu, G. (1999). Skeletal cells and regenerative myogenesis. Curr. Top. Dev. Biol. 71, 113–130.
myogenic progenitors originating from embryonic dorsal aorta coex- Megeney, L.A., Kablar, B., Garrett, K., Anderson, J.E., and Rudnicki,
press endothelial and myogenic markers and contribute to postnatal M.A. (1996). MyoD is required for myogenic stem cell function in adult
muscle growth and regeneration. J. Cell Biol. 147, 869–878. skeletal muscle. Genes Dev. 10, 1173–1183.

Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc. 1009


Minoguchi, S., Taniguchi, Y., Kato, H., Okazaki, T., Strobl, L.J., Zim- limb muscle satellite and side population cells. Proc. Natl. Acad. Sci.
ber-Strobl, U., Bornkamm, G.W., and Honjo, T. (1997). RBP-L, a tran- USA 103, 945–950.
scription factor related to RBP-Jkappa. Mol. Cell. Biol. 17, 2679–2687.
Schultz, E. (1996). Satellite cell proliferative compartments in growing
Montarras, D., Morgan, J., Collins, C., Relaix, F., Zaffran, S., Cumano, skeletal muscles. Dev. Biol. 175, 84–94.
A., Partridge, T., and Buckingham, M. (2005). Direct isolation of satel-
lite cells for skeletal muscle regeneration. Science 309, 2064–2067. Seale, P., Sabourin, L.A., Girgis-Gabardo, A., Mansouri, A., Gruss, P.,
and Rudnicki, M.A. (2000). Pax7 is required for the specification of
Moore, K.A., and Lemischka, I.R. (2006). Stem cells and their niches.
myogenic satellite cells. Cell 102, 777–786.
Science 311, 1880–1885.
Polesskaya, A., Seale, P., and Rudnicki, M.A. (2003). Wnt signaling in- Shinin, V., Gayraud-Morel, B., Gomes, D., and Tajbakhsh, S. (2006).
duces the myogenic specification of resident CD45+ adult stem cells Asymmetric division and cosegregation of template DNA strands in
during muscle regeneration. Cell 113, 841–852. adult muscle satellite cells. Nat. Cell Biol. 8, 677–687.
Qu, Z., Balkir, L., van Deutekom, J.C., Robbins, P.D., Pruchnic, R., and Srinivas, S., Watanabe, T., Lin, C.S., William, C.M., Tanabe, Y., Jessell,
Huard, J. (1998). Development of approaches to improve cell survival T.M., and Costantini, F. (2001). Cre reporter strains produced by tar-
in myoblast transfer therapy. J. Cell Biol. 142, 1257–1267. geted insertion of EYFP and ECFP into the ROSA26 locus. BMC
Rando, T.A., and Blau, H.M. (1994). Primary mouse myoblast purifica- Dev. Biol. 1, 4.
tion, characterization, and transplantation for cell-mediated gene ther-
Tajbakhsh, S., Rocancourt, D., and Buckingham, M. (1996). Muscle
apy. J. Cell Biol. 125, 1275–1287.
progenitor cells failing to respond to positional cues adopt non-
Relaix, F., Rocancourt, D., Mansouri, A., and Buckingham, M. (2005). A myogenic fates in myf-5 null mice. Nature 384, 266–270.
Pax3/Pax7-dependent population of skeletal muscle progenitor cells.
Nature 435, 948–953. Tallquist, M.D., Weismann, K.E., Hellstrom, M., and Soriano, P. (2000).
Early myotome specification regulates PDGFA expression and axial
Rhyu, M.S., Jan, L.Y., and Jan, Y.N. (1994). Asymmetric distribution of
skeleton development. Development 127, 5059–5070.
numb protein during division of the sensory organ precursor cell con-
fers distinct fates to daughter cells. Cell 76, 477–491. Venters, S.J., and Ordahl, C.P. (2005). Asymmetric cell divisions are
Rosenblatt, J.D., Lunt, A.I., Parry, D.J., and Partridge, T.A. (1995). concentrated in the dermomyotome dorsomedial lip during epaxial pri-
Culturing satellite cells from living single muscle fiber explants. In Vitro mary myotome morphogenesis. Anat. Embryol. (Berl.) 209, 449–460.
Cell. Dev. Biol. 31, 773–779. Yablonka-Reuveni, Z., and Anderson, J.E. (2006). Satellite cells from
Schienda, J., Engleka, K.A., Jun, S., Hansen, M.S., Epstein, J.A., dystrophic (mdx) mice display accelerated differentiation in primary
Tabin, C.J., Kunkel, L.M., and Kardon, G. (2006). Somitic origin of cultures and in isolated myofibers. Dev. Dyn. 235, 203–212.

1010 Cell 129, 999–1010, June 1, 2007 ª2007 Elsevier Inc.

You might also like