You are on page 1of 13

REVIEW ARTICLE

Differential modulation of cell cycle progression


distinguishes members of the myogenic regulatory factor
family of transcription factors
Kulwant Singh1 and F. Jeffrey Dilworth1,2
1 Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, ON, Canada
2 Department of Cellular and Molecular Medicine, University of Ottawa, ON, Canada

Keywords The muscle-specific basic helix–loop–helix proteins MyoD, Myf5, myogenin


cell cycle; DNA repair; gene expression; (Myog) and MRF4 constitute the myogenic regulatory factor (MRF) fam-
MRF4; Myf5; MyoD; myogenesis; myogenin
ily of transcription factors that drive muscle gene expression during myo-
(Myog)
genesis. Having evolved from a single ancestral gene, the spatial and
Correspondence temporal specificity of expression for each family member has been used to
F. J. Dilworth, Ottawa Hospital Research define a hierarchical relationship between the four MRFs. Molecular char-
Institute, 501 Smyth Rd, Mailbox 511, acterization of two of the MRFs (MyoD and Myog) suggests an important
Ottawa, ON, Canada K1H 8L6 distinction between these factors, whereby MyoD establishes an open chro-
Fax: +1 613 739 6294 matin structure at muscle-specific genes, whereas Myog drives high levels
Tel: +1 613 737 8899 ext 70339
of transcription of genes within this open chromatin state. Furthermore,
E-mail: jdilworth@ohri.ca
recent data have provided an additional distinction between MRF function
(Received 7 January 2013, revised 1 with respect to cell cycle regulation. Indeed, MyoD has been shown to
February 2013, accepted 5 February 2013) directly activate genes involved in cell cycle progression, leading to myo-
blast proliferation. In contrast, Myog has antiproliferative activity through
doi:10.1111/febs.12188 the activation of genes that shut down the cell proliferation machinery,
leading to cell cycle exit and myoblast differentiation. Although the tran-
scriptional activities of MyoD and Myog synergize to drive muscle differ-
entiation, it is the expression of Myog that sets in motion a gene
expression program that constitutes a ‘point of no return’, leading to cell
cycle exit. In this review, we compare and contrast the current literature
with respect to MRF function, with a particular emphasis on the differen-
tial role of MRFs in modulating the cell cycle.

Introduction
Developmental gene expression programs are estab- that is permissive to binding of the transcriptional
lished through the combined activity of tissue- machinery at specific genes within a cell type [3]. The
restricted and ubiquitously expressed transcription fac- paradigm for such a model has been the transcrip-
tors that allow communication between distal and tional regulatory network that governs vertebrate skel-
proximal regulatory regions of genes [1,2]. According etal myogenesis. Indeed, expression of the skeletal
to current models, tissue-restricted transcription fac- muscle-specific transcription factor MyoD was long
tors help to establish a remodeled chromatin structure ago shown to be sufficient to mediate lineage repro-

Abbreviations
bHLH, basic helix–loop–helix; C/H, cysteine/histidine-rich; ChIP, chromatin immunoprecipitation; E, embryonic day; KO, knockout; MAPK,
mitogen-activated protein kinase; MRF, myogenic regulatory factor; Myog, myogenin; pRB, retinoblastoma protein; TAD, transactivation
domain.

FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS 3991
Modulation of cell cycle by MRFs K. Singh and F. Jeffrey Dilworth

gramming through activation of the muscle gene most studies examining the mechanisms of transactiva-
expression program in multiple cell types [4]. This sem- tion have focused on the activities of individual
inal finding led to the cloning of three related skeletal MRFs. Thus, we currently lack a good understanding
muscle-specific transcription factors [5]. Collectively, of the specific functional roles played by individual
MyoD, Myf5, myogenin (Myog), and MRF4 consti- MRFs in the processes of muscle development, repair,
tute a family of transcription factors that have been and homeostasis. Here, we review the current literature
termed myogenic regulatory factors (MRFs). Charac- concerning MRF functions that supports the notion
teristic of MRFs, these proteins share a highly con- that MRFs play differential roles in mediating cell
served variant of the basic helix–loop–helix (bHLH) cycle progression.
domain [6] that confers their myogenic potential [7].
Whereas the expression of MRFs is skeletal muscle-
Understanding the role of MRFs
specific, their function requires heterodimerization with
through mouse genetics
a member of the ubiquitously expressed E-protein fam-
ily of bHLH proteins. Upon dimerization, the MRF– Studies over the last few decades have made significant
E-protein heterodimeric complexes are able to bind the advances in demarcating the functions of individual
E-box consensus sequence (CANNTG), which is pres- MRFs in myogenesis. Importantly, these studies have
ent in the regulatory regions of muscle-specific genes shown that these muscle-specific transcription factors
[8]. Although E-boxes are well represented in the gen- have evolved a hierarchical relationship that retains a
ome, with more than 14 million sites, MRFs bind only certain degree of functional redundancy, while high-
a small fraction of these sequences [9,10]. Further- lighting the fact that each MRF also possess unique
more, chromatin immunoprecipitation (ChIP) sequenc- properties that subtly affect the myogenic process.
ing analysis has shown that MRFs bind both Mouse genetic studies have played a key role in identi-
overlapping and distinct loci within the genome fying the hierarchical relationship between MRFs,
[10,11]. This suggests that the binding of MRFs to whereby MyoD and Myf5 are generally viewed as fac-
their target E-box is limited by chromatin accessibility, tors involved in the determination of myogenic cells,
but also through properties intrinsic to the individual whereas Myog and MRF4 are more closely associated
family members. with terminal differentiation and homeostasis of myofi-
Phylogenetic analysis indicates that the four verte- bers [5]. The definition of MyoD and Myf5 as determi-
brate MRF genes have evolved from a single ancestral nation factors comes from genetic studies in mice,
MRF gene as a result of gene duplication events and where the double knockout (KO) of these two MRFs
subsequent divergent mutations [12]. Indeed, many resulted in postnatal lethality, owing to a complete
invertebrates, such as Caenorhabditis elegans, Drosoph- absence of skeletal myoblasts or myofibers [20–22].
ila, sea urchins, and acidians, continue to develop their The absence of an overt phenotype in mice lacking
musculature in the presence of a single MRF gene [13– either MyoD or Myf5 provides strong evidence for
15]. However, the more complex musculature of functional overlap between these two determination
vertebrates has forced the evolution of four MRFs to factors [20–22]. However, further exploration of myo-
regulate the complex gene expression program in myo- genesis in the absence of MyoD or Myf5 has revealed
genesis. To establish this complex regulation of gene subtle difference between these two transcription fac-
expression, MRFs have retained high conservation of tors. Developmental studies of the single KO mice
their DNA-binding domain (bHLH), while the trans- showed delayed myogenesis in the epaxial myotome in
activation domains of each transcription factor have the absence of Myf5, whereas myogenesis in the hyp-
diverges. This has allowed MyoD, Myf5, Myog and axial myotome was delayed in mice lacking MyoD
MRF4 to retain a certain degree of functional overlap [23]. Further evidence for MyoD and Myf5 determin-
while acquiring the unique properties required for ing distinct muscle populations in the myotome comes
development of the mammalian musculature. This has from studies showing that ablation of Myf5-expressing
been clearly demonstrated in genetic models, where cells failed to prevent MyoD-dependent skeletal muscle
knock-in studies have shown that MRFs display a cer- differentiation [24,25]. This suggests that MyoD and
tain degree of functional redundancy, but are not com- Myf5 are responsible for determining distinct myo-
pletely interchangeable [16,17]. Although elegant genic populations in the myotome that have the poten-
experiments involving swapping between MyoD and tial to compensate for each other in development.
Myog have provided some insights into the roles of Mice that lack Myog continue to specify the muscle
specific protein domains in mediating differential lineage through the formation of myoblasts. However,
transactivation by these transcription factors [18,19], these mice show perinatal lethality, because of severe

3992 FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS
K. Singh and F. Jeffrey Dilworth Modulation of cell cycle by MRFs

disruption of myoblast differentiation and muscle fiber structure called the dermomyotome [35]. Within these
formation, leading to the idea that Myog is a differen- structures, expression of MRFs is first detected at
tiation factor in the myogenic process [26,27]. This around embryonic day (E)8.0, as sonic hedgehog sig-
functional distinction between Myog and its related naling from the notochord and floor plate induces ep-
family members MyoD and Myf5 is further high- axial expression of Myf5 in the dorsal lips of the
lighted by studies showing that the Myog/MyoD or dermomyotome, committing these cells to become the
Myog/Myf5 double KO mice specify the muscle line- epaxial myotome [36,37]. At E10.5, Wnt (from the
age but do not form muscle fibers – a phenotype simi- dorsal ectoderm) and Bmp4 (from the lateral meso-
lar to that of the Myog KO mouse [28]. These findings derm) signaling establishes MyoD expression in the
demonstrate that the function of Myog does not over- hypaxial dermomyotome, causing these cells to estab-
lap with those of MyoD and Myf5, and that Myog lish the hypaxial myotome [38,39]. Transcripts for
acts downstream of MyoD and Myf5 in skeletal mus- Myog and MRF4 are first detected at E8.5 and E9.0,
cle development as a differentiation factor. respectively, and their expression is evenly distributed
Studies have demonstrated that MRF4 can act as throughout the myotome [39–42]. Consistent with a
both a determination and a differentiation factor [29]. dual role in both determination and differentiation of
Similar to the effect of KO of the determination fac- the muscle lineage, MRF4 transcripts show biphasic
tors MyoD and Myf5, MRF4 null mice do not show expression, whereby they are downregulated by E11.5,
any overt muscle phenotype [30]. This would suggest but reappear at E16.0 in differentiated muscle fibers
that the determination role of MRF4 is functionally [40]. These MRFs execute the myogenic differentiation
redundant with MyoD and Myf5, whereas its differen- program, via expression of downstream targets, that
tiation role is redundant with that of Myog. Indeed, results in the development of trunk muscle from the
MRF4 KO mice did show strong upregulation of epaxial myotome, whereas limbs, diaphragm and body
Myog expression, suggesting that MRF4 probably wall muscle develop from the hypaxial myotome.
functions to downregulate Myog expression in the Whereas in vivo studies have provided important
mature myofiber [30]. Interestingly, MRF4/MyoD KO spatial information about the expression of MRFs, the
mice died at birth, showing a phenotype highly similar hierarchical relationship regulating the expression of
to that of the Myog null mutant mice [31]. In MRF4/ the different family members has been defined through
MyoD double KO mice, Myog was expressed, but this ex vivo studies using satellite cells. Satellite cells (SCs)
expression was insufficient to support normal myogen- are resident muscle stem cells that are mitotically qui-
esis in vivo. This suggests that MRF4 and MyoD play escent in healthy adult muscle and become activated
a redundant role in mediating skeletal muscle differen- upon damage to the muscle fiber [43]. Overall, these
tiation during development. Taken together, these studies have shown that the progression of activated
genetic studies establish the need for a minimum of satellite cells towards the myogenic lineage is mainly
two MRFs to generate functional muscle during devel- controlled by expression of Myf5 and MyoD [44].
opment: one to determine the muscle lineage, and a Recent work suggests that Myf5 sits at the top of the
second to permit terminal differentiation. hierarchy, as it was shown that the majority of quies-
cent satellite cells transcribe the Myf5 gene and are
poised to enter the myogenic program. To maintain
Spatial and temporal control of MRF
these poised cells in the quiescent state, mRNAs
expression
encoding Myf5 remain untranslated, owing to miR-31-
The four members of the MRF family can act as mas- dependent sequestration of the transcripts in mRNP
ter regulators of skeletal myogenesis, whereby their granules. Upon satellite cell activation, mRNP granule
exogenous expression can hijack the inherent gene dissociation results in the release of sequestered tran-
expression program of a nonmuscle cell and drive it scripts and rapid translation of the Myf5 mRNAs [45].
towards a myogenic fate [4,32–34]. Therefore, the loca- Soon afterwards, MyoD expression is initiated through
tion, timing and expression levels of the MRFs during the activity of Pax3/7, Six1/4, and FoxO3, to permit
embryonic development are tightly regulated to ensure expansion of a cell population that is committed to the
the accurate progression of the developmental process. myogenic lineage [46,47]. In the proliferating myo-
In the murine model, precursor cells for myogenesis blasts, the MyoD gene is transcribed through a
originate in the segmentally arranged nascent somites TFIID-dependent mechanism, whereby a moderate
that flank both side of the neural tube and notochord. level of expression is ensured through spatial localiza-
As embryonic development progresses (Fig. 1), a tion near the periphery of the nucleus [48,49]. As myo-
portion of the somites transforms into a transient blasts initiate differentiation towards myotubes, the

FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS 3993
Modulation of cell cycle by MRFs K. Singh and F. Jeffrey Dilworth

Fig. 1. A schematic representation of embryonic and postnatal skeletal myogenesis. During vertebrate myogenesis, somites differentiate
and subdivide to give rise to the dermomyotome (DM) and sclerotome (SCL) in response to signals from the neural tube (NT) and notochord
(N). Cells from the dorsomedial lip (DML) migrate under the DM to form the epaxial myotome, which is the main source of the deep back
musculature. Similarly, cells originating in the ventrolateral lip (VLL) migrate under the DM to generate the hypaxial myotome, which gives
rise to the lateral trunk musculature. A proportion of cells from the VLL undergoing epithelial–mesenchymal transition delaminate and
migrate to the region of presumptive limb muscle development (migrating limb precursors). The paired homeobox gene Pax3 has been
demonstrated to be a key regulator of embryonic skeletal myogenesis, and sufficient to activate the expression of Myf5 and MyoD and
initiate the myogenic program that leads to the development of myoblasts. Although myogenic precursor cells express both Pax3 and Pax7,
Pax7-expressing cells are more prominent in the central region of the DM, and this is the population that gives rise to the major pool of
Pax7+ satellite cells (SC) in the body. SCs are mitotically quiescent in adult muscle, and are located underneath the basal lamina. In
response to muscle injury, SCs are activated and give rise to Myf5 /MyoD cells, which return to the quiescent stage, and a Myf5+/MyoD+
myoblast population. Myoblasts generated from both embryonic and postnatal myogenesis undergo extensive proliferation, which leads to
the generation of Myog+ myocytes in response to appropriate differentiation cues. Finally, these myocytes fuse to form myotubes and
subsequently myofibers, which continue to express the terminal differentiation marker MRF4, structural and metabolic genes such as the
myosin heavy chain (MHC) and the muscle creatinine kinase (MCK) genes.

MyoD gene moves towards the lumen of the nucleus, Myog to rescue the absence of skeletal muscle [16].
where a TAF3/TRF3-dependent transcriptional mech- The formation of healthy myofibers in these knock-in
anism results in a high level of MyoD expression mice demonstrated that Myog can act to specify the
[48,49]. Under these conditions, MyoD induces Myog muscle lineage in the proper context. However, it is
expression, which results in downregulation of Myf5 noted that these mice died at birth, owing to reduced
expression [50]. This switch in expression from Myf5 skeletal muscle formation. Thus, whereas Myog was
to Myog coincides with cell cycle exit and a commit- able to specify the muscle lineage, it appears that
ment to differentiate [2,51]. The combined activity of Myog may not have allowed for the expansion of the
MyoD and Myog leads to the expression of the muscle progenitor population to generate sufficient
MRF4 gene and other late muscle differentiation genes cells to establish a complete musculature. Therefore,
to permit the formation of multinucleated fibers. In these findings demonstrate that there are functional
mature muscle fibers, expression of MyoD and Myog differences inherent to MRF family members beyond
is then downregulated, whereas MRF4 continues to be their specific spatial and temporal expression patterns
expressed at high levels to act as the predominant during development.
MRF in adult muscle [41].
The sequential expression of MRFs during muscle
Divergence within structural domains
differentiation, combined with their functional redun-
of MRFs
dancy, would suggest that the timing of MRF expres-
sion could be responsible for the differential roles of Although all four MRFs are able to induce lineage
family members in muscle formation. Supporting such reprogramming of fibroblasts towards a skeletal mus-
a possibility, insertion of the Myog cDNA into the cle lineage, studies in cultured cells have demonstrated
Myf5 locus in Myf5/MyoD double KO mice allowed differential efficiency between MRFs in activating the

3994 FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS
K. Singh and F. Jeffrey Dilworth Modulation of cell cycle by MRFs

expression of silent muscle genes [18,52]. To explain MyoD to E-boxes within repressive chromatin through
this divergent ability to activate gene expression within an interaction with the homeodomain protein Pbx1
chromatin, differences in protein structure have been [19]. Once tethered to the chromatin by Pbx1, MyoD
explored. Examination of the protein sequences shows can then recruit the SWI/SNF ATP-dependent chro-
that MRFs vary in size from 224 amino acids (Myog) matin remodeling complex [54] and the p300 histone
to 319 amino acids (MyoD). Structurally, the MRFs acetyltransferase [55,56] to muscle-specific genes for
are highly similar, in that they possess a conserved the establishment of an open chromatin structure.
bHLH domain for DNA binding that is flanked by Importantly, the C/H and helix 3 domains of MyoD
less conserved N-terminal and C-terminal domains and Myf5 are highly conserved, whereas the domains
that mediate transcriptional activation (Fig. 2). How- diverge in the determination factors Myog and MRF4
ever, the divergent amino acid sequences in the trans- [18]. Genome-wide binding analyses have demon-
activation domains (TADs) of MRFs raises the strated that MyoD and Myf5 share a significant pro-
possibility that they may be responsible for the inher- portion of their targets in proliferating myoblasts [10].
ent differences between family members. This hypothe- This would suggest that a shared ability to remodel
sis was tested by Ishibashi et al. [53], who chromatin might be at the heart of the redundant abil-
interchanged the TADs between MyoD and Myf5. ity of MyoD and Myf5 to efficiently specify the muscle
These experiments demonstrated that the N-terminal lineage. Importantly, the C/H and helix 3 domains of
and C-terminal domains of MyoD and Myf5 were Myog cannot mediate remodeling of chromatin within
interchangeable for the activation of gene expression the promoter of muscle genes [18,52]. Instead, Myog is
involved in myoblast proliferation. However, in condi- a strong activator of transcription at loci with an open
tions of differentiation, the N-terminal and C-terminal chromatin structure previously established by MyoD
TADs of MyoD act in a synergistic manner to induce [57]. That being said, Myog can function in concert
the expression of muscle differentiation genes, and this with transcription factors such as Mef2D to recruit
synergy could not be achieved in combination with chromatin remodeling machinery at muscle genes to
either of the Myf5 TADs [53]. permit transcriptional activation [58]. This observation
Within the TADs of MyoD, two structural domains would explain how Myog could act as a specification
have been identified that allow MyoD to activate the factor when expressed from the Myf5 locus in the
expression of silent genes during myogenesis [18]. MyoD/Myf5 double KO background [16]. Thus,
These domains are termed the cysteine/histidine-rich MyoD and Myf5 possess an activity that allows for
(C/H) domain and helix 3 domain, and they lie within the opening of chromatin that is not shared by Myog.
the N-terminal and C-terminal TADs respectively. Domain swapping studies have also revealed func-
Interestingly, swapping of the C/H and helix 3 tional differences between MyoD and MRF4, whereby
domains of MyoD into Myog allows Myog to effi- the N-terminal domain of MRF4 can act as either a
ciently activate the expression of silent muscle genes transcriptional activator or as a repressor, depending
[18]. Characterization of the C/H and helix 3 domains on the promoter context [59]. Among the target genes
showed that this structure is important for targeting repressed by MRF4 is the cardiac a-actin gene, which

N-terminal TAD C-terminal

C = conserved
NC = not conserved
p = phosphorylation
Ac = acetylation

Fig. 2. A schematic diagram of the structure of MyoD as a representative of the MRF family. The amino acids encompassed by the
different domains are indicated. Various known post-translational modifications (PTMs) are presented in table below, and their conservation
is determined on the basis of multiple sequence alignment.

FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS 3995
Modulation of cell cycle by MRFs K. Singh and F. Jeffrey Dilworth

is activated by MyoD expression [59]. Direct competi- myoblasts through direct binding to the promoter
tion studies on the cardiac a-actin promoter showed [9,10]. Gene ontology analysis of these different target
that the repressive property of MRF4 predominates genes revealed that MyoD displayed enhanced binding
over MyoD-mediated transactivation, suggesting that in myotubes at genes involved in muscle development,
the relative levels of different MRFs may modulate the whereas genes involved in the regulation of cell cycle
transcriptional output of specific muscle genes [59]. were preferentially bound by MyoD in proliferating
Interestingly, MRF4 activity is modulated by the p38 myoblasts [9,61]. Thus, MyoD binding facilitates the
mitogen-activated protein kinase (MAPK) signaling expression of genes that promote proliferation or dif-
pathway during the course of myoblast differentiation ferentiation of myoblasts, depending on the cellular
[60]. Phosphorylation of MRF4 by p38 MAPK at environment.
Ser31 and Ser42 within the N-terminal transactivation Genome-wide occupancy of Myf5 has been reported
domain inhibits its function, permitting activation of recently in proliferating myoblasts [10]. Comparative
the cardiac a-actin gerne while blocking CKm gene analysis of MyoD and Myf5 binding sites showed a
expression [60]. Thus, the N-terminal TAD of MRF4 highly significant overlap of ~ 30% [10]. This result is
cooperates with p38 MAPK to selectively modulate consistent with the notion that MyoD and Myf5 share
the expression of the late myogenic transcriptional a role in defining myoblast identity. Unfortunately, the
program. Taken together, these results suggest that study did not report the ontology of Myf5 target
divergent TADs within the MRF family play a role in genes, and thus it remains to be seen whether Myf5
modulating their targeting to specific genomic loci. modulates myoblast proliferation through direct upreg-
ulation of genes involved in cell cycle regulation.
Analysis of the genome-wide binding of Myog that
MRFs and their transcriptional targets
was performed within the ENCODE project has not
The binding of several MRFs has been explored in yet been published [63]. Thus, our understanding of
cultured cell systems with high-throughput technolo- Myog binding is shaped mostly by ChIP array studies
gies such as ChIP microarray and ChIP sequencing performed to identify associations with defined pro-
[9,10,57,61]. Among these, genome-wide binding of moter regions [57,61]. These studies revealed that
MyoD has been the most thoroughly explored. The Myog is bound to 75% of the promoters that are tar-
consensus among these different studies is that MyoD geted by MyoD in differentiating myotubes [61]. This
binds a large number of genes in muscle cells while finding is consistent with the view that MyoD estab-
modifying gene expression at only a fraction of its tar- lishes an open chromatin structure that, in turn, per-
gets [9,10,57,61]. However, an important finding of the mits binding of Myog to establish a high level of
study by Cao et al. was the observation that binding transcriptional activation [57]. Examination of the
of MyoD within the genome correlated well with genes co-bound by MyoD and Myog identified genes
opening of the chromatin structure through acetylation involved in muscle development [57,61]. Interestingly,
of histones [9]. This opening of chromatin has recently microarray studies examining the role of Myog in dif-
been shown to establish enhancer elements that can ferentiation recently identified genes involved in cell
control the expression of both coding and noncoding cycle progression as key transcriptional targets that
RNAs [62]. This would suggest that a major role of are downregulated by Myog during differentiation
MyoD in the myogenic process is the opening of chro- [51]. This suggests that Myog is an important modula-
matin at specific loci to establish the muscle-specific tor of cell cycle exit during differentiation. Thus, in
chromatin state. Consistent with this idea, studies in contrast to MyoD, which promotes proliferation in
the Tapscott laboratory demonstrated that MyoD- growing myoblasts, Myog attenuates the expression of
dependent activation of late muscle genes required the genes that mediate cell cycle progression.
activity of Myog [57]. On the basis of these findings, it
was proposed that the transcriptional activation
MRFs as modulators of the cell cycle
domain of Myog drives high-level expression at genes
that have an open chromatin structure established by Studies from cultured cell systems support the notion
MyoD [57]. Nevertheless, MyoD can also directly that MyoD and Myog have opposing roles in modu-
modulate the activation of gene expression. During lating the cell cycle. In fact, both MyoD and Myf5
differentiation, MyoD-dependent activation of immedi- have been shown to promote expansion of the muscle
ate-early genes occurs in the absence of Myog [57]. progenitor population [64–66]. In contrast, Myog
Similarly, MyoD has been shown to directly regulate appears to possess intrinsic activity that is required to
the expression of a subset of genes in proliferating mediate cell cycle exit [51]. Indeed, it was recently

3996 FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS
K. Singh and F. Jeffrey Dilworth Modulation of cell cycle by MRFs

demonstrated that ectopic expression of Myog in pro- from studies showing the distinct and contrasting
liferating myoblasts leads to exit from the cell cycle expression patterns of MyoD and Myf5 during the dif-
[51]. This finding provides an explanation for the phe- ferent phases of the cell cycle [67]. Myf5 protein levels
notype observed in the studies where Myog is peak in G0, decrease during G1, and then rise again at
expressed from the Myf5 locus in the Myf5/MyoD the end of G1 where they remain stable through mito-
double KO mouse [16]. These mice had specified the sis (Fig. 3). In contrast, MyoD has been shown to
muscle lineage and established healthy muscle fibers. block the G1/S transition [68]. Thus, MyoD protein
However, the mice died at birth, owing to insufficient levels peak in mid-G1, are reduced to their minimum
musculature. Thus, expression of Myog from the Myf5 level on G1/S transition, and are reaugmented from S
locus is sufficient to determine the muscle lineage, to M [67]. These changes in MyoD and Myf5 protein
although it probably caused precocious exit from the levels during the cell cycle are modulated through
cell cycle, leading to impaired expansion of the progen- post-translational modifications that signal their degra-
itor pool, resulting in less muscle. Furthermore, this dation. Although not all of the mechanisms regulating
finding might explain why stable expression of Myog MRF stability have been established, several studies
in primary myoblasts has never been reported. Taken have highlighted modifications that lead to degrada-
together, these findings support the notion that MyoD tion of MyoD or Myf5. In particular, the Myf5 pro-
and Myf5 promote expansion of muscle progenitor tein level is modulated by its phosphorylation and
pools, whereas Myog induces cell cycle exit. subsequent degradation at mitosis [69,70]. In the case
Characterization of satellite cells from MyoD KO of MyoD, degradation at late G1 is mediated by the
[65] and Myf5 KO [66] mice showed that they both ubiquitin proteasome system, which is triggered by cy-
had proliferation defects. This suggests that the two clin E/CDK2-dependent phosphorylation of MyoD on
determination MRF factors play nonredundant roles Ser200 [71,72]. The transcriptional activity of MRFs
in the cell cycle. Further evidence for this notion come can also be modulated during the cell cycle, as it has

Fig. 3. A schematic overview of crosstalk between MRFs and cell cycle regulation. MyoD and Myf5 show distinct and contrasting
expression patterns during the different phases of the cell cycle. The Myf5 protein level peaks in G0, decreases during G1, and then rise
again at the end of G1 where they remain stable through mitosis. In contrast, the MyoD protein level peaks in mid-G1, decreases to its
minimum level in the G1/S transition, and is reaugmented from S to M. The levels of both proteins during the different phases of the cell
cycle are regulated by phosphorylation-dependent degradation via the 26S proteasome. In proliferating myoblasts, MyoD initiates the
expression of two genes, CDC6 and MCM2, which are primarily involved in making chromatin operational for DNA replication and
progression of cells through S-phase. In response to appropriate differentiation signals, MyoD induces the expression of Myog, and
establishes a transcriptionally permissive chromatin structure at the p21cip, p57kip and pRB genes. Myog and MyoD then synergize to
upregulate the expression of these key regulators of cell cycle exit – p21cip and p57kip repress the activity of Cdks and cyclins, whereas
pRB targets E2F family members, which are major regulators of the expression of Cdks and cyclins. Myog also interfers with cell cycle
progression by upregulating the expression of miR-20a and LATS2. miR-20a is a microRNA that is well characterized for its ability to
downregulate the transcription factors E2F1 and E2F3 through targeting the 3′-UTRs of their mRNAs. LATS2 is a protein kinase that has
been implicated in targeting of the transcriptional repressor complex DREAM to E2F target genes to block cell cycle progression.

FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS 3997
Modulation of cell cycle by MRFs K. Singh and F. Jeffrey Dilworth

been shown that MyoD is phosphorylated at Ser5 and extensive literature has supported a role for MyoD in
Ser200 by cyclin B/Cdc2 in mitosis to inhibit its DNA- the induction of cell cycle exit during terminal differen-
binding activity and transcriptional activation ability tiation (reviewed in [78]). Indeed, it has been shown
[73]. Thus, the activities of Myf5 and MyoD are that MyoD / myoblasts fail to exit the cell cycle, as
dynamically regulated throughout the cell cycle via the transcriptional regulator nuclear factor-jB (NF-jB)
modulation of transcriptional activity and protein maintains a nuclear localization to activate the tran-
abundance. scription of key cell cycle regulators [79]. However,
Although distinctions in the expression patterns recent studies examining bromodeoxyuridine staining in
have been observed, the relative importance of MyoD differentiating myoblasts have demonstrated that
and Myf5 to cell cycle progression has not been estab- MyoD expression in promyogenic conditions is not suf-
lished. Genome-wide binding studies have previously ficient for cell cycle exit [51]. Instead, it appears that the
established that MyoD binds to the transcriptional activation of Myog expression by MyoD constitutes a
regulatory region of genes with critical roles in the cell ‘point of no return’, where Myog initiates a gene expres-
cycle [9,57,61]. Characterization of specific promoters sion program that commits the differentiating myoblast
in growth-stimulated quiescent myoblasts showed that to exit the cell cycle [51]. Consistent with the existing lit-
MyoD directly activates expression of the CDC6 and erature, Myog-induced cell cycle exit appears to occur
MCM2 genes, which are involved in preparing chro- through the activation of CDKN1a (p21cip), as well as
matin for DNA replication, and consequently progres- pathways that inhibit the expression of E2F target
sion of cell through S-phase [64]. We note that, genes – cell cycle targets that have previously been
whereas MyoD shows greater efficiency in establishing attributed to MyoD [45,78,80–82]. Indeed, exogenous
transcription at two genes that modulate the cell cycle, expression of Myog in proliferating myoblasts leads to
Myf5 can also perform this function, showing some upregulation of p21cip, which triggers cell cycle exit [51].
degree of functional redundancy [64]. Future mecha- This appears to be a direct modulation of p21cip expres-
nistic studies based on the plethora of genome-wide sion, as a binding site for Myog has been identified at
data should prove highly informative with respect to the CDKN1a promoter (unpublished observation based
the transcriptional activation of specific genes by Myf5 on the ENCODE data from CalTech, K. Singh and F.
and MyoD in proliferating myoblasts. J. Dilworth). In the case of E2F target genes, Myog
An interesting distinction between MyoD and Myf5 takes a multipronged approach to ensure efficient silenc-
is their ability to modulate cell cycle progression in ing of these loci. In a first mechanism, Myog upregulates
response to DNA damage [74]. The cellular response transcription of the miR17-92 cluster of microRNAs
to DNA damage is to block cell cycle progression at [51]. Among these microRNAs, miR-20a has been
specific checkpoints, which allows DNA repair to pre- shown to target the mRNA of E2F1, E2F2 and E2F3 to
vent the propagation of genomic mutations in daugh- inhibit their translation [83–85]. Therefore Myog can
ter cells [75,76]. In the case of skeletal myogenesis, indirectly downregulate the cellular protein levels of
MyoD is a genuine target for the differentiation check- E2F family members. A second mechanism used by
point. In response to genotoxic stress, the transcrip- Myog to inhibit E2F target gene expression is upregula-
tional activity of MyoD is repressed to prevent tion of Lats2 kinase [51], which has been shown to inhi-
myotube formation in cells that have arrested the cell bit transactivation by E2F proteins. Lats2 was identified
cycle because of DNA damage. This transcriptional in a small hairpin RNA screen of factors that function
inhibition requires phosphorylation of MyoD at Tyr30 with retinoblastoma protein (pRB) to mediate cell cycle
by c-Abl tyrosine kinase [77]. Interestingly, the consen- exit [86]. There, it was shown that Lats2, a component
sus site for Abl-mediated tyrosine phosphorylation is of the Hippo pathway [87], is required to initiate a phos-
absent in Myf5, making this protein insensitive to the phorylation cascade that leads to pRB-dependent bind-
DNA damage response. The differential susceptibility ing of the repressive complex DREAM to E2F target
of MyoD and Myf5 to c-Abl-mediated repression is genes [86]. This finding is particularly interesting, as it
not surprising, as MyoD levels peak in mid-G1, where has been shown that downregulation of pRB and Arf
myoblasts exit the cell cycle, whereas Myf5 levels are allows Myog-expressing myocytes to re-enter the cell
minimal at this stage of the cell cycle. Interestingly, cycle [88]. Furthermore, MyoD-induced myogenesis
the c-Abl consensus binding site (YDDP) is also leads to upregulation of pRB to mediate cell cycle exit
absent in Myog and MRF4, suggesting that this role [89] through the establishment of repressive histone
in facilitating DNA repair is unique to MyoD [74,77]. methylation at cell cycle genes [90]. Although we have
For successful execution of the myogenic differentia- not examined whether Myog can directly lead to upreg-
tion program, myoblasts must exit the cell cycle. An ulation of pRB and ARF, we note that MyoD and

3998 FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS
K. Singh and F. Jeffrey Dilworth Modulation of cell cycle by MRFs

Myog both bind to the promoter and an intronic enhan- technologies that permit us to pursue unique target
cer of the Rb1 gene (unpublished observation based on genes as well as changes in gene expression and chro-
the ENCODE data from Caltech). On the basis of this matin structure at a global level have begun to provide
observation, we suggest that MyoD may create an open novel insights into the importance of the multiple
chromatin structure at the Rb1 gene that facilitates MRFs in mediating myogenesis. These studies have
strong transactivation by Myog. However, this possibil- identified a role for the determination MRFs in speci-
ity still needs to be formally tested. Nevertheless, it fying the muscle lineage through the opening of chro-
appears that MyoD and Myog act in a coordinated matin structure at genes involved in muscle
manner to ensure cell cycle exit through the activation development, but also in expanding the muscle progen-
of key genes involved in suppressing E2F activity. The itor population. In addition, they have identified a role
mechanism by which NF-jB activity is inhibited during for the differentiation MRF, Myog, in activating high-
cell cycle exit, and whether Myog participates in this level expression of muscle genes that lie in an open
process, remain to be determined. chromatin structure while also activating signaling cas-
It is important to note that the mechanisms cades to ensure cell cycle withdrawal. Further exami-
described above for Myog-mediated cell cycle exit are nation of the plethora of information present in the
all indirect, acting by controlling the upregulation of high-throughput datasets is certain to provide us with
target genes that are directly involved in blocking cell further insights into the subtle difference between the
cycle progression. This distinction is important, as it various MRFs. Unfortunately, our knowledge of
explains observations made by the Walsh group MRF4 function in myogenesis remains poorly charac-
showing that a small percentage of differentiating terized, owing to the absence of good cellular models
myoblasts staining positive for Myog can proceed for study of its function ex vivo. The current develop-
through a round of cell cycle to incorporate bromo- ment of more sensitive techniques to exploit ChIP
deoxyuridine [91]. Thus, we propose that expression sequencing and RNA sequencing technologies will
of Myog during myogenic differentiation represents a hopefully soon allow for the isolation of MRF4-
‘point of no return’, where the MRF initiates a gene expressing cells from the developing mouse, so that we
expression program that commits the myoblast to can explore the distinct functional features of this
withdraw from the cell cycle. In this model, cells that MRF, which displays both determination and differen-
had committed to division prior to achieving suffi- tiation characteristics.
cient expression of Myog target genes would complete
a final round of replication before permanently exit-
Acknowledgements
ing the cell cycle.
The molecular characterization of cultured cell sys- We would like to thank M. Brand for critically read-
tems has thus provided some mechanistic insights into ing the manuscript. Work in the Dilworth laboratory
the differential roles of MRFs in cell cycle progression on MRF function is supported by a grant from the
as defined through ex vivo cellular studies of myo- Canadian Institutes of Health Research (MOP-77778).
blasts from KO mice. Combining these findings, we F. J. Dilworth is the Canada Research Chair in Epige-
propose the following generalization: the MRFs that netic Regulation of Transcription.
play a role in determination (MyoD and Myf5) facili-
tate cell cycle progression, whereas the MRFs that
References
mediate differentiation (Myog and MRF4) induce cell
cycle exit. Confirmation of this generalization will 1 Aziz A, Liu QC & Dilworth FJ (2010) Regulating a
require studies to examine the role of MRF4 in the master regulator: establishing tissue-specific gene
cell cycle. expression in skeletal muscle. Epigenetics 5,
691–695.
2 Dilworth FJ & Blais A (2011) Epigenetic regulation of
Conclusions and perspective satellite cell activation during muscle regeneration.
The functional redundancy within the MRF family is Stem Cell Res Ther 2, 18. doi:10.1186/scrt59.
evident from genetic studies in mice showing that the 3 Spitz F & Furlong EE (2012) Transcription factors:
from enhancer binding to developmental control. Nat
development of the murine skeletal musculature can be
Rev Genet 13, 613–626.
directed through the activities of as few as two MRFs
4 Weintraub H, Tapscott SJ, Davis RL, Thayer MJ,
– a determination MRF and a differentiation MRF.
Adam MA, Lassar AB & Miller AD (1989) Activation
Although it has been difficult to unravel the subtle dif-
of muscle-specific genes in pigment, nerve, fat, liver,
ferences in function between MRFs, high-throughput

FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS 3999
Modulation of cell cycle by MRFs K. Singh and F. Jeffrey Dilworth

and fibroblast cell lines by forced expression of MyoD. 19 Berkes CA, Bergstrom DA, Penn BH, Seaver KJ,
Proc Natl Acad Sci USA 86, 5434–5438. Knoepfler PS & Tapscott SJ (2004) Pbx marks genes
5 Tapscott SJ (2005) The circuitry of a master switch: for activation by MyoD indicating a role for a
Myod and the regulation of skeletal muscle gene homeodomain protein in establishing myogenic
transcription. Development 132, 2685–2695. potential. Mol Cell 14, 465–477.
6 Davis RL & Weintraub H (1992) Acquisition of 20 Rudnicki MA, Braun T, Hinuma S & Jaenisch R
myogenic specificity by replacement of three amino acid (1992) Inactivation of MyoD in mice leads to up-
residues from MyoD into E12. Science 256, 1027–1030. regulation of the myogenic HLH gene Myf-5 and
7 Tapscott SJ, Davis RL, Thayer MJ, Cheng PF, results in apparently normal muscle development.
Weintraub H & Lassar AB (1988) MyoD1: a nuclear Cell 71, 383–390.
phosphoprotein requiring a Myc homology region to 21 Braun T, Rudnicki MA, Arnold HH & Jaenisch R
convert fibroblasts to myoblasts. Science 242, 405–411. (1992) Targeted inactivation of the muscle regulatory
8 Berkes CA & Tapscott SJ (2005) MyoD and the gene Myf-5 results in abnormal rib development and
transcriptional control of myogenesis. Semin Cell Dev perinatal death. Cell 71, 369–382.
Biol 16, 585–595. 22 Rudnicki MA, Schnegelsberg PN, Stead RH, Braun T,
9 Cao Y, Yao Z, Sarkar D, Lawrence M, Sanchez GJ, Arnold HH & Jaenisch R (1993) MyoD or Myf-5 is
Parker MH, MacQuarrie KL, Davison J, Morgan MT, required for the formation of skeletal muscle. Cell 75,
Ruzzo WL et al. (2010) Genome-wide MyoD binding in 1351–1359.
skeletal muscle cells: a potential for broad cellular 23 Kablar B, Krastel K, Ying C, Asakura A, Tapscott SJ
reprogramming. Dev Cell 18, 662–674. & Rudnicki MA (1997) MyoD and Myf-5 differentially
10 Soleimani VD, Yin H, Jahani-Asl A, Ming H, Kockx regulate the development of limb versus trunk skeletal
CE, van Ijcken WF, Grosveld F & Rudnicki MA muscle. Development 124, 4729–4738.
(2012) Snail regulates MyoD binding-site occupancy to 24 Haldar M, Karan G, Tvrdik P & Capecchi MR (2008)
direct enhancer switching and differentiation-specific Two cell lineages, myf5 and myf5-independent,
transcription in myogenesis. Mol Cell 47, 457–468. participate in mouse skeletal myogenesis. Dev Cell 14,
11 Blais A & Dynlacht BD (2005) Constructing 437–445.
transcriptional regulatory networks. Genes Dev 19, 25 Gensch N, Borchardt T, Schneider A, Riethmacher D
1499–1511. & Braun T (2008) Different autonomous myogenic cell
12 Atchley WR, Fitch WM & Bronner-Fraser M (1994) populations revealed by ablation of Myf5-expressing
Molecular evolution of the MyoD family of cells during mouse embryogenesis. Development 135,
transcription factors. Proc Natl Acad Sci USA 91, 1597–1604.
11522–11526. 26 Hasty P, Bradley A, Morris JH, Edmondson DG,
13 Krause M, Fire A, Harrison SW, Priess J & Weintraub Venuti JM, Olson EN & Klein WH (1993) Muscle
H (1990) CeMyoD accumulation defines the body wall deficiency and neonatal death in mice with a targeted
muscle cell fate during C. elegans embryogenesis. Cell mutation in the myogenin gene. Nature 364, 501–506.
63, 907–919. 27 Nabeshima Y, Hanaoka K, Hayasaka M, Esumi E,
14 Venuti JM, Goldberg L, Chakraborty T, Olson EN & Li S, Nonaka I & Nabeshima Y (1993) Myogenin gene
Klein WH (1991) A myogenic factor from sea urchin disruption results in perinatal lethality because of severe
embryos capable of programming muscle differentiation muscle defect. Nature 364, 532–535.
in mammalian cells. Proc Natl Acad Sci USA 88, 28 Rawls A, Morris JH, Rudnicki M, Braun T, Arnold
6219–6223. HH, Klein WH & Olson EN (1995) Myogenin’s
15 Michelson AM, Abmayr SM, Bate M, Arias AM & functions do not overlap with those of MyoD or Myf-5
Maniatis T (1990) Expression of a MyoD family during mouse embryogenesis. Dev Biol 172, 37–50.
member prefigures muscle pattern in Drosophila 29 Kassar-Duchossoy L, Gayraud-Morel B, Gomes D,
embryos. Genes Dev 4, 2086–2097. Rocancourt D, Buckingham M, Shinin V & Tajbakhsh
16 Wang Y & Jaenisch R (1997) Myogenin can substitute S (2004) Mrf4 determines skeletal muscle identity in
for Myf5 in promoting myogenesis but less efficiently. Myf5:Myod double-mutant mice. Nature 431, 466–471.
Development 124, 2507–2513. 30 Zhang W, Behringer RR & Olson EN (1995)
17 Zhu Z & Miller JB (1997) MRF4 can substitute for Inactivation of the myogenic bHLH gene MRF4 results
myogenin during early stages of myogenesis. Dev Dyn in up-regulation of myogenin and rib anomalies. Genes
209, 233–241. Dev 9, 1388–1399.
18 Bergstrom DA & Tapscott SJ (2001) Molecular 31 Rawls A, Valdez MR, Zhang W, Richardson J, Klein
distinction between specification and differentiation in WH & Olson EN (1998) Overlapping functions of the
the myogenic basic helix–loop–helix transcription factor myogenic bHLH genes MRF4 and MyoD revealed in
family. Mol Cell Biol 21, 2404–2412. double mutant mice. Development 125, 2349–2358.

4000 FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS
K. Singh and F. Jeffrey Dilworth Modulation of cell cycle by MRFs

32 Braun T, Buschhausen-Denker G, Bober E, Tannich E 46 Hu P, Geles KG, Paik JH, DePinho RA & Tjian R
& Arnold HH (1989) A novel human muscle factor (2008) Codependent activators direct myoblast-specific
related to but distinct from MyoD1 induces myogenic MyoD transcription. Dev Cell 15, 534–546.
conversion in 10T1/2 fibroblasts. EMBO J 8, 701–709. 47 Grifone R, Demignon J, Houbron C, Souil E, Niro C,
33 Edmondson DG & Olson EN (1989) A gene with Seller MJ, Hamard G & Maire P (2005) Six1 and Six4
homology to the myc similarity region of MyoD1 is homeoproteins are required for Pax3 and Mrf
expressed during myogenesis and is sufficient to activate expression during myogenesis in the mouse embryo.
the muscle differentiation program. Genes Dev 3, Development 132, 2235–2249.
628–640. 48 Deato MD & Tjian R (2007) Switching of the core
34 Miner JH & Wold B (1990) Herculin, a fourth member transcription machinery during myogenesis. Genes Dev
of the MyoD family of myogenic regulatory genes. Proc 21, 2137–2149.
Natl Acad Sci USA 87, 1089–1093. 49 Yao J & Tjian R (2011) Sub-nuclear
35 Yusuf F & Brand-Saberi B (2006) The eventful somite: compartmentalization of core promoter factors and
patterning, fate determination and cell division in the target genes. Cell Cycle 10, 2405–2406.
somite. Anat Embryol (Berl) 211 (Suppl 1), 21–30. 50 Deato MD, Marr MT, Sottero T, Inouye C, Hu P &
36 Borycki AG, Brunk B, Tajbakhsh S, Buckingham M, Tjian R (2008) MyoD targets TAF3/TRF3 to activate
Chiang C & Emerson CP Jr (1999) Sonic hedgehog myogenin transcription. Mol Cell 32, 96–105.
controls epaxial muscle determination through Myf5 51 Liu QC, Zha X, Faralli H, Yin H, Louis-Jeune C,
activation. Development 126, 4053–4063. Perdiguero E, Pranckeviciene E, Munoz-Canoves P,
37 Ott MO, Bober E, Lyons G, Arnold H & Buckingham Rudnicki MA, Brand M et al. (2012) Comparative
M (1991) Early expression of the myogenic regulatory expression profiling identifies differential roles for
gene, myf-5, in precursor cells of skeletal muscle in the Myogenin and p38alpha MAPK signaling in
mouse embryo. Development 111, 1097–1107. myogenesis. J Mol Cell Biol 4, 386–397.
38 Cheng L, Alvares LE, Ahmed MU, El-Hanfy AS & 52 Gerber AN, Klesert TR, Bergstrom DA & Tapscott SJ
Dietrich S (2004) The epaxial-hypaxial subdivision of (1997) Two domains of MyoD mediate transcriptional
the avian somite. Dev Biol 274, 348–369. activation of genes in repressive chromatin: a
39 Sassoon D, Lyons G, Wright WE, Lin V, Lassar A, mechanism for lineage determination in myogenesis.
Weintraub H & Buckingham M (1989) Expression of Genes Dev 11, 436–450.
two myogenic regulatory factors myogenin and MyoD1 53 Ishibashi J, Perry RL, Asakura A & Rudnicki MA
during mouse embryogenesis. Nature 341, 303–307. (2005) MyoD induces myogenic differentiation through
40 Bober E, Lyons GE, Braun T, Cossu G, Buckingham cooperation of its NH2- and COOH-terminal regions.
M & Arnold HH (1991) The muscle regulatory gene, J Cell Biol 171, 471–482.
Myf-6, has a biphasic pattern of expression during early 54 de la Serna IL, Ohkawa Y, Berkes CA, Bergstrom DA,
mouse development. J Cell Biol 113, 1255–1265. Dacwag CS, Tapscott SJ & Imbalzano AN (2005)
41 Hinterberger TJ, Sassoon DA, Rhodes SJ & Konieczny MyoD targets chromatin remodeling complexes to the
SF (1991) Expression of the muscle regulatory factor myogenin locus prior to forming a stable DNA-bound
MRF4 during somite and skeletal myofiber complex. Mol Cell Biol 25, 3997–4009.
development. Dev Biol 147, 144–156. 55 Dilworth FJ, Seaver KJ, Fishburn AL, Htet SL &
42 Smith TH, Kachinsky AM & Miller JB (1994) Somite Tapscott SJ (2004) In vitro transcription system
subdomains, muscle cell origins, and the four muscle delineates the distinct roles of the coactivators pCAF
regulatory factor proteins. J Cell Biol 127, 95–105. and p300 during MyoD/E47-dependent transactivation.
43 Brack AS & Rando TA (2012) Tissue-specific stem Proc Natl Acad Sci USA 101, 11593–11598.
cells: lessons from the skeletal muscle satellite cell. Cell 56 Puri PL, Sartorelli V, Yang XJ, Hamamori Y, Ogryzko
Stem Cell 10, 504–514. VV, Howard BH, Kedes L, Wang JY, Graessmann A,
44 Zammit PS, Heslop L, Hudon V, Rosenblatt JD, Nakatani Y et al. (1997) Differential roles of p300 and
Tajbakhsh S, Buckingham ME, Beauchamp JR & PCAF acetyltransferases in muscle differentiation. Mol
Partridge TA (2002) Kinetics of myoblast proliferation Cell 1, 35–45.
show that resident satellite cells are competent to fully 57 Cao Y, Kumar RM, Penn BH, Berkes CA, Kooperberg
regenerate skeletal muscle fibers. Exp Cell Res 281, C, Boyer LA, Young RA & Tapscott SJ (2006) Global
39–49. and gene-specific analyses show distinct roles for Myod
45 Crist CG, Montarras D & Buckingham M (2012) and Myog at a common set of promoters. EMBO J 25,
Muscle satellite cells are primed for myogenesis but 502–511.
maintain quiescence with sequestration of Myf5 mRNA 58 Ohkawa Y, Marfella CG & Imbalzano AN (2006)
targeted by microRNA-31 in mRNP granules. Cell Skeletal muscle specification by myogenin and Mef2D
Stem Cell 11, 118–126. via the SWI/SNF ATPase Brg1. EMBO J 25, 490–501.

FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS 4001
Modulation of cell cycle by MRFs K. Singh and F. Jeffrey Dilworth

59 Moss JB, Olson EN & Schwartz RJ (1996) The 71 Song A, Wang Q, Goebl MG & Harrington MA (1998)
myogenic regulatory factor MRF4 represses the cardiac Phosphorylation of nuclear MyoD is required for its
alpha-actin promoter through a negative-acting rapid degradation. Mol Cell Biol 18, 4994–4999.
N-terminal protein domain. J Biol Chem 271, 72 Tintignac LA, Leibovitch MP, Kitzmann M, Fernandez
31688–31694. A, Ducommun B, Meijer L & Leibovitch SA (2000)
60 Suelves M, Lluis F, Ruiz V, Nebreda AR & Munoz- Cyclin E-cdk2 phosphorylation promotes late G1-phase
Canoves P (2004) Phosphorylation of MRF4 degradation of MyoD in muscle cells. Exp Cell Res
transactivation domain by p38 mediates repression of 259, 300–307.
specific myogenic genes. EMBO J 23, 365–375. 73 Batonnet-Pichon S, Tintignac LJ, Castro A, Sirri V,
61 Blais A, Tsikitis M, Acosta-Alvear D, Sharan R, Leibovitch MP, Lorca T & Leibovitch SA (2006)
Kluger Y & Dynlacht BD (2005) An initial MyoD undergoes a distinct G2/M-specific regulation in
blueprint for myogenic differentiation. Genes Dev 19, muscle cells. Exp Cell Res 312, 3999–4010.
553–569. 74 Innocenzi A, Latella L, Messina G, Simonatto M,
62 Chakravarthy AB, Catalano PJ, Mondschein JK, Marullo F, Berghella L, Poizat C, Shu CW, Wang JY,
Rosenthal DI, Haller DG, Whittington R, Spitz FR, Puri PL et al. (2011) An evolutionarily acquired
Wagner H, Sigurdson ER, Tschetter LK et al. (2012) genotoxic response discriminates MyoD from Myf5,
Phase II trial of paclitaxel/cisplatin followed by surgery and differentially regulates hypaxial and epaxial
and adjuvant radiation therapy and 5-fluorouracil/ myogenesis. EMBO Rep 12, 164–171.
leucovorin for gastric cancer (ECOG E7296). 75 Rouse J & Jackson SP (2002) Interfaces between the
Gastrointest Cancer Res 5, 191–197. detection, signaling, and repair of DNA damage.
63 Landt SG, Marinov GK, Kundaje A, Kheradpour P, Science 297, 547–551.
Pauli F, Batzoglou S, Bernstein BE, Bickel P, Brown 76 Zhou BB & Elledge SJ (2000) The DNA damage
JB, Cayting P et al. (2012) ChIP-seq guidelines and response: putting checkpoints in perspective. Nature
practices of the ENCODE and modENCODE 408, 433–439.
consortia. Genome Res 22, 1813–1831. 77 Puri PL, Bhakta K, Wood LD, Costanzo A, Zhu J &
64 Zhang K, Sha J & Harter ML (2010) Activation of Wang JY (2002) A myogenic differentiation checkpoint
Cdc6 by MyoD is associated with the expansion activated by genotoxic stress. Nat Genet 32, 585–593.
of quiescent myogenic satellite cells. J Cell Biol 188, 78 De Falco G, Comes F & Simone C (2006) pRb: master
39–48. of differentiation. Coupling irreversible cell cycle
65 Megeney LA, Kablar B, Garrett K, Anderson JE & withdrawal with induction of muscle-specific
Rudnicki MA (1996) MyoD is required for myogenic transcription. Oncogene 25, 5244–5249.
stem cell function in adult skeletal muscle. Genes Dev 79 Parker MH, von Maltzahn J, Bakkar N, Al-Joubori B,
10, 1173–1183. Ishibashi J, Guttridge D & Rudnicki MA (2012)
66 Ustanina S, Carvajal J, Rigby P & Braun T (2007) The MyoD-dependent regulation of NF-kappaB activity
myogenic factor Myf5 supports efficient skeletal muscle couples cell-cycle withdrawal to myogenic
regeneration by enabling transient myoblast differentiation. Skelet Muscle 2, 6, doi:10.1186/2044-
amplification. Stem Cells 25, 2006–2016. 5040-2-6.
67 Kitzmann M, Carnac G, Vandromme M, Primig M, 80 Zhang P, Wong C, Liu D, Finegold M, Harper JW &
Lamb NJ & Fernandez A (1998) The muscle regulatory Elledge SJ (1999) p21(CIP1) and p57(KIP2) control
factors MyoD and myf-5 undergo distinct cell muscle differentiation at the myogenin step. Genes Dev
cycle-specific expression in muscle cells. J Cell Biol 142, 13, 213–224.
1447–1459. 81 Figliola R & Maione R (2004) MyoD induces the
68 Venkat A, Jauch E, Russell WS, Crist CR & Farrell R expression of p57Kip2 in cells lacking p21Cip1/Waf1:
(2012) Care of the patient with an autism spectrum overlapping and distinct functions of the two cdk
disorder by the general physician. Postgrad Med J 88, inhibitors. J Cell Physiol 200, 468–475.
472–481. 82 Wright KJ & Tjian R (2009) Wnt signaling targets
69 Lindon C, Montarras D & Pinset C (1998) Cell cycle- ETO coactivation domain of TAF4/TFIID in vivo.
regulated expression of the muscle determination factor Proc Natl Acad Sci USA 106, 55–60.
Myf5 in proliferating myoblasts. J Cell Biol 140, 83 O’Donnell KA, Wentzel EA, Zeller KI, Dang CV &
111–118. Mendell JT (2005) c-Myc-regulated microRNAs
70 Doucet C, Gutierrez GJ, Lindon C, Lorca T, Lledo G, modulate E2F1 expression. Nature 435, 839–843.
Pinset C & Coux O (2005) Multiple phosphorylation 84 Sylvestre Y, De Guire V, Querido E, Mukhopadhyay
events control mitotic degradation of the muscle UK, Bourdeau V, Major F, Ferbeyre G & Chartrand P
transcription factor Myf5. BMC Biochem 6, 27, doi:10. (2007) An E2F/miR-20a autoregulatory feedback loop.
1186/1471-2091-6-27. J Biol Chem 282, 2135–2143.

4002 FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS
K. Singh and F. Jeffrey Dilworth Modulation of cell cycle by MRFs

85 Nagel S, Venturini L, Przybylski GK, Grabarczyk P, yields regenerative cells from postmitotic mammalian
Schmidt CA, Meyer C, Drexler HG, Macleod RA & muscle. Cell Stem Cell 7, 198–213.
Scherr M (2009) Activation of miR-17-92 by NK-like 89 Deato MD & Tjian R (2008) An unexpected role of
homeodomain proteins suppresses apoptosis via TAFs and TRFs in skeletal muscle differentiation:
reduction of E2F1 in T-cell acute lymphoblastic switching core promoter complexes. Cold Spring Harbor
leukemia. Leuk Lymphoma 50, 101–108. Symp Quant Biol 73, 217–225.
86 Tschop K, Conery AR, Litovchick L, Decaprio JA, 90 Blais A, van Oevelen CJ, Margueron R, Acosta-Alvear
Settleman J, Harlow E & Dyson N (2011) A kinase D & Dynlacht BD (2007) Retinoblastoma tumor
shRNA screen links LATS2 and the pRB tumor suppressor protein-dependent methylation of
suppressor. Genes Dev 25, 814–830. histone H3 lysine 27 is associated with irreversible cell
87 Avruch J, Zhou D, Fitamant J, Bardeesy N, Mou F & cycle exit. J Cell Biol 179, 1399–1412.
Barrufet LR (2012) Protein kinases of the Hippo 91 Andres V & Walsh K (1996) Myogenin expression, cell
pathway: regulation and substrates. Semin Cell Dev Biol cycle withdrawal, and phenotypic differentiation are
23, 770–784. temporally separable events that precede cell fusion
88 Pajcini KV, Corbel SY, Sage J, Pomerantz JH & Blau upon myogenesis. J Cell Biol 132, 657–666.
HM (2010) Transient inactivation of Rb and ARF

FEBS Journal 280 (2013) 3991–4003 ª 2013 The Authors Journal compilation ª 2013 FEBS 4003

You might also like