You are on page 1of 13

REVIEWS

Transcriptional mechanisms regulating


skeletal muscle differentiation,
growth and homeostasis
Thomas Braun* and Mathias Gautel‡
Abstract | Skeletal muscle is the dominant organ system in locomotion and energy
metabolism. Postnatal muscle grows and adapts largely by remodelling pre-existing fibres,
whereas embryonic muscle grows by the proliferation of myogenic cells. Recently, the
genetic hierarchies of the myogenic transcription factors that control vertebrate muscle
development — by myoblast proliferation, migration, fusion and functional adaptation into
fast-twitch and slow-twitch fibres — have become clearer. The transcriptional mechanisms
controlling postnatal hypertrophic growth, remodelling and functional differentiation
redeploy myogenic factors in concert with serum response factor (SRF), JUNB and forkhead
box protein O3A (FOXO3A). It has also emerged that there is extensive post-transcriptional
regulation by microRNAs in development and postnatal remodelling.

Myofibril
Movement is a defining feature of all animals, even if it factors and epigenetic regulatory mechanisms to control
The structural unit of striated is sometimes restricted to specific developmental stages. muscle development and postnatal remodelling. During
muscle fibres, which is formed Larval migration, feeding, flight, reproduction and blood embryonic development, these genetic programmes
from longitudinally joined circulation all rely on the directional and coordinated determine the primary differentiation and also the
sarcomeres. Several myofibrils
movement that is afforded by muscles. The evolutionary future metabolic and contractile properties of tissues in
form each fibre.
advantages of efficient locomotion led to several solutions specific anatomical compartments, thus controlling the
Myoblasts for the construction of the motile organs (the muscles) commit­ment of future slow-twitch or fast-twitch mus­
Embryonic cells that will in all animal phyla. In all muscle cells, myosin II motor cles. Postnatally, the degree of muscle use and the specifi­c
become a muscle cell or
proteins and actin filaments generate force and move­ innervation patterns determine the composition and
part of a muscle cell.
ment. In the striated muscles that are used for locomotion, turnover rates of muscle contractile proteins, as well as
acto­myosin contraction is amplified in serial and parallel of the supporting metabolic enzymes, ion channels and
arrangements of numerous contractile units, called sarco­ signal transduction proteins. This remodelling of differ­
*Max-Planck-Institute for
meres. These are made up of actin and myosin filaments entiated muscle determines the contractile properties and
Heart and Lung Research,
Department for Cardiac arranged in highly ordered, almost crystalline arrange­ the preferred sources of energy. It is governed by signal­
Development and ments, as well as hundreds of regulatory proteins such ling cascades (modulated by growth factors and cytokines,
Remodelling, as the troponin–tropomyosin complex, and scaffolding steroid hormones and mechanical activity) that control
Benekestrasse 2, and cytoskeletal crosslinking proteins such as α‑actinin, the transcriptional activit­y of muscle-specific and pleio­
61231 Bad Nauheim,
Germany.
myomesi­n and the kinase titin1,2 (FIG. 1). tropic transcription factors. These genetic programmes

King’s College London, In vertebrates, striated muscle cells are found in two also regulate changes in muscle mass. During embryonic
Randall Division for Cell tissues: skeletal and heart muscle (FIG. 1). Although they development, muscle mass increases predominantly by
and Molecular Biophysics both have highly ordered myofibril structures, they have proliferative growth of myoblasts. Postnatally, the contri­
and Cardiovascular Division,
distinct embryonic origins and are tailored for particular bution of cell proliferation decreases, and hypertrophic
Muscle Signalling and
Development Section, purposes by different genetic programmes. Furthermore, growth and remodelling of pre-existing muscle fibres
New Hunt’s House, in vertebrates specialized skeletal muscles with different dominates; the resident stem cells (the satellite cells) are
Guy’s Campus, contractile (slow-twitch or fast-twitch) and metabolic then mostly engaged in damage repair 5–7.
London SE1 1UL, UK. properties coexist3,4. Recent research has shed important light on the
e-mails: thomas.braun@
mpi-bn.mpg.de;
Such distinctions are determined by the activity of spe­ basic mechanisms that lead to the commitment of pre­
mathias.gautel@kcl.ac.uk cific transcription factors, the myogenic regulatory factors cursor cells to the muscle lineage (myogenesis). As a
doi:10.1038/nrm3118 (MRFs), which act together with pleiotropic transcription result, we now have a better understanding of how the

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 12 | JUNE 2011 | 349

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

remodelling uses myogenic transcription factors that are


0WENGWU also active during early development. In this Review, we
outline how tight developmental regulation, mechanical
functions and the homeostasis of the organism are inter­
5CTEQOGTG
linked during development and postnatal muscle adapta­
+DCPF #DCPF +DCPF tion. We focus on recent insights into the transcriptional
<FKUM /DCPF <FKUM and post-transcriptiona­l control that commits myogenic
#EVKPȮNCOGPV cells to proliferation or differentiation, determines their
αCEVKPKP /[QUKPȮNCOGPV /[QOGUKP lineage fate as slow-twitch or fast-twitch striated mus­
cles and regulates the postnatal adaptive remodelling
6KVKP of committed muscle cells. Where appropriate, we con­
sider relevant similarities between skeletal and cardiac
striated muscles.

Transcriptional control of myogenesis


Skeletal muscle cells of higher vertebrates arise during
midgestation (in mice between embryonic day 9 (E9)
/#42U 5356/0$4 .&$/74(%.1%- and E12) from three different locations within the middle
CPF/74( /.2CPF/;1<U
layer of cells in the primitive embryo: the segmented som­
Figure 1 | Striated muscle structure. The contractile machinery of skeletal muscle itic paraxial mesoderm, the unsegmented cranial paraxial
syncytial myotubes (left) and single cardiomyocytes (right) is formed from long arrays
0CVWTG4GXKGYU^/QNGEWNCT%GNN$KQNQI[ mesoderm and the prechordal mesoderm; these repre­
of sarcomere units, which are joined into myofibrils. The sarcomere (bottom) is sent different parts of the mesoderm along the rostrocauda­l
constructed from interdigitating, antiparallel filaments of actin and myosin, the elastic axis (reviewed in REF. 8). The skeletal muscles of the trunk
titin filaments and the crosslinker proteins for actin — α‑actinin, myosin and myomesin. and limbs are derived from cells of the segmented paraxial
Sarcomeres contain many other accessory components, including proteins involved in
mesoderm (known as somites), which form on either side
transcriptional regulation and turnover control. The transcription factor CLOCK, the
of the neural tube in vertebrate embryos. Somites further
transcriptional cofactors muscle LIM protein (MLP), muscle ankyrin-repeat proteins
(MARPs) and LIM domain-binding protein 3 (LDB3) are found at the Z‑disk and/or differentiate into a dorsal part, called the dermomyo­
the I‑band. Multifunctional components of the protein turnover machinery include tome (which retains an epithelial structure) and a ventra­l
sequestosome 1 (SQSTM1), NBR1 and the muscle-upregulated RING finger proteins part, called the sclerotome (which breaks up into the
(MURFs). MYOZs, myozenins. mesenchyme that contributes to the axial skeleto­n of the
embryo). The first muscle cells are formed in the myotome,
which is located directly underneath the dermomyotome
functional diversity of different striated muscles evolves and is formed by cells that separate by delamination from
on the develop­mental level, and how physiological stimuli the dermomyotome (reviewed in REF. 9) (BOX 1).
are translated into changes in muscle gene expression In all the anatomical sites where skeletal muscle forms,
patterns, metabolic flow and protein turnover. This determination and terminal differentiation of muscle
recent research has also revealed how postnatal muscle cells are governed by a network of four MRFs: myogenic

Paraxial mesoderm
The mesodermal areas that Box 1 | Skeletal muscle cell specification and differentiation during embryogenesis
form directly lateral to the
Formation of trunk muscles
neural tube.
The morphogenetic events that are involved in the formation of the myotome have been extensively studied. Cell tracing
Rostrocaudal axis methods have shown that a first wave of myogenic factor 5 (MYF5)-expressing muscle progenitor cells, which
A description of anatomical progressively withdraw from the cell cycle, delaminate and migrate towards the rostral somite, making up the medial
location in animals. Rostral region of epithelial somites. Myofibres differentiate in rostrocaudal (head to tail) and mediolateral (centre outwards)
(from the latin rostrum directions (reviewed in REF. 133). This initial wave of myogenesis is followed by a second wave of myoblasts. These cells
meaning beak) refers to the come from all four lips of the dermomyotome, which is the epithelial cell layer that comprises all of the mesodermal
anterior (‘nose-end’) of somites (but not the sclerotome) and that gives rise to the axial skeleton. The dorso–medial lip is initially the sole
the animal and caudal
contributor to myogenesis, followed by the posterior lip, until eventually the anterior and lateral borders begin to
(from the latin caudum
contribute to myogenesis134. It should be emphasized that the myotome that is formed by the epithelial borders of
meaning tail) refers to the
posterior (‘tail or feet end’).
the dermomyotome is composed of postmitotic cells, which do not contribute to further muscle growth. Only the
continuous addition of proliferating muscle progenitor cells allows the primary muscle compartment to expand.
Somites Formation of head muscles
Mesodermal structures found The formation of head muscles differs significantly from the formation of their counterparts in the trunk and limbs.
on either side of the neural
The head musculature originates from the cranial paraxial mesoderm (CPM), which is located anterior to the somites;
tube in vertebrate embryos
additional input to head muscles comes from the lateral splanchnic mesoderm (SpM). Cells from the CPM migrate to
that eventually give rise to
muscle, skin and vertebrae. the proximal part of the central region of the branchial arches, and SpM-derived cells contribute to the distal region.
Later in development, CPM-derived myogenic cells of the first branchial arch will form the masseter muscle, whereas
Delamination SpM- derived myogenic cells generate the lower jaw muscles (reviewed in REF. 135). Formation of head muscles is also
A process in embryology in strongly influenced by cranial neural crest cells136. Deletion of neural crest cells leads to severe reduction of jaw muscles
which cells from a single layer after the onset of muscle specification and differentiation135,137, and this is probably due to interactions between
separate to form two different muscles and neural crest-derived tendons138.
layers, or laminae.

350 | JUNE 2011 | VOLUME 12 www.nature.com/reviews/molcellbio

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

factor 5 (MYF5), muscle-specific regulatory factor 4 'ZVTCQEWNCT 6QPIWGCPF $TCPEJKCNCTEJGU


(MRF4; also known as MYF6), myoblast determination OWUENG NCT[PIGCNOWUENG
protein (MYOD) and myogenin. MRFs are transcription 2+6: 2+6: 2+6: 6$:
factors that activate many downstream genes to initiate
muscle cell differentiation. MYOD and MYF5 are muscle- /;(QT/4( /;( /;1& /;( /;1&
specific transcription factors and constitute a cross- /;1& /[QIGPKP /[QIGPKP
regulator­y transcriptional network that is at the core of
muscle cell determination and differentiation (FIG. 2); /[QIGPKP
disruption of this network completely abrogates skel­
etal muscle formation. MYF5 and MYOD are generally
thought to act as determination genes, whereas myogenin
is essential for the terminal differentiation of committed
myoblasts (reviewed in REF. 10). MRF4 seems to have a
dual role: it is thought to be a differentiation gene acting
.KODOWUENGU 6TWPMOWUENGU
in postmitotic maturating cells, but it is also expressed by
5+:QT5+:CPF 2#:
undifferentiated proliferating cells in which it might act as ';#QT';# 2#: /;( /4(
a determination gene11. The upstream signals (transcrip­
/;(QT/4(
/;(/;( /;1&
tion factors and extracellular signals (BOX 2)) that activate
MRFs differ significantly at various anatomical locations /;1& /[QIGPKP /[QIGPKP
(FIG. 2), although some molecular cues are shared.
Figure 2 | Different ways to activate the genetic
programme of muscle 0CVWTG4GXKGYU^/QNGEWNCT%GNN$KQNQI[
differentiation. All muscle cells
Regulation of trunk skeletal muscle formation. Numerous express a core set of myogenic factors (for example,
transcription factors, such as paired box, homeobox and myogenic factor 5 (MYF5), muscle-specific regulatory
T-box proteins, have been identified as binding upstream factor 4 (MRF4), myoblast determination protein (MYOD)
of MRF genes. None of these factors is exclusively and myogenin), which are required for myogenic
expressed in the muscle progenitor cells that give rise differentiation. Other transcription factors reflect
to differentiated muscl­e cells, which suggests that these lineage-specific differences and are necessary for the
trans­cription factor­s prepare the stage for additional actors activation of myogenic factors and/or proliferation and
(such as MRFs) to initiate myogenesis. Alternatively, they survival of muscle progenitor cells. Muscle groups from the
may act together with other transcription factors to allow head, including extraocular, tongue and laryngeal muscles,
and branchial arches, are derived from occipital somites,
the activation of MRFs. In both cases, instructive or per­
cranial paraxial mesoderm, splanchnic mesoderm and
missive inductional cues are necessary to achieve stable prechordal mesoderm. In these cells, pituitary homeobox 2
self-sustained activation of MRFs in muscle progenitor (PITX2) predominantly controls the myogenic hierarchy,
cells and muscle cell formation. leading to the activation of MYF5 and MYOD and
The differentiation of hypaxial muscles and epaxial eventually terminal differentiation induced by myogenin.
muscles seems to differ with regard to the transcriptional By contrast, muscles from the limbs and the trunk are all
network that activates MRFs. Indeed, the target genes of derived from trunk somites. In limb muscles, sine oculis
paired box protein 3 (PAX3), which is upstream of the homeobox homologue (SIX) and eyes absent (EYA) proteins
MRFs, differ in hypaxial and epaxial muscles. regulate paired box protein 3 (PAX3), which in turn controls
In mouse epaxial muscles, PAX3 seems to activate the proliferative myogenic cell pool, the differentiation
of which is induced by a cascade involving MYF5, MRF4,
MYF5 by controlling the expression of Dmrt2, which in
MYOD and myogenin. In trunk muscles, MYF5 or MRF4 can
turn activates the epaxial enhancer of Myf5 in somites12. show parallel activation of MYOD and myogenin, whereas
Neural crest Furthermore, analysis of mice depleted of PAX3 and PAX3 acts upstream of MYOD. Solid lines represent direct
A group of embryonic cells that MYF5 showed almost complete loss of trunk muscles control and dashed lines represent indirect control.
separate from the embryonic
and a loss of MyoD expression; this indicates that MyoD TBX1, T-box transcription factor. Figure modified, with
neural plate and migrate,
giving rise to the spinal and expression depends on either PAX3 or MYF5 (REF. 13). permission, from REF. 150 © (2009) Elsevier.
autonomic ganglia, peripheral At present, it is not known precisely how PAX3 activates
glia, chromaffin cells, MyoD expression, although experiments in presomitic
melanocytes and some explant cultures suggest that WNT signalling regulates activation of myogenesis in vitro and transient induc­
haematopoietic cells.
MyoD expression in a PAX3‑dependent manner 14. Taken tion of myogenic determination genes in vivo in chicken
Hypaxial muscles together, current evidence suggests that PAX3 medi­ embryos13,16. Interestingly, mutants of the Pax3 paralogue
Muscles that usually lie ventral ates the activation of MYOD and MYF5 in a tight inter­ Pax7 do not show overt muscle defects during mouse
to the vertebrae and are play with muscle-inducing signals. Early experiments development17, and the combined loss of PAX3 and PAX7
innervated by the ventral
showed that ectopic expression of PAX3 is sufficient to yields a phenotype that is similar to PAX3 mutants, with
ramus of the spinal nerves.
induce the expression of MYOD, MYF5 and myogenin the initial formation of skeletal muscle occurring in the
Epaxial muscles in the absence of inducing tissues (that is, neural tube myotome until E10.5. This indicates that the initial form­
Muscles that usually lie dorsal and paraxial surface epithelium) in both the paraxial and ation of skeletal muscle cells in the myotome occurring
to the vertebrae (in fish and latera­l plate mesoderm of chicken embryo, and in the up to E10.5 is not directly under the control of PAX3 and
amphibiae they lie dorsal to
the septum). They are
neural tube15,16; however, directed expression of PAX3 PAX7. As compound PAX3 and PAX7 mutants show a
innervated by the dorsal in vivo did not induce a stable myogenic fate. Similarly, severe disruption of muscle development at later stages,
ramus of the spinal nerves. expression of LBX1, a PAX3 target gene, resulted in the with very few differentiated muscle cells18, it is possible

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 12 | JUNE 2011 | 351

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 2 | Extracellular signals directing muscle development a putative paired domain-binding site within the –58 to
–56 kb distal Myf5 enhancer, which directs Myf5 expres­
The activation of the network of transcription factors that controls skeletal muscle sion in myogenic progenitor cells in limbs. Both PAX3
development depends on paracrine factors that are released by adjacent tissues, such as and mesenchyme homeobox gene 2 (MEOX2) transcrip­
the neural tube, notochord, surface ectoderm and lateral mesoderm. Several secreted
tion factors could bind these consensus sites in vitro, so
factors have been identified that determine the spatial and temporal onset of
they are potential regulators23. However, there is no valid
myogenesis. Surprisingly, no consensus has been reached as to whether these molecules
instruct naive cells (instructive induction), amplify a pool of committed progenitors genetic evidence showing that MEOX2 has a signifi­
and/‌or enable a default differentiation pathway (permissive induction) or primarily cant effect on Myf5 expression in the limb, despite some
prevent programmed cell death of muscle progenitor cells (reviewed in REF. 139). early reports claiming a reduced expression of MYF5 in
From numerous studies, it is clear that sonic hedgehog (SHH) and WNT signalling have MEOX2‑mutant limbs. It seems more likely that sine oculis
pivotal roles in the induction of myogenesis. Moreover, other signalling molecules, such homeobox homologue 1 (SIX1) and SIX4 (see below)
as Noggin and bone morphogenetic proteins (BMPs) — which inactivate and activate occupy the homeobox-binding site in the 145 bp element.
receptors of the transforming growth factor-β (TGFβ) superfamily, respectively — play In fact, it was recently shown that SIX1 and SIX4 regulate
an important part in orchestrating the activation of myogenesis140. the transcription of MYF5 in the limb together with PAX3,
A study in mice showed that the canonical β‑catenin-mediated WNT signalling
by binding to the 145 bp element –57.5 kb from Myf5. An
pathway acts co-operatively with SHH, its receptor Patched and its downstream
additional regulatory element seems to contribute to the
target, GLI (a zinc-finger transcription factor), to regulate the expression of myogenic
factor 5 (Myf5). According to this model, the SHH–Patched–GLI pathway is not expression of Myf5 in the hindlimbs, and this element
sufficient to induce myogenesis if the local concentration of β‑catenin does not might be responsible for some of the remaining expressio­n
suffice to support full transcriptional activity of lymphoid enhancer-bracking factor 1 of Myf5 in the hindlimb buds of SIX1 mutants24.
LEF1 or other transcription factors, which facilitate the localization of β‑catenin to SIX proteins are also involved in the regulation of
specific cis-regulatory elements. The transcription of the first myogenic transcription myogenesis in the limb. SIX proteins act, at least in part,
factor, MYF5, is activated in the epaxial domain only when both SHH and canonical upstream of myogenic regulatory factors and PAX pro­
WNT signalling pathways are activated at the onset of somitogenesis141. Despite teins (FIG. 2). The human and mouse genome contains
compelling evidence for the decisive role of canonical β‑catenin-mediated WNT 6 SIX genes (SIX1–SIX6), 3 of which (SIX1, SIX4 and
signalling in the induction of myogenesis, other authors reported that adenylyl
SIX5) are expressed from E8 in overlapping expression
cyclase signalling through protein kinase A (PKA) and its target transcription factor,
patterns in somites, limb buds, dorsal root ganglia and
cAMP-responsive element-binding protein (CREB), are required for WNT-directed
myogenic gene expression142. It is possible, however, that the WNT signalling pathway branchial arches. SIX proteins form complexes with their
that involves PKA and CREB acts in parallel to canonical β‑catenin-mediated WNT transcriptional co-activators, eyes absent (EYA) proteins,
signalling and becomes limiting only under specific conditions. to stimulate transcription synergistically 25. SIX1 mutants
die at birth and show selective muscle hypoplasia in the
diaphragm, forelimb, distal ventral hindlimb and abdo­
that PAX3 and/or PAX7 are responsible for the enlarge­ men26. The muscle phenotype is aggravated in compound
ment of muscle precursor cell populations. This would mutants of SIX1 and SIX4, which do not have myogenic
increase the bias towards myogenic differentiation and progenitor cells in their limb buds, resulting in legs with
thus enable myogenic cells to respond to environmental no muscles27. Furthermore, neither double SIX1 and
cues16. This view is further supported by the observation SIX4 mutants nor double EYA1 and EYA mutants express
that PAX3 directly regulates components of the fibroblast Pax3 in the hypaxial dermomyotome, indicating that Six
growth factor (FGF) signalling pathway, which have a role and Eya lie upstream of Pax3 in the genetic hierarchy
in the expansion of the myogenic progenitor cell pool19. of hypaxial myogenesis. SIX1 and SIX4 double mutants
In hypaxial muscle cells, PAX3 seems to directly activate also show a reduced expression of MyoD, myogenin and
MYF5 but not MYOD. Similarly, a study in muscle stem other myotomal markers, although the early activation
cell-derived myoblasts showed that expression of MYF5 of MYF5 in the epaxial somite is un­affected27. However,
is regulated by PAX7 (REF. 20). However, despite having a later in development the expression of SIX genes seem
role in the activation of MYF5 and the morpho­genesis to depend on MRFs, indicating a dual role for SIX pro­
of the hypaxial dermomyotome, PAX3 does not seem to teins both upstream and downstream of MRFs during
be essential for the development of hypaxial muscles, as myogenesis28. The role of SIX proteins in the differentia­
the lateral halves of somites from E9.25 PAX3‑mutant tion of muscle fibres intro slow-twitch or fast-twitch is
embryos transplanted into the limb bud of chicken host discussed below.
embryos differentiated normally 21. Furthermore, recent MRFs are also assisted by many other factors such
studies using a PAX3–engrailed fusion protein, which acts as PBX and MEIS proteins29, which function as hetero­
as a transcriptional repressor, suggested that PAX3 directly dimers and act as cofactors with basic helix–loop–helix
regulates Myf5 in the hypaxial somite and its limb mus­ (bHLH) proteins such as MRFs and various homeobox
cle derivatives. In these mice, the expression of Myf5, but transcription factors. PBX and MEIS proteins participate
not of MyoD, was compromised in hypaxial muscles. The in the feedforward mechanisms that enable MRFs to acti­
authors postulated that the regulation of Myf5 by PAX3 vate ‘early’ genes of the muscle differentiation programme
in hypaxial muscles was achieved by a PAX3 consensus immediately, whereas genes activated later on in the dif­
site located in a 145 bp regulatory element –57.5 kb from ferentiation programme need both MRFs and the addi­
Myf5. Mutation of the PAX3‑binding site in the context tional participation of one of the earlier MRF targets29
of the 145 bp element abolished all expression in trans­ (reviewed in REF.10). Myocyte enhancer factor 2 (MEF2)
genic mouse embryos22. Similarly, sequences have been proteins are also important members of this regulatory
described that contain a predicted homeobox adjacent to circuit. MEF2 proteins interact directly with MYOD

352 | JUNE 2011 | VOLUME 12 www.nature.com/reviews/molcellbio

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

in vitro and synergistically activate reporter genes that miR‑1, miR‑133, miR‑206 and miR‑208 seems to be
contain E-boxes and MEF2‑binding sites30. E-boxes and under the control of a core muscle transcriptional net­
MEF2‑binding sites are often located close to one another work, which involves the pleiotropic serum response
within the promoters and enhancers of muscle-specific factor (SRF), MYOD, and the bHLH transcription factor
genes, suggesting a model in which MRFs and MEF2 bind Twist in co­operation with MEF2 (REFS 40–43). Chromatin
DNA and activate transcription in a cooperative manner. immuno­precipitation followed by microarray (ChIP–
Three of the four MEF2 proteins (MEF2A, MEF2C and chip) analysis indicated that MYOD and myogenin bind
MEF2D) are expressed in skeletal muscle, and expression sequences upstream of miR‑1 and miR‑133 (REF. 41).
and splicing of these isoforms is altered in response to Furthermore, MEF2 has a crucial role in the control of an
MYOD, which further supports a role for MYOD in the intragenic enhancer located in the miR‑1–2 locus (which
feedforward mechanism that is driven by MRFs29. contains miR‑1 and miR‑2)44. Recently, analysis of mice
deficient in MYF5 and MYOD revealed a surprisingly
The transcriptional regulation of head muscles. Recent specific requirement of MYF5 for miR‑1 and miR‑206
evidence suggests that head muscle development follows expression. At early developmental stages, the expression
a distinct programme that does not require the action of of both miR‑1 and miR‑206 was almost entirely absent in
PAX3 and PAX7. Initial specification of mouse massete­r the somites of MYF5‑mutant embryos, whereas mouse
muscles depends on the bHLH genes MyoR and capsu­ embryos lacking MYOD showed an apparently normal
lin31. Additional players are pituitary homeobox 2 (Pitx2) expression of both miRNAs45. Whereas miR‑133 and
and T-box transcription factor 1 (Tbx1), which collabo­ miR‑206 are expressed as independent transcriptional
rate with the core myogenic programme to gener­ate head units, miR‑208 is encoded by an intron of its host gene,
muscles. Both Pitx2 and Tbx1 are expressed widely in α‑myosin heavy chain (αMHC)46. Both miR‑208 and
the developing mouse embryo and play an important αMHC are heart specific and concurrently expressed
part in the formation of head muscles (FIG. 2). Indeed, during development, suggesting that their expression is
PITX2 mutants do not properly develop the muscles that controlled by a common regulatory element46.
are derived from the first branchial arch32 and they lack Some experiments suggest that miRNAs act as modu­
extraocular muscles33. TBX1 mutants also suffer from lators of myogenic differentiation, in particular because
impaired myogenesis in the first branchial arch and lack some miRNAs, such as miR‑1 and miR‑133 (FIG. 3a), are
the muscles that normally originate from other arches absent from undifferentiated myoblasts and are strongly
owing to severe malformations of these structures34,35. upregulated upon differentiation47. An increase of miR‑1
It has recently been shown that TBX1 and MYF5 act expression in tissue culture accelerates myoblast dif­
syner­gistically to govern myogenesis in pharyngeal muscle ferentiation by downregulating histone deacetylase 4
progenitor­s, thereby acting as a complementary pathway (HDAC4), a repressor of muscle differentiation, and
to that involving PAX3 and MYF5 in the body36. It is poss­ knockdown of miR‑1 impedes myogenic differentia­
ible that PITX2 and TBX1 regulate the quiescence and tion40. Although a similar role for miR‑1 in facilitating
self-renewal of muscle progenitors in the head similarl­y differentiation has been shown in the developing mouse
to PAX3 and PAX7 in trunk skeletal muscles. heart, where a tissue-specific overexpression of miR‑1
induces premature differentiation of cardiomyocytes43,
Post-transcriptional control by microRNAs conclusive evidence for an indispensable role of miR‑1 in
The human genome contains thousands of non-coding the regulation of myogenic differentiation is still missing.
RNAs, the best-studied class of which are microRNAs Recent studies, however, suggest that re-expression of
(miRNAs) (reviewed in REF. 37), which regulate gene miR‑206 in human rhabdomyosarcoma cells promotes
expression at the transcriptional and post-transcriptiona­l myogenic differentiation and blocks tumour growth
levels. miRNAs suppress gene expression through in xenografted mice by switching the global mRNA
their complementarity to the sequence of one or more expression profile to one that resembles mature mus­
mRNAs, usually at a site in the 3′ untranslated region. cle48. This finding supports an important modulatory
The formation of an miRNA–target complex results role for miR‑206 in the control of myogenesis (FIG. 3b),
either in inhibition of protein translation or in degrada­ although miR‑206‑knockout mice show no major arrest
tion of the mRNA transcript through a process similar to of myogeni­c differentiation49.
RNA interference38. There is no doubt that the formation, The modulatory role of miRNAs on muscle dif­
maintenance, and physiological and pathophysiological ferentiation is underscored by several observations on
responses of skeletal muscles, with all their complex regu­ the role of miR‑1, miR‑27, miR‑206 and miR‑486 in the
latory circuits, are subject to regulation by non-coding regulation of PAX3 and PAX7 (FIG. 3c). miR-1, miR‑27
Masseter muscles RNAs. In fact, the increase of complexity provided by and miR‑206 were found to inhibit PAX3 and to thereby
A specific subset of
the extent of genomic non-coding sequences provides a release the inhibitory effect of PAX3 on terminal muscle
branchiomeric muscles that
are derived from the first satisfying explanation for the intricate layers of regulation differentiation50,51; an analogous role was described for
branchial arch and are found in skeletal muscle cells. the regu­lation of PAX7, which is repressed by miR–1 and
involved in mastication. Many miRNAs are expressed in skeletal and cardiac miR‑206 (REFS 52,53). Therefore, the feedback loop ampli­
muscle. Some of them are found specifically in skeletal fying term­inal MYOD-induced muscle differentiation
Pharyngeal muscle
A subgroup of head muscles
and/or cardiac muscle, or at least are highly enriched involves the transcriptional switch towards late factors
acting on the pharynx to in these tissues, suggesting specific roles in myogen­ such as MEF2C, but also the epigenetic downregulation
control swallowing. esis39. The expression of the muscle-specific miRNAs of muscle stem cell factors such as PAX3 and PAX7.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 12 | JUNE 2011 | 353

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

C D Postnatal control of muscle phenotype


/;( /'(% /;1& $/2U Skeletal muscle has a remarkable ability to rapidly adjust
to changes in physiological requirements, by changes
OK4QTOK4 *&#% OK4 (QNNKUVCVKP in excitability, contractile characteristics, metabolism,
and fibre size and mass (see below). These sweeping
/[QDNCUVU /[QVWDGU 7VTQRJKP changes involve the concerted control of transcription
&KȭGTGPVKCVKQP
#NVGTPCVKXG regulation, protein synthesis, protein degradation and
54( URNKEKPI /[QDNCUVU /[QVWDGU metabolic flow, the details of which have only recently
&KȭGTGPVKCVKQP
emerged. During postnatal adaptation of muscle, or
P26$
muscle remodelling, aspects of early developmental
E programmes can be reactivated and can cooperate with
/;1&
specific factors to determine fibre-type characteristics58;
MEF2, nuclear factor of activated T cells (NFAT), myo­
OK4OK4
OK4CPFOK4 genin, JUNB and SRF have prominent roles in these
/[QDNCUVU /[QVWDGU processes59–64. Post-translational modifiers of histones
&KȭGTGPVKCVKQP (histone acetylases and HDACs)64,65 and of transcription
2#:CPF2#:
factors (protein kinases and phosphatases, and ubiquitin
and SUMO transferases) cooperate in the complex inter­
Figure 3 | Regulation of major myogenic pathways by microRNAs. a | Regulation of
0CVWTG4GXKGYU^/QNGEWNCT%GNN$KQNQI[ actions between chromatin structure and transcriptional
myogenic differentiation by miR‑1 and miR‑133. miR‑1 and miR‑133 are downstream
of the myogenic transcription factors myogenic factor 5 (MYF5) and myocyte enhancer
machinery. Many of these responses are modulated
factor 2C (MEF2C) and control the expression of differentiation genes: first, through by muscle activity, either by sensing neuronal activity
the pleiotropic transcription factor serum response factor (SRF); second, by inhibiting the though intra­cellular Ca2+ levels or directly by mechani­
assembly of splicing-regulatory protein complexes by neural polypyrimidine tract-binding cal strain. In addition, various hormonal and cytokine
protein 2 (nPTB2), which controls the splicing of many mRNAs in muscles; and third, by signalling mechanisms feed into the same pathways.
inhibiting histone deacetylase 4 (HDAC4), which blocks myogenic differentiation. The adult skeletal musculature contains highly dif­
b | miR‑206 is downstream of the myogenic master transcription factor myoblast ferentiated fibre types: one slow-twitch, oxidative fibre
determination protein (MYOD) and feeds back on MyoD expression by inhibiting type (type I) and three fast-twitch, glycolytic fibre types
follistatin and bone morphogenetic proteins (BMPs), ultimately determining the (type 2)4. Fast-twitch fibre development follows defined
expression of differentiation markers such as utrophin. c | Regulation of proliferation and
patterns, involving control by SIX1 and SIX4 (see
differentiation by the myoblast transcription factors paired box protein 3 (PAX3) and PAX7
is controlled upstream by miR‑1, miR‑27, miR‑206 and miR‑486, which repress PAX3 and
above)66, which also play an important part postnatally
PAX7 and thus promote the terminal differentiation of myoblasts downstream of MYOD. in the adaptation to the fast muscle phenotype25. By con­
trast, slow-twitch fibres are determined by the transcrip­
tional repressor BLIMP1 (REF. 67), which suppresses the
The regulation of myogenesis by miR‑1 and miR‑133 transcription factor SOX6 (REFS 68,69). The distinct origin
is unclear, as the human and mouse genomes each con­ of fast-twitch and slow-twitch fibres is also highlighted
tain two bicistronic loci, which both express miR‑1 and by the observation that the developmental lineages
miR‑133. It has been reported that the selective disruption and the regeneration of slow- and fast-twitch muscles
of the miR‑1–2 locus in mice results in a severe cardiac stems from the recruitment of intrinsically different
phenotype, including 50% embryonic lethality with fre­ myogenic precursors70 or satellite cells71. Postnatally,
quent ventricular septum defects54, which is surprising SIX1 co­operates with EYA1 in fibre type differentiation.
given that the sequences of miR‑1 and miR‑133 are identi­ Indeed, forced co-expression of SIX1 and EYA1 in the
cal and that both loci are expressed in skeletal and cardiac slow-twitch fibres of soleus muscle induced a transition
muscle. Combined knockouts of the miR‑1‑2–miR‑133a1 to a fast-twitch fibre type, with the replacement of the
and miR‑1‑1–miR‑133a2 clusters are still pending and will slower myosin heavy chain isoforms I and IIA by the
provide more definitive answers. Nevertheless, it seems faster IIB and/or IIX, accompanied by the activation
likely that miR‑1 and miR‑133 are required to delineate of other fast-twitch fibre-specific genes and a switch to
the phenotype of differentiated muscl­e cells, which is glycolyti­c metabolism.
consistent with the conclusions drawn from the miR‑1 For physiological, activity-dependent adaptation of
overexpression experiment55. existing fibres, the type of neuronal activity acting on
Currently views on miRNA functions remain dispa­ a fibre is probably the most important factor determin­
rate and are complicated by some underlying technical ing fibre type. The firing pattern of neurons innervating
problems. For example, massive overexpression might fast-twitch and slow-twitch muscle fibres has markedly
Antagomirs not necessarily reflect a physiological function of an indi­ different frequencies and temporal patterns, leading
Synthetic or genetically
vidual miRNA. Instead, the non-physiological presence to different membrane potentials and ultimately to net
engineered oligonucleotides
used to silence endogenous of large amounts of an exogenous miRNA might cause differences in intracellular Ca2+ levels. These changes
microRNAs. numerous off-target effects, as also demonstrated for small can be sensed by the Ca2+-activated Ser phosphatase
interfering RNAs, and could lead to wrong conclusions56. calcineurin (also known as PP2B), which dephosphoryl­
Soleus muscle Similarly, knockdown approaches using antagomir­s, ates and thereby activates NFAT. This can then act as a
A muscle in the calf of the leg
that flexes the ankle; in rodents
although highly appealing because of their relative ease calcineurin-dependent sensor and can translate nerve
it is predominantly composed of use and wide applicability, might hold some pitfalls that activity in skeletal muscle into altered fibre-type-specific
of slow-twitch fibres. need to be explored57. gene expression programmes (FIG. 4). The nuclear version

354 | JUNE 2011 | VOLUME 12 www.nature.com/reviews/molcellbio

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

of NFAT (NFATC1) cooperates with numerous muscle- factors4. The coordinated changes of transcriptional and
specific cofactors such as MYF5 (REF. 72) to modulate splice mechanisms, protein turnover and cell fate inte­
muscle-specific gene expression 59, thus controlling grates signalling pathways from hormone and cytokine
the expression of fast and slow myosin isoforms 73. receptors, as well as the sarcomere itself.
The activit­y-dependent nuclear import and export of Satellite cells play important parts in skeletal muscle
NFATC1 is a rapid event, occurring within minutes repair 7,77,78 and thus, in the long-term, homeostasis of
of the appropriate slow-type stimulation pattern74. Its muscl­e tissue. They are also thought to be responsible for
activity is counteracted by the calcineurin inhibitor early postnatal muscle growth. By contrast, their role in
CAIN (also known as CABIN1), which promotes nuclear eliciting hypertrophic growth is disputed, as hypertrophic
export of NFATC1 both in soleus and stimulated tibialis growth in adults seems to occur without satellite cell acti­
anterior muscle. vation79. Owing to the large number of recent studies on
Further regulation of calcineurin activity is mediated satellite cells, which would require a separate review, and
by a family of sarcomere-associated proteins known as their unclear role in regulation of postnatal muscle mass,
myozenins (MOYZs; also known as FATZs and cal­ we do not discuss satellite cells further.
sarcins), which are encoded by the three MYOZ genes.
MYOZ2 is expressed in adult cardiac and slow-twitch Transcriptional control of muscle protein turnover.
skeletal muscle, whereas MYOZ1 is restricted to fast- Recently, the expression of catabolic genes was found
twitch skeletal muscle. MYOZs interact with calcineurin to be regulated by the transcription factor forkhead box
and the PDZ domain and LIM domain-containing O1 (FOXO1) and FOXO3A80,81. The activity of FOXO
protein LIM domain-binding protein 3 (LDB3; also famil­y proteins is predominantly regulated by phospho­
known as ZASP, cypher and oracle) (reviewed in REF. 2). rylation: dephosphorylated FOXOs translocate to the
MYOZ2‑knockout mice show increased calcineurin nucleus and are transcriptionally active, whereas phos­
signalling in striated muscles, suggesting that MYOZ2 phorylated FOXOs are sequestered in the cytoplasm.
acts as a brake on calcineurin activity 75. As a result of Signalling through several receptor Tyr kinases activates
the abnormal calcineurin activation, MYOZ2‑knockout the phospho­inositide 3‑kinase (PI3K)–AKT pathway,
mice have an excess of slow-twitch skeletal muscle fibres, a key regulator of muscle mass and metabolism that
which is consistent with the established roles of calci­ directly stimulates protein synthesis by activating mam­
neurin and NFAT in activity-dependent regulation of malian target of rapamycin (mTOR) and its downstream
fibre types59. Similarly, MYOZ2‑knockout mice activate targets82 (FIG. 4). Apart from its anabolic effects on protein
a cardiac hypertrophic gene programme in the absence synthesis and carbohydrate metabolism, AKT-mediated
of hypertrophy and show enhanced cardiac growth in phosphoryl­ation also blocks FOXO1 and FOXO3A
Tibialis anterior muscle
A muscle in the front muscle
response to pressure overload75. nuclear import and transcriptional activity.
compartment of the lower leg Owing to the probable function of the sarcomere Genes depending on FOXO proteins for their expres­
that helps to extend the ankle; Z‑disk (FIG. 1) as a sensor for muscle mechanical strain sion in muscle include a range of atrophy-related genes
in rodents it is predominantly (reviewed in REF. 76), calcineurin activity might also be known as atrogenes, such as atrogin 1 (also known as
composed of fast-twitch fibres.
mechanically modulated by the MYOZs in the Z‑disk. FBXO32 and MAFBX1) and the muscle-upregulated RING
Microgravity The interplay of transcriptional regulation by calci­ finger (MURF) ubiquitin ligases82. The protein products
Gravity below the 1Gal neurin, SIX1 and EYA, and MRFs, which seems possible of atrogenes target myofibrillar, metabolic and transcrip­
experienced on Earth; for from these observations, and the control of SIX1 activity tional proteins for degradation by both the ubiquiti­n–
example, during space flight. in fibre-type transition is currently unclear. proteasome system and the autophagy–lysosomal system83.

Ubiquitin–proteasome
FOXO3A, which seems to be the dominant FOXO protein
system Postnatal control of muscle mass in muscles, also controls the transcription of ubiquitous
A system of selective, The adaptive changes of muscle in response to changes autophagy-related genes (which can also be regarded as
ATP-dependent protein in activity not only determine the contractile phenotype atrogenes), such as those genes encodin­g microtubule-
degradation, in which target
but also importantly affect muscle protein turnover and associated protein 1 light chain 3 (LC3), sequestosome 1
proteins that have been
conjugated by ubiquitin thus muscle mass. Muscle mass increases by hypertrophy (SQSTM1) and probably the related protein NBR1, and
are degraded by the 26S (increased cellular protein content) and is controlled by BNIP3 — BNIP3 seems to mediate the effect of FOXO3
proteasome. The ubiquitin anabolic and catabolic mechanisms, which regulate the on autophagy 84,85. Therefore, repressing the transcription
conjugation step requires increased synthesis of muscle proteins or their degra­ of atrogenes augments the AKT-mediated increase of pro­
the activity of highly specific
ubiquitin ligases.
dation, respectively. The atrophic loss of muscle mass tein translation by shutting down protein breakdown, thus
often responds to inverse stimuli and can be triggered increasing muscle mass.
Autophagy by disuse, microgravity, catabolic steroids such as gluco­ Apart from AKT signalling, many pathways with input
A catabolic process involving corticoids, cytokines such as tumour necrosis factor from both catabolic and anabolic signals affect FOXO-
the engulfment of (usually
(TNF), genetic factors, acidosis and catabolic nutritional mediated transcription and therefore atrogene expression
damaged) organelles and
long-lived proteins or states. Hypertrophy and atrophy are associated with (FIG. 4). A direct amplification seems to be provided by
protein aggregates by changes in sarcomeric protein composition (BOX 3), meta­ atrogin 1, which triggers the degradation of calcineurin86,
double-membrane vesicles bolic enzymes and contractile phenotype, and atroph­y thus inhibiting the anabolic transcriptional signals medi­
(autophagosomes) that fuse especially can be regarded as an extreme end-stage of ated by the calcineurin–NFAT pathway (see above).
with lysosomes to form
autolysosomes, in which their
activity-regulated adaptation. In fact, disuse­d muscle loss Furthermore, although the transfer of Lys48‑linked
contents are degraded by is associated with a shift towards a fast-twitch pheno­ polyubiquitin chains generally targets proteins for protea­
acidic lysosomal hydrolases. type and the reactivation of developmental transcription somal degradation, atrogin 1 in cardiac muscle mediates

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 12 | JUNE 2011 | 355

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Lys63‑linked polyubiquitylation of FOXO1 and FOXO3A, although these mechanisms have been linked mainly to
which leads to their activation independently of AKT muscle atrophy, it is now emerging that autophagy has
and provides an atrogin 1-dependen­t positive feedback an important role not only under the extreme condi­
mechanism for atrophic gene expression87. However, tions of atrophic muscle remodelling but also in muscle

C +)( /[QUVCVKP .GRVKP 60(

2NCUOCOGODTCPG
OK4 O614
2+- +--
0WENGWU
26'0 *&#%
%.1%- 5/#& -.( )4
OK4 #4 /.2 ,70$ /;1)

#-6 (1:1 0(κ$


R %$(β /'(%
5356/
/#42
/;( 54( /74(
O614 0(#6

OK4 %CNEKPGWTKP

D
2TQVGKPU[PVJGUKU 2TQVGKPFGITCFCVKQP
%C 
#VTQIKP .%
E /74( $0+2
/.2 /;1<U
αCEVKPKP 6KVKP /[QUKPȮNCOGPV 5356/
6KVKP
#EVKPȮNCOGPV 0$4
%.1%- 0$4
5356/
/#42
/74(
%$(β
/[QOGUKP

<FKUM +DCPF /DCPF <FKUM


5CTEQOGTG
Figure 4 | Control of postnatal muscle transcription and protein turnover. a | Protein synthesis is transcriptionally
0CVWTG4GXKGYU^/QNGEWNCT%GNN$KQNQI[
regulated by anabolic transcription factors (shown in green), including myogenic regulatory factors, transcription factors
such as serum response factor (SRF), JUNB, CLOCK and nuclear factor of activated T cells (NFAT), and transcriptional
cofactors and modifiers such as muscle LIM protein (MLP) and the muscle ankyrin repeat proteins (MARPs). Several of these
are multi-compartment proteins shuttling to the nucleus from the sarcomere, where they mostly associate with the
Z‑disk or I‑band and can thus respond to mechanical strain-induced conformational changes in the sarcomere. Protein
degradation is controlled by a set of catabolic transcription factors (shown in magenta) that include forkhead box protein
O1 (FOXO1), FOXO3A, myogenin (MYOG), KLF14, nuclear factor-κB (NF‑κB), SMAD2 and SMAD3. FOXO proteins and
NF‑κB regulate the transcription of atrogenes (shown in orange): atrogin 1, muscle-upregulated RING finger (MURF)
ubiquitin ligases, and components of the autophagy pathway such as microtubule-associated protein 1 light chain 3 (LC3),
BNIP3, sequestosome 1 (SQSTM1) and possibly NBR1. In addition, the atrogenes MURF1, MURF2 and SQSTM1 translocate
to the nucleus and repress SRF activity, presumably by MURF ubiquitin ligase activity. The anabolic regulation of protein
synthesis by the phosphoinositide 3‑kinase (PI3K)–AKT cascade represses the degradation signals mediated by FOXO
proteins. The postnatal pro-atrophic action of myogenin is downstream and redundant with histone deacetylase 4 (HDAC4)
and HDAC5. Several amplifying feedback mechanisms exist that either repress or augment anabolic or catabolic signals,
including SRF-mediated expression of miR‑486, which represses FOXO proteins in hypertrophy, and FOXO protein-
mediated expression of miR‑1, which represses insulin-like growth factor 1 (IGF1) in atrophy. Additional inputs exist
through the activity of hormones and cytokines, especially anabolic signalling through IGF1, leptin and androgen receptor
(AR), and catabolic signals through tumour necrosis factor receptor (TNFR), myostatin and glucocorticoid receptor (GR).
b | Fibre-type remodelling depends largely on the sensing of Ca2+ levels by calcineurin and the resultant modulation of
NFAT and myogenic factor 5 (MYF5) activity; this is antagonized by the myozenins (MYOZs), which are sarcomeric
calcineurin inhibitors. c | Ca2+, the activator of muscle contraction, also indirectly regulates the mechanical feedback
systems embedded in the sarcomere. Titin has emerged as a sarcomeric strain-sensor at the Z‑disk, I‑band and M‑band
that feeds into the activity and localization of transcription factors and atrogenes. Solid lines represent direct actions
and dashed lines represent indirect actions. Bold dashed arrows represent receptor-linked signals. CBFβ, core-binding
factor β; IKK, inhibitor of NF‑κB kinase; MEF2C, myocyte enhancer factor 2C; mTOR, mammalian target of rapamycin;
PTEN, phosphatase and tensin homologue.

356 | JUNE 2011 | VOLUME 12 www.nature.com/reviews/molcellbio

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | Transcriptional control of sarcomere assembly of the differentiated pheno­typ­­e (FIG. 4). Of the pleiotropic
factors, SRF is required for postnatal hypertrophy, and
The concerted interplay of transcription factors and the modulation of their action on JUNB promotes muscle growth partly by directly inter­
the transcript level by microRNAs orchestrate the final functional differentiation of acting with and repressing FOXO proteins 63. Given the
muscle cells: the formation of contractile structures in sarcomeres and myofibrils
extensive involvement of miRNAs in feedback regulation
(FIG. 1). The assembly (and disassembly) of these multiprotein complexes (sarcomere
during development, it is not surprising that miRNAs
assembly or sarcomerogenesis) follows ordered pathways143, regulated on the
transcriptional, translational and post-translational levels. Furthermore, myofibril also have roles in muscle maintenance and remodellin­g.
assembly involves the participation of transient scaffolds and adaptors, notably the In fact, miR‑486, which induces myoblast differentiation
microtubule network, which seems to be necessary both for the directional dispersion by inhibiting PAX7 (see above), promotes PI3K–AKT
of protein intermediates and for that of mRNAs (reviewed in REF. 1). The highly ordered signalling by downregulating phosphatase and tensin
assembly of sarcomeres also requires the giant molecular ruler protein titin, which has homologue (PTEN; which inhibits PI3K) and directly
several functions: it forms a giant scaffold that interacts with almost all other inhibits FOXO1A itself 92. miR‑1 also amplifies the
sarcomeric proteins; it acts as an elastic spring; and it is a signal transduction protein atrophic response; it is trans­criptionally regu­lated by
acting through its carboxy-terminal protein kinase domain94,144. Therefore, it is FOXO proteins and suppresses insulin growth factor 1
unsurprising that titin depletion or truncation disrupts sarcomere assembly
(IGF1) expression, thus inhibitin­g AKT signalling 93
(reviewed in REF. 145).
(FIG. 4). These pathways are extensively modulated by
The appearance of sarcomeric proteins, notably the contractile proteins actin and
myosin and the structural proteins of the Z‑disk and M‑line (myomesin and α‑actinin, mechanical stress and cytokine and hormona­l signals
respectively) follows a strictly ordered programme of sequential transcription, (see below).
translation and incorporation into nascent cytoskeletal structures, with many
components only transiently associating with intermediates of the assembly process. Mechanical control of protein turnover. Slow- and fast-
The actin filament system and actin-associated cytoskeletal proteins — such as twitch fibres show distinct mechanical behaviour, but the
α‑actinin and the Z‑disk portion of titin — assemble first, followed by the formation mechanisms regulating their differentiation and adaption
of an M‑line scaffold of titin and myomesin and the integration of myosin filaments are also partly sensitive to mechanical stress. The adap­
(reviewed in REF. 1). This ordered sequence of the appearance and incorporation tive changes of muscle to changes in mechanical load and
of sarcomeric proteins seems to be due to both transcriptional and translational
activity not only affect the contractile phenotype but also
mechanisms (which regulate the sequential appearance of sarcomeric proteins) as well
have an impact on muscle mass.
as post-translational control mechanisms that coordinate their ordered incorporation
into assembling sarcomeres. Recently, it emerged that the expression of genes for Recent progress shows that hypertrophic and atrophic
different parts of the sarcomere is regulated by different transcription factors. signalling pathways communicate with several ‘hubs’
Although early myogenic differentiation requires, among other factors, myocyte within the sarcomere. Mechanical forces seem to have
enhancer factor 2A (MEF2A), the expression of myosin filament and M‑line proteins important roles in modulating the conformation and thus
is under the control of the late factor MEF2C146,147. The actin filament proteins are the activity of protein complexes94,95. Several transcription
regulated independently and require serum response factor (SRF), which is necessary factors and transcriptional modifiers communicate with
for de novo sarcomere assembly in the embryo because it mediates the transcription mechanosensors embedded in the sarcomere. The Z‑disk
of other of key sarcomeric proteins such as troponin C148. SRF also has a pivotal role in coordinates several direct links to mechanically modulated
postnatal hypertrophic growth of differentiated cardiac and skeletal muscle61,62,149.
transcriptional regulation. MOYZ2, a negative regulator of
calcineurin activity, seems to be mechanically sensitive75.
The transcriptional co-regulator muscle LIM protein
maintenance, by removing defective organelles and con­ (MLP; also known as CRP3) weakly associates with the
tributing to energy homeostasis88. The expression of sarcomeric cytoskeleton, but translocates to the nucleus
some atrogenes is also regulated by nuclear factor‑κB in response to mechanical strain, suggesting that it acts
(NF‑κB) (reviewed in REF. 89), thereby linking catabolic as a transducer of mechanical strain, although whether
signalling and cytokine signalling (for example, through it is a direct mechanosensor is still controversial96–98.
TNF) (FIG. 4). How FOXO proteins and NF‑κB signal­ CLOCK, another transcription factor that is also located
ling are coordinated is currently unclear — the deletion at the Z‑disk, has also been implicated in the mechanically
of the upstream activating kinase of NF‑κB, inhibitor of modulated interplay of circadian rhythm, mechanical
NF‑κΒ kinase B (IKKB), protects muscle from atroph­y 90, activity and energy demand regulation99 (FIG. 4). Similarly
whereas constitutively active FOXO protein­s are sufficien­t to MLP, CLOCK seems to shuttle between Z‑disks and the
to induce atrophy 80,81. nucleus in response to mechanical strain and to contribute
Myogenin, which as discussed above is a key regu­lator to MYOD-dependent gene expression in the daily cycling
of muscle development, also has a key role in muscle atro­ maintenance of adult skeletal muscle100. This activity may
phy by directly regulating the transcription of MURF1 be important to coordinate the actual disuse atrophy
(also known as TRIM63) and atrogin 1 (REFS 64,91). programmes with circadian rhythms, to prevent muscle
Myogenin is rapidly upregulated in denervated muscle, atrophy initiation during sleep. The recent identification
and its trans­criptional actions on atrogenes are appar­ of core binding factor-β (CBFβ), a key element of JUNB-
ently controlled by HDAC4 and HDAC5 (REF. 64). This mediated gene expression, at the Z‑disk101 emphasizes the
provides a bridge between early muscle development fact that links between cytoskeletal mechanical stress and
and postnatal adaptation of muscle. The catabolic pro­ anabolic gene expression seem to be a prominent feature
grammes controlled by FOXO proteins, myogenin and in postnatal muscle growth and maintenance control.
NF‑κB are in balance with a network of muscle-specific Thus, several mechanically responsive direct transcrip­
and pleiotropic transcription factors that cooperate in tional links exist between the sarcomeric Z‑disk and
only partly understood ways to control the maintenance the nucleus.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 12 | JUNE 2011 | 357

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Mechanosensitive signal complexes are also found at variant 119,120, or by the liver. Signalling through IGF1
the I‑band (FIG 1). Ankyrin-repeat domain 2 (ANKRD2), receptors activates the PI3K–AKT signalling pathway
cardiac ankyrin repeat protein (CARP) and diabetes- (FIG. 4) and thus represses FOXO protein activity, pro­
associated ankyrin repeat protein (DARP) — collectively moting muscle growth. The activity of IGF1 on the AKT
known as the muscle ankyrin repeat proteins (MARPs) pathway is antagonized by myostatin (also known as
— can form a complex with the I‑band proteins titin, GDF8) (reviewed in REF. 121) primarily in differentiated
myopalladin and calpain 3 (REF. 102). This complex is fibres122–124. The importance of myostatin as a negative
sensitive to stretch and muscle injury and links to the regulator of muscle mass is highlighted by its increased
transcriptional pathways that control cell survival and serum levels in patients with heart failure, which leads
muscle gene expression103,104 (FIG. 4). Owing to the very to skeletal muscle cachexia that is completely blocked
elastic nature of the I‑band region of titin, it is perhaps by genetic deletion of the gene encoding myostatin in
not surprising to find mechanically modulated transcrip­ heart tissue125,126. The myostatin-activated transcrip­
tional links in this region, which may predominantl­y tion factors SMAD2 and SMAD3 relay downstream
sense the passive strain on the sarcomere. myostatin signalling into an atrophy programme that
At the M‑band, the mechanically modulated kinase also depends on FOXO proteins 123 and that is amplified
domain of titin interacts with a complex of the protein by the inhibition of AKT and mTOR signalling 122. These
products of the atrogenes NBR1, SQSTM1 and the observations suggest that the myostatin and AKT path­
MURFs105,106. This complex dissociates under mechanica­l ways crosstalk at the cell signalling and transcriptional
arrest, and MURF1 and MURF2 (MURF3 has not yet levels (FIG. 4). Intriguingly, FOXO protein-dependent
been analysed) translocate to the cytoplasm and the atrophy through SMAD2 and SMAD3 is independent
nucleus105,107. SQSTM1 was also recently shown to trans­ of MURF1 and atrogin 1, but also leads to a decrease
locate together with MURF1 and MURF2 to the nucleus in the levels of these atrogenes122,123. Whether atrophy
in mechanically arrested skeletal muscle­s107. One of the is then promoted by the activation of other E3 ubiquitin
probable nuclear targets of MURFs is SRF105,108,109, suggest­ ligases or by the repression of pro-hypertrophic pathway­s
ing that the MURF-induced nuclear export and transcrip­ remains unclear.
tional repression of SRF may contribute to amplifying the Another hormone that affects muscle mass is leptin,
transcriptional atrophy programme105,107,110; for example, a major regulator of energy intake and expenditure.
by suppressing SRF-dependent miR‑486 expression Leptin was shown to positively regulate muscle mass
(FIG. 4). The multiple sarcomeric and nuclear localiza­ by suppressing the activity of FOXO3A, further dem­
tions of MURFs suggest their intracellular localization onstrating how muscle and fat tissue metabolism are
may also be important in coordinating their diverse interlinked127 (FIG. 4).
functions in protein turnover regulation. Titin might A further hormonal input feeding into the AKT–
act as an activity-dependent brake on protein degrada­ FOXO protein nexus occurs by signalling through the
tion by suppressing the expression or stability of atro­ cytoplasmic steroid receptors, the anabolic androgen
genes (FIG. 4). Both NBR1 and SQSTM1 promote the receptor and the catabolic glucocorticoid receptor. The
autophagy of poly­ubiquitylated proteins by interacting muscle anabolic effects of androgens have long been
with LC3 (REFS 111–114), but the substrates they recruit known; however, it has only recently been shown, using
for degradation remain to be identified. In addition, these muscle-specific androgen receptor‑knockout mice,
proteins interact with several protein kinases apart from that the myocytic androgen receptor is required for
titin and seem to have major signalling functions115,116. the production of the androgen-induced IGF1 isoform
MURF1 also seems to be required for TNF-induced IGF1EA128 and can thus regulate autocrine or paracrine
reduction in skeletal muscle force development 117, sug­ activation of the muscle AKT signalling pathway (FIG. 4).
gesting that MURF signalling might be involved in the Conversely, the glucocorticoid receptor acts upstream
NF‑κB-mediated atrophy response89 that is a hallmark of of FOXO proteins and is itself regulated in a feedback
cachexia118. Direct links to hormone and cytokine recep­ loop by the IGF1–AKT-activated mTOR129, thus pro­
tors, such as those between the atrogene SQSTM1 and viding a crosstalk loop between anabolic and catabolic
TNF receptor signalling, suggest a complex interplay hormonal signals. This feedback loop also involves the
of mechanical and cell surface receptor-linked signal­ transcription factor KLF15, which is implicated in sev­
ling 116. Thus, the differentiated sarcomere serves as a eral skeletal muscle metabolic processes. Exploiting such
complex feedback device for the adaptive remodelling cross-connection­s to suppress the unwanted atrophic
Cachexia of the contractile machinery and its supporting energy effects of corticoid treatment might be an interesting
A syndrome of muscle loss that metabolism in response to mechanical load, and several practical application of this observation.
is usually caused by increased mechanically active regions feed back to distinct trans­
catabolic metabolism.
criptional programmes. It seems likely that further such Conclusions and perspectives
E3 ubiquitin ligases mechano–transcriptional links will emerge. The versatility and plasticity of striated muscles is due
A group of proteins that to finely tuned networks of transcription factors and
mediate the transfer of Hormonal control of muscle growth. Muscle mass can their regulation by extracellular and intracellular cyto­
ubiquitin, often by linking the also be regulated by hormones. One of the most promi­ skeletal signals. The embryonic origin and the functional
catalytic activity of an E2
transferase to recognition
nent known muscle growth factors is IGF1, which remodelling of slow-twitch oxidative or fast-twitch
and binding of the specific is secreted by myocytes in an autocrine manner in glyco­lytic muscle fibres has become mechanistically
substrates. response to mechanical strain as a muscle-specific splice clear. Similarly, the control of muscle mass in response

358 | JUNE 2011 | VOLUME 12 www.nature.com/reviews/molcellbio

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Sarcopenia to activity has become clearer by the identification of circadian rhythm transcription factors in coordinating
The degenerative loss of the major pathways controlling atrogene expression muscle growth. This information will be increasingl­y
skeletal muscle mass and and the cytokine and mechanical stimuli that modu­ important for the development of new muscle
strength associated with ageing late their activity on the gene and protein levels. Model therapeutics.
and pathological processes.
organisms from nematodes to vertebrates have proved The emerging pivotal role of miRNAs in muscle devel­
invaluable in outlining these molecular mechanisms. It opment and postnatal remodelling adds additional layers
will now be important to identify how the mechanisms of complexity, if not even complications, to the analysis
specifying embryonic fibre fates are postnatally modu­ and understanding of the molecular networks of these
lated by activity-regulate­d pathways. The redeployment processes. However, exploiting miRNAs in pathological
of embryonic factors in postnatal muscle remodelling states — for example, miR‑206 in rhabdomyo­sarcoma
is an exciting recent development, both for our mecha­ or miR‑1 and miR‑486 in muscle atrophy — could open
nistic understanding of postnatal adaptation and for the door to a whole new class of therapeutics with a wide
the identification of pathways amenable to pharmaco­ range of applications. The technical problems of imple­
logical intervention. It is still an open question how menting nucleic acids as therapeutic agents are by no
muscl­e stem cells are specified, and how this popula­ means trivial, and the promise of miRNAs in therapy
tion is maintained to ensure proper muscle homeostasis will therefore probably face practical challenges. It seems
under different physio­logical conditions. Understanding logical and tempting to target the downstream effectors
these mechanisms in more detail may lead to the iden­ of atrophic pathways, the protein products of the atro­
tification of molecular targets that could be exploited to genes, in the pursuit of small-molecule drugs against
direct cellular therapies for muscular dystrophies, which atrophy. Indeed, a small-molecule inhibitor of MURF1
are partly manifested by depletion of the satellite cell that is active in cultured muscle cells was recently
pool. Similarly, preventing dysfunction or decline of the reported, suggesting that MURFs might indeed be
number of satellite cells during ageing could ameliorate accessible pharmacological targets130. However, targeting
sarcopenia, a major cause of age-related disability. MURFs may lead to unexpected physiological effects,
Many of the pathways controlling postnatal muscle as MURFs are expressed early at the onset of muscle dif­
adaptation and remodelling also operate in the heart. ferentiation109,131, show high homology and have a prob­
Indeed, research on skeletal muscle has spearheaded able role in protein homeostasis. Furthermore, a recent
the research on muscle remodelling and brought the report showed that mutations in MURF1 cause cardiac
molecular analysis of cardiac atrophy and remodelling disease132. Harnessing the past research on muscle devel­
to a new level. However, we still need to understan­d the opment and signalling for therapeutic purposes is there­
complex interplay of hypertrophic and atrophic factors, fore likely to depend on further insights into the basic
the nuclear roles of atrogenes and the intriguing role of mechanisms of these complex regulatory networks.

1. Ehler, E. & Gautel, M. The sarcomere and 11. Kassar-Duchossoy, L. et al. Mrf4 determines skeletal 20. McKinnell, I. W. et al. Pax7 activates myogenic genes
sarcomerogenesis. Adv. Exp. Med. Biol. 642, 1–14 muscle identity in Myf5:MyoD double-mutant mice. by recruitment of a histone methyltransferase
(2008). Nature 431, 466–471 (2004). complex. Nature Cell Biol. 10, 77–84 (2008).
2. Lange, S., Ehler, E. & Gautel, M. From A to Z and 12. Sato, T., Rocancourt, D., Marques, L., 21. Daston, G., Lamar, E., Olivier, M. & Goulding, M.
back? Multicompartment proteins in the sarcomere. Thorsteinsdottir, S. & Buckingham, M. A Pax3/Dmrt2/ Pax‑3 is necessary for migration but not differentiation
Trends Cell Biol. 16, 11–18 (2006). Myf5 regulatory cascade functions at the onset of of limb muscle precursors in the mouse. Development
3. Pette, D. & Staron, R. S. Myosin isoforms, muscle fiber myogenesis. PLoS Genet. 6, e1000897 (2010). 122, 1017–1027 (1996).
types, and transitions. Microsc. Res. Tech. 50, 13. Tajbakhsh, S., Rocancourt, D., Cossu, G. & 22. Bajard, L. et al. A novel genetic hierarchy functions
500–509 (2000). Buckingham, M. Redefining the genetic hierarchies during hypaxial myogenesis: Pax3 directly activates
4. Schiaffino, S. Fibre types in skeletal muscle: controlling skeletal myogenesis: Pax‑3 and Myf‑5 act Myf5 in muscle progenitor cells in the limb.
a personal account. Acta. Physiol. (Oxf.) 199, upstream of MyoD. Cell 89, 127–138 (1997). Genes Dev. 20, 2450–2464 (2006).
451–463 (2010). 14. Brunelli, S., Relaix, F., Baesso, S., Buckingham, M. & The direct activation of Myf5 by PAX3 in limb
An authoritative review of skeletal muscle Cossu, G. β-catenin-independent activation of MyoD in muscle cells is described.
phenotypes and the molecular mechanisms presomitic mesoderm requires PKC and depends on 23. Buchberger, A., Freitag, D. & Arnold, H. H. A homeo-
controlling muscle fibre types. Pax3 transcriptional activity. Dev. Biol. 304, 604–614 paired domain-binding motif directs Myf5 expression
5. Zammit, P. S. All muscle satellite cells are equal, but (2007). in progenitor cells of limb muscle. Development 134,
are some more equal than others? J. Cell Sci. 121, 15. Maroto, M. et al. Ectopic Pax‑3 activates MyoD and 1171–1180 (2007).
2975–2982 (2008). Myf‑5 expression in embryonic mesoderm and neural 24. Giordani, J. et al. Six proteins regulate the activation
6. Ten Broek, R. W., Grefte, S. & Von den Hoff, J. W. tissue. Cell 89, 139–148 (1997). of Myf5 expression in embryonic mouse limbs.
Regulatory factors and cell populations involved in 16. Mennerich, D. & Braun, T. Activation of myogenesis by Proc. Natl Acad. Sci. USA 104, 11310–11315
skeletal muscle regeneration. J. Cell. Physiol. 224, the homeobox gene Lbx1 requires cell proliferation. (2007).
7–16 (2010). EMBO J. 20, 7174–7183 (2001). 25. Grifone, R. et al. Six1 and Eya1 expression can
7. Tedesco, F. S., Dellavalle, A., Diaz-Manera, J., 17. Mansouri, A., Stoykova, A., Torres, M. & Gruss, P. reprogram adult muscle from the slow-twitch
Messina, G. & Cossu, G. Repairing skeletal muscle: Dysgenesis of cephalic neural crest derivatives in phenotype into the fast-twitch phenotype. Mol. Cell.
regenerative potential of skeletal muscle stem cells. Pax7‑/- mutant mice. Development 122, 831–838 Biol. 24, 6253–6267 (2004).
J. Clin. Invest. 120, 11–19 (2010). (1996). 26. Laclef, C. et al. Altered myogenesis in Six1‑deficient
A timely review on the role of satellite cells in 18. Relaix, F., Rocancourt, D., Mansouri, A. & mice. Development 130, 2239–2252 (2003).
muscle damage repair. Buckingham, M. A Pax3/Pax7‑dependent population 27. Grifone, R. et al. Six1 and Six4 homeoproteins are
8. Bryson-Richardson, R. J. & Currie, P. D. The genetics of skeletal muscle progenitor cells. Nature 435, required for Pax3 and Mrf expression during
of vertebrate myogenesis. Nature Rev. Genet. 9, 948–953 (2005). myogenesis in the mouse embryo. Development 132,
632–646 (2008). This work defines the major populations of 2235–2249 (2005).
A comprehensive review of the embryological progenitor cells in muscle development. 28. Grifone, R. et al. Eya1 and Eya2 proteins are required
processes leading to the formation of skeletal 19. Lagha, M. et al. Pax3 regulation of FGF signaling for hypaxial somitic myogenesis in the mouse embryo.
muscles. affects the progression of embryonic progenitor cells Dev. Biol. 302, 602–616 (2007).
9. Buckingham, M. Making muscle in mammals. into the myogenic program. Genes Dev. 22, 29. Penn, B. H., Bergstrom, D. A., Dilworth, F. J.,
Trends Genet. 8, 144–149 (1992). 1828–1837 (2008). Bengal, E. & Tapscott, S. J. A MyoD-generated feed-
10. Berkes, C. A. & Tapscott, S. J. MyoD and the The role of PAX3 in the lineage commitment of forward circuit temporally patterns gene expression
transcriptional control of myogenesis. Semin. Cell Dev. myogenic progenitor cells in concert with other during skeletal muscle differentiation. Genes Dev. 18,
Biol. 16, 585–595 (2005). myogenic factors is described. 2348–2353 (2004).

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 12 | JUNE 2011 | 359

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

30. Molkentin, J. D., Black, B. L., Martin, J. F. & 53. Dey, B. K., Gagan, J. & Dutta, A. miR‑206 and ‑486 74. Tothova, J. et al. NFATc1 nucleocytoplasmic shuttling
Olson, E. N. Cooperative activation of muscle gene induce myoblast differentiation by downregulating is controlled by nerve activity in skeletal muscle.
expression by MEF2 and myogenic bHLH proteins. Pax7. Mol. Cell. Biol. 31, 203–214 (2011). J. Cell Sci. 119, 1604–1611 (2006).
Cell 83, 1125–1136 (1995). 54. Zhao, Y. et al. Dysregulation of cardiogenesis, cardiac Distinct neuronal activity patterns modulate
31. Lu, J. R. et al. Control of facial muscle development by conduction, and cell cycle in mice lacking miRNA‑1‑2. cytoplasmic Ca2+ levels, and thus the activity
MyoR and capsulin. Science 298, 2378–2381 Cell 129, 303–317 (2007). of the transcription factor NFATC1 by modulating
(2002). 55. Lim, L. P. et al. Microarray analysis shows that some the activity of its activator, calcineurin.
One of the first papers to delineate the microRNAs downregulate large numbers of target 75. Frey, N. et al. Mice lacking calsarcin‑1 are sensitized to
transcriptional network that drives head muscle mRNAs. Nature 433, 769–773 (2005). calcineurin signaling and show accelerated
development. A seminal paper demonstrating the regulation of cardiomyopathy in response to pathological
32. Dong, F. et al. Pitx2 promotes development of multiple targets by miRNAs. biomechanical stress. Nature Med. 10, 1336–1343
splanchnic mesoderm-derived branchiomeric muscle. 56. Kleinman, M. E. et al. Sequence- and target- (2004).
Development 133, 4891–4899 (2006). independent angiogenesis suppression by siRNA via 76. Hoshijima, M. Mechanical stress-strain sensors
33. Gage, P. J., Suh, H. & Camper, S. A. Dosage TLR3. Nature 452, 591–597 (2008). embedded in cardiac cytoskeleton: Z disk, titin, and
requirement of Pitx2 for development of multiple 57. Krutzfeldt, J. et al. Specificity, duplex degradation and associated structures. Am. J. Physiol. Heart Circ.
organs. Development 126, 4643–4651 (1999). subcellular localization of antagomirs. Nucleic Acids Physiol. 290, H1313–H1325 (2006).
34. Jerome, L. A. & Papaioannou, V. E. DiGeorge Res. 35, 2885–2892 (2007). 77. Zammit, P. S., Partridge, T. A. & Yablonka-Reuveni, Z.
syndrome phenotype in mice mutant for the T‑box 58. Bassel-Duby, R. & Olson, E. N. Signaling pathways in The skeletal muscle satellite cell: the stem cell that
gene, Tbx1. Nature Genet. 27, 286–291 (2001). skeletal muscle remodeling. Annu. Rev. Biochem. 75, came in from the cold. J. Histochem. Cytochem. 54,
35. Kelly, R. G., Jerome-Majewska, L. A. & Papaioannou, 19–37 (2006). 1177–1191 (2006).
V. E. The del22q11.2 candidate gene Tbx1 regulates 59. Schiaffino, S., Sandri, M. & Murgia, M. Activity- 78. Sacco, A., Doyonnas, R., Kraft, P., Vitorovic, S. &
branchiomeric myogenesis. Hum. Mol. Genet. 13, dependent signaling pathways controlling muscle Blau, H. M. Self-renewal and expansion of single
2829–2840 (2004). diversity and plasticity. Physiology (Bethesda) 22, transplanted muscle stem cells. Nature 456,
36. Sambasivan, R. et al. Distinct regulatory cascades 269–278 (2007). 502–506 (2008).
govern extraocular and pharyngeal arch muscle 60. Potthoff, M. J. et al. Histone deacetylase degradation 79. Blaauw, B. et al. Inducible activation of Akt increases
progenitor cell fates. Dev. Cell 16, 810–821 (2009). and MEF2 activation promote the formation of skeletal muscle mass and force without satellite cell
This paper addresses the distinct regulatory slow-twitch myofibers. J. Clin. Invest. 117, activation. FASEB J. 23, 3896–3905 (2009).
circuits that determine head muscle fates. 2459–2467 (2007). 80. Sandri, M. et al. Foxo transcription factors induce
37. Ying, S. Y., Chang, D. C. & Lin, S. L. The microRNA The identification of regulatory circuits that are the atrophy-related ubiquitin ligase atrogin‑1 and
(miRNA): overview of the RNA genes that modulate involved in slow-twitch muscle fibre formation. cause skeletal muscle atrophy. Cell 117, 399–412
gene function. Mol. Biotechnol. 38, 257–268 (2008). 61. Li, S. et al. Requirement for serum response factor for (2004).
38. Bushati, N. & Cohen, S. M. microRNA functions. skeletal muscle growth and maturation revealed by 81. Stitt, T. N. et al. The IGF‑1/PI3K/Akt pathway prevents
Annu. Rev. Cell Dev. Biol. 23, 175–205 (2007). tissue-specific gene deletion in mice. Proc. Natl Acad. expression of muscle atrophy-induced ubiquitin ligases
39. Ge, Y. & Chen, J. MicroRNAs in skeletal myogenesis. Sci. USA 102, 1082–1087 (2005). by inhibiting FOXO transcription factors. Mol. Cell. 14,
Cell Cycle 10, 441–448 (2011). 62. Parlakian, A. et al. Targeted inactivation of serum 395–403 (2004).
40. Chen, J. F. et al. The role of microRNA‑1 and response factor in the developing heart results in FOXO proteins are identified as the major
microRNA‑133 in skeletal muscle proliferation and myocardial defects and embryonic lethality. Mol. Cell. transcription factors regulating the expression
differentiation. Nature Genet. 38, 228–233 (2006). Biol. 24, 5281–5289 (2004). of MURF atrogenes in atrophy.
41. Rao, P. K., Kumar, R. M., Farkhondeh, M., These two papers identify the essential role of 82. Sandri, M. Signaling in muscle atrophy and
Baskerville, S. & Lodish, H. F. Myogenic factors that SRF for postnatal muscle growth. hypertrophy. Physiology (Bethesda) 23, 160–170
regulate expression of muscle-specific microRNAs. 63. Raffaello, A. et al. JunB transcription factor maintains (2008).
Proc. Natl Acad. Sci. USA 103, 8721–8726 (2006). skeletal muscle mass and promotes hypertrophy. A comprehensive review of the mechanisms
42. Sokol, N. S. & Ambros, V. Mesodermally expressed J. Cell Biol. 191, 101–113 (2010). regulating muscle protein turnover and
Drosophila microRNA‑1 is regulated by Twist and is JUNB directly represses the activity of FOXO metabolism.
required in muscles during larval growth. Genes Dev. proteins and thus augments anti-atrophic 83. Willis, M. S., Schisler, J. C., Portbury, A. L. &
19, 2343–2354 (2005). transcriptional pathways. Patterson, C. Build it up – tear it down: protein quality
43. Zhao, Y., Samal, E. & Srivastava, D. Serum response 64. Moresi, V. et al. Myogenin and class II HDACs control control in the cardiac sarcomere. Cardiovasc. Res. 81,
factor regulates a muscle-specific microRNA that neurogenic muscle atrophy by inducing E3 ubiquitin 439–448 (2009).
targets Hand2 during cardiogenesis. Nature 436, ligases. Cell 143, 35–45 (2010). 84. Mammucari, C. et al. FoxO3 controls autophagy in
214–220 (2005). Myogenin is redeployed in postnatal muscle under skeletal muscle in vivo. Cell Metab. 6, 458–471
The identification of upstream regulators and hierarchical control by HDACs to drive the (2007).
downstream targets of muscle-specific miRNAs. expression of atrogenes. 85. Zhao, J. et al. FoxO3 coordinately activates protein
44. Liu, N. et al. An intragenic MEF2‑dependent enhancer 65. Molkentin, J. D. Dichotomy of Ca2+ in the heart: degradation by the autophagic/lysosomal and
directs muscle-specific expression of microRNAs 1 and contraction versus intracellular signaling. J. Clin. proteasomal pathways in atrophying muscle cells.
133. Proc. Natl Acad. Sci. USA 104, 20844–20849 Invest. 116, 623–626 (2006). Cell Metab. 6, 472–483 (2007).
(2007). 66. Niro, C. et al. Six1 and Six4 gene expression is References 84 and 85 identify a crucial pathway
45. Sweetman, D. et al. Specific requirements of MRFs for necessary to activate the fast-type muscle gene that controls muscle mass and homeostatic
the expression of muscle specific microRNAs, miR‑1, program in the mouse primary myotome. Dev. Biol. regulation through the transcription of autophagy
miR‑206 and miR‑133. Dev. Biol. 321, 491–499 338, 168–182 (2010). and lysosomal proteins under the control of FOXO
(2009). 67. Baxendale, S. et al. The B‑cell maturation factor proteins.
46. van Rooij, E. et al. Control of stress-dependent Blimp‑1 specifies vertebrate slow-twitch muscle fiber 86. Li, H. H. et al. Atrogin‑1/muscle atrophy F‑box inhibits
cardiac growth and gene expression by a microRNA. identity in response to Hedgehog signaling. calcineurin-dependent cardiac hypertrophy by
Science 316, 575–579 (2007). Nature Genet. 36, 88–93 (2004). participating in an SCF ubiquitin ligase complex.
47. Boutz, P. L., Chawla, G., Stoilov, P. & Black, D. L. References 66 and 67 identify the major J. Clin. Invest. 114, 1058–1071 (2004).
MicroRNAs regulate the expression of the alternative developmental programmes determining 87. Li, H. H. et al. Atrogin‑1 inhibits Akt-dependent
splicing factor nPTB during muscle development. future slow- or fast-twitch muscle types. cardiac hypertrophy in mice via ubiquitin-dependent
Genes Dev. 21, 71–84 (2007). 68. Hagiwara, N., Yeh, M. & Liu, A. Sox6 is required for coactivation of forkhead proteins. J. Clin. Invest. 117,
The regulation of alternative splicing by miRNAs normal fiber type differentiation of fetal skeletal 3211–3223 (2007).
through control of a differential splicing factor. muscle in mice. Dev. Dyn. 236, 2062–2076 88. Masiero, E. et al. Autophagy is required to maintain
48. Taulli, R. et al. The muscle-specific microRNA miR‑206 (2007). muscle mass. Cell Metab. 10, 507–515 (2009).
blocks human rhabdomyosarcoma growth in 69. von Hofsten, J. et al. Prdm1- and Sox6‑mediated Autophagy is required for the homeostasis of
xenotransplanted mice by promoting myogenic transcriptional repression specifies muscle fibre type muscle mass and is not per se detrimental, as its
differentiation. J. Clin. Invest. 119, 2366–2378 in the zebrafish embryo. EMBO Rep. 9, 683–689 inhibition is shown here to lead to a paradoxical
(2009). (2008). loss of muscle mass.
49. Williams, A. H. et al. MicroRNA‑206 delays ALS 70. Hinits, Y., Osborn, D. P. & Hughes, S. M. Differential 89. Mourkioti, F. & Rosenthal, N. NF‑κB signaling in
progression and promotes regeneration of requirements for myogenic regulatory factors skeletal muscle: prospects for intervention in muscle
neuromuscular synapses in mice. Science 326, distinguish medial and lateral somitic, cranial and fin diseases. J. Mol. Med. 86, 747–759 (2008).
1549–1554 (2009). muscle fibre populations. Development 136, 90. Mourkioti, F. et al. Targeted ablation of IKK2 improves
50. Crist, C. G. et al. Muscle stem cell behavior is modified 403–414 (2009). skeletal muscle strength, maintains mass, and
by microRNA‑27 regulation of Pax3 expression. 71. Kalhovde, J. M. et al. ‘Fast’ and ‘slow’ muscle fibres in promotes regeneration. J. Clin. Invest. 116,
Proc. Natl Acad. Sci. USA 106, 13383–13387 hindlimb muscles of adult rats regenerate from 2945–2954 (2006).
(2009). intrinsically different satellite cells. J. Physiol. 562, Transcription of MURF atrogenes is controlled by
51. Hirai, H. et al. MyoD regulates apoptosis of myoblasts 847–857 (2005). NF-κB activity.
through microRNA-mediated down-regulation of Pax3. 72. Perez-Ruiz, A., Gnocchi, V. F. & Zammit, P. S. Control 91. Macpherson, P. C., Wang, X. & Goldman, D.
J. Cell Biol. 191, 347–365 (2010). of Myf5 activation in adult skeletal myonuclei Myogenin regulates denervation-dependent muscle
52. Chen, J. F. et al. microRNA‑1 and microRNA‑206 requires ERK signalling. Cell Signal. 19, 1671–1680 atrophy in mouse soleus muscle. J. Cell. Biochem.
regulate skeletal muscle satellite cell proliferation and (2007). 4 Apr 2011(doi: 10.1002/jcb.23136).
differentiation by repressing Pax7. J. Cell Biol. 190, 73. Calabria, E. et al. NFAT isoforms control activity- 92. Small, E. M. et al. Regulation of PI3‑kinase/Akt
867–879 (2010). dependent muscle fiber type specification. signaling by muscle-enriched microRNA‑486.
The discovery of the role of muscle-specific miRNAs Proc. Natl Acad. Sci. USA 106, 13335–13340 Proc. Natl Acad. Sci. USA 107, 4218–4223
in the control of satellite cells. (2009). (2010).

360 | JUNE 2011 | VOLUME 12 www.nature.com/reviews/molcellbio

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

miR‑486 is transcriptionally controlled by SRF 112. Pankiv, S. et al. p62/SQSTM1 binds directly to atg8/ 133. Kalcheim, C. & Ben-Yair, R. Cell rearrangements during
and negatively regulates FOXO protein expression, lc3 to facilitate degradation of ubiquitinated protein development of the somite and its derivatives.
providing an amplifying feedback loop in aggregates by autophagy. J. Biol. Chem. 282, Curr. Opin. Genet. Dev. 15, 371–380 (2005).
hypertrophying muscle. 24131–24145 (2007). 134. Gros, J., Scaal, M. & Marcelle, C. A two-step
93. Elia, L. et al. Reciprocal regulation of microRNA‑1 and 113. Waters, S., Marchbank, K., Solomon, E., mechanism for myotome formation in chick. Dev. Cell
insulin-like growth factor‑1 signal transduction Whitehouse, C. & Gautel, M. Interactions with LC3 and 6, 875–882 (2004).
cascade in cardiac and skeletal muscle in physiological polyubiquitin chains link nbr1 to autophagic protein 135. Tzahor, E. Heart and craniofacial muscle development:
and pathological conditions. Circulation 120, turnover. FEBS Lett. 583, 1846–1852 (2009). a new developmental theme of distinct myogenic
2377–2385 (2009). 114. Kirkin, V. et al. A role for NBR1 in autophagosomal fields. Dev. Biol. 327, 273–279 (2009).
94. Gautel, M. The sarcomeric cytoskeleton: who picks degradation of ubiquitinated substrates. Mol. Cell 33, 136. Noden, D. M. The role of the neural crest in patterning
up the strain? Curr. Opin. Cell Biol. 23, 39–46 505–516 (2009). of avian cranial skeletal, connective, and muscle
(2011). 115. Moscat, J., Diaz-Meco, M. T. & Wooten, M. W. Signal tissues. Dev. Biol. 96, 144–165 (1983).
95. Kruger, M. & Linke, W. A. The giant protein titin: a integration and diversification through the p62 137. Rinon, A. et al. Cranial neural crest cells regulate head
regulatory node that integrates myocyte signaling scaffold protein. Trends Biochem. Sci. 32, 95–100 muscle patterning and differentiation during
pathways. J. Biol. Chem. 21 Jan 2011 (doi:10.1074/ (2007). vertebrate embryogenesis. Development 134,
jbc.R110.173260). 116. Gautel, M. Cytoskeletal protein kinases: titin and its 3065–3075 (2007).
96. Boateng, S. Y., Senyo, S. E., Qi, L., Goldspink, P. H. & relations in strain sensing. Pflugers Arch. 18 Mar 138. Grenier, J., Teillet, M. A., Grifone, R., Kelly, R. G. &
Russell, B. Myocyte remodelling in response to 2011(doi:10.1007/s00424‑011‑0946–0941). Duprez, D. Relationship between neural crest cells and
hypertrophic stimuli requires nucleocytoplasmic 117. Adams, V. et al. Induction of MuRF1 is essential for cranial mesoderm during head muscle development.
shuttling of muscle LIM protein. J. Mol. Cell. Cardiol. TNF‑α‑induced loss of muscle function in mice. J. Mol. PLoS ONE 4, e4381 (2009).
47, 426–435 (2009). Biol. 384, 48–59 (2008). 139. Borycki, A. G. & Emerson, C. P., Jr. Multiple tissue
97. Geier, C. et al. Beyond the sarcomere: CSRP3 118. Tisdale, M. J. Mechanisms of cancer cachexia. Physiol. interactions and signal transduction pathways control
mutations cause hypertrophic cardiomyopathy. Rev. 89, 381–410 (2009). somite myogenesis. Curr. Top. Dev. Biol. 48, 165–224
Hum. Mol. Genet. 17, 2753–2765 (2008). 119. Goldspink, G. Changes in muscle mass and phenotype (2000).
98. Knoll, R. et al. The cardiac mechanical stretch sensor and the expression of autocrine and systemic growth 140. Reshef, R., Maroto, M. & Lassar, A. B. Regulation of
machinery involves a Z disc complex that is defective factors by muscle in response to stretch and overload. dorsal somitic cell fates: BMPs and Noggin control the
in a subset of human dilated cardiomyopathy. J. Anat. 194, 323–334 (1999). timing and pattern of myogenic regulator expression.
Cell 111, 943–955 (2002). 120. Vinciguerra, M., Musaro, A. & Rosenthal, N. Genes Dev. 12, 290–303 (1998).
99. Qi, L. & Boateng, S. Y. The circadian protein Clock Regulation of muscle atrophy in aging and disease. 141. Borello, U. et al. The Wnt/β-catenin pathway regulates
localizes to the sarcomeric Z‑disk and is a sensor of Adv. Exp. Med. Biol. 694, 211–233 (2010). Gli-mediated Myf5 expression during somitogenesis.
myofilament cross-bridge activity in cardiac myocytes. 121. Matsakas, A. & Patel, K. Intracellular signalling Development 133, 3723–3732 (2006).
Biochem. Biophys. Res. Commun. 351, 1054–1059 pathways regulating the adaptation of skeletal muscle 142. Chen, A. E., Ginty, D. D. & Fan, C. M. Protein kinase A
(2006). to exercise and nutritional changes. Histol. signalling via CREB controls myogenesis induced by
100. Andrews, J. L. et al. CLOCK and BMAL1 regulate Histopathol. 24, 209–222 (2009). Wnt proteins. Nature 433, 317–322 (2005).
MyoD and are necessary for maintenance of skeletal 122. Trendelenburg, A. U. et al. Myostatin reduces 143. Sanger, J. W., Wang, J., Fan, Y., White, J. & Sanger,
muscle phenotype and function. Proc. Natl Acad. Sci. AKT/TORC1/p70S6K signaling, inhibiting myoblast J. M. Assembly and dynamics of myofibrils. J. Biomed.
USA 107, 19090–19095 (2010). differentiation and myotube size. Am. J. Physiol. Cell Biotechnol. 2010, 858606 (2010).
Circadian rhythm, mechanical activity and myogenic Physiol. 296, C1258–C1270 (2009). 144. Linke, W. A. & Kruger, M. The giant protein titin as an
transcription regulation are linked in references 99 123. Sartori, R. et al. Smad2 and 3 transcription factors integrator of myocyte signaling pathways. Physiology
and 100. control muscle mass in adulthood. Am. J. Physiol. Cell (Bethesda) 25, 186–198 (2010).
101. Meder, B. et al. JunB-CBFβ signaling is essential to Physiol. 296, C1248–C1257 (2009). 145. Kontrogianni-Konstantopoulos, A., Ackermann, M. A.,
maintain sarcomeric Z‑disc structure and when 124. Morissette, M. R., Cook, S. A., Buranasombati, C., Bowman, A. L., Yap, S. V. & Bloch, R. J. Muscle giants:
defective leads to heart failure. J. Cell Sci. 123, Rosenberg, M. A. & Rosenzweig, A. Myostatin inhibits molecular scaffolds in sarcomerogenesis. Physiol. Rev.
2613–2620 (2010). IGF‑I‑induced myotube hypertrophy through Akt. 89, 1217–1267 (2009).
102. Hayashi, C. et al. Multiple molecular interactions Am. J. Physiol. Cell Physiol. 297, C1124–C1132 146. Hinits, Y. & Hughes, S. M. Mef2s are required for
implicate the connectin/titin N2A region as a (2009). thick filament formation in nascent muscle fibres.
modulating scaffold for p94/calpain 3 activity in 125. George, I. et al. Myostatin activation in patients Development 134, 2511–2519 (2007).
skeletal muscle. J. Biol. Chem. 283, 14801–14814 with advanced heart failure and after mechanical 147. Potthoff, M. J. et al. Regulation of skeletal muscle
(2008). unloading. Eur. J. Heart Fail. 12, 444–453 sarcomere integrity and postnatal muscle function
103. Tsukamoto, Y. et al. Arpp/Ankrd2, a member of the (2010). by Mef2c. Mol. Cell Biol. 27, 8143–8151(2007).
muscle ankyrin repeat proteins (MARPs), translocates 126. Heineke, J. et al. Genetic deletion of myostatin from References 146 and 147 identify MEF2C as the
from the I‑band to the nucleus after muscle injury. the heart prevents skeletal muscle atrophy in heart main transcription factor that directs the formation
Histochem. Cell Biol. 129, 55–64 (2008). failure. Circulation 121, 419–425 (2010). of mature sarcomeres by driving the expression of
104. Kojic, S. et al. The Ankrd2 protein, a link between The secretion of myostatin by failing hearts is a myosin and myomesin.
the sarcomere and the nucleus in skeletal muscle. major factor in heart failure-associated muscle 148. Niu, Z. et al. Serum response factor orchestrates
J. Mol. Biol. 339, 313–325 (2004). loss. nascent sarcomerogenesis and silences the
105. Lange, S. et al. The kinase domain of titin controls 127. Sainz, N. et al. Leptin administration favors muscle biomineralization gene program in the heart.
muscle gene expression and protein turnover. Science mass accretion by decreasing FoxO3a and Proc. Natl Acad. Sci. USA 105, 17824–17829
308, 1599–1603 (2005). increasing PGC‑1α in ob/ob mice. PLoS ONE 4, (2008).
A mechanically modulated pathway acting through e6808 (2009). 149. Niu, Z., Li, A., Zhang, S. X. & Schwartz, R. J. Serum
the giant protein kinase titin regulates the activity 128. Chambon, C. et al. Myocytic androgen receptor response factor micromanaging cardiogenesis.
of the atrogenes MURF, SQSTM1 and NBR1, controls the strength but not the mass of limb Curr. Opin. Cell Biol. 19, 618–627 (2007).
with MURF2 acting as a repressor of SRF activity. muscles. Proc. Natl Acad. Sci. USA 107, 150. Rios, A. C., & Marcelle C. Head muscles: aliens who
Disruption of this titin link leads to human 14327–14332 (2010). came in from the cold? Dev. Cell 16,
myopathy. 129. Shimizu, N. et al. Crosstalk between glucocorticoid 779–780 (2009).
106. Puchner, E. et al. Mechanoenzymatics of titin kinase. receptor and nutritional sensor mTOR in skeletal
Proc. Natl Acad. Sci. USA 105, 13385–13390 muscle. Cell Metab. 13, 170–182 (2011). Acknowledgements
(2008). AR, a well-known hypertrophic factor, augments in We acknowledge the contributions that have also advanced
107. Ochala, J. et al. Preferential skeletal muscle myosin feedback loops the anabolic signals of the the field by those authors whose work we could not cite owing
loss in response to mechanical silencing in a novel rat IGF1–AKT pathway. to space constraints. Work in the authors’ laboratories was
intensive care unit model: underlying mechanisms. 130. Eddins, M. J. et al. Targeting the ubiquitin E3 ligase funded by: the UK Medical Research Council; the British
J. Physiol. 589, 2007–2026 (2011). MuRF1 to inhibit muscle atrophy. Cell Biochem. Heart Foundation; The Excellence Cluster Cardio-Pulmonary
108. Willis, M. S. et al. Muscle ring finger 1, but not muscle Biophys. 30 Mar 2011 (doi:10.1007/ System (ECCPS) of the Justus-Liebig-University, Giessen,
ring finger 2, regulates cardiac hypertrophy in vivo. s12013‑011‑9175–9177). Germany, the Goethe University, Frankfurt, Germany and the
Circ. Res. 100, 456–459 (2007). Small-molecule inhibitors of the atrogene MURF1 Max-Planck-Institute for Heart and Lung Research (DFG)
109. Spencer, J. A., Eliazer, S., Ilaria, R. L. Jr, Richardson, ubiquitin ligase are active in cells and suggest that Germany; and the University of Giessen and the University of
J. A. & Olson, E. N. Regulation of microtubule atrogene inhibition might be a viable route to Marburg Lung Center (UGMLC), funded by the government
dynamics and myogenic differentiation by MURF, a therapies for muscle loss. of the state of Hessen, Germany.
striated muscle RING-finger protein. J. Cell Biol. 150, 131. Perera, S., Holt, M. R., Mankoo, B. S. & Gautel, M.
771–784 (2000). Developmental regulation of MURF ubiquitin ligases Competing interests statement
Identification of the E3 ubiquitin ligase MURF3 and and autophagy proteins nbr1, p62/SQSTM1 and LC3 The authors declare no competing financial interests.
its dual roles in muscle development and protein during cardiac myofibril assembly and turnover. Dev.
degradation, and linkage of these roles to the Biol. 351, 46–61 (2011).
transcriptional control of SRF. The atrogenes MURF1, MURF2 and MURF3

FURTHER INFORMATION
110. Gautel, M. The sarcomere and the nucleus: functional show tight developmental regulation and are Thomas Braun’s homepage:
links to hypertrophy, atrophy and sarcopenia. Adv. expressed in developing muscle, suggesting that http://www.mpg.de/369653/herz_lungenforschung_wissM10
Med. Biol. Exp. 642, 176–191 (2008). their main function is not simply control of muscle Matthias Gautel’s homepage:
111. Komatsu, M. et al. Homeostatic levels of p62 mass loss. http://www.kcl.ac.uk/schools/biohealth/research/randall/
control cytoplasmic inclusion body formation in 132. Chen, S. N. et al. TRIM63, encoding MuRF1, is a novel res-sections/musclesig/gautel/mgautel
autophagy-deficient mice. Cell 131, 1149–1163 gene for familial hypertrophic cardiomyopathy. ALL LINKS ARE ACTIVE IN THE ONLINE PDF
(2007). Circulation 122 (Suppl.), A21194 (2010).

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 12 | JUNE 2011 | 361

© 2011 Macmillan Publishers Limited. All rights reserved

You might also like