Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.

With more than 600 individual muscles, skeletal muscle represents the largest tissue mass of the body and is essential for motion and support. Skeletal muscles are distinct from both cardiac and smooth muscles in that they can be voluntarily controlled by the organism. They are composed of bundles of striated myofibers that consist of elongated multinucleated syncytia. These fibers are surrounded by a basal lamina and are filled with a highly organized cytoskeleton composed of myofibrils. Skeletal muscles of the body arise from the somites, transient embryonic structures that originate from the paraxial mesoderm. By contrast, muscles of the head and neck derive from the anterior paraxial mesoderm, which does not form somites, and which will not be discussed in this Review.

Despite its regenerative capability, compromised muscle function is a hallmark of a number of conditions, ranging from developmental disorders to rhabdomyosarcoma and muscular dystrophies (Emery, 2002). In the adult, acute muscle injury such as volumetric loss, late-onset muscle dystrophies, neuromuscular degenerative diseases, cachexia as well as aging (sarcopenia) can result in significant muscular impairment leading to a severely compromised quality of life. There is an acute need for novel therapies to treat muscle diseases, and understanding the developmental basis of skeletal muscle formation and function holds value for the elucidation and possible treatment of muscle pathologies. Notably, it is well known that rhabdomyosarcomas exhibit undifferentiated myogenic features (Dagher and Helman, 1999), and that during adult muscle regeneration, developmental programs are partially reactivated in the injured tissue (Dumont et al., 2015). Therefore, a better understanding of myogenesis can also shed light on the mechanisms of muscle disease and regeneration.

Understanding how muscle is formed in vivo will also help pave the way for recreating muscle tissue in vitro from pluripotent stem cells (PSCs), either embryonic (ESCs) or induced (iPSCs). Although this field has lagged behind other lineages such as cardiac, neural and endodermal, the in vitro transposition of early signaling events as they occur during paraxial mesoderm specification in the embryo has led to recent success in this area (Chal et al., 2015; Crist, 2017; Hicks and Pyle, 2015). This was made possible thanks to the wealth of knowledge accumulated over decades of developmental studies in model organisms. The ability to generate skeletal muscle in vitro opens up new avenues for deciphering essential but poorly understood aspects of skeletal myogenesis such as myoblast fusion and satellite cell differentiation, and might also lead to important breakthroughs in disease modeling, drug screening and cell therapeutics. In this Review, we first provide a comprehensive overview of skeletal myogenesis in vivo, from its earliest developmental origin in the paraxial mesoderm to the formation of mature myofibers. We then focus on recent efforts to recapitulate muscle specification and skeletal myogenesis from PSCs in vitro, covering both directed differentiation and direct reprogramming approaches.

Skeletal muscles originate from the paraxial mesoderm, a tissue that forms in the primitive streak/blastopore during gastrulation and later in the tail bud during embryonic axis elongation (Fig. 1). The nascent paraxial mesoderm constitutes the presomitic mesoderm at the posterior tip of the embryo. The presomitic mesoderm is a transient tissue that can be further subdivided into an immature posterior and a committed anterior region, the latter of which segments to form the somites. It is within the somites that skeletal myogenesis is initiated with the specification of the premyogenic progenitors and skeletal myoblasts. Several phases of proliferation and differentiation lead to the formation of multinucleated myofibers from the fusion of mononucleated myocytes. In the following sections, we discuss the key cellular and molecular events that regulate the progression through these developmental steps in the embryo.

Fig. 1.

The formationand differentiation of the paraxial mesoderm. (A) Spatial organization of mesoderm fate in the posterior region of an amniote embryo. Mesoderm forms by ingression of epiblast cells at the level of the primitive streak (PS). Mesoderm subtypes (color-coded) are distinguished by their mediolateral position, whereby the axial mesoderm corresponds to the notochord. Progressively more lateral domains of the paraxial mesoderm (PM), intermediate mesoderm (IM) and lateral plate mesoderm (LPM) are shown and the corresponding marker genes are indicated. The nascent mesoderm is patterned by specific signaling pathways – in particular BMP, Wnt, FGF and retinoic acid (RA) signaling – the activities of which are distributed in gradients in the developing embryo (as shown to the right). During axis elongation (arrow), paraxial mesoderm progenitors are, at early stages, located in the anterior primitive streak posterior to the node, and they become incorporated into the tail bud later on. These progenitors include the neuromesodermal progenitors (NMPs). Dorsal view, anterior to the top. (B) Diagram recapitulating the differentiation of paraxial mesoderm toward skeletal muscle. From left to right, the developmental sequence (top) and the intermediate cell types with their marker genes (bottom) are shown. Cell types are color-coded according to the tissue types shown in A. aPSM, anterior presomitic mesoderm; pPSM, posterior presomitic mesoderm; Emb., embryonic.

Fig. 1.

The formationand differentiation of the paraxial mesoderm. (A) Spatial organization of mesoderm fate in the posterior region of an amniote embryo. Mesoderm forms by ingression of epiblast cells at the level of the primitive streak (PS). Mesoderm subtypes (color-coded) are distinguished by their mediolateral position, whereby the axial mesoderm corresponds to the notochord. Progressively more lateral domains of the paraxial mesoderm (PM), intermediate mesoderm (IM) and lateral plate mesoderm (LPM) are shown and the corresponding marker genes are indicated. The nascent mesoderm is patterned by specific signaling pathways – in particular BMP, Wnt, FGF and retinoic acid (RA) signaling – the activities of which are distributed in gradients in the developing embryo (as shown to the right). During axis elongation (arrow), paraxial mesoderm progenitors are, at early stages, located in the anterior primitive streak posterior to the node, and they become incorporated into the tail bud later on. These progenitors include the neuromesodermal progenitors (NMPs). Dorsal view, anterior to the top. (B) Diagram recapitulating the differentiation of paraxial mesoderm toward skeletal muscle. From left to right, the developmental sequence (top) and the intermediate cell types with their marker genes (bottom) are shown. Cell types are color-coded according to the tissue types shown in A. aPSM, anterior presomitic mesoderm; pPSM, posterior presomitic mesoderm; Emb., embryonic.

Specification of paraxial mesoderm progenitors

The paraxial mesoderm is composed of two bilateral strips of tissue flanking the neural tube and notochord. Posteriorly, these strips are unsegmented and form the presomitic mesoderm, while anteriorly they are composed of somites which define the embryonic segments (Fig. 1A). In amniotes, such as mouse or chicken, the paraxial mesoderm forms by ingression of the epiblast at the level of the anterior streak and later on from the tail bud. Formation of the primitive streak and activation of Wnt3 and the early mesoderm marker brachyury (T) depend on several signaling factors including Nodal and BMP4 (Beddington and Robertson, 1999; Liu et al., 1999; Ramkumar and Anderson, 2011; Tam and Loebel, 2007). Detailed fate mapping and grafting studies have identified several types of progenitors that give rise to the paraxial mesoderm. One type corresponds to a resident cell population able to give rise to both paraxial mesoderm and neural tube derivatives, the so-called neuromesodermal progenitors, which co-express the genes T and Sox2 (Garriock et al., 2015; Takemoto et al., 2011; Tzouanacou et al., 2009). A second type of progenitor gives rise only to paraxial mesoderm, while a third type can give rise to both paraxial mesoderm and lateral plate derivatives (Iimura et al., 2007; Stern and Canning, 1990; Wymeersch et al., 2016), and a fourth type can give rise to paraxial mesoderm and notochord (Selleck and Stern, 1991).

Paraxial mesoderm specification is concomitant to precursor exit from the progenitor zone and their entry to the posterior presomitic mesoderm, which are processes largely controlled by the Wnt and fibroblast growth factor (FGF) signaling pathways (Fig. 1A) (Ciruna and Rossant, 2001; Takada et al., 1994; Yamaguchi et al., 1994). Key Wnt/FGF targets including the transcription factors T, Tbx6 and Msgn1 are expressed in partially overlapping domains of the presomitic mesoderm (Chapman et al., 1996; Ciruna and Rossant, 2001; Yamaguchi et al., 1999; Yoon et al., 2000). These transcription factors are essential for paraxial mesoderm specification and patterning (Chapman et al., 1996; Nowotschin et al., 2012; van der Velden et al., 2006; Wilkinson et al., 1990; Yoon and Wold, 2000). In the Msgn1 mouse mutant, embryos develop an enlarged tail bud that fails to differentiate further into posterior presomitic mesoderm (Chalamalasetty et al., 2014; Yoon and Wold, 2000). In the absence of Wnt or FGF signaling, the mouse embryo is truncated and ectopic neural tissue forms in place of the posterior paraxial mesoderm (Boulet and Capecchi, 2012; Ciruna and Rossant, 2001; Takada et al., 1994). Thus, Wnt and FGF signaling are required for the differentiation of neuromesodermal progenitors toward a paraxial mesoderm fate at the expense of neural fate (Garriock et al., 2015; Jurberg et al., 2014). Downregulation of FGF and Wnt signaling in the tail bud signals the arrest of paraxial mesoderm production and the end of axis elongation (Cambray and Wilson, 2007; Denans et al., 2015; Olivera-Martinez and Storey, 2007; Tenin et al., 2010). Wnt3a is required for expression of Fgf8 in the tail bud (Aulehla et al., 2003), whereas Fgf4 and Fgf8 are necessary for Wnt signaling in the nascent presomitic mesoderm (Boulet and Capecchi, 2012; Naiche et al., 2011). Thus, the Wnt and FGF pathways form a closed regulatory loop that controls the specification and formation of the paraxial mesoderm.

BMP signaling also plays an important role in the specification and fate of mesoderm, both along the anterior-posterior axis at the early primitive streak stage and later along the mediolateral axis during late primitive streak and tail bud stages (Winnier et al., 1995). BMP4 is secreted by the posterior primitive streak and lateral tissues such as the lateral plate and extraembryonic mesoderm. BMP action is counteracted by opposite gradients of noggin and other BMP antagonists produced by the axial structures of the embryo (McMahon et al., 1998; Pourquié et al., 1996; Reshef et al., 1998; Tonegawa et al., 1997). This results in the establishment of a BMP signaling gradient that controls the mediolateral fates of mesoderm. Each mesodermal type, from the notochord to the extraembryonic mesoderm, requires progressively higher levels of BMP signaling for their specification (Kishigami and Mishina, 2005). Not surprisingly, formation of the paraxial mesoderm is exquisitely sensitive to changes in BMP signaling. For example, when a bead producing the BMP inhibitor noggin is grafted in the posterior primitive streak, which normally gives rise to lateral plate, these cells are converted to a paraxial mesoderm fate, leading to the formation of ectopic somitic columns (Tonegawa et al., 1997). In another study, a graft of quail posterior primitive streak, which would normally give rise to the lateral plate mesoderm, together with noggin-producing cells led to the formation of ectopic somites in a chicken embryo host (Streit and Stern, 1999). Also, mouse embryos chimeric for a BMP receptor null mutation show an expansion of the paraxial mesoderm domain, evidenced by the formation of ectopic rows of somites (Miura et al., 2006). Strikingly, this phenotype can be rescued by inhibiting FGF signaling, suggesting that BMP and FGF may antagonize each other. Together, this argues in favor of some plasticity of the paraxial mesoderm precursors of the primitive streak, which can still be induced to a lateral plate mesoderm fate if exposed to BMP signaling. In addition, these data indicate that, in vivo, BMP signaling needs to be suppressed for cells to acquire and maintain paraxial mesoderm fate.

Differentiation of the presomitic mesoderm

The specification of the future pairs of embryonic segments – the somites – is the result of highly dynamic molecular processes within the presomitic mesoderm. This involves a molecular oscillator known as the segmentation clock, which generates pulses of Notch, FGF and Wnt signaling to control the periodic production of somites (reviewed by Hubaud and Pourquié, 2014). The posterior domain of the paraxial mesoderm is composed of four consecutive transcriptional domains: the tail bud, the posterior presomitic mesoderm, the anterior presomitic mesoderm, and the forming somite (S0) (Chal et al., 2015). The tail bud domain contains the paraxial mesoderm progenitors and is exposed to the highest Wnt/FGF signaling activity (Aulehla et al., 2003; Chal et al., 2015; Naiche et al., 2011). More anteriorly, cells of the posterior presomitic mesoderm are characterized by the expression of genes such as Msgn1 (Fig. 1). In this domain, the Wnt/FGF pathways are still highly active and are essential for maintenance of paraxial mesoderm identity and for segmentation clock oscillations (Aulehla and Pourquie, 2008; Chal et al., 2015; Dunty et al., 2008; Jurberg et al., 2014). The Wnt/FGF activity gradient along the presomitic mesoderm is proposed to define a threshold, at which level cells become competent to respond to the clock by activating the expression of segmentation genes such as Mesp2 (Aulehla et al., 2003; Dubrulle et al., 2001). This specific threshold is called the determination front or wavefront and is roughly positioned at the posterior boundary of the anterior third of the presomitic mesoderm (Fig. 1A). Cells of the posterior presomitic mesoderm undergo abrupt signaling, metabolic and transcriptional changes as they enter the anterior presomitic mesoderm (Chal et al., 2015; Oginuma et al., 2017; Ozbudak et al., 2010) (Fig. 1A), including downregulation of Msgn1 and activation of Mesp2, Pax3, Foxc1/2 and Meox1/2 (Goulding et al., 1991; Kume et al., 2001; Mankoo et al., 2003; Saga et al., 1997) (Fig. 1B). In the anterior presomitic mesoderm, the posterior Wnt/FGF gradients are counteracted by retinoic acid (RA) produced by the somitic region (Fig. 1A). The tail bud containing the paraxial mesoderm precursors is protected from the differentiating action of RA by the expression of the RA-degrading enzyme Cyp26 (Abu-Abed et al., 2001; Iulianella et al., 1999; Sakai et al., 2001).

The bilateral stripes of Mesp2 define the anterior and posterior boundaries of the future segment (Oginuma et al., 2008; Takahashi et al., 2000). Anterior to the determination front, the newly formed segment acquires its posterior and anterior identities in response to a dynamic process largely controlled by Notch signaling (Chal and Pourquie, 2009). The cells of the posterior presomitic mesoderm are mesenchymal and highly motile (Bénazéraf et al., 2010; Chalamalasetty et al., 2014; Delfini et al., 2005), whereas the anterior presomitic mesoderm undergoes a mesenchymal-to-epithelial transition (MET), becoming progressively organized into dorsal and ventral epithelial layers surrounding a mesenchymal core (Duband et al., 1987; Martins et al., 2009). This process is controlled by Tcf15 (paraxis), a transcription factor activated in the anterior presomitic mesoderm by Wnt6 from the dorsal ectoderm (Burgess et al., 1996; Linker et al., 2005; Šošić et al., 1997). In the anteriormost presomitic mesoderm, a posterior fissure forms at the boundary between Mesp2-positive and -negative cells, resulting in the formation of an epithelial block of tissue – a new somite. This tissue remodeling involves Eph-ephrins (Barrios et al., 2003; Nakajima et al., 2006; Watanabe et al., 2009), cadherins (Chal et al., 2017b; Horikawa et al., 1999) and the small G proteins Cdc42 and Rac1 (Nakaya et al., 2004). Even so, in mouse or chicken embryos with severe segmentation and/or epithelialization defects, timely differentiation of the paraxial mesoderm as well as muscle and cartilage formation is observed, although improperly patterned (Bessho et al., 2003; Burgess et al., 1996; Dale et al., 2003; Saga et al., 1997). Thus, the differentiation and patterning of the paraxial mesoderm can be genetically uncoupled, suggesting that the underlying molecular mechanisms are relatively independent.

Compartmentalization of somites

Soon after their formation, somites become compartmentalized along the dorsoventral axis into a dorsal epithelial dermomyotome and a ventral mesenchymal sclerotome. The dermomyotome gives rise to skeletal muscle, brown fat and dermis of the back, whereas the ventral sclerotome produces the axial skeleton and tendons. At the time of their formation, each somite is composed of an anterior Tbx18+ and a posterior Uncx+ compartment with distinct derivatives (reviewed by Chal and Pourquie, 2009). In the chicken embryo, newly formed somites can also be further subdivided into a medial and a lateral compartment (Olivera-Martinez et al., 2000; Ordahl and Le Douarin, 1992; Selleck and Stern, 1991), which exhibit different fates and express different sets of genes (Martins et al., 2009; Pourquie et al., 1996). Cells of the lateral somite give rise to the hypaxial muscles of the limbs or the intercostals, whereas the medial somite forms the sclerotome, dermis of the back and paraxial muscles (Fig. 2) (Olivera-Martinez et al., 2000; Ordahl and Le Douarin, 1992).

Fig. 2.

Somite patterning and myotome formation. (A) Spatial relationship between the epithelial somite and the surrounding structures. The mesodermal subtypes are shown, as well as the future epaxial and hypaxial domains. Each epithelial somite is patterned into dorsoventral, mediolateral and anteroposterior compartments by signaling factors secreted by the surrounding tissues. Dorsally, Wnt signaling is required for dermomyotome specification, while BMP signaling produced by the lateral plate mesoderm (LPM) inhibits the differentiation of somitic lineages. Ventrally, Shh secreted from the midline plays a major role in sclerotome induction. (B) Spatial relationship between the differentiated somite and the surrounding structures. Dorsally, the somite differentiates into the dermomyotome (DM, red), which can be further subdivided into central dermomyotome (cDM), dorsomedial lip (DML) and ventrolateral lip (VLL). The dermomyotome also gives rise to the myotome (m, orange), which forms beneath from the four DM lips. Cells delaminate from the VLL to give rise to the myogenic progenitors of the limbs that migrate into the LPM. The ventral somite undergoes an epithelial-to-mesenchyme transition to form the sclerotome (scl, blue). BMP signaling (not shown) produced by the LPM transiently inhibits somitic lineage differentiation. Concomitantly, neural crest (nc, gray) delaminates from the dorsal neural tube and, while migrating ventrally, contacts dermomyotomal cells to promote myogenic induction through Notch activation and β-catenin stabilization (asterisk). The respective contributions of the various mesodermal subtypes to the adult tissues are listed beneath each tissue type; for example, the notochord contributes to the formation of the nucleus pulposus in the adult. ao, dorsal aorta; IM, intermediate mesoderm; smt, somatopleura; spl, splanchnopleura; n, notochord; nt, neural tube; D, dorsal; L, lateral; M, medial; V, ventral.

Fig. 2.

Somite patterning and myotome formation. (A) Spatial relationship between the epithelial somite and the surrounding structures. The mesodermal subtypes are shown, as well as the future epaxial and hypaxial domains. Each epithelial somite is patterned into dorsoventral, mediolateral and anteroposterior compartments by signaling factors secreted by the surrounding tissues. Dorsally, Wnt signaling is required for dermomyotome specification, while BMP signaling produced by the lateral plate mesoderm (LPM) inhibits the differentiation of somitic lineages. Ventrally, Shh secreted from the midline plays a major role in sclerotome induction. (B) Spatial relationship between the differentiated somite and the surrounding structures. Dorsally, the somite differentiates into the dermomyotome (DM, red), which can be further subdivided into central dermomyotome (cDM), dorsomedial lip (DML) and ventrolateral lip (VLL). The dermomyotome also gives rise to the myotome (m, orange), which forms beneath from the four DM lips. Cells delaminate from the VLL to give rise to the myogenic progenitors of the limbs that migrate into the LPM. The ventral somite undergoes an epithelial-to-mesenchyme transition to form the sclerotome (scl, blue). BMP signaling (not shown) produced by the LPM transiently inhibits somitic lineage differentiation. Concomitantly, neural crest (nc, gray) delaminates from the dorsal neural tube and, while migrating ventrally, contacts dermomyotomal cells to promote myogenic induction through Notch activation and β-catenin stabilization (asterisk). The respective contributions of the various mesodermal subtypes to the adult tissues are listed beneath each tissue type; for example, the notochord contributes to the formation of the nucleus pulposus in the adult. ao, dorsal aorta; IM, intermediate mesoderm; smt, somatopleura; spl, splanchnopleura; n, notochord; nt, neural tube; D, dorsal; L, lateral; M, medial; V, ventral.

Cells of the newly formed somites are not yet committed to a specific lineage (Aoyama and Asamoto, 1988). Grafting experiments and in vitro explant cultures have shown that somitic domains are progressively specified in response to factors secreted by the surrounding tissues, namely the dorsal ectoderm, neural tube, notochord and lateral plate (Fig. 2A) (reviewed by Christ and Scaal, 2008). Wnt, BMP and Shh represent the major signaling pathways for the induction of different somitic fates (reviewed by Marcelle et al., 2002; Yusuf and Brand-Saberi, 2006). Dorsally, local inhibition of BMP signaling is also essential for proper dermomyotome specification, while Wnt signals produced from the dorsal neural tube and ectoderm act to maintain the dermomyotome fate (reviewed by Hirsinger et al., 2000). Moreover, lineage-tracing studies in the mouse embryo have shown that the central engrailed-positive domain of the dermomyotome contains early progenitors positive for Pax7, Pax3 and/or Myf5, which will give rise to brown fat (Atit et al., 2006; Lepper and Fan, 2010; Sanchez-Gurmaches and Guertin, 2014; Seale et al., 2008). Ventrally, Shh signaling from the notochord and floor plate specifies the sclerotomal compartment, which downregulates Pax3 and upregulates Pax1 and Nkx3.2 expression (Fig. 2A) (Fan and Tessier-Lavigne, 1994; Johnson et al., 1994; Murtaugh et al., 1999). However, Shh can also stimulate the formation of myotomal cells (Borycki et al., 1999).

Myogenesis in the embryo and the adult

Initiation of myogenesis in the somite

In mouse and chicken embryos, the first sign of myogenesis is the activation of the myogenic factor Myf5 in cells of the dorsomedial part of the newly formed somite (Ott et al., 1991; Pownall and Emerson, 1992) (Fig. 1B). The dorsal epithelial dermomyotome, which expresses Pax3, becomes subdivided into a central domain, a dorsomedial lip, anterior and posterior lips, and a ventrolateral lip (Gros et al., 2004). Soon after dermomyotome formation, cells in the dorsomedial lip begin to express Myf5 and to downregulate Pax3 (Bober et al., 1994; Ott et al., 1991; Williams and Ordahl, 1994) (Fig. 2B). The primary myotome forms as a cell layer sandwiched between the dermomyotome dorsally and the sclerotome ventrally (Ordahl, 1993).

The first postmitotic skeletal muscle cells formed in the embryo are the myocytes of the myotome (Fig. 3A). These cells express specialized cytoskeletal proteins including slow (type I, Myh7) and embryonic (Myh3) myosin heavy chains (MyHC), α-actins [cardiac (Actc1) and skeletal (Acta1)] and desmin (Babai et al., 1990; Furst et al., 1989; Lyons et al., 1991a, 1990; Sassoon et al., 1988), as well as the Notch ligand jagged 2 (Hayashi et al., 1996; Hirsinger et al., 2001) and metabolic enzymes such as β-enolase and carbonic anhydrase III (CAIII) (Condon et al., 1990; Lyons et al., 1991b; Tweedie et al., 1991). The newly formed mononucleated myocytes elongate along the anterior-posterior axis to span the entire somite length, a process controlled by Wnt11 signaling (Christ et al., 1983; Denetclaw et al., 1997; Gros et al., 2009). More cells are progressively added to the myotome by the other dermomyotomal lips (Gros et al., 2004) and these cells fuse to existing myocytes leading to the formation of slow MyHC+ myofibers (Sieiro-Mosti et al., 2014) (Fig. 3A). After formation of the myotome, the central dermomyotome loses its epithelial character and its Pax3+ cells translocate to populate the myotome, providing the myogenic precursors involved in later phases of myogenesis (Fig. 3) (Gros et al., 2005; Kahane et al., 2001; Relaix et al., 2005). Myogenesis progresses as a rostral-to-caudal wave of maturation as the embryo elongates and as new pairs of somites are sequentially added (Cossu et al., 1995; Kato and Gurdon, 1993). Limb muscles derive from cells migrating from the lateral dermomyotome into the developing limb buds (Chevallier, 1979; Jacob et al., 1979). These cells progressively organize into muscle masses and form myofibers (reviewed by Buckingham et al., 2003). In the trunk and limbs, the myogenic program is controlled by a core network of transcription factors, including Pax3 and a set of muscle regulatory factors (MRFs) consisting of Myf5, MyoD (Myod1), MRF4 (Myf6) and myogenin (Berkes and Tapscott, 2005; Pownall et al., 2002; Rudnicki et al., 1993; Tapscott, 2005). In the embryo, myogenin controls the terminal differentiation of myoblasts into myocytes (Hasty et al., 1993; Nabeshima et al., 1993; Venuti et al., 1995). Genetic studies have also identified a set of transcription factors that are upstream regulators of skeletal myogenesis, which include Rp58 (Zfp238 or Zbtb18), Meox1/2, Six1/4, Eya1/2 and Nfix (Biressi et al., 2007; Grifone et al., 2007, 2005; Mankoo et al., 1999, 2003; Yokoyama et al., 2009).

Fig. 3.

Stages of skeletal myogenesis from the embryo to the adult. (A) Developmental sequence of muscle formation from the dermomyotome. The early myotome (left, yellow) is composed of primary myocytes, which are aligned along the anteroposterior axis and span each somitic compartment. During primary myogenesis (middle), Pax3+ progenitors (yellow cytoplasm, green nuclei) delaminate from the dorsal side of the dermomyotome and contribute to the formation of large primary myofibers (yellow). Some Pax3+ progenitors also migrate from the ventral lip to populate the body wall and limb buds (hypaxial domain). During secondary myogenesis (right), Pax7+ myogenic progenitors (red cytoplasm, brown nuclei) contribute to secondary (red) fiber formation, using the primary fibers as a scaffold and contributing to the growth of fetal muscles. During this phase, satellite cell precursors (purple cytoplasm, brown nuclei) localize under the basal lamina (dotted line) of the fibers where they can be found in adult muscles. Key processes associated with each stage are listed above. nt, neural tube; n, notochord; DM, dermomyotome; MTJ, myotendinous junction; NMJ, neuromuscular junction. (B) Differentiation of somitic progenitors toward skeletal muscles and adult satellite cells. Myogenic stem cells contribute to fetal myogenesis while maintaining a pool of progenitors, which eventually become located on mature myofibers in the satellite cell niche. For each step, markers for the intermediates and differentiated skeletal myofibers are shown. Additional markers are also shown in smaller font. Differentiation stages along the myogenic lineages are color-coded according to A. ‘Myocytes' encompasses also myotubes and myofibers. Emb., embryonic. Glut1, 2 and 4 are also known as Slc2a1, Slc2a2 and Slc2a4, respectively; for other symbols and synonyms see the main text.

Fig. 3.

Stages of skeletal myogenesis from the embryo to the adult. (A) Developmental sequence of muscle formation from the dermomyotome. The early myotome (left, yellow) is composed of primary myocytes, which are aligned along the anteroposterior axis and span each somitic compartment. During primary myogenesis (middle), Pax3+ progenitors (yellow cytoplasm, green nuclei) delaminate from the dorsal side of the dermomyotome and contribute to the formation of large primary myofibers (yellow). Some Pax3+ progenitors also migrate from the ventral lip to populate the body wall and limb buds (hypaxial domain). During secondary myogenesis (right), Pax7+ myogenic progenitors (red cytoplasm, brown nuclei) contribute to secondary (red) fiber formation, using the primary fibers as a scaffold and contributing to the growth of fetal muscles. During this phase, satellite cell precursors (purple cytoplasm, brown nuclei) localize under the basal lamina (dotted line) of the fibers where they can be found in adult muscles. Key processes associated with each stage are listed above. nt, neural tube; n, notochord; DM, dermomyotome; MTJ, myotendinous junction; NMJ, neuromuscular junction. (B) Differentiation of somitic progenitors toward skeletal muscles and adult satellite cells. Myogenic stem cells contribute to fetal myogenesis while maintaining a pool of progenitors, which eventually become located on mature myofibers in the satellite cell niche. For each step, markers for the intermediates and differentiated skeletal myofibers are shown. Additional markers are also shown in smaller font. Differentiation stages along the myogenic lineages are color-coded according to A. ‘Myocytes' encompasses also myotubes and myofibers. Emb., embryonic. Glut1, 2 and 4 are also known as Slc2a1, Slc2a2 and Slc2a4, respectively; for other symbols and synonyms see the main text.

Primary and secondary myogenesis

Myogenesis during development can be separated into two phases: an early embryonic or primary phase (E10.5-E12.5 in mouse, E3-7 in chicken) and a later fetal or secondary phase (E14.5-17.5 in mouse, E8+ in chicken) (Biressi et al., 2007; Stockdale, 1992). The first phase results in the production of the primary myofibers, which derive from Pax3+ (mouse) or Pax3+/Pax7+ (chicken) dermomyotomal progenitors (Horst et al., 2006; Hutcheson et al., 2009; Otto et al., 2006). These primary myofibers form the early myotomes and limb muscles, providing the templates upon which adult muscles will be built (Fig. 3) (Murphy and Kardon, 2011). They express a specific set of proteins, such as the slow MyHC and myosin light chain 1 (MyLC1, Myl1) (Kelly et al., 1997). During the second phase of myogenesis in mouse, a subset of the Pax3+ myogenic progenitors begins to express Pax7 and downregulates Pax3. These Pax7+ myogenic precursors fuse among themselves or to the primary fibers and give rise to secondary or fetal fibers that express specific markers such as β-enolase, Nfix or MyLC3 (Myl3) (Fougerousse et al., 2001; Keller et al., 1992; Kelly et al., 1997; Messina et al., 2010). At this time, the fibers also start to express fast MyHC isoforms (Van Horn and Crow, 1989). During secondary myogenesis, muscle growth is sustained essentially by cell fusion and the addition of myonuclei from proliferating Pax7+ progenitors (White et al., 2010). This is in contrast to postnatal muscle growth, which mostly results from individual fiber hypertrophy through the addition of novel myofibrils (Gokhin et al., 2008; Sparrow and Schöck, 2009). A subset of the Pax7+ progenitors will also form the pool of adult muscle stem cells – the satellite cells (Gros et al., 2005; Kassar-Duchossoy et al., 2005; Relaix et al., 2005).

Signaling controlling myogenesis

Embryonic tissues surrounding the somite provide key inductive signals for skeletal myogenesis. The notochord, neural tube and dorsal ectoderm, which produce Shh and Wnt signals, are crucial for this process (Münsterberg et al., 1995; Münsterberg and Lassar, 1995; Rios et al., 2011). The neural crest cells that migrate between the neural tube and somites also play a key role in the onset of myogenesis by triggering Myf5 activation in dermomyotome cells. Neural crest cells, which carry Wnt1 at their surface and also express delta-like 1 (Dll1), activate Notch signaling in the cells they contact in the dorsomedial lip. Notch activation in the lip cells in turn inhibits GSK3β activity and leads to the stabilization of Snail, resulting in the delamination of cells into the nascent myotome (Rios et al., 2011; Serralbo and Marcelle, 2014; Sieiro et al., 2016) (Fig. 2B). Another signaling molecule, hepatocyte growth factor (HGF), supports different aspects of myogenesis during development. In particular, HGF produced by the lateral plate mesoderm is essential for the proper migration of myoblasts (Andermarcher et al., 1996; Bladt et al., 1995; Brand-Saberi et al., 1996; Takayama et al., 1996). Insulin signaling has been shown to act with Wnt signaling to promote myogenesis and myoblast fusion (Charge and Rudnicki, 2004; van der Velden et al., 2006), while FGF signaling promotes myoblast proliferation while blocking differentiation (Itoh et al., 1996; Milasincic et al., 1996). In the context of adult regeneration, the FGF, insulin-like growth factor (IGF), HGF and Wnt signaling pathways have all been shown to play a role in satellite cell activation (Charge and Rudnicki, 2004; Flanagan-Steet et al., 2000; von Maltzahn et al., 2012). Wnt7a/Fzd7 planar cell polarity signaling is involved in the control of satellite cell self-renewal versus differentiation (Bentzinger et al., 2014, 2013; Le Grand et al., 2009).

Another important protein for myogenesis is myostatin (Gdf8), which represses muscle hypertrophy (Amthor et al., 2009; McPherron et al., 1997). In the absence of myostatin, mutants show a striking hypertrophic phenotype (Lee, 2004; reviewed by Rodriguez et al., 2014). Additionally, a number of microRNAs (miRNAs) are highly expressed, often specifically, in muscle cells (Chen et al., 2006; Kim et al., 2006; Sweetman et al., 2008). Although the identification of the signaling upstream and downstream of these miRNAs is still in its infancy (Motohashi et al., 2013), miRNAs regulate almost every aspect of myogenesis (reviewed by Ge and Chen, 2011). Finally, two additional pathways have been shown to play key roles in regulating myogenesis. In vitro work has revealed an important role for p38 MAPK intracellular signaling in the control of myogenesis, whereby inhibition of p38α/β (Mapk14/11) was shown to block myogenic differentiation and cell fusion (Cuenda and Cohen, 1999; Wu et al., 2000; Zetser et al., 1999). The nuclear factor of activated T-cells (NFAT)/calcineurin pathway is also active in skeletal muscle and is involved in the control of myogenic differentiation, myotube formation and fiber type specification (Abbott et al., 1998; Chin et al., 1998; Dunn et al., 1999; Musaro et al., 1999; Semsarian et al., 1999).

Formation of the skeletal myofibers

Myoblast fusion, myofibrillogenesis and basal lamina

The mature skeletal myofiber contains a highly organized cytoskeleton composed of aligned myofibrils. These fibers form by fusion of myoblasts to produce multinucleated myotubes, which further mature into myofibers (reviewed by Abmayr and Pavlath, 2012). The processes of myoblast-myoblast fusion and myoblast-myotube fusion are highly regulated during development but are still poorly understood in vertebrates. In flies, muscle fusion is orchestrated by two distinct types of myoblasts, with a limited number of founder cells initiating fusion with surrounding fusion-competent myoblasts (Rochlin et al., 2010). Fusion-competent myoblasts produce actin-based podosome-like structures that invade the muscle founder cell and form pores, allowing the transfer of the cytoplasm and nucleus from the myoblast to the founder cell (Kim et al., 2015b). In vertebrates, myoblast fusion starts with a recognition and an adhesion phase involving surface receptors. There is evidence to suggest that actin dynamics might play an important role (Laurin et al., 2008), and proteins involved in endocytosis and in membrane repair pathways have also been implicated (Demonbreun et al., 2015; Kim et al., 2015a). In vivo, elongation of the myofibers takes place by fusion of new myoblasts at their extremities and is dependent on TGFβ signaling (Gu et al., 2016; Williams and Goldspink, 1971). The cytokine IL4 has also been shown to control myoblast fusion, acting downstream of NFATC2 to control the fusion of myoblasts to myotubes (Horsley et al., 2003).

Myofibrillogenesis refers to the formation of the myofibrils that fill most of the fiber sarcoplasm, extending to both extremities of the fibers where they anchor to the myotendinous junction (reviewed by Sparrow and Schöck, 2009; Lemke and Schnorrer, 2017). Myofibrils are composed of a regular array of contractile modules – the sarcomeres. Each myofiber is also individually surrounded by a specialized basal lamina, called the endomysium (Bowman, 1840; Sanes, 2003), and harbors a specialized plasma membrane called the sarcolemma, which provides structural stability and allows for neuronal signal transduction. The sarcolemma anchors to the basal lamina through a complex of transmembrane proteins called the dystrophin-associated glycoprotein complex (DGC), which connects the myofiber cytoskeleton to the extracellular matrix (ECM) (Rahimov and Kunkel, 2013). The excitation-contraction coupling that is integral to myofiber function is achieved through the formation of a network of plasma membrane invaginations called T-tubules. In the sarcoplasm, the apposition of the cisternae, a specialized sarcoplasmic reticulum that acts as a calcium store, to the T-tubules, forms the triads (Flucher, 1992; Flucher et al., 1992). The triads, which are established during perinatal development (Flucher et al., 1993), are closely associated to myofibrils and allow for the transduction of the sarcolemmal depolarization upon neural excitation.

Myofiber types through developmental stages

Adult myofibers express specific isoforms of MyHC, which are associated with distinct electrophysiological properties and metabolism. Thus, oxidative slow twitch fibers express slow MyHC (type I, Myh7), whereas glycolytic fast twitch fibers express fast MyHC [types IIa (Myh2), IIb (Myh4) and IIx (Myh1)]. The expression of MyHC isoforms follows a developmental sequence, with the embryonic and slow MyHC being the first to be expressed (reviewed by Schiaffino and Reggiani, 2011). Fetal and neonatal fibers transiently express perinatal MyHC (Myh8), while the fast isoforms start to be expressed during late fetal myogenesis (Fig. 3B). Fiber type specificity is controlled in part by the transcription factors Six and Eya, which promote fast-type fiber diversity (Grifone et al., 2004; Richard et al., 2011). Fiber type also depends on neural input, which is established during fetal stages – as early as E14.5 in mouse limb (Hurren et al., 2015; Martin, 1990). Thus, the final fiber type of a given muscle is the product of its developmental history, its innervation and, during postnatal life, the physiological demands placed upon it (Schiaffino and Reggiani, 2011).

The origin of satellite cells

Satellite cells, which make up the main population of tissue-specific stem cells found in adult muscle, can be traced back to the Pax7+ myogenic progenitors of the dermomyotome central domain (Gros et al., 2005; Kassar-Duchossoy et al., 2005; Relaix et al., 2005). These Pax7+ embryonic progenitors become the source of the adult Pax7+ satellite cells (Gros et al., 2005; Lepper and Fan, 2010; Seale et al., 2000), although in some specific muscles, such as the diaphragm, prospective satellite cells also maintain Pax3 expression (Relaix et al., 2004, 2005). Notch signaling is essential to maintain the pool of Pax3/7+ progenitors, and in its absence no satellite cells can form (Vasyutina et al., 2007). During the peak of muscle mass growth, which is during the fetal and perinatal stages, the pool of satellite cell progenitors actively divides and represents up to 30% of the mononucleated cells in the mouse muscle tissue (Allbrook et al., 1971; Hellmuth and Allbrook, 1971). However, within 2 months after birth, these progenitors are reduced in number to a small pool of quiescent Pax7+ satellite cells, which account for only a few percent of the mononucleated cells in adult muscle (Allbrook et al., 1971; Cardasis and Cooper, 1975; Schmalbruch and Hellhammer, 1976). In mouse, satellite cells become enclosed under the basal lamina of myofibers during late fetal stages, at about E18.5 (Relaix et al., 2005). Although Pax7 is not essential for the specification of adult satellite cells per se, these cells are progressively lost in Pax7 mutant mice (Günther et al., 2013; Oustanina et al., 2004; von Maltzahn et al., 2013). A majority of the adult Pax7+ satellite cells also expresses Myf5, but these are distinct from proliferating Myf5+ myoblasts which also express MyoD (Beauchamp et al., 2000; Cornelison and Wold, 1997; Zammit et al., 2004) (Fig. 3).

Skeletal muscle has been generated in vitro from PSCs using two main approaches: directed differentiation and direct reprogramming, and sometimes a combination thereof. Direct reprogramming is achieved by the overexpression of selected transcription factors to reprogram cells to the myogenic lineage (Table S1). Conversely, directed differentiation approaches have employed several methodologies, each aimed at recapitulating the early differentiation stages that take place in the embryo to generate skeletal muscle (Table S2). Reviews of the field have so far been mostly comparative, focusing on the different methods used to generate skeletal muscles in vitro and their relative success (Abujarour and Valamehr, 2015; Baker and Lyons, 1996; Smith et al., 2016). In the following sections, we will discuss these studies in the context of developmental and cell biology and consider the extent to which they recapitulate in vivo myogenesis.

Directed differentiation of PSCs into skeletal muscle

The first report of skeletal myogenesis in vitro was provided by Rohwedel et al. (1994), who differentiated mouse ESCs (mESCs) into embryoid bodies (EBs) and observed the expression of MRFs and the formation of myogenin+ skeletal myocytes. EBs are three-dimensional (3D) cell aggregates that differentiate spontaneously into the three germ layers; however, they are usually very heterogeneous and lineage specification cannot be properly controlled (Doetschman et al., 1985; Robbins et al., 1990; Yamada et al., 1994). More recently, adherent monolayer cultures of PSCs have been used to generate more homogenous differentiation. The general principle of directed differentiation approaches is to present specific signaling molecules to the differentiating cells in vitro, as they would experience them in vivo, thus recapitulating normal development.

Primitive streak and mesoderm induction in vitro

The earliest stage of muscle differentiation in vivo occurs when paraxial mesoderm precursors in the epiblast activate the pan-mesodermal marker T in the primitive streak. Several studies aimed at recapitulating these early stages of mesoderm induction and specification using PSCs have led to the identification of the BMP, activin/Nodal, FGF, insulin (PI3K) and Wnt pathways as major signals able to induce the specification of various T+ mesodermal subpopulations (reviewed by Murry and Keller, 2008). Initial studies focused mostly on the differentiation of posterior primitive streak derivatives such as lateral plate and extraembryonic mesoderm, which produce hematopoietic and cardiogenic progenitors (Kaufman et al., 2001; Mummery et al., 2003). These studies identified BMP signaling as a key requirement for hematopoietic progenitor specification and they established the role of Wnt signaling for mesoderm induction in vitro (Chadwick et al., 2003; Gadue et al., 2006; Johansson and Wiles, 1995; Kattman et al., 2011; Lindsley et al., 2006; Nakanishi et al., 2009; Yang et al., 2008; Zhang et al., 2008). More recent studies have shown that treatment of PSCs with activin favors the differentiation of anterior primitive streak derivatives such as endoderm, whereas treatment with BMP triggers the differentiation of posterior streak derivatives such as lateral plate and extraembryonic mesoderm (Bernardo et al., 2011; Mendjan et al., 2014). Differentiation of late primitive streak that generates paraxial mesoderm could be achieved by treating PSCs first with activin, FGF and PI3K inhibitors to induce anterior primitive streak fate and then with GSK3 inhibitors (to activate Wnt signaling) and FGF to recapitulate the late primitive streak environment (Mendjan et al., 2014). Alternatively, by treating both mouse and human PSCs with BMP and Wnt activators sequentially, several groups were able to generate paraxial mesoderm progenitors (Chal et al., 2015; Mendjan et al., 2014; Sakurai et al., 2009, 2012). Recent studies also highlighted the importance of cell-cell and cell-ECM interactions in controlling early stages of mesoderm differentiation: human PSCs (hPSCs) exposed to identical media will adopt distinct cell fate depending on their precise in vitro organization and volumetric cell density (Kempf et al., 2016; Warmflash et al., 2014). Early stages of gastrulation have also been recapitulated in vitro in 3D mouse PSC cultures, termed gastruloids, where Tbx6 expression is observed (ten Berge et al., 2008; van den Brink et al., 2014).

Induction of a presomitic mesoderm fate in vitro

Production of mouse or human paraxial mesoderm progenitors characterized by expression of the presomitic mesoderm markers Msgn1 or Tbx6 can be achieved by treating PSC cultures with Wnt activators alone or in combination with other factors, such as FGF, without prior primitive streak induction (Borchin et al., 2013; Gouti et al., 2014; Mendjan et al., 2014; Shelton et al., 2014). Xu et al. (2013) also added forskolin to the Wnt/FGF combination, whereas others used only FGF [in a 3D system (Hosoyama et al., 2014)], only Wnt (Hwang et al., 2014), or Wnt followed by a Notch inhibition step (Choi et al., 2016). In most instances, these protocols require purification of the paraxial mesoderm progenitors by flow cytometry using cell surface markers or lineage-specific fluorescent reporters (for more details, see Box 1). In the mouse tail bud, Wnt activates expression of Fgf8 (Aulehla et al., 2003), and thus treating cultures with a Wnt pathway activator is also likely to result in FGF activation. Accordingly, in mouse FGF treatment is dispensable for paraxial mesoderm induction from mESCs (Chal et al., 2015). Wnt is also required for the induction of the neuromesodermal (T+/Sox2+) progenitors from mouse and human PSCs in vitro (reviewed by Henrique et al., 2015).

Box 1. Tools for characterizing PSC-derived myogenic progenitors

Surface markers. A number of surface markers have been used to purify myogenic precursors from PSCs induced to differentiate to the muscle lineage by various protocols. Among them, PDGFRα (CD140a) and VEGFR2 (Flk1, KDR, CD309) have been used to enrich for various subtypes of early mesoderm produced in vitro (Nishikawa et al., 1998). Notably, PDGFRα+/VEGFR2 cells isolated from differentiating PSC cultures have been proposed to correspond to putative paraxial mesoderm progenitors, including premyogenic cells (Darabi et al., 2012, 2008; Sakurai et al., 2009, 2012). Myogenic progenitors have also been isolated from mouse or human PSC cultures using CD106 (VCAM1, SM/C2.6) (Chang et al., 2009; Mizuno et al., 2010), CD34 (Demestre et al., 2015), CD56 (NCAM1) (Young et al., 2016), CD56 and CD73 (Barberi et al., 2007), CD82 (Uezumi et al., 2016), CXCR4 (CD184) and c-Met (Borchin et al., 2013). However, the usefulness of these markers is limited by their overall poor lineage specificity. Notably, PDGFRα and VEGFR2 are dynamically expressed on several mesodermal populations including paraxial mesoderm and lateral plate mesoderm and thus are not lineage-specific markers (Ding et al., 2013; Ema et al., 2006; Motoike et al., 2003; Takebe et al., 2006).

Fluorescent reporters. Lineage-specific promoters have also been used to drive the expression of a fluorescent protein (chiefly GFP) to generate reporter cell lines. One of the first examples of this was the brachyury-GFP mESC reporter line, which was used to analyze mesoderm induction in vitro (Fehling et al., 2003; Gadue et al., 2006). More recently, ESC lines that harbor fluorescent reporters for the expression of key paraxial mesoderm/myogenic lineage markers, namely Msgn1, Pax3, myogenin (Myog) and Pax7, have been used to track the sequential differentiation of PSCs toward paraxial mesoderm and skeletal muscle (Chal et al., 2015; Sudheer et al., 2016).

The generation of cells with a posterior presomitic mesoderm fate from PSCs in vitro was analyzed using mESC reporter lines for Msgn1 expression (Chal et al., 2015; Sudheer et al., 2016) (Box 1). Optimal induction of posterior presomitic mesoderm fate was observed when mESCs were pretreated with BMP4 to differentiate them into epiblast-like cells, followed by treatment combining a Wnt activator (R-spondin 3 or the GSK3β inhibitor CHIR99021) and a BMP inhibitor [LDN-193189 (Cuny et al., 2008)], which prevented the induced presomitic mesoderm cells from drifting to a lateral plate mesoderm fate (Chal et al., 2015). In the absence of BMP inhibition, Msgn1+ cells started to activate Bmp4 and to upregulate lateral plate markers in vitro (Chal et al., 2017a preprint). Paraxial mesoderm and lateral plate mesoderm share common precursors in the primitive streak, and grafts of the anterior streak (territory fated to give rise to the paraxial mesoderm) into the mid-streak level (territory fated to give rise to the lateral plate) is enough to change the fate of the cells according to their new position (Garcia-Martinez and Schoenwolf, 1992). These data indicate that the fate of the precursors is not determined at the primitive streak stage. Furthermore, exposure of paraxial mesoderm precursors to BMP4 in vivo leads them to switch to a lateral plate mesoderm fate, indicating that BMP4 acts as a lateral plate inducer (Tonegawa et al., 1997). BMP signaling also has a well-established role in promoting the formation of lateral plate mesoderm derivatives such as hematopoietic and cardiovascular cell types both in vivo and in vitro (Adelman et al., 2002; Tonegawa et al., 1997; reviewed by Murry and Keller, 2008; Orlova et al., 2015). Sakurai et al. (2012) also reported that high levels of BMP4 inhibit presomitic mesoderm and myogenic differentiation from PSCs.

Posterior presomitic mesoderm identity of mESCs induced with Wnt activation and BMP inhibition was confirmed using microarrays comparing their transcriptome with that of microdissected posterior presomitic mesoderm from E9.5 mouse embryos (Chal et al., 2015). Remarkably, these posterior presomitic mesoderm cells induced in vitro also expressed cyclic genes such as Hes7, suggesting that the segmentation machinery is activated in vitro (Chal et al., 2015). Efficient differentiation to a posterior presomitic mesoderm fate was also achieved by exposing hPSCs to a medium that included the small molecules CHIR99021 and LDN-193189, followed by FGF addition to the medium after 3 days of differentiation (Chal et al., 2016, 2015). In these conditions, cells acquired a posterior presomitic mesoderm fate with over 90% of cells positive for Tbx6 (Chal et al., 2016). Faster presomitic mesoderm induction was also obtained by pretreating cells with activators of the Wnt, FGF and TGFβ pathways combined with PI3K inhibition for 1 day prior to treatment with Wnt activator and BMP inhibitor (Loh et al., 2016).

In mouse embryos, Msgn1 expression is downregulated at the determination front where embryonic segments are first specified. This marks the level where the first stripes of segmentation gene expression, such as Mesp2 or Ripply1/2, are observed and where Pax3 is first activated. Differentiation of mouse ESCs to anterior presomitic mesoderm was studied using a Pax3 reporter line, demonstrating that expression of segmentation genes such as Ripply2 or those involved in somite rostrocaudal patterning such as Uncx or Tbx18 can be activated by maintaining Wnt activation and BMP inhibition in cultured cells (Chal et al., 2015). Similar activation of the segmentation program was also reported in hPSCs using Wnt and FGF inhibition (Loh et al., 2016).

Generation of skeletal myoblasts

The proliferating myoblasts that fuse to form adult myofibers in vivo are derived from Pax3+ precursors found in the dermomyotome. Induction of such Pax3+ precursors has been achieved by maintaining mESC cultures in a medium containing a Wnt activator and a BMP inhibitor (Chal et al., 2015). The RA biosynthetic enzyme Aldh1a2 (Raldh2) is expressed in mouse Pax3+ cells induced in vitro (Chal et al., 2015), suggesting that the cultures are able to produce RA in sufficient amounts to promote presomitic mesoderm-like cell maturation without the need for external supply (Kennedy et al., 2009; Ryan et al., 2012). Subsequent treatment of the cultures with HGF, IGF and FGF led to efficient induction of myogenin+ myocytes both from mouse and human PSCs after 1 to 2 weeks of differentiation (Chal et al., 2015, 2016) (Fig. 4). In mESC cultures, these myocytes appear as elongated mononucleated cells expressing slow MyHC and primary myotome cells. The rapid increase in myogenin+ myocytes was accompanied by a concomitant decrease of Pax3+ myogenic progenitors. These were progressively replaced by a pool of Pax7+ myogenic progenitors, as observed in vivo (Chal et al., 2015; Kassar-Duchossoy et al., 2005; Relaix et al., 2005). Several studies have aimed at deciphering the respective contribution of individual transcription factors including Pax3/7 and the MRFs during myogenic differentiation of mESCs using engineered mutations or siRNA-based knockdown (Table S2). Other studies with hPSCs have reported different induction kinetics of the Pax3/7+ progenitors. Awaya et al. (2012) described the late differentiation (50 days) of Pax3+ and Pax7+ cells that can be isolated with CD56 (NCAM1) and CD73 (NT5E) surface markers. Myosphere culture of differentiating hPSCs has also been described, which produces Pax7 expression after 6 weeks and myotube formation after 8 weeks of culture (Hosoyama et al., 2014). Recently, Caron et al. (2016) described a serum-based protocol to differentiate human myotubes within 26 days; although some cells in these cultures express Pax3, no Pax7+ cells were generated.

Fig. 4.

Skeletal myogenesis from pluripotent stem cells. Comparison of strategies to generate skeletal muscles from PSCs. The sequence of the differentiation stages of mESCs and hPSCs into skeletal muscle and their corresponding markers is shown from left to right (top). A comparison of the approaches to generating skeletal muscle from PSCs is shown in the middle. Directed differentiation approaches aim to recapitulate the developmental stages of paraxial mesoderm specification and differentiation by manipulating signaling pathways such as Wnt and BMP. The sequence of developmental stages and important signaling pathways associated with each stage are shown. Direct reprogramming approaches aim to bypass early developmental stages by overexpressing a myogenic regulator, chiefly Pax3/7 or MyoD (iPAX3/7, iMYOD). In some instances, the cellular events occurring during differentiation remain largely unclear (dashed line). Directed differentiation into skeletal muscle occurs according to slightly different timelines [days (d) of in vitro differentiation are indicated] using mESCs as compared with hPSCs (bottom).

Fig. 4.

Skeletal myogenesis from pluripotent stem cells. Comparison of strategies to generate skeletal muscles from PSCs. The sequence of the differentiation stages of mESCs and hPSCs into skeletal muscle and their corresponding markers is shown from left to right (top). A comparison of the approaches to generating skeletal muscle from PSCs is shown in the middle. Directed differentiation approaches aim to recapitulate the developmental stages of paraxial mesoderm specification and differentiation by manipulating signaling pathways such as Wnt and BMP. The sequence of developmental stages and important signaling pathways associated with each stage are shown. Direct reprogramming approaches aim to bypass early developmental stages by overexpressing a myogenic regulator, chiefly Pax3/7 or MyoD (iPAX3/7, iMYOD). In some instances, the cellular events occurring during differentiation remain largely unclear (dashed line). Directed differentiation into skeletal muscle occurs according to slightly different timelines [days (d) of in vitro differentiation are indicated] using mESCs as compared with hPSCs (bottom).

Direct reprogramming strategies using transcription factors

The discovery that treatment of fibroblast cultures with 5-azacytidine, a demethylating agent, could induce cells to differentiate into myoblasts (Constantinides et al., 1977; Taylor and Jones, 1979) led to the demonstration that MyoD, whose locus is a target of 5-azacytidine, could reprogram fibroblasts to a muscle fate (Davis et al., 1987; Weintraub et al., 1989). This in turn led to direct reprogramming strategies whereby overexpression of a myogenic factor – often MyoD – in PSCs forces their differentiation toward the skeletal muscle lineage, bypassing de facto early developmental stages (Dekel et al., 1992; Shani et al., 1992; reviewed by Comai and Tajbakhsh, 2014) (Fig. 4, Table S1). MyoD overexpression is often triggered after EB formation, and MyoD expression directly in PSCs is inefficient unless BAF60c (SMARCD3) is co-expressed (Albini et al., 2013). MyoD-reprogrammed hPSCs can generate myotubes within 10 days (Rao et al., 2012) and are also able to engraft when transplanted in mouse muscle (Goudenege et al., 2012; Ozasa et al., 2007; Zheng et al., 2006) (Table S1). Moreover, MyoD-reprogrammed myotubes have been used to model muscle diseases such as Duchenne or Miyoshi muscular dystrophies (Shoji et al., 2015; Yasuno et al., 2014; Young et al., 2016) (Table S1).

In the chicken embryo, Pax3 overexpression can lead to ectopic activation of the myogenic program in lateral plate or neural tube cells (Maroto et al., 1997). Using both mouse and human PSCs, Perlingeiro and colleagues developed direct reprogramming methods using Pax3/7 overexpression combined with EB formation to induce myogenic differentiation (Darabi et al., 2012, 2008, 2011; Filareto et al., 2013) (Fig. 4). As Pax3/7 are also expressed in neural derivatives in vivo (Fougerousse et al., 2002; Gerard et al., 1995; Goulding et al., 1991; Jostes et al., 1990), a purification step using surface markers such as PDGFRα (CD140a) or CD56 was needed to enrich in myogenic cells (Darabi et al., 2008, 2011; Quattrocelli et al., 2011) (Box 1). Pax3 overexpression acts on the early PDGFRα+ population, upregulating transcription factors Tcf15 and Meox1 and preventing the cells from undergoing cardiomyogenesis (Magli et al., 2013, 2014). Both Pax3- and Pax7-reprogrammed mouse progenitors were shown to engraft when transplanted in a mouse model. Pax3- or Pax7-reprogrammed hPSCs can generate myogenic progenitors that are CD29 (ITGβ1)+/CD44+/CD56+ and CXCR4/CD106 (VCAM1) and which can engraft into mouse muscle (Darabi et al., 2012; Filareto et al., 2015) (Box 1). Overexpression of Mesp1 has also been used to produce putative cardio-pharyngeal progenitors with the capacity to differentiate into both cardiac and cranial paraxial mesoderms (Chan et al., 2013; Lescroart et al., 2014). Other factors used for direct reprogramming of PSCs into myogenic cells include other MRFs and a mutated version of HMGA2, as well as the expression of growth factors such as IGF2 or a modified HGF (Table S1). Despite some success, direct reprogramming methods are nevertheless often undefined, as they rely on EB formation in serum-containing media and often require further cell sorting (Table S1, Box 1).

Myofiber maturation in vitro

Sarcomeres and structural assembly

Maturation of PSC-derived cell types has proven a major challenge for most cell lineages, with differentiated cells often retaining a fetal-like phenotype (Robertson et al., 2013). Mature striated myotubes have been generated in vitro from mESCs spontaneously differentiated from EBs (Hirsch et al., 1998; Rohwedel et al., 1998b), as well as from Pax7-reprogrammed hPSCs (Skoglund et al., 2014). Demestre et al. (2015) compared these two methods in their ability to produce human actinin+ myotubes and reported that myotubes generated by Pax7-mediated reprogramming were slower to mature. Millimeter-long myofibers exhibiting MyHC+ and titin+ striations and containing large numbers of nuclei have also been generated from mouse and human PSCs in adherent, serum-free conditions (Chal et al., 2015) (Fig. 5). The maturation of the myofibers was evidenced by the transition from slow to perinatal MyHC expression, the formation of highly organized myofibrils, the expression of Nfix, which is a secondary myogenesis marker (Messina et al., 2010), and the assembly of individual basal lamina (Fig. 5). Based on their morphological characteristics, the authors suggested that these myofibers resemble perinatal fibers (Chal et al., 2015; White et al., 2010). Remarkably, the formation of such long striated myofibers is rarely observed when primary myoblasts, myogenic lines (such as C2C12) or satellite cells are differentiated in vitro, as they tend to form poorly organized multinucleated myosacs.

Fig. 5.

Cellular features of PSC-derived skeletal myofibers. Schematic of a myofiber (center) and the corresponding representative features detectable by immunohistochemistry on skeletal myofibers differentiated from mESCs. In the central scheme, the elongated myofiber contains aligned striated myofibril bundles composed of various MyHCs (yellows), dystrophin in a subsarcolemmal position (thin irregular black line), aligned mitochondria (green), myonuclei (gray) and nascent T-tubule network (light blue lines). The fiber extremities form a specialized contact surface reminiscent of a myotendinous junction (MTJ), while at the equatorial level the sarcolemma (solid black line) can harbor a postsynaptic element (brown) indicating post-neuromuscular junction (NMJ) assembly. Satellite cells (purple) can be located under the basal lamina (black dotted line). Images show the various markers (green) and the location of the features illustrated in the central scheme. In some cases, nuclei are counterstained with DAPI (blue). In the NMJ/post-synapse image, fibers are counterstained for MyHC (red). In the satellite-like cells panel, fibers are visible in a merged phase-contrast image. Scale bars: 100 μm.

Fig. 5.

Cellular features of PSC-derived skeletal myofibers. Schematic of a myofiber (center) and the corresponding representative features detectable by immunohistochemistry on skeletal myofibers differentiated from mESCs. In the central scheme, the elongated myofiber contains aligned striated myofibril bundles composed of various MyHCs (yellows), dystrophin in a subsarcolemmal position (thin irregular black line), aligned mitochondria (green), myonuclei (gray) and nascent T-tubule network (light blue lines). The fiber extremities form a specialized contact surface reminiscent of a myotendinous junction (MTJ), while at the equatorial level the sarcolemma (solid black line) can harbor a postsynaptic element (brown) indicating post-neuromuscular junction (NMJ) assembly. Satellite cells (purple) can be located under the basal lamina (black dotted line). Images show the various markers (green) and the location of the features illustrated in the central scheme. In some cases, nuclei are counterstained with DAPI (blue). In the NMJ/post-synapse image, fibers are counterstained for MyHC (red). In the satellite-like cells panel, fibers are visible in a merged phase-contrast image. Scale bars: 100 μm.

Electron microscopy analysis of mouse and human PSC-derived myofibers revealed that they exhibit highly organized sarcomeric units, reminiscent of the cytoarchitecture found in vivo (Hirsch et al., 1998; Skoglund et al., 2014). Myofibrils generated in these fibers can exhibit the appropriate organization of MyHC, troponin T, nebulin and titin, and undergo the stage-specific developmental transitions through α-actins (cardiac to skeletal) and MyHC (embryonic, fetal, and perinatal/fast) isoforms (Chal et al., 2015; Hirsch et al., 1998; Mizuno et al., 2009; Rohwedel et al., 1998b). Furthermore, desmin null mESCs have been used to demonstrate the important role of desmin in the early formation of myotubes (Höllrigl et al., 2002; Weitzer et al., 1995). While these studies are encouraging, ultrastructural analyses have shown that hPSC-derived myofibers have myofibrils that are less regularly organized than those found in vivo, featuring irregular and misaligned Z-disks. They also fail to form a mature T-tubule network, a hallmark of adult myofibers (Skoglund et al., 2014). Although triad-like structures could be identified, they were limited to the subsarcolemmal space, lacking the orthogonal alignment to myofibrils observed in vivo. This incomplete maturation could be explained by the immature character of the fibers and the lack of innervation in these cultures.

The formation of a basal lamina (endomysium) has been shown to occur in vitro, as evidenced by the deposition of a continuous sheath of laminin along individual fibers (Chal et al., 2015). The dystrophin-associated glycoprotein complex (DGC), which links the myofiber cytoskeleton to the basal lamina through the sarcolemma, is also assembled in vitro. DGC components, including dystrophin (Chal et al., 2015; Ozasa et al., 2007), dystroglycan and integrins (Hirsch et al., 1998; Jacobson et al., 2001; Tremblay and Carbonetto, 2006), were found distributed at the sarcolemma of PSC-derived myofibers (Fig. 5, Tables S1 and S2). During development, integrins α4 and α7 play a key role in skeletal myogenesis and associate with integrin β1 to form receptors for ECM proteins such as laminin (Menko and Boettiger, 1987; Rosen et al., 1992). Intriguingly, differentiation of Itgb1 null mESCs showed that mutant myoblasts can still fuse and form myotubes with apparently normal sarcomeres and costameres (Hirsch et al., 1998; Rohwedel et al., 1998a). Moreover, myofibers differentiated from Dag1 null mESCs exhibit defects in the assembly of postsynaptic elements including acetylcholine receptor (AchR) clusters (Jacobson et al., 2001). Dmdmdx mPSC lines were shown to exhibit defects in myogenic differentiation and increased apoptosis (Chen et al., 2012), while Chal et al. (2015) reported a branching defect characterized by an increased number of split fibers, a phenotype also seen in Duchenne muscular dystrophy (DMD) patients and the mdx mouse (Chan and Head, 2011). Finally, myoblasts derived from DMD human iPSCs were shown to exhibit fusion defects that could be corrected by dual inhibition of TGFβ and BMP signalings (Choi et al., 2016).

Excitation-contraction coupling

During development, motoneuron input is important for proper myofiber maturation and viability. However, in vitro, cultures of PSC-derived muscle that do not receive motoneuron input nevertheless achieve a relatively high level of functional maturation, as evidenced by myofibrils and spontaneous contractions (Caron et al., 2016; Chal et al., 2015; Choi et al., 2016; Shelton et al., 2014). PSC-derived myofibers have been further characterized by electrophysiological studies focusing on calcium handling, ions currents and AchR cluster properties. In vivo, early embryonic myofibers exhibit transient, low voltage-activated calcium currents (T-type, ICaT), which are progressively replaced during fetal stages by slow, high voltage-activated currents (L-type, ICaL) (Beam et al., 1986; Cognard et al., 1986). Myofibers derived from Pax7-reprogrammed hPSCs exhibit excitable properties (Skoglund et al., 2014) but have immature ion handling characterized by slow calcium buffering and a small potassium conductance, which could explain the spontaneous fiber twitches observed in these cultures (Skoglund et al., 2014).

In vivo, as myotubes mature, they upregulate AchR expression and switch from the fetal γ-subunit to the adult ε-subunit (Buonanno and Merlie, 1986; Mishina et al., 1986). Patch clamp and gene expression analyses of mESC-derived myotubes have shown that this developmental transition can also occur in vitro, as these cells start to express the adult AchRε after 2 weeks of differentiation (Guan et al., 1999; Jacobson et al., 2001; Rohwedel et al., 1998b, 1994; Tremblay and Carbonetto, 2006) (Fig. 5B). However, it has been shown that postsynaptic elements can form in vitro on myotubes in the absence of neural input, and that these cells remain immature (reviewed by Sanes and Lichtman, 1999). For this reason, there have been several efforts to establish a co-culture system of PSC-derived myogenic cells with rat embryonic spine or with PSC-derived motoneurons. This system improved the maturation of PSC-derived myofibers, as evidenced by AchR clustering and electrophysiological recordings (Demestre et al., 2015; Puttonen et al., 2015; Rohwedel et al., 1998b); however, electrophysiological analysis suggested immaturity of neuromuscular junction activity (Puttonen et al., 2015; Vyskocˇil and Vrbová, 1993).

Generation of satellite-like cells in vitro

In adult muscle, the pool of quiescent Pax7+ satellite cells is closely associated with the myofibers (reviewed by Brack and Rando, 2012; Dumont et al., 2015), an arrangement that aides muscle repair during regeneration (Lepper et al., 2011; Murphy et al., 2011; Sambasivan et al., 2011). However, satellite cells are found in very limited numbers and they cannot be amplified in vitro as they differentiate and lose their regenerative properties in culture (Gilbert et al., 2010; Montarras et al., 2005). The generation of Pax7+ cells from PSCs has been reported in several of the protocols discussed previously but in most cases their characterization has remained preliminary (Tables S1 and S2). Notably, the association of Pax7+ cells with mature myofibers and the location of the cells under the basal lamina, as it is seen in vivo, has only been reported by Chal et al. (2015) (Fig. 5). One of the key characteristics of the Pax7+ satellite cells is their capacity to regenerate myofibers while being able to self-renew and reconstitute the satellite stem cell compartment when grafted in adult injured muscles. Several of the myogenic differentiation protocols discussed above have reported the production of myofibers when the progenitors obtained in vitro were transplanted into injured adult muscle (Tables S1 and S2). In some cases these produced donor-derived Pax7+ cells resembling satellite cells; however, in all cases the transplanted cells were either the result of ʻbulk' differentiation or were isolated using surface markers resulting in a mixed population of cells containing a fraction of putative Pax7+ cells. The in vivo potential of pure populations of PSC-derived Pax7+ cells produced in vitro was evaluated using a Pax7-fluorescent reporter mESC line (Fig. 5). In differentiated cultures of this line, Pax7+ cells first experienced a proliferative phase between 2 and 3 weeks of differentiation, followed by a reduction in the number of Pax7+ progenitors, with the remaining cells found closely associated to mature fibers, as expected for satellite cells (Chal et al., 2015). Similar dynamics could be observed in differentiated hPSCs, where Pax7+ cells could also be found closely associated to myofibers. Furthermore, a significant proportion of the Pax7+ cells differentiated in vitro were found to be Ki67, suggesting that they are quiescent (Chal et al., 2015). When activated, satellite cells are able to divide asymmetrically, thus allowing for self-renewal and for the generation of a myoblast that will proliferate and contribute to muscle regeneration. Pax7+ cells generated in vitro from mESCs can give rise to both myofibers and Pax7+ satellite cells when grafted in vivo in an mdx mouse model, suggesting that these cells can behave like bona fide satellite cells (Chal et al., 2015). The myofibers differentiated in vitro from mESCs exhibit characteristics of perinatal fibers, suggesting that the associated Pax7+ cells are likely to correspond to perinatal satellite cells (Tierney et al., 2016).

The potential for PSCs to serve as developmental models, including for skeletal muscle development, has been recognized for more than 20 years (Baker and Lyons, 1996; Keller, 1995; Rohwedel et al., 2001; Wobus et al., 2001). However, these systems have been criticized for their artificial nature and their simplicity compared with the full complexity of organisms (Moretti et al., 2013). Moreover, it is still unclear if in vitro differentiation encounters the same developmental constraints as in vivo. A similar concern has been raised about the physiological relevance and the proper maturation of the cell types generated in vitro, which in most cases reach only a fetal state (Robertson et al., 2013; Satin et al., 2008). For skeletal muscle, this is evidenced by the neonatal properties of the in vitro myofibers, including smaller size, immature metabolism and a weak physiological response (Bursac et al., 2015). Parameters that control the maturation of PSC-derived myofibers remain largely unexplored and the use of integrated co-culture systems or biomaterials has been seldom investigated (Chal et al., 2016; Leung et al., 2013). Nevertheless, the recent availability of more efficient myogenic differentiation protocols for PSCs will allow the study of skeletal myogenesis with unprecedented accessibility and resolution. A number of questions remain to be answered about the nature of the muscle cell generated in vitro, in particular about their axial/anatomical identities and fiber composition, and how these relate to what is seen in vivo (Donoghue et al., 1992; Jarad and Miner, 2009; Kieny et al., 1972; Nowicki and Burke, 2000; Rosenthal et al., 1989).

A number of translational applications for PSC-derived skeletal muscle cells can be envisioned. Although satellite cells represent the ideal target population for muscle regenerative medicine (Collins et al., 2005), their accessibility, number and manipulability remain very limited. Furthermore, cell therapy approaches still face significant challenges, including transplantation routes, scale up, host immune response and functional integration (Tremblay and Skuk, 2008). If these limitations can be overcome, then PSC-derived myogenic cultures that recreate the satellite cell niche in vitro may be a valuable source of cells for transplantation (Cosgrove et al., 2009). In the near future, hPSC-based myogenic cultures are likely to become an important preclinical model with which to study disease mechanisms in place of primary cultures generated from patient biopsies, which are subject to variability and limited accessibility. Combined with the relative ease in deriving novel hPSCs and progress in genome engineering, disease-specific models can now be generated for a large spectrum of muscle pathologies (Avior et al., 2016; Merkle and Eggan, 2013; Rüegg and Glass, 2011), facilitating the development of in vitro therapeutic approaches (Kazuki et al., 2010; Rohwedel et al., 2001; Tran et al., 2013; van Deutekom et al., 2007).

Decades of developmental studies have provided a compelling picture of skeletal muscle formation in vertebrate embryos; however, important myogenic processes remain poorly studied due to limited in vivo accessibility. PSC-based systems herald a new area of ʻdevelopment-in-a-dish', with the disciplines of developmental and stem cell biology now merging closer than ever. Directed differentiation methods can recapitulate many aspects of bona fide skeletal myogenesis, allowing us to probe key developmental mechanisms in an unprecedented manner. One of the most exciting opportunities that PSC-based systems provide is to understand the regulation of skeletal myogenesis as it occurs during human development. In turn, this will help to provide more relevant tissue models for diseases and therapeutic investigations. Several key challenges remain on this front, the first of which is the generation of physiologically relevant myofibers. This is likely to be solved by rational design in combining the factors that control skeletal muscle maturation with advances in bioengineering. The second key challenge is the recreation of the satellite cell niche. While there is evidence that coexistence of myofibers and Pax7+ progenitors allows for the occurrence of Pax7+ satellite-like cells in vitro (Chal et al., 2015), a precise understanding of the niche environment and the molecular mechanisms at play remains to be achieved. A sustained effort to better characterize the regulation of skeletal myogenesis during development and in the adult is therefore crucial for success in the clinic. Although PSC-derived skeletal myogenesis is still a nascent field, this is a time of exciting opportunities for the developmental biology, drug discovery and regenerative medicine communities.

We are grateful to members of the O.P. laboratory for helpful discussions.

Funding

This work was supported by advanced grant ERC-2009-AdG 249931 from the European Research Council to O.P., by the EU Seventh Framework Programme grant Plurimes (agreement no. 602423), by a strategic grant from the French Muscular Dystrophy Association (AFM-Téléthon) to O.P., and by the INGESTEM project (Agence Nationale de la Recherche).

Abbott
,
K. L.
,
Friday
,
B. B.
,
Thaloor
,
D.
,
Murphy
,
T. J.
and
Pavlath
,
G. K.
(
1998
).
Activation and cellular localization of the cyclosporine A-sensitive transcription factor NF-AT in skeletal muscle cells
.
Mol. Biol. Cell
9
,
2905
-
2916
.
Abmayr
,
S. M.
and
Pavlath
,
G. K.
(
2012
).
Myoblast fusion: lessons from flies and mice
.
Development
139
,
641
-
656
.
Abu-Abed
,
S.
,
Dolle
,
P.
,
Metzger
,
D.
,
Beckett
,
B.
,
Chambon
,
P.
and
Petkovich
,
M.
(
2001
).
The retinoic acid-metabolizing enzyme, CYP26A1, is essential for normal hindbrain patterning, vertebral identity, and development of posterior structures
.
Genes Dev.
15
,
226
-
240
.
Abujarour
,
R.
and
Valamehr
,
B.
(
2015
).
Generation of skeletal muscle cells from pluripotent stem cells: advances and challenges
.
Front. Cell Dev. Biol.
3
,
29
.
Adelman
,
C. A.
,
Chattopadhyay
,
S.
and
Bieker
,
J. J.
(
2002
).
The BMP/BMPR/Smad pathway directs expression of the erythroid-specific EKLF and GATA1 transcription factors during embryoid body differentiation in serum-free media
.
Development
129
,
539
-
549
.
Albini
,
S.
,
Coutinho
,
P.
,
Malecova
,
B.
,
Giordani
,
L.
,
Savchenko
,
A.
,
Forcales
,
S. V.
and
Puri
,
P. L.
(
2013
).
Epigenetic reprogramming of human embryonic stem cells into skeletal muscle cells and generation of contractile myospheres
.
Cell Rep.
3
,
661
-
670
.
Allbrook
,
D. B.
,
Han
,
M. F.
and
Hellmuth
,
A. E.
(
1971
).
Population of muscle satellite cells in relation to age and mitotic activity
.
Pathology
3
,
233
-
243
.
Amthor
,
H.
,
Otto
,
A.
,
Vulin
,
A.
,
Rochat
,
A.
,
Dumonceaux
,
J.
,
Garcia
,
L.
,
Mouisel
,
E.
,
Hourde
,
C.
,
Macharia
,
R.
,
Friedrichs
,
M.
, et al. 
(
2009
).
Muscle hypertrophy driven by myostatin blockade does not require stem/precursor-cell activity
.
Proc. Natl. Acad. Sci. USA
106
,
7479
-
7484
.
Andermarcher
,
E.
,
Surani
,
M. A.
and
Gherardi
,
E.
(
1996
).
Co-expression of the HGF/SF and c-met genes during early mouse embryogenesis precedes reciprocal expression in adjacent tissues during organogenesis
.
Dev. Genet.
18
,
254
-
266
.
Aoyama
,
H.
and
Asamoto
,
K.
(
1988
).
Determination of somite cells: independence of cell differentiation and morphogenesis
.
Development
104
,
15
-
28
.
Atit
,
R.
,
Sgaier
,
S. K.
,
Mohamed
,
O. A.
,
Taketo
,
M. M.
,
Dufort
,
D.
,
Joyner
,
A. L.
,
Niswander
,
L.
and
Conlon
,
R. A.
(
2006
).
Beta-catenin activation is necessary and sufficient to specify the dorsal dermal fate in the mouse
.
Dev. Biol.
296
,
164
-
176
.
Aulehla
,
A.
and
Pourquie
,
O.
(
2008
).
Oscillating signaling pathways during embryonic development
.
Curr. Opin. Cell Biol.
20
,
632
-
637
.
Aulehla
,
A.
,
Wehrle
,
C.
,
Brand-Saberi
,
B.
,
Kemler
,
R.
,
Gossler
,
A.
,
Kanzler
,
B.
and
Herrmann
,
B. G.
(
2003
).
Wnt3a plays a major role in the segmentation clock controlling somitogenesis
.
Dev. Cell
4
,
395
-
406
.
Avior
,
Y.
,
Sagi
,
I.
and
Benvenisty
,
N.
(
2016
).
Pluripotent stem cells in disease modelling and drug discovery
.
Nat. Rev. Mol. Cell Biol.
17
,
170
-
182
.
Awaya
,
T.
,
Kato
,
T.
,
Mizuno
,
Y.
,
Chang
,
H.
,
Niwa
,
A.
,
Umeda
,
K.
,
Nakahata
,
T.
and
Heike
,
T.
(
2012
).
Selective development of myogenic mesenchymal cells from human embryonic and induced pluripotent stem cells
.
PLoS ONE
7
,
e51638
.
Babai
,
F.
,
Musevi-Aghdam
,
J.
,
Schurch
,
W.
,
Royal
,
A.
and
Gabbiani
,
G.
(
1990
).
Coexpression of alpha-sarcomeric actin, alpha-smooth muscle actin and desmin during myogenesis in rat and mouse embryos I. Skeletal muscle
.
Differentiation
44
,
132
-
142
.
Baker
,
R. K.
and
Lyons
,
G. E.
(
1996
).
Embryonic stem cells and in vitro muscle development
.
Curr. Top. Dev. Biol.
33
,
263
-
279
.
Barberi
,
T.
,
Bradbury
,
M.
,
Dincer
,
Z.
,
Panagiotakos
,
G.
,
Socci
,
N. D.
and
Studer
,
L.
(
2007
).
Derivation of engraftable skeletal myoblasts from human embryonic stem cells
.
Nat. Med.
13
,
642
-
648
.
Barrios
,
A.
,
Poole
,
R. J.
,
Durbin
,
L.
,
Brennan
,
C.
,
Holder
,
N.
and
Wilson
,
S. W.
(
2003
).
Eph/Ephrin signaling regulates the mesenchymal-to-epithelial transition of the paraxial mesoderm during somite morphogenesis
.
Curr. Biol.
13
,
1571
-
1582
.
Beam
,
K. G.
,
Knudson
,
C. M.
and
Powell
,
J. A.
(
1986
).
A lethal mutation in mice eliminates the slow calcium current in skeletal muscle cells
.
Nature
320
,
168
-
170
.
Beauchamp
,
J. R.
,
Heslop
,
L.
,
Yu
,
D. S. W.
,
Tajbakhsh
,
S.
,
Kelly
,
R. G.
,
Wernig
,
A.
,
Buckingham
,
M. E.
,
Partridge
,
T. A.
and
Zammit
,
P. S.
(
2000
).
Expression of CD34 and Myf5 defines the majority of quiescent adult skeletal muscle satellite cells
.
J. Cell Biol.
151
,
1221
-
1234
.
Beddington
,
R. S. P.
and
Robertson
,
E. J.
(
1999
).
Axis development and early asymmetry in mammals
.
Cell
96
,
195
-
209
.
Bénazéraf
,
B.
,
Francois
,
P.
,
Baker
,
R. E.
,
Denans
,
N.
,
Little
,
C. D.
and
Pourquié
,
O.
(
2010
).
A random cell motility gradient downstream of FGF controls elongation of an amniote embryo
.
Nature
466
,
248
-
252
.
Bentzinger
,
C. F.
,
Wang
,
Y. X.
,
von Maltzahn
,
J.
,
Soleimani
,
V. D.
,
Yin
,
H.
and
Rudnicki
,
M. A.
(
2013
).
Fibronectin regulates Wnt7a signaling and satellite cell expansion
.
Cell Stem Cell
12
,
75
-
87
.
Bentzinger
,
C. F.
,
von Maltzahn
,
J.
,
Dumont
,
N. A.
,
Stark
,
D. A.
,
Wang
,
Y. X.
,
Nhan
,
K.
,
Frenette
,
J.
,
Cornelison
,
D. D. W.
and
Rudnicki
,
M. A.
(
2014
).
Wnt7a stimulates myogenic stem cell motility and engraftment resulting in improved muscle strength
.
J. Cell Biol.
205
,
97
-
111
.
Berkes
,
C. A.
and
Tapscott
,
S. J.
(
2005
).
MyoD and the transcriptional control of myogenesis
.
Semin. Cell Dev. Biol.
16
,
585
-
595
.
Bernardo
,
A. S.
,
Faial
,
T.
,
Gardner
,
L.
,
Niakan
,
K. K.
,
Ortmann
,
D.
,
Senner
,
C. E.
,
Callery
,
E. M.
,
Trotter
,
M. W.
,
Hemberger
,
M.
,
Smith
,
J. C.
, et al. 
(
2011
).
BRACHYURY and CDX2 mediate BMP-induced differentiation of human and mouse pluripotent stem cells into embryonic and extraembryonic lineages
.
Cell Stem Cell
9
,
144
-
155
.
Bessho
,
Y.
,
Hirata
,
H.
,
Masamizu
,
Y.
and
Kageyama
,
R.
(
2003
).
Periodic repression by the bHLH factor Hes7 is an essential mechanism for the somite segmentation clock
.
Genes Dev.
17
,
1451
-
1456
.
Biressi
,
S.
,
Molinaro
,
M.
and
Cossu
,
G.
(
2007
).
Cellular heterogeneity during vertebrate skeletal muscle development
.
Dev. Biol.
308
,
281
-
293
.
Bladt
,
F.
,
Riethmacher
,
D.
,
Isenmann
,
S.
,
Aguzzi
,
A.
and
Birchmeier
,
C.
(
1995
).
Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud
.
Nature
376
,
768
-
771
.
Bober
,
E.
,
Franz
,
T.
,
Arnold
,
H. H.
,
Gruss
,
P.
and
Tremblay
,
P.
(
1994
).
Pax-3 is required for the development of limb muscles: a possible role for the migration of dermomyotomal muscle progenitor cells
.
Development
120
,
603
-
612
.
Borchin
,
B.
,
Chen
,
J.
and
Barberi
,
T.
(
2013
).
Derivation and FACS-mediated purification of PAX3+/PAX7+ skeletal muscle precursors from human pluripotent stem cells
.
Stem Cell Rep.
1
,
620
-
631
.
Borycki
,
A. G.
,
Brunk
,
B.
,
Tajbakhsh
,
S.
,
Buckingham
,
M.
,
Chiang
,
C.
and
Emerson
,
C. P.
, Jr
. (
1999
).
Sonic hedgehog controls epaxial muscle determination through Myf5 activation
.
Development
126
,
4053
-
4063
.
Boulet
,
A. M.
and
Capecchi
,
M. R.
(
2012
).
Signaling by FGF4 and FGF8 is required for axial elongation of the mouse embryo
.
Dev. Biol.
371
,
235
-
245
.
Bowman
,
W.
(
1840
).
On the minute structure and movements of voluntary muscle
.
Philos. Trans. R. Soc. Lond.
130
,
457
-
501
.
Brack
,
A. S.
and
Rando
,
T. A.
(
2012
).
Tissue-specific stem cells: lessons from the skeletal muscle satellite cell
.
Cell Stem Cell
10
,
504
-
514
.
Brand-Saberi
,
B.
,
Müller
,
T. S.
,
Wilting
,
J.
,
Christ
,
B.
and
Birchmeier
,
C.
(
1996
).
Scatter factor/hepatocyte growth factor (SF/HGF) induces emigration of myogenic cells at interlimb level in vivo
.
Dev. Biol.
179
,
303
-
308
.
Buckingham
,
M.
,
Bajard
,
L.
,
Chang
,
T.
,
Daubas
,
P.
,
Hadchouel
,
J.
,
Meilhac
,
S.
,
Montarras
,
D.
,
Rocancourt
,
D.
and
Relaix
,
F.
(
2003
).
The formation of skeletal muscle: from somite to limb
.
J. Anat.
202
,
59
-
68
.
Buonanno
,
A.
and
Merlie
,
J. P.
(
1986
).
Transcriptional regulation of nicotinic acetylcholine receptor genes during muscle development
.
J. Biol. Chem.
261
,
11452
-
11455
.
Burgess
,
R.
,
Rawls
,
A.
,
Brown
,
D.
,
Bradley
,
A.
and
Olson
,
E. N.
(
1996
).
Requirement of the paraxis gene for somite formation and musculoskeletal patterning
.
Nature
384
,
570
-
573
.
Bursac
,
N.
,
Juhas
,
M.
and
Rando
,
T. A.
(
2015
).
Synergizing engineering and biology to treat and model skeletal muscle injury and disease
.
Annu. Rev. Biomed. Eng.
17
,
217
-
242
.
Cambray
,
N.
and
Wilson
,
V.
(
2007
).
Two distinct sources for a population of maturing axial progenitors
.
Development
134
,
2829
-
2840
.
Cardasis
,
C. A.
and
Cooper
,
G. W.
(
1975
).
An analysis of nuclear numbers in individual muscle fibers during differentiation and growth: a satellite cell-muscle fiber growth unit
.
J. Exp. Zool.
191
,
347
-
357
.
Caron
,
L.
,
Kher
,
D.
,
Lee
,
K. L.
,
McKernan
,
R.
,
Dumevska
,
B.
,
Hidalgo
,
A.
,
Li
,
J.
,
Yang
,
H.
,
Main
,
H.
,
Ferri
,
G.
, et al. 
(
2016
).
A human pluripotent stem cell model of facioscapulohumeral muscular dystrophy-affected skeletal muscles
.
Stem Cells Transl. Med.
5
,
1145
-
1161
.
Chadwick
,
K.
,
Wang
,
L.
,
Li
,
L.
,
Menendez
,
P.
,
Murdoch
,
B.
,
Rouleau
,
A.
and
Bhatia
,
M.
(
2003
).
Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells
.
Blood
102
,
906
-
915
.
Chal
,
J.
and
Pourquie
,
O.
(
2009
).
Patterning and differentiation of the vertebrate spine
. In
The Skeletal System: Cold Spring Harbor Monographs Vol.
53
(ed.
O.
Pourquie
), pp.
41
-
116
.
Cold Spring Harbor, NY
:
Cold Spring Harbor Laboratory Press
.
Chal
,
J.
,
Oginuma
,
M.
,
Al Tanoury
,
Z.
,
Gobert
,
B.
,
Sumara
,
O.
,
Hick
,
A.
,
Bousson
,
F.
,
Zidouni
,
Y.
,
Mursch
,
C.
,
Moncuquet
,
P.
, et al. 
(
2015
).
Differentiation of pluripotent stem cells to muscle fiber to model Duchenne muscular dystrophy
.
Nat. Biotechnol.
33
,
962
-
969
.
Chal
,
J.
,
Al Tanoury
,
Z.
,
Hestin
,
M.
,
Gobert
,
B.
,
Aivio
,
S.
,
Hick
,
A.
,
Cherrier
,
T.
,
Nesmith
,
A. P.
,
Parker
,
K. K.
and
Pourquié
,
O.
(
2016
).
Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro
.
Nat. Protoc.
11
,
1833
-
1850
.
Chal
,
J.
,
Al Tanoury
,
Z.
,
Oginuma
,
M.
,
Moncuquet
,
P.
,
Myanari
,
A.
,
Gobert
,
B.
and
Pourquie
,
O.
(
2017a
).
Recapitulating early development of mouse musculo-skeletal precursors of the paraxial mesoderm in vitro
.
BioRxiv
,
https://doi.org/10.1101/140574
.
Chal
,
J.
,
Guillot
,
C.
and
Pourquié
,
O.
(
2017b
).
PAPC couples the segmentation clock to somite morphogenesis by regulating N-cadherin-dependent adhesion
.
Development
144
,
664
-
676
.
Chalamalasetty
,
R. B.
,
Garriock
,
R. J.
,
Dunty
,
W. C.
, Jr
,
Kennedy
,
M. W.
,
Jailwala
,
P.
,
Si
,
H.
and
Yamaguchi
,
T. P.
(
2014
).
Mesogenin 1 is a master regulator of paraxial presomitic mesoderm differentiation
.
Development
141
,
4285
-
4297
.
Chan
,
S.
and
Head
,
S. I.
(
2011
).
The role of branched fibres in the pathogenesis of Duchenne muscular dystrophy
.
Exp. Physiol.
96
,
564
-
571
.
Chan
,
S. S.-K.
,
Shi
,
X.
,
Toyama
,
A.
,
Arpke
,
R. W.
,
Dandapat
,
A.
,
Iacovino
,
M.
,
Kang
,
J.
,
Le
,
G.
,
Hagen
,
H. R.
,
Garry
,
D. J.
, et al. 
(
2013
).
Mesp1 patterns mesoderm into cardiac, hematopoietic, or skeletal myogenic progenitors in a context-dependent manner
.
Cell Stem Cell
12
,
587
-
601
.
Chang
,
H.
,
Yoshimoto
,
M.
,
Umeda
,
K.
,
Iwasa
,
T.
,
Mizuno
,
Y.
,
Fukada
,
S.
,
Yamamoto
,
H.
,
Motohashi
,
N.
,
Miyagoe-Suzuki
,
Y.
,
Takeda
,
S.
, et al. 
(
2009
).
Generation of transplantable, functional satellite-like cells from mouse embryonic stem cells
.
FASEB J.
23
,
1907
-
1919
.
Chapman
,
D. L.
,
Agulnik
,
I.
,
Hancock
,
S.
,
Silver
,
L. M.
and
Papaioannou
,
V. E.
(
1996
).
Tbx6, a mouse T-Box gene implicated in paraxial mesoderm formation at gastrulation
.
Dev. Biol.
180
,
534
-
542
.
Charge
,
S. B. P.
and
Rudnicki
,
M. A.
(
2004
).
Cellular and molecular regulation of muscle regeneration
.
Physiol. Rev.
84
,
209
-
238
.
Chen
,
J.-F.
,
Mandel
,
E. M.
,
Thomson
,
J. M.
,
Wu
,
Q.
,
Callis
,
T. E.
,
Hammond
,
S. M.
,
Conlon
,
F. L.
and
Wang
,
D.-Z.
(
2006
).
The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation
.
Nat. Genet.
38
,
228
-
233
.
Chen
,
F.
,
Cao
,
J.
,
Liu
,
Q.
,
Qin
,
J.
,
Kong
,
J.
,
Wang
,
Y.
,
Li
,
Y.
,
Geng
,
J.
,
Li
,
Q.
,
Yang
,
L.
, et al. 
(
2012
).
Comparative study of myocytes from normal and mdx mice iPS cells
.
J. Cell. Biochem.
113
,
678
-
684
.
Chevallier
,
A.
(
1979
).
Role of the somitic mesoderm in the development of the thorax in bird embryos. II. Origin of thoracic and appendicular musculature
.
J. Embryol. Exp. Morphol.
49
,
73
-
88
.
Chin
,
E. R.
,
Olson
,
E. N.
,
Richardson
,
J. A.
,
Yang
,
Q.
,
Humphries
,
C.
,
Shelton
,
J. M.
,
Wu
,
H.
,
Zhu
,
W.
,
Bassel-Duby
,
R.
and
Williams
,
R. S.
(
1998
).
A calcineurin-dependent transcriptional pathway controls skeletal muscle fiber type
.
Genes Dev.
12
,
2499
-
2509
.
Choi
,
I. Y.
,
Lim
,
H.
,
Estrellas
,
K.
,
Mula
,
J.
,
Cohen
,
T. V.
,
Zhang
,
Y.
,
Donnelly
,
C. J.
,
Richard
,
J.-P.
,
Kim
,
Y. J.
,
Kim
,
H.
, et al. 
(
2016
).
Concordant but varied phenotypes among duchenne muscular dystrophy patient-specific myoblasts derived using a human iPSC-based model
.
Cell Rep.
15
,
2301
-
2312
.
Christ
,
B.
and
Scaal
,
M.
(
2008
).
Formation and differentiation of avian somite derivatives
.
Adv. Exp. Med. Biol.
638
,
1
-
41
.
Christ
,
B.
,
Jacob
,
M.
and
Jacob
,
H. J.
(
1983
).
On the origin and development of the ventrolateral abdominal muscles in the avian embryo. An experimental and ultrastructural study
.
Anat. Embryol.
166
,
87
-
101
.
Ciruna
,
B.
and
Rossant
,
J.
(
2001
).
FGF signaling regulates mesoderm cell fate specification and morphogenetic movement at the primitive streak
.
Dev. Cell
1
,
37
-
49
.
Cognard
,
C.
,
Lazdunski
,
M.
and
Romey
,
G.
(
1986
).
Different types of Ca2+ channels in mammalian skeletal muscle cells in culture
.
Proc. Natl. Acad. Sci. USA
83
,
517
-
521
.
Collins
,
C. A.
,
Olsen
,
I.
,
Zammit
,
P. S.
,
Heslop
,
L.
,
Petrie
,
A.
,
Partridge
,
T. A.
and
Morgan
,
J. E.
(
2005
).
Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche
.
Cell
122
,
289
-
301
.
Comai
,
G.
and
Tajbakhsh
,
S.
(
2014
).
Molecular and cellular regulation of skeletal myogenesis
.
Curr. Top. Dev. Biol.
110
,
1
-
73
.
Condon
,
K.
,
Silberstein
,
L.
,
Blau
,
H. M.
and
Thompson
,
W. J.
(
1990
).
Development of muscle fiber types in the prenatal rat hindlimb
.
Dev. Biol.
138
,
256
-
274
.
Constantinides
,
P. G.
,
Jones
,
P. A.
and
Gevers
,
W.
(
1977
).
Functional striated muscle cells from non-myoblast precursors following 5-azacytidine treatment
.
Nature
267
,
364
-
366
.
Cornelison
,
D. D. W.
and
Wold
,
B. J.
(
1997
).
Single-cell analysis of regulatory gene expression in quiescent and activated mouse skeletal muscle satellite cells
.
Dev. Biol.
191
,
270
-
283
.
Cosgrove
,
B. D.
,
Sacco
,
A.
,
Gilbert
,
P. M.
and
Blau
,
H. M.
(
2009
).
A home away from home: challenges and opportunities in engineering in vitro muscle satellite cell niches
.
Differentiation
78
,
185
-
194
.
Cossu
,
G.
,
Kelly
,
R.
,
Di Donna
,
S.
,
Vivarelli
,
E.
and
Buckingham
,
M.
(
1995
).
Myoblast differentiation during mammalian somitogenesis is dependent upon a community effect
.
Proc. Natl. Acad. Sci. USA
92
,
2254
-
2258
.
Crist
,
C.
(
2017
).
Emerging new tools to study and treat muscle pathologies: genetics and molecular mechanisms underlying skeletal muscle development, regeneration, and disease
.
J. Pathol.
241
,
264
-
272
.
Cuenda
,
A.
and
Cohen
,
P.
(
1999
).
Stress-activated protein kinase-2/p38 and a rapamycin-sensitive pathway are required for C2C12 myogenesis
.
J. Biol. Chem.
274
,
4341
-
4346
.
Cuny
,
G. D.
,
Yu
,
P. B.
,
Laha
,
J. K.
,
Xing
,
X.
,
Liu
,
J.-F.
,
Lai
,
C. S.
,
Deng
,
D. Y.
,
Sachidanandan
,
C.
,
Bloch
,
K. D.
and
Peterson
,
R. T.
(
2008
).
Structure-activity relationship study of bone morphogenetic protein (BMP) signaling inhibitors
.
Bioorg. Med. Chem. Lett.
18
,
4388
-
4392
.
Dagher
,
R.
and
Helman
,
L.
(
1999
).
Rhabdomyosarcoma: an overview
.
Oncologist
4
,
34
-
44
.
Dale
,
J. K.
,
Maroto
,
M.
,
Dequeant
,
M.-L.
,
Malapert
,
P.
,
McGrew
,
M.
and
Pourquie
,
O.
(
2003
).
Periodic Notch inhibition by Lunatic Fringe underlies the chick segmentation clock
.
Nature
421
,
275
-
278
.
Darabi
,
R.
,
Gehlbach
,
K.
,
Bachoo
,
R. M.
,
Kamath
,
S.
,
Osawa
,
M.
,
Kamm
,
K. E.
,
Kyba
,
M.
and
Perlingeiro
,
R. C. R.
(
2008
).
Functional skeletal muscle regeneration from differentiating embryonic stem cells
.
Nat. Med.
14
,
134
-
143
.
Darabi
,
R.
,
Santos
,
F. N. C.
,
Filareto
,
A.
,
Pan
,
W.
,
Koene
,
R.
,
Rudnicki
,
M. A.
,
Kyba
,
M.
and
Perlingeiro
,
R. C. R.
(
2011
).
Assessment of the myogenic stem cell compartment following transplantation of Pax3/Pax7-induced embryonic stem cell-derived progenitors
.
Stem Cells
29
,
777
-
790
.
Darabi
,
R.
,
Arpke
,
R. W.
,
Irion
,
S.
,
Dimos
,
J. T.
,
Grskovic
,
M.
,
Kyba
,
M.
and
Perlingeiro
,
R. C. R.
(
2012
).
Human ES- and iPS-derived myogenic progenitors restore DYSTROPHIN and improve contractility upon transplantation in dystrophic mice
.
Cell Stem Cell
10
,
610
-
619
.
Davis
,
R. L.
,
Weintraub
,
H.
and
Lassar
,
A. B.
(
1987
).
Expression of a single transfected cDNA converts fibroblasts to myoblasts
.
Cell
51
,
987
-
1000
.
Dekel
,
I.
,
Magal
,
Y.
,
Pearson-White
,
S.
,
Emerson
,
C. P.
and
Shani
,
M.
(
1992
).
Conditional conversion of ES cells to skeletal muscle by an exogenous MyoD1 gene
.
New Biologist
4
,
217
-
224
.
Delfini
,
M.-C.
,
Dubrulle
,
J.
,
Malapert
,
P.
,
Chal
,
J.
and
Pourquie
,
O.
(
2005
).
Control of the segmentation process by graded MAPK/ERK activation in the chick embryo
.
Proc. Natl. Acad. Sci. USA
102
,
11343
-
11348
.
Demestre
,
M.
,
Orth
,
M.
,
Föhr
,
K. J.
,
Achberger
,
K.
,
Ludolph
,
A. C.
,
Liebau
,
S.
and
Boeckers
,
T. M.
(
2015
).
Formation and characterisation of neuromuscular junctions between hiPSC derived motoneurons and myotubes
.
Stem Cell Res.
15
,
328
-
336
.
Demonbreun
,
A. R.
,
Biersmith
,
B. H.
and
McNally
,
E. M.
(
2015
).
Membrane fusion in muscle development and repair
.
Semin. Cell Dev. Biol.
45
,
48
-
56
.
Denans
,
N.
,
Iimura
,
T.
and
Pourquie
,
O.
(
2015
).
Hox genes control vertebrate body elongation by collinear Wnt repression
.
eLife
4
,
doi: 10.7554/eLife.04379
.
Denetclaw
,
W. F. J.
,
Christ
,
B.
and
Ordahl
,
C. P.
(
1997
).
Location and growth of epaxial myotome precursor cells
.
Development
124
,
1601
-
1610
.
Ding
,
G.
,
Tanaka
,
Y.
,
Hayashi
,
M.
,
Nishikawa
,
S.-I.
and
Kataoka
,
H.
(
2013
).
PDGF receptor alpha+ mesoderm contributes to endothelial and hematopoietic cells in mice
.
Dev. Dyn.
242
,
254
-
268
.
Doetschman
,
T. C.
,
Eistetter
,
H.
,
Katz
,
M.
,
Schmidt
,
W.
and
Kemler
,
R.
(
1985
).
The in vitro development of blastocyst-derived embryonic stem cell lines: formation of visceral yolk sac, blood islands and myocardium
.
J. Embryol. Exp. Morphol.
87
,
27
-
45
.
Donoghue
,
M. J.
,
Morris-Valero
,
R.
,
Johnson
,
Y. R.
,
Merlie
,
J. P.
and
Sanes
,
J. R.
(
1992
).
Mammalian muscle cells bear a cell-autonomous, heritable memory of their rostrocaudal position
.
Cell
69
,
67
-
77
.
Duband
,
J. L.
,
Dufour
,
S.
,
Hatta
,
K.
,
Takeichi
,
M.
,
Edelman
,
G. M.
and
Thiery
,
J. P.
(
1987
).
Adhesion molecules during somitogenesis in the avian embryo
.
J. Cell Biol.
104
,
1361
-
1374
.
Dubrulle
,
J.
,
McGrew
,
M. J.
and
Pourquié
,
O.
(
2001
).
FGF signaling controls somite boundary position and regulates segmentation clock control of spatiotemporal Hox gene activation
.
Cell
106
,
219
-
232
.
Dumont
,
N. A.
,
Wang
,
Y. X.
and
Rudnicki
,
M. A.
(
2015
).
Intrinsic and extrinsic mechanisms regulating satellite cell function
.
Development
142
,
1572
-
1581
.
Dunn
,
S. E.
,
Burns
,
J. L.
and
Michel
,
R. N.
(
1999
).
Calcineurin is required for skeletal muscle hypertrophy
.
J. Biol. Chem.
274
,
21908
-
21912
.
Dunty
,
W. C.
, Jr
,
Biris
,
K. K.
,
Chalamalasetty
,
R. B.
,
Taketo
,
M. M.
,
Lewandoski
,
M.
and
Yamaguchi
,
T. P.
(
2008
).
Wnt3a/beta-catenin signaling controls posterior body development by coordinating mesoderm formation and segmentation
.
Development
135
,
85
-
94
.
Ema
,
M.
,
Takahashi
,
S.
and
Rossant
,
J.
(
2006
).
Deletion of the selection cassette, but not cis-acting elements, in targeted Flk1-lacZ allele reveals Flk1 expression in multipotent mesodermal progenitors
.
Blood
107
,
111
-
117
.
Emery
,
A. E. H.
(
2002
).
The muscular dystrophies
.
Lancet
359
,
687
-
695
.
Fan
,
C.-M.
and
Tessier-Lavigne
,
M.
(
1994
).
Patterning of mammalian somites by surface ectoderm and notochord: evidence for sclerotome induction by a hedgehog homolog
.
Cell
79
,
1175
-
1186
.
Fehling
,
H. J.
,
Lacaud
,
G.
,
Kubo
,
A.
,
Kennedy
,
M.
,
Robertson
,
S.
,
Keller
,
G.
and
Kouskoff
,
V.
(
2003
).
Tracking mesoderm induction and its specification to the hemangioblast during embryonic stem cell differentiation
.
Development
130
,
4217
-
4227
.
Filareto
,
A.
,
Parker
,
S.
,
Darabi
,
R.
,
Borges
,
L.
,
Iacovino
,
M.
,
Schaaf
,
T.
,
Mayerhofer
,
T.
,
Chamberlain
,
J. S.
,
Ervasti
,
J. M.
,
McIvor
,
R. S.
, et al. 
(
2013
).
An ex vivo gene therapy approach to treat muscular dystrophy using inducible pluripotent stem cells
.
Nat. Commun.
4
,
1549
.
Filareto
,
A.
,
Rinaldi
,
F.
,
Arpke
,
R. W.
,
Darabi
,
R.
,
Belanto
,
J. J.
,
Toso
,
E. A.
,
Miller
,
A. Z.
,
Ervasti
,
J. M.
,
McIvor
,
R. S.
,
Kyba
,
M.
, et al. 
(
2015
).
Pax3-induced expansion enables the genetic correction of dystrophic satellite cells
.
Skeletal Muscle
5
,
36
.
Flanagan-Steet
,
H.
,
Hannon
,
K.
,
McAvoy
,
M. J.
,
Hullinger
,
R.
and
Olwin
,
B. B.
(
2000
).
Loss of FGF receptor 1 signaling reduces skeletal muscle mass and disrupts myofiber organization in the developing limb
.
Dev. Biol.
218
,
21
-
37
.
Flucher
,
B. E.
(
1992
).
Structural analysis of muscle development: transverse tubules, sarcoplasmic reticulum, and the triad
.
Dev. Biol.
154
,
245
-
260
.
Flucher
,
B. E.
,
Phillips
,
J. L.
,
Powell
,
J. A.
,
Andrews
,
S. B.
and
Daniels
,
M. P.
(
1992
).
Coordinated development of myofibrils, sarcoplasmic reticulum and transverse tubules in normal and dysgenic mouse skeletal muscle, in vivo and in vitro
.
Dev. Biol.
150
,
266
-
280
.
Flucher
,
B. E.
,
Andrews
,
S. B.
,
Fleischer
,
S.
,
Marks
,
A. R.
,
Caswell
,
A.
and
Powell
,
J. A.
(
1993
).
Triad formation: organization and function of the sarcoplasmic reticulum calcium release channel and triadin in normal and dysgenic muscle in vitro
.
J. Cell Biol.
123
,
1161
-
1174
.
Fougerousse
,
F.
,
Edom-Vovard
,
F.
,
Merkulova
,
T.
,
Ott
,
M. O.
,
Durand
,
M.
,
Butler-Browne
,
G.
and
Keller
,
A.
(
2001
).
The muscle-specific enolase is an early marker of human myogenesis
.
J. Muscle Res. Cell Motil.
22
,
535
-
544
.
Fougerousse
,
F.
,
Durand
,
M.
,
Lopez
,
S.
,
Suel
,
L.
,
Demignon
,
J.
,
Thornton
,
C.
,
Ozaki
,
H.
,
Kawakami
,
K.
,
Barbet
,
P.
,
Beckmann
,
J. S.
, et al. 
(
2002
).
Six and Eya expression during human somitogenesis and MyoD gene family activation
.
J. Muscle Res. Cell Motil.
23
,
255
-
264
.
Furst
,
D. O.
,
Osborn
,
M.
and
Weber
,
K.
(
1989
).
Myogenesis in the mouse embryo: differential onset of expression of myogenic proteins and the involvement of titin in myofibril assembly
.
J. Cell Biol.
109
,
517
-
527
.
Gadue
,
P.
,
Huber
,
T. L.
,
Paddison
,
P. J.
and
Keller
,
G. M.
(
2006
).
Wnt and TGF-beta signaling are required for the induction of an in vitro model of primitive streak formation using embryonic stem cells
.
Proc. Natl. Acad. Sci. USA
103
,
16806
-
16811
.
Garcia-Martinez
,
V.
and
Schoenwolf
,
G. C.
(
1992
).
Positional control of mesoderm movement and fate during avian gastrulation and neurulation
.
Dev. Dyn.
193
,
249
-
256
.
Garriock
,
R. J.
,
Chalamalasetty
,
R. B.
,
Kennedy
,
M. W.
,
Canizales
,
L. C.
,
Lewandoski
,
M.
and
Yamaguchi
,
T. P.
(
2015
).
Lineage tracing of neuromesodermal progenitors reveals novel Wnt-dependent roles in trunk progenitor cell maintenance and differentiation
.
Development
142
,
1628
-
1638
.
Ge
,
Y.
and
Chen
,
J.
(
2011
).
MicroRNAs in skeletal myogenesis
.
Cell Cycle
10
,
441
-
448
.
Gerard
,
M.
,
Abitbol
,
M.
,
Delezoide
,
A. L.
,
Dufier
,
J. L.
,
Mallet
,
J.
and
Vekemans
,
M.
(
1995
).
PAX-genes expression during human embryonic development, a preliminary report
.
C. R. Acad. Sci. III
318
,
57
-
66
.
Gilbert
,
P. M.
,
Havenstrite
,
K. L.
,
Magnusson
,
K. E. G.
,
Sacco
,
A.
,
Leonardi
,
N. A.
,
Kraft
,
P.
,
Nguyen
,
N. K.
,
Thrun
,
S.
,
Lutolf
,
M. P.
and
Blau
,
H. M.
(
2010
).
Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture
.
Science
329
,
1078
-
1081
.
Gokhin
,
D. S.
,
Ward
,
S. R.
,
Bremner
,
S. N.
and
Lieber
,
R. L.
(
2008
).
Quantitative analysis of neonatal skeletal muscle functional improvement in the mouse
.
J. Exp. Biol.
211
,
837
-
843
.
Goudenege
,
S.
,
Lebel
,
C.
,
Huot
,
N. B.
,
Dufour
,
C.
,
Fujii
,
I.
,
Gekas
,
J.
,
Rousseau
,
J.
and
Tremblay
,
J. P.
(
2012
).
Myoblasts derived from normal hESCs and dystrophic hiPSCs efficiently fuse with existing muscle fibers following transplantation
.
Mol. Ther.
20
,
2153
-
2167
.
Goulding
,
M. D.
,
Chalepakis
,
G.
,
Deutsch
,
U.
,
Erselius
,
J. R.
and
Gruss
,
P.
(
1991
).
Pax-3, a novel murine DNA binding protein expressed during early neurogenesis
.
EMBO J.
10
,
1135
-
1147
.
Gouti
,
M.
,
Tsakiridis
,
A.
,
Wymeersch
,
F. J.
,
Huang
,
Y.
,
Kleinjung
,
J.
,
Wilson
,
V.
and
Briscoe
,
J.
(
2014
).
In vitro generation of neuromesodermal progenitors reveals distinct roles for wnt signalling in the specification of spinal cord and paraxial mesoderm identity
.
PLoS Biol.
12
,
e1001937
.
Grifone
,
R.
,
Laclef
,
C.
,
Spitz
,
F.
,
Lopez
,
S.
,
Demignon
,
J.
,
Guidotti
,
J.-E.
,
Kawakami
,
K.
,
Xu
,
P.-X.
,
Kelly
,
R.
,
Petrof
,
B. J.
, et al. 
(
2004
).
Six1 and Eya1 expression can reprogram adult muscle from the slow-twitch phenotype into the fast-twitch phenotype
.
Mol. Cell. Biol.
24
,
6253
-
6267
.
Grifone
,
R.
,
Demignon
,
J.
,
Houbron
,
C.
,
Souil
,
E.
,
Niro
,
C.
,
Seller
,
M. J.
,
Hamard
,
G.
and
Maire
,
P.
(
2005
).
Six1 and Six4 homeoproteins are required for Pax3 and Mrf expression during myogenesis in the mouse embryo
.
Development
132
,
2235
-
2249
.
Grifone
,
R.
,
Demignon
,
J.
,
Giordani
,
J.
,
Niro
,
C.
,
Souil
,
E.
,
Bertin
,
F.
,
Laclef
,
C.
,
Xu
,
P.-X.
and
Maire
,
P.
(
2007
).
Eya1 and Eya2 proteins are required for hypaxial somitic myogenesis in the mouse embryo
.
Dev. Biol.
302
,
602
-
616
.
Gros
,
J.
,
Scaal
,
M.
and
Marcelle
,
C.
(
2004
).
A two-step mechanism for myotome formation in chick
.
Dev. Cell
6
,
875
-
882
.
Gros
,
J.
,
Manceau
,
M.
,
Thomé
,
V.
and
Marcelle
,
C.
(
2005
).
A common somitic origin for embryonic muscle progenitors and satellite cells
.
Nature
435
,
954
-
958
.
Gros
,
J.
,
Serralbo
,
O.
and
Marcelle
,
C.
(
2009
).
WNT11 acts as a directional cue to organize the elongation of early muscle fibres
.
Nature
457
,
589
-
593
.
Gu
,
J.-M.
,
Wang
,
D. J.
,
Peterson
,
J. M.
,
Shintaku
,
J.
,
Liyanarachchi
,
S.
,
Coppola
,
V.
,
Frakes
,
A. E.
,
Kaspar
,
B. K.
,
Cornelison
,
D. D.
and
Guttridge
,
D. C.
(
2016
).
An NF-kappaB - EphrinA5-dependent communication between NG2(+) interstitial cells and myoblasts promotes muscle growth in neonates
.
Dev. Cell
36
,
215
-
224
.
Guan
,
K.
,
Rohwedel
,
J.
and
Wobus
,
A. M.
(
1999
).
Embryonic stem cell differentiation models: cardiogenesis, myogenesis, neurogenesis, epithelial and vascular smooth muscle cell differentiation in vitro
.
Cytotechnology
30
,
211
-
226
.
Günther
,
S.
,
Kim
,
J.
,
Kostin
,
S.
,
Lepper
,
C.
,
Fan
,
C.-M.
and
Braun
,
T.
(
2013
).
Myf5-positive satellite cells contribute to Pax7-dependent long-term maintenance of adult muscle stem cells
.
Cell Stem Cell
13
,
590
-
601
.
Hasty
,
P.
,
Bradley
,
A.
,
Morris
,
J. H.
,
Edmondson
,
D. G.
,
Venuti
,
J. M.
,
Olson
,
E. N.
and
Klein
,
W. H.
(
1993
).
Muscle deficiency and neonatal death in mice with a targeted mutation in the myogenin gene
.
Nature
364
,
501
-
506
.
Hayashi
,
H.
,
Mochii
,
M.
,
Kodama
,
R.
,
Hamada
,
Y.
,
Mizuno
,
N.
,
Eguchi
,
G.
and
Tachi
,
C.
(
1996
).
Isolation of a novel chick homolog of Serrate and its coexpression with C-Notch-1 in chick development
.
Int. J. Dev. Biol.
40
,
1089
-
1096
.
Hellmuth
,
A. E.
and
Allbrook
,
D. B.
(
1971
).
Muscle satellite cell numbers during the postnatal period
.
J. Anat.
110
,
503
.
Henrique
,
D.
,
Abranches
,
E.
,
Verrier
,
L.
and
Storey
,
K. G.
(
2015
).
Neuromesodermal progenitors and the making of the spinal cord
.
Development
142
,
2864
-
2875
.
Hicks
,
M.
and
Pyle
,
A.
(
2015
).
The path from pluripotency to skeletal muscle: developmental myogenesis guides the way
.
Cell Stem Cell
17
,
255
-
257
.
Hirsch
,
E.
,
Lohikangas
,
L.
,
Gullberg
,
D.
,
Johansson
,
S.
and
Fassler
,
R.
(
1998
).
Mouse myoblasts can fuse and form a normal sarcomere in the absence of beta1 integrin expression
.
J. Cell Sci.
111
,
2397
-
2409
.
Hirsinger
,
E.
,
Jouve
,
C.
,
Dubrulle
,
J.
and
Pourquié
,
O.
(
2000
).
Somite formation and patterning
.
Int. Rev. Cytol.
198
,
1
-
65
.
Hirsinger
,
E.
,
Malapert
,
P.
,
Dubrulle
,
J.
,
Delfini
,
M. C.
,
Duprez
,
D.
,
Henrique
,
D.
,
Ish-Horowicz
,
D.
and
Pourquie
,
O.
(
2001
).
Notch signalling acts in postmitotic avian myogenic cells to control MyoD activation
.
Development
128
,
107
-
116
.
Höllrigl
,
A.
,
Puz
,
S.
,
Al-Dubai
,
H.
,
Kim
,
J. U.
,
Capetanaki
,
Y.
and
Weitzer
,
G.
(
2002
).
Amino-terminally truncated desmin rescues fusion of des(−/−) myoblasts but negatively affects cardiomyogenesis and smooth muscle development
.
FEBS Lett.
523
,
229
-
233
.
Horikawa
,
K.
,
Radice
,
G.
,
Takeichi
,
M.
and
Chisaka
,
O.
(
1999
).
Adhesive subdivisions intrinsic to the epithelial somites
.
Dev. Biol.
215
,
182
-
189
.
Horsley
,
V.
,
Jansen
,
K. M.
,
Mills
,
S. T.
and
Pavlath
,
G. K.
(
2003
).
IL-4 acts as a myoblast recruitment factor during mammalian muscle growth
.
Cell
113
,
483
-
494
.
Horst
,
D.
,
Ustanina
,
S.
,
Sergi
,
C.
,
Mikuz
,
G.
,
Juergens
,
H.
,
Braun
,
T.
and
Vorobyov
,
E.
(
2006
).
Comparative expression analysis of Pax3 and Pax7 during mouse myogenesis
.
Int. J. Dev. Biol.
50
,
47
-
54
.
Hosoyama
,
T.
,
McGivern
,
J. V.
,
Van Dyke
,
J. M.
,
Ebert
,
A. D.
and
Suzuki
,
M.
(
2014
).
Derivation of myogenic progenitors directly from human pluripotent stem cells using a sphere-based culture
.
Stem Cells Transl. Med.
3
,
564
-
574
.
Hubaud
,
A.
and
Pourquié
,
O.
(
2014
).
Signalling dynamics in vertebrate segmentation
.
Nat. Rev. Mol. Cell Biol.
15
,
709
-
721
.
Hurren
,
B.
,
Collins
,
J. J. P.
,
Duxson
,
M. J.
and
Deries
,
M.
(
2015
).
First neuromuscular contact correlates with onset of primary myogenesis in rat and mouse limb muscles
.
PLoS ONE
10
,
e0133811
.
Hutcheson
,
D. A.
,
Zhao
,
J.
,
Merrell
,
A.
,
Haldar
,
M.
and
Kardon
,
G.
(
2009
).
Embryonic and fetal limb myogenic cells are derived from developmentally distinct progenitors and have different requirements for beta-catenin
.
Genes Dev.
23
,
997
-
1013
.
Hwang
,
Y.
,
Suk
,
S.
,
Shih
,
Y.-R. V.
,
Seo
,
T.
,
Du
,
B.
,
Xie
,
Y.
,
Li
,
Z.
and
Varghese
,
S.
(
2014
).
WNT3A promotes myogenesis of human embryonic stem cells and enhances in vivo engraftment
.
Sci. Rep.
4
,
5916
.
Iimura
,
T.
,
Yang
,
X.
,
Weijer
,
C. J.
and
Pourquie
,
O.
(
2007
).
Dual mode of paraxial mesoderm formation during chick gastrulation
.
Proc. Natl. Acad. Sci. USA
104
,
2744
-
2749
.
Itoh
,
N.
,
Mima
,
T.
and
Mikawa
,
T.
(
1996
).
Loss of fibroblast growth factor receptors is necessary for terminal differentiation of embryonic limb muscle
.
Development
122
,
291
-
300
.
Iulianella
,
A.
,
Beckett
,
B.
,
Petkovich
,
M.
and
Lohnes
,
D.
(
1999
).
A molecular basis for retinoic acid-induced axial truncation
.
Dev. Biol.
205
,
33
-
48
.
Jacob
,
M.
,
Christ
,
B.
and
Jacob
,
H. J.
(
1979
).
The migration of myogenic cells from the somites into the leg region of avian embryos. An ultrastructural study
.
Anat. Embryol.
157
,
291
-
309
.
Jacobson
,
C.
,
Côté
,
P. D.
,
Rossi
,
S. G.
,
Rotundo
,
R. L.
and
Carbonetto
,
S.
(
2001
).
The dystroglycan complex is necessary for stabilization of acetylcholine receptor clusters at neuromuscular junctions and formation of the synaptic basement membrane
.
J. Cell Biol.
152
,
435
-
450
.
Jarad
,
G.
and
Miner
,
J. H.
(
2009
).
The Pax3-Cre transgene exhibits a rostrocaudal gradient of expression in the skeletal muscle lineage
.
Genesis
47
,
1
-
6
.
Johansson
,
B. M.
and
Wiles
,
M. V.
(
1995
).
Evidence for involvement of activin A and bone morphogenetic protein 4 in mammalian mesoderm and hematopoietic development
.
Mol. Cell. Biol.
15
,
141
-
151
.
Johnson
,
R. L.
,
Laufer
,
E.
,
Riddle
,
R. D.
and
Tabin
,
C.
(
1994
).
Ectopic expression of Sonic hedgehog alters dorsal-ventral patterning of somites
.
Cell
79
,
1165
-
1173
.
Jostes
,
B.
,
Walther
,
C.
and
Gruss
,
P.
(
1990
).
The murine paired box gene, Pax7, is expressed specifically during the development of the nervous and muscular system
.
Mech. Dev.
33
,
27
-
37
.
Jurberg
,
A. D.
,
Aires
,
R.
,
Nóvoa
,
A.
,
Rowland
,
J. E.
and
Mallo
,
M.
(
2014
).
Compartment-dependent activities of Wnt3a/beta-catenin signaling during vertebrate axial extension
.
Dev. Biol.
394
,
253
-
263
.
Kahane
,
N.
,
Cinnamon
,
Y.
,
Bachelet
,
I.
and
Kalcheim
,
C.
(
2001
).
The third wave of myotome colonization by mitotically competent progenitors: regulating the balance between differentiation and proliferation during muscle development
.
Development
128
,
2187
-
2198
.
Kassar-Duchossoy
,
L.
,
Giacone
,
E.
,
Gayraud-Morel
,
B.
,
Jory
,
A.
,
Gomes
,
D.
and
Tajbakhsh
,
S.
(
2005
).
Pax3/Pax7 mark a novel population of primitive myogenic cells during development
.
Genes Dev.
19
,
1426
-
1431
.
Kato
,
K.
and
Gurdon
,
J. B.
(
1993
).
Single-cell transplantation determines the time when Xenopus muscle precursor cells acquire a capacity for autonomous differentiation
.
Proc. Natl. Acad. Sci. USA
90
,
1310
-
1314
.
Kattman
,
S. J.
,
Witty
,
A. D.
,
Gagliardi
,
M.
,
Dubois
,
N. C.
,
Niapour
,
M.
,
Hotta
,
A.
,
Ellis
,
J.
and
Keller
,
G.
(
2011
).
Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines
.
Cell Stem Cell
8
,
228
-
240
.
Kaufman
,
D. S.
,
Hanson
,
E. T.
,
Lewis
,
R. L.
,
Auerbach
,
R.
and
Thomson
,
J. A.
(
2001
).
Hematopoietic colony-forming cells derived from human embryonic stem cells
.
Proc. Natl. Acad. Sci. USA
98
,
10716
-
10721
.
Kazuki
,
Y.
,
Hiratsuka
,
M.
,
Takiguchi
,
M.
,
Osaki
,
M.
,
Kajitani
,
N.
,
Hoshiya
,
H.
,
Hiramatsu
,
K.
,
Yoshino
,
T.
,
Kazuki
,
K.
,
Ishihara
,
C.
, et al. 
(
2010
).
Complete genetic correction of ips cells from Duchenne muscular dystrophy
.
Mol. Ther.
18
,
386
-
393
.
Keller
,
G. M.
(
1995
).
In vitro differentiation of embryonic stem cells
.
Curr. Opin. Cell Biol.
7
,
862
-
869
.
Keller
,
A.
,
Ott
,
M.-O.
,
Lamandé
,
N.
,
Lucas
,
M.
,
Gros
,
F.
,
Buckingham
,
M.
and
Lazar
,
M.
(
1992
).
Activation of the gene encoding the glycolytic enzyme beta-enolase during early myogenesis precedes an increased expression during fetal muscle development
.
Mech. Dev.
38
,
41
-
54
.
Kelly
,
R. G.
,
Zammit
,
P. S.
,
Schneider
,
A.
,
Alonso
,
S.
,
Biben
,
C.
and
Buckingham
,
M. E.
(
1997
).
Embryonic and fetal myogenic programs act through separate enhancers at the MLC1F/3F locus
.
Dev. Biol.
187
,
183
-
199
.
Kempf
,
H.
,
Olmer
,
R.
,
Haase
,
A.
,
Franke
,
A.
,
Bolesani
,
E.
,
Schwanke
,
K.
,
Robles-Diaz
,
D.
,
Coffee
,
M.
,
Göhring
,
G.
,
Dräger
,
G.
, et al. 
(
2016
).
Bulk cell density and Wnt/TGFbeta signalling regulate mesendodermal patterning of human pluripotent stem cells
.
Nat. Commun.
7
,
13602
.
Kennedy
,
K. A. M.
,
Porter
,
T.
,
Mehta
,
V.
,
Ryan
,
S. D.
,
Price
,
F.
,
Peshdary
,
V.
,
Karamboulas
,
C.
,
Savage
,
J.
,
Drysdale
,
T. A.
,
Li
,
S.-C.
, et al. 
(
2009
).
Retinoic acid enhances skeletal muscle progenitor formation and bypasses inhibition by bone morphogenetic protein 4 but not dominant negative beta-catenin
.
BMC Biol.
7
,
67
.
Kieny
,
M.
,
Mauger
,
A.
and
Sengel
,
P.
(
1972
).
Early regionalization of the somitic mesoderm as studied by the development of the axial skeleton of the chick embryo
.
Dev. Biol.
28
,
142
-
161
.
Kim
,
H. K.
,
Lee
,
Y. S.
,
Sivaprasad
,
U.
,
Malhotra
,
A.
and
Dutta
,
A.
(
2006
).
Muscle-specific microRNA miR-206 promotes muscle differentiation
.
J. Cell Biol.
174
,
677
-
687
.
Kim
,
J. H.
,
Jin
,
P.
,
Duan
,
R.
and
Chen
,
E. H.
(
2015a
).
Mechanisms of myoblast fusion during muscle development
.
Curr. Opin. Genet. Dev.
32
,
162
-
170
.
Kim
,
J. H.
,
Ren
,
Y.
,
Ng
,
W. P.
,
Li
,
S.
,
Son
,
S.
,
Kee
,
Y.-S.
,
Zhang
,
S.
,
Zhang
,
G.
,
Fletcher
,
D. A.
,
Robinson
,
D. N.
, et al. 
(
2015b
).
Mechanical tension drives cell membrane fusion
.
Dev. Cell
32
,
561
-
573
.
Kishigami
,
S.
and
Mishina
,
Y.
(
2005
).
BMP signaling and early embryonic patterning
.
Cytokine Growth Factor Rev.
16
,
265
-
278
.
Kume
,
T.
,
Jiang
,
H.
,
Topczewska
,
J. M.
and
Hogan
,
B. L.
(
2001
).
The murine winged helix transcription factors, Foxc1 and Foxc2, are both required for cardiovascular development and somitogenesis
.
Genes Dev.
15
,
2470
-
2482
.
Laurin
,
M.
,
Fradet
,
N.
,
Blangy
,
A.
,
Hall
,
A.
,
Vuori
,
K.
and
Cote
,
J.-F.
(
2008
).
The atypical Rac activator Dock180 (Dock1) regulates myoblast fusion in vivo
.
Proc. Natl. Acad. Sci. USA
105
,
15446
-
15451
.
Le Grand
,
F.
,
Jones
,
A. E.
,
Seale
,
V.
,
Scimè
,
A.
and
Rudnicki
,
M. A.
(
2009
).
Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells
.
Cell Stem Cell
4
,
535
-
547
.
Lee
,
S.-J.
(
2004
).
Regulation of muscle mass by myostatin
.
Annu. Rev. Cell Dev. Biol.
20
,
61
-
86
.
Lemke
,
S. B.
and
Schnorrer
,
F.
(
2017
).
Mechanical forces during muscle development
.
Mech. Dev.
144
,
92
-
101
.
Lepper
,
C.
and
Fan
,
C.-M.
(
2010
).
Inducible lineage tracing of Pax7-descendant cells reveals embryonic origin of adult satellite cells
.
Genesis
48
,
424
-
436
.
Lepper
,
C.
,
Partridge
,
T. A.
and
Fan
,
C.-M.
(
2011
).
An absolute requirement for Pax7-positive satellite cells in acute injury-induced skeletal muscle regeneration
.
Development
138
,
3639
-
3646
.
Lescroart
,
F.
,
Chabab
,
S.
,
Lin
,
X.
,
Rulands
,
S.
,
Paulissen
,
C.
,
Rodolosse
,
A.
,
Auer
,
H.
,
Achouri
,
Y.
,
Dubois
,
C.
,
Bondue
,
A.
, et al. 
(
2014
).
Early lineage restriction in temporally distinct populations of Mesp1 progenitors during mammalian heart development
.
Nat. Cell Biol.
16
,
829
-
840
.
Leung
,
M.
,
Cooper
,
A.
,
Jana
,
S.
,
Tsao
,
C.-T.
,
Petrie
,
T. A.
and
Zhang
,
M.
(
2013
).
Nanofiber-based in vitro system for high myogenic differentiation of human embryonic stem cells
.
Biomacromolecules
14
,
4207
-
4216
.
Lindsley
,
R. C.
,
Gill
,
J. G.
,
Kyba
,
M.
,
Murphy
,
T. L.
and
Murphy
,
K. M.
(
2006
).
Canonical Wnt signaling is required for development of embryonic stem cell-derived mesoderm
.
Development
133
,
3787
-
3796
.
Linker
,
C.
,
Lesbros
,
C.
,
Gros
,
J.
,
Burrus
,
L. W.
,
Rawls
,
A.
and
Marcelle
,
C.
(
2005
).
beta-Catenin-dependent Wnt signalling controls the epithelial organisation of somites through the activation of paraxis
.
Development
132
,
3895
-
3905
.
Liu
,
P.
,
Wakamiya
,
M.
,
Shea
,
M. J.
,
Albrecht
,
U.
,
Behringer
,
R. R.
and
Bradley
,
A.
(
1999
).
Requirement for Wnt3 in vertebrate axis formation
.
Nat. Genet.
22
,
361
-
365
.
Loh
,
K. M.
,
Chen
,
A.
,
Koh
,
P. W.
,
Deng
,
T. Z.
,
Sinha
,
R.
,
Tsai
,
J. M.
,
Barkal
,
A. A.
,
Shen
,
K. Y.
,
Jain
,
R.
,
Morganti
,
R. M.
, et al. 
(
2016
).
Mapping the pairwise choices leading from pluripotency to human bone, heart, and other mesoderm cell types
.
Cell
166
,
451
-
467
.
Lyons
,
G. E.
,
Ontell
,
M.
,
Cox
,
R.
,
Sassoon
,
D.
and
Buckingham
,
M.
(
1990
).
The expression of myosin genes in developing skeletal muscle in the mouse embryo
.
J. Cell Biol.
111
,
1465
-
1476
.
Lyons
,
G. E.
,
Buckingham
,
M. E.
and
Mannherz
,
H. G.
(
1991a
).
alpha-Actin proteins and gene transcripts are colocalized in embryonic mouse muscle
.
Development
111
,
451
-
454
.
Lyons
,
G. E.
,
Muhlebach
,
S.
,
Moser
,
A.
,
Masood
,
R.
,
Paterson
,
B. M.
,
Buckingham
,
M. E.
and
Perriard
,
J. C.
(
1991b
).
Developmental regulation of creatine kinase gene expression by myogenic factors in embryonic mouse and chick skeletal muscle
.
Development
113
,
1017
-
1029
.
Magli
,
A.
,
Schnettler
,
E.
,
Rinaldi
,
F.
,
Bremer
,
P.
and
Perlingeiro
,
R. C. R.
(
2013
).
Functional dissection of Pax3 in paraxial mesoderm development and myogenesis
.
Stem Cells
31
,
59
-
70
.
Magli
,
A.
,
Schnettler
,
E.
,
Swanson
,
S. A.
,
Borges
,
L.
,
Hoffman
,
K.
,
Stewart
,
R.
,
Thomson
,
J. A.
,
Keirstead
,
S. A.
and
Perlingeiro
,
R. C. R.
(
2014
).
Pax3 and Tbx5 specify whether PDGFRalpha+ cells assume skeletal or cardiac muscle fate in differentiating embryonic stem cells
.
Stem Cells
32
,
2072
-
2083
.
Mankoo
,
B. S.
,
Collins
,
N. S.
,
Ashby
,
P.
,
Grigorieva
,
E.
,
Pevny
,
L. H.
,
Candia
,
A.
,
Wright
,
C. V. E.
,
Rigby
,
P. W. J.
and
Pachnis
,
V.
(
1999
).
Mox2 is a component of the genetic hierarchy controlling limb muscle development
.
Nature
400
,
69
-
73
.
Mankoo
,
B. S.
,
Skuntz
,
S.
,
Harrigan
,
I.
,
Grigorieva
,
E.
,
Candia
,
A.
,
Wright
,
C. V. E.
,
Arnheiter
,
H.
and
Pachnis
,
V.
(
2003
).
The concerted action of Meox homeobox genes is required upstream of genetic pathways essential for the formation, patterning and differentiation of somites
.
Development
130
,
4655
-
4664
.
Marcelle
,
C.
,
Lesbros
,
C.
and
Linker
,
C.
(
2002
).
Somite patterning: a few more pieces of the puzzle
.
Results Probl. Cell Differ.
38
,
81
-
108
.
Maroto
,
M.
,
Reshef
,
R.
,
Münsterberg
,
A. E.
,
Koester
,
S.
,
Goulding
,
M.
and
Lassar
,
A. B.
(
1997
).
Ectopic Pax-3 activates MyoD and Myf-5 expression in embryonic mesoderm and neural tissue
.
Cell
89
,
139
-
148
.
Martin
,
P.
(
1990
).
Tissue patterning in the developing mouse limb
.
Int. J. Dev. Biol.
34
,
323
-
336
.
Martins
,
G. G.
,
Rifes
,
P.
,
Amândio
,
R.
,
Rodrigues
,
G.
,
Palmeirim
,
I.
and
Thorsteinsdóttir
,
S.
(
2009
).
Dynamic 3D cell rearrangements guided by a fibronectin matrix underlie somitogenesis
.
PLoS ONE
4
,
e7429
.
McMahon
,
J. A.
,
Takada
,
S.
,
Zimmerman
,
L. B.
,
Fan
,
C.-M.
,
Harland
,
R. M.
and
McMahon
,
A. P.
(
1998
).
Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite
.
Genes Dev.
12
,
1438
-
1452
.
McPherron
,
A. C.
,
Lawler
,
A. M.
and
Lee
,
S.-J.
(
1997
).
Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member
.
Nature
387
,
83
-
90
.
Mendjan
,
S.
,
Mascetti
,
V. L.
,
Ortmann
,
D.
,
Ortiz
,
M.
,
Karjosukarso
,
D. W.
,
Ng
,
Y.
,
Moreau
,
T.
and
Pedersen
,
R. A.
(
2014
).
NANOG and CDX2 pattern distinct subtypes of human mesoderm during exit from pluripotency
.
Cell Stem Cell
15
,
310
-
325
.
Menko
,
A. S.
and
Boettiger
,
D.
(
1987
).
Occupation of the extracellular matrix receptor, integrin, is a control point for myogenic differentiation
.
Cell
51
,
51
-
57
.
Merkle
,
F. T.
and
Eggan
,
K.
(
2013
).
Modeling human disease with pluripotent stem cells: from genome association to function
.
Cell Stem Cell
12
,
656
-
668
.
Messina
,
G.
,
Biressi
,
S.
,
Monteverde
,
S.
,
Magli
,
A.
,
Cassano
,
M.
,
Perani
,
L.
,
Roncaglia
,
E.
,
Tagliafico
,
E.
,
Starnes
,
L.
,
Campbell
,
C. E.
, et al. 
(
2010
).
Nfix regulates fetal-specific transcription in developing skeletal muscle
.
Cell
140
,
554
-
566
.
Milasincic
,
D. J.
,
Calera
,
M. R.
,
Farmer
,
S. R.
and
Pilch
,
P. F.
(
1996
).
Stimulation of C2C12 myoblast growth by basic fibroblast growth factor and insulin-like growth factor 1 can occur via mitogen-activated protein kinase-dependent and -independent pathways
.
Mol. Cell. Biol.
16
,
5964
-
5973
.
Mishina
,
M.
,
Takai
,
T.
,
Imoto
,
K.
,
Noda
,
M.
,
Takahashi
,
T.
,
Numa
,
S.
,
Methfessel
,
C.
and
Sakmann
,
B.
(
1986
).
Molecular distinction between fetal and adult forms of muscle acetylcholine receptor
.
Nature
321
,
406
-
411
.
Miura
,
S.
,
Davis
,
S.
,
Klingensmith
,
J.
and
Mishina
,
Y.
(
2006
).
BMP signaling in the epiblast is required for proper recruitment of the prospective paraxial mesoderm and development of the somites
.
Development
133
,
3767
-
3775
.
Mizuno
,
Y.
,
Suzuki
,
M.
,
Nakagawa
,
H.
,
Ninagawa
,
N.
and
Torihashi
,
S.
(
2009
).
Switching of actin isoforms in skeletal muscle differentiation using mouse ES cells
.
Histochem. Cell Biol.
132
,
669
-
672
.
Mizuno
,
Y.
,
Chang
,
H.
,
Umeda
,
K.
,
Niwa
,
A.
,
Iwasa
,
T.
,
Awaya
,
T.
,
Fukada
,
S.
,
Yamamoto
,
H.
,
Yamanaka
,
S.
,
Nakahata
,
T.
, et al. 
(
2010
).
Generation of skeletal muscle stem/progenitor cells from murine induced pluripotent stem cells
.
FASEB J.
24
,
2245
-
2253
.
Montarras
,
D.
,
Morgan
,
J.
,
Collins
,
C.
,
Relaix
,
F.
,
Zaffran
,
S.
,
Cumano
,
A.
,
Partridge
,
T.
and
Buckingham
,
M.
(
2005
).
Direct isolation of satellite cells for skeletal muscle regeneration
.
Science
309
,
2064
-
2067
.
Moretti
,
A.
,
Laugwitz
,
K.-L.
,
Dorn
,
T.
,
Sinnecker
,
D.
and
Mummery
,
C.
(
2013
).
Pluripotent stem cell models of human heart disease
.
Cold Spring Harb. Perspect. Med.
3
,
pii: a014027
.
Motohashi
,
N.
,
Alexander
,
M. S.
,
Shimizu-Motohashi
,
Y.
,
Myers
,
J. A.
,
Kawahara
,
G.
and
Kunkel
,
L. M.
(
2013
).
Regulation of IRS1/Akt insulin signaling by microRNA-128a during myogenesis
.
J. Cell Sci.
126
,
2678
-
2691
.
Motoike
,
T.
,
Markham
,
D. W.
,
Rossant
,
J.
and
Sato
,
T. N.
(
2003
).
Evidence for novel fate of Flk1+ progenitor: contribution to muscle lineage
.
Genesis
35
,
153
-
159
.
Mummery
,
C.
,
Ward-van Oostwaard
,
D.
,
Doevendans
,
P.
,
Spijker
,
R.
,
van den Brink
,
S.
,
Hassink
,
R.
,
van der Heyden
,
M.
,
Opthof
,
T.
,
Pera
,
M.
,
de la Riviere
,
A. B.
, et al. 
(
2003
).
Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells
.
Circulation
107
,
2733
-
2740
.
Münsterberg
,
A. E.
and
Lassar
,
A. B.
(
1995
).
Combinatorial signals from the neural tube, floor plate and notochord induce myogenic bHLH gene expression in the somite
.
Development
121
,
651
-
660
.
Münsterberg
,
A. E.
,
Kitajewski
,
J.
,
Bumcrot
,
D. A.
,
McMahon
,
A. P.
and
Lassar
,
A. B.
(
1995
).
Combinatorial signaling by Sonic hedgehog and Wnt family members induces myogenic bHLH gene expression in the somite
.
Genes Dev.
9
,
2911
-
2922
.
Murphy
,
M.
and
Kardon
,
G.
(
2011
).
Origin of vertebrate limb muscle: the role of progenitor and myoblast populations
.
Curr. Top. Dev. Biol.
96
,
1
-
32
.
Murphy
,
M. M.
,
Lawson
,
J. A.
,
Mathew
,
S. J.
,
Hutcheson
,
D. A.
and
Kardon
,
G.
(
2011
).
Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration
.
Development
138
,
3625
-
3637
.
Murry
,
C. E.
and
Keller
,
G.
(
2008
).
Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development
.
Cell
132
,
661
-
680
.
Murtaugh
,
L. C.
,
Chyung
,
J. H.
and
Lassar
,
A. B.
(
1999
).
Sonic hedgehog promotes somitic chondrogenesis by altering the cellular response to BMP signaling
.
Genes Dev.
13
,
225
-
237
.
Musaro
,
A.
,
McCullagh
,
K. J. A.
,
Naya
,
F. J.
,
Olson
,
E. N.
and
Rosenthal
,
N.
(
1999
).
IGF-1 induces skeletal myocyte hypertrophy through calcineurin in association with GATA-2 and NF-ATc1
.
Nature
400
,
581
-
585
.
Nabeshima
,
Y.
,
Hanaoka
,
K.
,
Hayasaka
,
M.
,
Esuml
,
E.
,
Li
,
S.
,
Nonaka
,
I.
and
Nabeshima
,
Y.
(
1993
).
Myogenin gene disruption results in perinatal lethality because of severe muscle defect
.
Nature
364
,
532
-
535
.
Naiche
,
L. A.
,
Holder
,
N.
and
Lewandoski
,
M.
(
2011
).
FGF4 and FGF8 comprise the wavefront activity that controls somitogenesis
.
Proc. Natl. Acad. Sci. USA
108
,
4018
-
4023
.
Nakajima
,
Y.
,
Morimoto
,
M.
,
Takahashi
,
Y.
,
Koseki
,
H.
and
Saga
,
Y.
(
2006
).
Identification of Epha4 enhancer required for segmental expression and the regulation by Mesp2
.
Development
133
,
2517
-
2525
.
Nakanishi
,
M.
,
Kurisaki
,
A.
,
Hayashi
,
Y.
,
Warashina
,
M.
,
Ishiura
,
S.
,
Kusuda-Furue
,
M.
and
Asashima
,
M.
(
2009
).
Directed induction of anterior and posterior primitive streak by Wnt from embryonic stem cells cultured in a chemically defined serum-free medium
.
FASEB J.
23
,
114
-
122
.
Nakaya
,
Y.
,
Kuroda
,
S.
,
Katagiri
,
Y. T.
,
Kaibuchi
,
K.
and
Takahashi
,
Y.
(
2004
).
Mesenchymal-epithelial transition during somitic segmentation is regulated by differential roles of Cdc42 and Rac1
.
Dev. Cell
7
,
425
-
438
.
Nishikawa
,
S. I.
,
Nishikawa
,
S.
,
Hirashima
,
M.
,
Matsuyoshi
,
N.
and
Kodama
,
H.
(
1998
).
Progressive lineage analysis by cell sorting and culture identifies FLK1+VE-cadherin+ cells at a diverging point of endothelial and hemopoietic lineages
.
Development
125
,
1747
-
1757
.
Nowicki
,
J. L.
and
Burke
,
A. C.
(
2000
).
Hox genes and morphological identity: axial versus lateral patterning in the vertebrate mesoderm
.
Development
127
,
4265
-
4275
.
Nowotschin
,
S.
,
Ferrer-Vaquer
,
A.
,
Concepcion
,
D.
,
Papaioannou
,
V. E.
and
Hadjantonakis
,
A.-K.
(
2012
).
Interaction of Wnt3a, Msgn1 and Tbx6 in neural versus paraxial mesoderm lineage commitment and paraxial mesoderm differentiation in the mouse embryo
.
Dev. Biol.
367
,
1
-
14
.
Oginuma
,
M.
,
Niwa
,
Y.
,
Chapman
,
D. L.
and
Saga
,
Y.
(
2008
).
Mesp2 and Tbx6 cooperatively create periodic patterns coupled with the clock machinery during mouse somitogenesis
.
Development
135
,
2555
-
2562
.
Oginuma
,
M.
,
Moncuquet
,
P.
,
Xiong
,
F.
,
Karoly
,
E.
,
Chal
,
J.
,
Guevorkian
,
K.
and
Pourquie
,
O.
(
2017
).
A gradient of glycolytic activity coordinates FGF and Wnt signaling during elongation of the body axis in amniote embryos
.
Dev. Cell
40
,
342
-
353 e310
.
Olivera-Martinez
,
I.
and
Storey
,
K. G.
(
2007
).
Wnt signals provide a timing mechanism for the FGF-retinoid differentiation switch during vertebrate body axis extension
.
Development
134
,
2125
-
2135
.
Olivera-Martinez
,
I.
,
Coltey
,
M.
,
Dhouailly
,
D.
and
Pourquie
,
O.
(
2000
).
Mediolateral somitic origin of ribs and dermis determined by quail-chick chimeras
.
Development
127
,
4611
-
4617
.
Ordahl
,
C. P.
(
1993
).
Myogenic lineages within the developing somite
. In
Molecular Basis of Morphogenesis
(ed.
M.
Bernfield
), pp.
165
-
176
.
New York
:
John Wiley and Sons
.
Ordahl
,
C. P.
and
Le Douarin
,
N. M.
(
1992
).
Two myogenic lineages within the developing somite
.
Development
114
,
339
-
353
.
Orlova
,
V. V.
,
Chuva de Sousa Lopes
,
S.
and
Valdimarsdottir
,
G.
(
2015
).
BMP-SMAD signaling: from pluripotent stem cells to cardiovascular commitment
.
Cytokine Growth Factor. Rev.
27
,
55
-
63
.
Ott
,
M. O.
,
Bober
,
E.
,
Lyons
,
G.
,
Arnold
,
H.
and
Buckingham
,
M.
(
1991
).
Early expression of the myogenic regulatory gene, myf-5, in precursor cells of skeletal muscle in the mouse embryo
.
Development
111
,
1097
-
1107
.
Otto
,
A.
,
Schmidt
,
C.
and
Patel
,
K.
(
2006
).
Pax3 and Pax7 expression and regulation in the avian embryo
.
Anat. Embryol.
211
,
293
-
310
.
Oustanina
,
S.
,
Hause
,
G.
and
Braun
,
T.
(
2004
).
Pax7 directs postnatal renewal and propagation of myogenic satellite cells but not their specification
.
EMBO J.
23
,
3430
-
3439
.
Ozasa
,
S.
,
Kimura
,
S.
,
Ito
,
K.
,
Ueno
,
H.
,
Ikezawa
,
M.
,
Matsukura
,
M.
,
Yoshioka
,
K.
,
Araki
,
K.
,
Yamamura
,
K.-I.
,
Abe
,
K.
, et al. 
(
2007
).
Efficient conversion of ES cells into myogenic lineage using the gene-inducible system
.
Biochem. Biophys. Res. Commun.
357
,
957
-
963
.
Ozbudak
,
E. M.
,
Tassy
,
O.
and
Pourquie
,
O.
(
2010
).
Spatiotemporal compartmentalization of key physiological processes during muscle precursor differentiation
.
Proc. Natl. Acad. Sci. USA
107
,
4224
-
4229
.
Pourquié
,
O.
,
Fan
,
C.-M.
,
Coltey
,
M.
,
Hirsinger
,
E.
,
Watanabe
,
Y.
,
Bréant
,
C.
,
Francis-West
,
P.
,
Brickell
,
P.
,
Tessier-Lavigne
,
M.
and
Le Douarin
,
N. M.
(
1996
).
Lateral and axial signals involved in avian somite patterning: a role for BMP4
.
Cell
84
,
461
-
471
.
Pownall
,
M. E.
and
Emerson
,
C. P.
, Jr
. (
1992
).
Sequential activation of three myogenic regulatory genes during somite morphogenesis in quail embryos
.
Dev. Biol.
151
,
67
-
79
.
Pownall
,
M. E.
,
Gustafsson
,
M. K.
and
Emerson
,
C. P.
, Jr
. (
2002
).
Myogenic regulatory factors and the specification of muscle progenitors in vertebrate embryos
.
Annu. Rev. Cell Dev. Biol.
18
,
747
-
783
.
Puttonen
,
K. A.
,
Ruponen
,
M.
,
Naumenko
,
N.
,
Hovatta
,
O. H.
,
Tavi
,
P.
and
Koistinaho
,
J.
(
2015
).
Generation of functional neuromuscular junctions from human pluripotent stem cell lines
.
Front. Cell. Neurosci.
9
,
473
.
Quattrocelli
,
M.
,
Palazzolo
,
G.
,
Floris
,
G.
,
Schöffski
,
P.
,
Anastasia
,
L.
,
Orlacchio
,
A.
,
Vandendriessche
,
T.
,
Chuah
,
M. K. L.
,
Cossu
,
G.
,
Verfaillie
,
C.
, et al. 
(
2011
).
Intrinsic cell memory reinforces myogenic commitment of pericyte-derived iPSCs
.
J. Pathol.
223
,
593
-
603
.
Rahimov
,
F.
and
Kunkel
,
L. M.
(
2013
).
The cell biology of disease: cellular and molecular mechanisms underlying muscular dystrophy
.
J. Cell Biol.
201
,
499
-
510
.
Ramkumar
,
N.
and
Anderson
,
K. V.
(
2011
).
SnapShot: mouse primitive streak
.
Cell
146
,
488
-
488.e482
.
Rao
,
L.
,
Tang
,
W.
,
Wei
,
Y.
,
Bao
,
L.
,
Chen
,
J.
,
Chen
,
H.
,
He
,
L.
,
Lu
,
P.
,
Ren
,
J.
,
Wu
,
L.
, et al. 
(
2012
).
Highly efficient derivation of skeletal myotubes from human embryonic stem cells
.
Stem Cell Rev.
8
,
1109
-
1119
.
Relaix
,
F.
,
Rocancourt
,
D.
,
Mansouri
,
A.
and
Buckingham
,
M.
(
2004
).
Divergent functions of murine Pax3 and Pax7 in limb muscle development
.
Genes Dev.
18
,
1088
-
1105
.
Relaix
,
F.
,
Rocancourt
,
D.
,
Mansouri
,
A.
and
Buckingham
,
M.
(
2005
).
A Pax3/Pax7-dependent population of skeletal muscle progenitor cells
.
Nature
435
,
948
-
953
.
Reshef
,
R.
,
Maroto
,
M.
and
Lassar
,
A. B.
(
1998
).
Regulation of dorsal somitic cell fates: BMPs and Noggin control the timing and pattern of myogenic regulator expression
.
Genes Dev.
12
,
290
-
303
.
Richard
,
A.-F.
,
Demignon
,
J.
,
Sakakibara
,
I.
,
Pujol
,
J.
,
Favier
,
M.
,
Strochlic
,
L.
,
Le Grand
,
F.
,
Sgarioto
,
N.
,
Guernec
,
A.
,
Schmitt
,
A.
, et al. 
(
2011
).
Genesis of muscle fiber-type diversity during mouse embryogenesis relies on Six1 and Six4 gene expression
.
Dev. Biol.
359
,
303
-
320
.
Rios
,
A. C.
,
Serralbo
,
O.
,
Salgado
,
D.
and
Marcelle
,
C.
(
2011
).
Neural crest regulates myogenesis through the transient activation of NOTCH
.
Nature
473
,
532
-
535
.
Robbins
,
J.
,
Gulick
,
J.
,
Sanchez
,
A.
,
Howles
,
P.
and
Doetschman
,
T.
(
1990
).
Mouse embryonic stem cells express the cardiac myosin heavy chain genes during development in vitro
.
J. Biol. Chem.
265
,
11905
-
11909
.
Robertson
,
C.
,
Tran
,
D. D.
and
George
,
S. C.
(
2013
).
Concise review: maturation phases of human pluripotent stem cell-derived cardiomyocytes
.
Stem Cells
31
,
829
-
837
.
Rochlin
,
K.
,
Yu
,
S.
,
Roy
,
S.
and
Baylies
,
M. K.
(
2010
).
Myoblast fusion: when it takes more to make one
.
Dev. Biol.
341
,
66
-
83
.
Rodriguez
,
J.
,
Vernus
,
B.
,
Chelh
,
I.
,
Cassar-Malek
,
I.
,
Gabillard
,
J. C.
,
Hadj Sassi
,
A.
,
Seiliez
,
I.
,
Picard
,
B.
and
Bonnieu
,
A.
(
2014
).
Myostatin and the skeletal muscle atrophy and hypertrophy signaling pathways
.
Cell. Mol. Life Sci.
71
,
4361
-
4371
.
Rohwedel
,
J.
,
Maltsev
,
V.
,
Bober
,
E.
,
Arnold
,
H.-H.
,
Hescheler
,
J.
and
Wobus
,
A. M.
(
1994
).
Muscle cell differentiation of embryonic stem cells reflects myogenesis in vivo: developmentally regulated expression of myogenic determination genes and functional expression of ionic currents
.
Dev. Biol.
164
,
87
-
101
.
Rohwedel
,
J.
,
Guan
,
K.
,
Zuschratter
,
W.
,
Jin
,
S.
,
Ahnert-Hilger
,
G.
,
Fürst
,
D.
,
Fässler
,
R.
and
Wobus
,
A. M.
(
1998a
).
Loss of beta1 integrin function results in a retardation of myogenic, but an acceleration of neuronal, differentiation of embryonic stem cells in vitro
.
Dev. Biol.
201
,
167
-
184
.
Rohwedel
,
J.
,
Kleppisch
,
T.
,
Pich
,
U.
,
Guan
,
K.
,
Jin
,
S.
,
Zuschratter
,
W.
,
Hopf
,
C.
,
Hoch
,
W.
,
Hescheler
,
J.
,
Witzemann
,
V.
, et al. 
(
1998b
).
Formation of postsynaptic-like membranes during differentiation of embryonic stem cells in vitro
.
Exp. Cell Res.
239
,
214
-
225
.
Rohwedel
,
J.
,
Guan
,
K.
,
Hegert
,
C.
and
Wobus
,
A. M.
(
2001
).
Embryonic stem cells as an in vitro model for mutagenicity, cytotoxicity and embryotoxicity studies: present state and future prospects
.
Toxicol. In Vitro
15
,
741
-
753
.
Rosen
,
G. D.
,
Sanes
,
J. R.
,
LaChance
,
R.
,
Cunningham
,
J. M.
,
Roman
,
J.
and
Dean
,
D. C.
(
1992
).
Roles for the integrin VLA-4 and its counter receptor VCAM-1 in myogenesis
.
Cell
69
,
1107
-
1119
.
Rosenthal
,
N.
,
Kornhauser
,
J. M.
,
Donoghue
,
M.
,
Rosen
,
K. M.
and
Merlie
,
J. P.
(
1989
).
Myosin light chain enhancer activates muscle-specific, developmentally regulated gene expression in transgenic mice
.
Proc. Natl. Acad. Sci. USA
86
,
7780
-
7784
.
Rudnicki
,
M. A.
,
Schnegelsberg
,
P. N. J.
,
Stead
,
R. H.
,
Braun
,
T.
,
Arnold
,
H.-H.
and
Jaenisch
,
R.
(
1993
).
MyoD or Myf-5 is required for the formation of skeletal muscle
.
Cell
75
,
1351
-
1359
.
Rüegg
,
M. A.
and
Glass
,
D. J.
(
2011
).
Molecular mechanisms and treatment options for muscle wasting diseases
.
Annu. Rev. Pharmacol. Toxicol.
51
,
373
-
395
.
Ryan
,
T.
,
Liu
,
J.
,
Chu
,
A.
,
Wang
,
L.
,
Blais
,
A.
and
Skerjanc
,
I. S.
(
2012
).
Retinoic acid enhances skeletal myogenesis in human embryonic stem cells by expanding the premyogenic progenitor population
.
Stem Cell Rev.
8
,
482
-
493
.
Saga
,
Y.
,
Hata
,
N.
,
Koseki
,
H.
and
Taketo
,
M. M.
(
1997
).
Mesp2: a novel mouse gene expressed in the presegmented mesoderm and essential for segmentation initiation
.
Genes Dev.
11
,
1827
-
1839
.
Sakai
,
Y.
,
Meno
,
C.
,
Fujii
,
H.
,
Nishino
,
J.
,
Shiratori
,
H.
,
Saijoh
,
Y.
,
Rossant
,
J.
and
Hamada
,
H.
(
2001
).
The retinoic acid-inactivating enzyme CYP26 is essential for establishing an uneven distribution of retinoic acid along the anterio-posterior axis within the mouse embryo
.
Genes Dev.
15
,
213
-
225
.
Sakurai
,
H.
,
Inami
,
Y.
,
Tamamura
,
Y.
,
Yoshikai
,
T.
,
Sehara-Fujisawa
,
A.
and
Isobe
,
K.-I.
(
2009
).
Bidirectional induction toward paraxial mesodermal derivatives from mouse ES cells in chemically defined medium
.
Stem Cell Res.
3
,
157
-
169
.
Sakurai
,
H.
,
Sakaguchi
,
Y.
,
Shoji
,
E.
,
Nishino
,
T.
,
Maki
,
I.
,
Sakai
,
H.
,
Hanaoka
,
K.
,
Kakizuka
,
A.
and
Sehara-Fujisawa
,
A.
(
2012
).
In vitro modeling of paraxial mesodermal progenitors derived from induced pluripotent stem cells
.
PLoS ONE
7
,
e47078
.
Sambasivan
,
R.
,
Yao
,
R.
,
Kissenpfennig
,
A.
,
Van Wittenberghe
,
L.
,
Paldi
,
A.
,
Gayraud-Morel
,
B.
,
Guenou
,
H.
,
Malissen
,
B.
,
Tajbakhsh
,
S.
and
Galy
,
A.
(
2011
).
Pax7-expressing satellite cells are indispensable for adult skeletal muscle regeneration
.
Development
138
,
3647
-
3656
.
Sanchez-Gurmaches
,
J.
and
Guertin
,
D. A.
(
2014
).
Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed
.
Nat. Commun.
5
,
4099
.
Sanes
,
J. R.
(
2003
).
The basement membrane/basal lamina of skeletal muscle
.
J. Biol. Chem.
278
,
12601
-
12604
.
Sanes
,
J. R.
and
Lichtman
,
J. W.
(
1999
).
Development of the vertebrate neuromuscular junction
.
Annu. Rev. Neurosci.
22
,
389
-
442
.
Sassoon
,
D. A.
,
Garner
,
I.
and
Buckingham
,
M.
(
1988
).
Transcripts of alpha-cardiac and alpha-skeletal actins are early markers for myogenesis in the mouse embryo
.
Development
104
,
155
-
164
.
Satin
,
J.
,
Itzhaki
,
I.
,
Rapoport
,
S.
,
Schroder
,
E. A.
,
Izu
,
L.
,
Arbel
,
G.
,
Beyar
,
R.
,
Balke
,
C. W.
,
Schiller
,
J.
and
Gepstein
,
L.
(
2008
).
Calcium handling in human embryonic stem cell-derived cardiomyocytes
.
Stem Cells
26
,
1961
-
1972
.
Schiaffino
,
S.
and
Reggiani
,
C.
(
2011
).
Fiber types in mammalian skeletal muscles
.
Physiol. Rev.
91
,
1447
-
1531
.
Schmalbruch
,
H.
and
Hellhammer
,
U.
(
1976
).
The number of satellite cells in normal human muscle
.
Anat. Rec.
185
,
279
-
287
.
Seale
,
P.
,
Sabourin
,
L. A.
,
Girgis-Gabardo
,
A.
,
Mansouri
,
A.
,
Gruss
,
P.
and
Rudnicki
,
M. A.
(
2000
).
Pax7 is required for the specification of myogenic satellite cells
.
Cell
102
,
777
-
786
.
Seale
,
P.
,
Bjork
,
B.
,
Yang
,
W.
,
Kajimura
,
S.
,
Chin
,
S.
,
Kuang
,
S.
,
Scimè
,
A.
,
Devarakonda
,
S.
,
Conroe
,
H. M.
,
Erdjument-Bromage
,
H.
, et al. 
(
2008
).
PRDM16 controls a brown fat/skeletal muscle switch
.
Nature
454
,
961
-
967
.
Selleck
,
M. A.
and
Stern
,
C. D.
(
1991
).
Fate mapping and cell lineage analysis of Hensen's node in the chick embryo
.
Development
112
,
615
-
626
.
Semsarian
,
C.
,
Wu
,
M.-J.
,
Ju
,
Y.-K.
,
Marciniec
,
T.
,
Yeoh
,
T.
,
Allen
,
D. G.
,
Harvey
,
R. P.
and
Graham
,
R. M.
(
1999
).
Skeletal muscle hypertrophy is mediated by a Ca2+-dependent calcineurin signalling pathway
.
Nature
400
,
576
-
581
.
Serralbo
,
O.
and
Marcelle
,
C.
(
2014
).
Migrating cells mediate long-range WNT signaling
.
Development
141
,
2057
-
2063
.
Shani
,
M.
,
Faerman
,
A.
,
Emerson
,
C. P.
,
Pearson-White
,
S.
,
Dekel
,
I.
and
Magal
,
Y.
(
1992
).
The consequences of a constitutive expression of MyoD1 in ES cells and mouse embryos
.
Symp. Soc. Exp. Biol.
46
,
19
-
36
.
Shelton
,
M.
,
Metz
,
J.
,
Liu
,
J.
,
Carpenedo
,
R. L.
,
Demers
,
S.-P.
,
Stanford
,
W. L.
and
Skerjanc
,
I. S.
(
2014
).
Derivation and expansion of PAX7-positive muscle progenitors from human and mouse embryonic stem cells
.
Stem Cell Rep.
3
,
516
-
529
.
Shoji
,
E.
,
Sakurai
,
H.
,
Nishino
,
T.
,
Nakahata
,
T.
,
Heike
,
T.
,
Awaya
,
T.
,
Fujii
,
N.
,
Manabe
,
Y.
,
Matsuo
,
M.
and
Sehara-Fujisawa
,
A.
(
2015
).
Early pathogenesis of Duchenne muscular dystrophy modelled in patient-derived human induced pluripotent stem cells
.
Sci. Rep.
5
,
12831
.
Sieiro
,
D.
,
Rios
,
A. C.
,
Hirst
,
C. E.
and
Marcelle
,
C.
(
2016
).
Cytoplasmic NOTCH and membrane-derived beta-catenin link cell fate choice to epithelial-mesenchymal transition during myogenesis
.
Elife
5
,
pii: e14847
.
Sieiro-Mosti
,
D.
,
De La Celle
,
M.
,
Pele
,
M.
and
Marcelle
,
C.
(
2014
).
A dynamic analysis of muscle fusion in the chick embryo
.
Development
141
,
3605
-
3611
.
Skoglund
,
G.
,
Laine
,
J.
,
Darabi
,
R.
,
Fournier
,
E.
,
Perlingeiro
,
R.
and
Tabti
,
N.
(
2014
).
Physiological and ultrastructural features of human induced pluripotent and embryonic stem cell-derived skeletal myocytes in vitro
.
Proc. Natl. Acad. Sci. USA
111
,
8275
-
8280
.
Smith
,
A. S. T.
,
Davis
,
J.
,
Lee
,
G.
,
Mack
,
D. L.
and
Kim
,
D.-H.
(
2016
).
Muscular dystrophy in a dish: engineered human skeletal muscle mimetics for disease modeling and drug discovery
.
Drug Discov. Today
21
,
1387
-
1398
.
Šošić
,
D.
,
Brand-Saberi
,
B.
,
Schmidt
,
C.
,
Christ
,
B.
and
Olson
,
E. N.
(
1997
).
Regulation of paraxis expression and somite formation by ectoderm- and neural tube-derived signals
.
Dev. Biol.
185
,
229
-
243
.
Sparrow
,
J. C.
and
Schöck
,
F.
(
2009
).
The initial steps of myofibril assembly: integrins pave the way
.
Nat. Rev. Mol. Cell Biol.
10
,
293
-
298
.
Stern
,
C. D.
and
Canning
,
D. R.
(
1990
).
Origin of cells giving rise to mesoderm and endoderm in chick embryo
.
Nature
343
,
273
-
275
.
Stockdale
,
F. E.
(
1992
).
Myogenic cell lineages
.
Dev. Biol.
154
,
284
-
298
.
Streit
,
A.
and
Stern
,
C. D.
(
1999
).
Mesoderm patterning and somite formation during node regression: differential effects of chordin and noggin
.
Mech. Dev.
85
,
85
-
96
.
Sudheer
,
S.
,
Liu
,
J.
,
Marks
,
M.
,
Koch
,
F.
,
Anurin
,
A.
,
Scholze
,
M.
,
Dorothea Senft
,
A.
,
Wittler
,
L.
,
Macura
,
K.
,
Grote
,
P.
, et al. 
(
2016
).
Different concentrations of FGF ligands, FGF2 or FGF8 determine distinct states of WNT-induced presomitic mesoderm
.
Stem Cells
34
,
1790
-
1800
.
Sweetman
,
D.
,
Goljanek
,
K.
,
Rathjen
,
T.
,
Oustanina
,
S.
,
Braun
,
T.
,
Dalmay
,
T.
and
Münsterberg
,
A.
(
2008
).
Specific requirements of MRFs for the expression of muscle specific microRNAs, miR-1, miR-206 and miR-133
.
Dev. Biol.
321
,
491
-
499
.
Takada
,
S.
,
Stark
,
K. L.
,
Shea
,
M. J.
,
Vassileva
,
G.
,
McMahon
,
J. A.
and
McMahon
,
A. P.
(
1994
).
Wnt-3a regulates somite and tailbud formation in the mouse embryo
.
Genes Dev.
8
,
174
-
189
.
Takahashi
,
Y.
,
Koizumi
,
K.-I.
,
Takagi
,
A.
,
Kitajima
,
S.
,
Inoue
,
T.
,
Koseki
,
H.
and
Saga
,
Y.
(
2000
).
Mesp2 initiates somite segmentation through the Notch signalling pathway
.
Nat. Genet.
25
,
390
-
396
.
Takayama
,
H.
,
La Rochelle
,
W. J.
,
Anver
,
M.
,
Bockman
,
D. E.
and
Merlino
,
G.
(
1996
).
Scatter factor/hepatocyte growth factor as a regulator of skeletal muscle and neural crest development
.
Proc. Natl. Acad. Sci. USA
93
,
5866
-
5871
.
Takebe
,
A.
,
Era
,
T.
,
Okada
,
M.
,
Martin Jakt
,
L.
,
Kuroda
,
Y.
and
Nishikawa
,
S.-I.
(
2006
).
Microarray analysis of PDGFR alpha+ populations in ES cell differentiation culture identifies genes involved in differentiation of mesoderm and mesenchyme including ARID3b that is essential for development of embryonic mesenchymal cells
.
Dev. Biol.
293
,
25
-
37
.
Takemoto
,
T.
,
Uchikawa
,
M.
,
Yoshida
,
M.
,
Bell
,
D. M.
,
Lovell-Badge
,
R.
,
Papaioannou
,
V. E.
and
Kondoh
,
H.
(
2011
).
Tbx6-dependent Sox2 regulation determines neural or mesodermal fate in axial stem cells
.
Nature
470
,
394
-
398
.
Tam
,
P. P. L.
and
Loebel
,
D. A. F.
(
2007
).
Gene function in mouse embryogenesis: get set for gastrulation
.
Nat. Rev. Genet.
8
,
368
-
381
.
Tapscott
,
S. J.
(
2005
).
The circuitry of a master switch: Myod and the regulation of skeletal muscle gene transcription
.
Development
132
,
2685
-
2695
.
Taylor
,
S. M.
and
Jones
,
P. A.
(
1979
).
Multiple new phenotypes induced in and 3T3 cells treated with 5-azacytidine
.
Cell
17
,
771
-
779
.
ten Berge
,
D.
,
Koole
,
W.
,
Fuerer
,
C.
,
Fish
,
M.
,
Eroglu
,
E.
and
Nusse
,
R.
(
2008
).
Wnt signaling mediates self-organization and axis formation in embryoid bodies
.
Cell Stem Cell
3
,
508
-
518
.
Tenin
,
G.
,
Wright
,
D.
,
Ferjentsik
,
Z.
,
Bone
,
R.
,
McGrew
,
M. J.
and
Maroto
,
M.
(
2010
).
The chick somitogenesis oscillator is arrested before all paraxial mesoderm is segmented into somites
.
BMC Dev. Biol.
10
,
24
.
Tierney
,
M. T.
,
Gromova
,
A.
,
Boscolo Sesillo
,
F.
,
Sala
,
D.
,
Spenlé
,
C.
,
Orend
,
G.
and
Sacco
,
A.
(
2016
).
Autonomous extracellular matrix remodeling controls a progressive adaptation in muscle stem cell regenerative capacity during development
.
Cell Rep.
14
,
1940
-
1952
.
Tonegawa
,
A.
,
Funayama
,
N.
,
Ueno
,
N.
and
Takahashi
,
Y.
(
1997
).
Mesodermal subdivision along the mediolateral axis in chicken controlled by different concentrations of BMP-4
.
Development
124
,
1975
-
1984
.
Tran
,
T.
,
Andersen
,
R.
,
Sherman
,
S. P.
and
Pyle
,
A. D.
(
2013
).
Insights into skeletal muscle development and applications in regenerative medicine
.
Int. Rev. Cell Mol. Biol.
300
,
51
-
83
.
Tremblay
,
M. R.
and
Carbonetto
,
S.
(
2006
).
An extracellular pathway for dystroglycan function in acetylcholine receptor aggregation and laminin deposition in skeletal myotubes
.
J. Biol. Chem.
281
,
13365
-
13373
.
Tremblay
,
J. P.
and
Skuk
,
D.
(
2008
).
Another new “super muscle stem cell” leaves unaddressed the real problems of cell therapy for duchenne muscular dystrophy
.
Mol. Ther.
16
,
1907
-
1909
.
Tweedie
,
S.
,
Morrison
,
K.
,
Charlton
,
J.
and
Edwards
,
Y. H.
(
1991
).
CAIII a marker for early myogenesis: analysis of expression in cultured myogenic cells
.
Somat. Cell Mol. Genet.
17
,
215
-
228
.
Tzouanacou
,
E.
,
Wegener
,
A.
,
Wymeersch
,
F. J.
,
Wilson
,
V.
and
Nicolas
,
J.-F.
(
2009
).
Redefining the progression of lineage segregations during mammalian embryogenesis by clonal analysis
.
Dev. Cell
17
,
365
-
376
.
Uezumi
,
A.
,
Nakatani
,
M.
,
Ikemoto-Uezumi
,
M.
,
Yamamoto
,
N.
,
Morita
,
M.
,
Yamaguchi
,
A.
,
Yamada
,
H.
,
Kasai
,
T.
,
Masuda
,
S.
,
Narita
,
A.
, et al. 
(
2016
).
Cell-surface protein profiling identifies distinctive markers of progenitor cells in human skeletal muscle
.
Stem Cell Rep.
7
,
263
-
278
.
van den Brink
,
S. C.
,
Baillie-Johnson
,
P.
,
Balayo
,
T.
,
Hadjantonakis
,
A.-K.
,
Nowotschin
,
S.
,
Turner
,
D. A.
and
Martinez Arias
,
A.
(
2014
).
Symmetry breaking, germ layer specification and axial organisation in aggregates of mouse embryonic stem cells
.
Development
141
,
4231
-
4242
.
van der Velden
,
J. L. J.
,
Langen
,
R. C.
,
Kelders
,
M. C.
,
Wouters
,
E. F.
,
Janssen-Heininger
,
Y. M.
and
Schols
,
A. M.
(
2006
).
Inhibition of glycogen synthase kinase-3beta activity is sufficient to stimulate myogenic differentiation
.
Am. J. Physiol.
290
,
C453
-
C462
.
van Deutekom
,
J. C.
,
Janson
,
A. A.
,
Ginjaar
,
I. B.
,
Frankhuizen
,
W. S.
,
Aartsma-Rus
,
A.
,
Bremmer-Bout
,
M.
,
den Dunnen
,
J. T.
,
Koop
,
K.
,
van der Kooi
,
A. J.
,
Goemans
,
N. M.
, et al. 
(
2007
).
Local dystrophin restoration with antisense oligonucleotide PRO051
.
N. Engl. J. Med.
357
,
2677
-
2686
.
Van Horn
,
R.
and
Crow
,
M. T.
(
1989
).
Fast myosin heavy chain expression during the early and late embryonic stages of chicken skeletal muscle development
.
Dev. Biol.
134
,
279
-
288
.
Vasyutina
,
E.
,
Lenhard
,
D. C.
and
Birchmeier
,
C.
(
2007
).
Notch function in myogenesis
.
Cell Cycle
6
,
1451
-
1454
.
Venuti
,
J. M.
,
Morris
,
J. H.
,
Vivian
,
J. L.
,
Olson
,
E. N.
and
Klein
,
W. H.
(
1995
).
Myogenin is required for late but not early aspects of myogenesis during mouse development
.
J. Cell Biol.
128
,
563
-
576
.
von Maltzahn
,
J.
,
Chang
,
N. C.
,
Bentzinger
,
C. F.
and
Rudnicki
,
M. A.
(
2012
).
Wnt signaling in myogenesis
.
Trends Cell Biol.
22
,
602
-
609
.
von Maltzahn
,
J.
,
Jones
,
A. E.
,
Parks
,
R. J.
and
Rudnicki
,
M. A.
(
2013
).
Pax7 is critical for the normal function of satellite cells in adult skeletal muscle
.
Proc. Natl. Acad. Sci. USA
110
,
16474
-
16479
.
Vyskočil
,
F.
and
Vrbová
,
G.
(
1993
).
Non-quantal release of acetylcholine affects polyneuronal innervation on developing rat muscle fibres
.
Eur. J. Neurosci.
5
,
1677
-
1683
.
Warmflash
,
A.
,
Sorre
,
B.
,
Etoc
,
F.
,
Siggia
,
E. D.
and
Brivanlou
,
A. H.
(
2014
).
A method to recapitulate early embryonic spatial patterning in human embryonic stem cells
.
Nat. Methods
11
,
847
-
854
.
Watanabe
,
T.
,
Sato
,
Y.
,
Saito
,
D.
,
Tadokoro
,
R.
and
Takahashi
,
Y.
(
2009
).
EphrinB2 coordinates the formation of a morphological boundary and cell epithelialization during somite segmentation
.
Proc. Natl. Acad. Sci. USA
106
,
7467
-
7472
.
Weintraub
,
H.
,
Tapscott
,
S. J.
,
Davis
,
R. L.
,
Thayer
,
M. J.
,
Adam
,
M. A.
,
Lassar
,
A. B.
and
Miller
,
A. D.
(
1989
).
Activation of muscle-specific genes in pigment, nerve, fat, liver, and fibroblast cell lines by forced expression of MyoD
.
Proc. Natl. Acad. Sci. USA
86
,
5434
-
5438
.
Weitzer
,
G.
,
Milner
,
D. J.
,
Kim
,
J. U.
,
Bradley
,
A.
and
Capetanaki
,
Y.
(
1995
).
Cytoskeletal control of myogenesis: a desmin null mutation blocks the myogenic pathway during embryonic stem cell differentiation
.
Dev. Biol.
172
,
422
-
439
.
White
,
R. B.
,
Biérinx
,
A.-S.
,
Gnocchi
,
V. F.
and
Zammit
,
P. S.
(
2010
).
Dynamics of muscle fibre growth during postnatal mouse development
.
BMC Dev. Biol.
10
,
21
.
Wilkinson
,
D. G.
,
Bhatt
,
S.
and
Herrmann
,
B. G.
(
1990
).
Expression pattern of the mouse T gene and its role in mesoderm formation
.
Nature
343
,
657
-
659
.
Williams
,
P. E.
and
Goldspink
,
G.
(
1971
).
Longitudinal growth of striated muscle fibres
.
J. Cell Sci.
9
,
751
-
767
.
Williams
,
B. A.
and
Ordahl
,
C. P.
(
1994
).
Pax-3 expression in segmental mesoderm marks early stages in myogenic cell specification
.
Development
120
,
785
-
796
.
Winnier
,
G.
,
Blessing
,
M.
,
Labosky
,
P. A.
and
Hogan
,
B. L.
(
1995
).
Bone morphogenetic protein-4 is required for mesoderm formation and patterning in the mouse
.
Genes Dev.
9
,
2105
-
2116
.
Wobus
,
A. M.
,
Guan
,
K.
and
Pich
,
U.
(
2001
).
In vitro differentiation of embryonic stem cells and analysis of cellular phenotypes
.
Methods Mol. Biol.
158
,
263
-
286
.
Wu
,
Z.
,
Woodring
,
P. J.
,
Bhakta
,
K. S.
,
Tamura
,
K.
,
Wen
,
F.
,
Feramisco
,
J. R.
,
Karin
,
M.
,
Wang
,
J. Y. J.
and
Puri
,
P. L.
(
2000
).
p38 and extracellular signal-regulated kinases regulate the myogenic program at multiple steps
.
Mol. Cell. Biol.
20
,
3951
-
3964
.
Wymeersch
,
F. J.
,
Huang
,
Y.
,
Blin
,
G.
,
Cambray
,
N.
,
Wilkie
,
R.
,
Wong
,
F. C.
and
Wilson
,
V.
(
2016
).
Position-dependent plasticity of distinct progenitor types in the primitive streak
.
Elife
5
,
e10042
.
Xu
,
C.
,
Tabebordbar
,
M.
,
Iovino
,
S.
,
Ciarlo
,
C.
,
Liu
,
J.
,
Castiglioni
,
A.
,
Price
,
E.
,
Liu
,
M.
,
Barton
,
E. R.
,
Kahn
,
C. R.
, et al. 
(
2013
).
A zebrafish embryo culture system defines factors that promote vertebrate myogenesis across species
.
Cell
155
,
909
-
921
.
Yamada
,
G.
,
Kioussi
,
C.
,
Schubert
,
F. R.
,
Eto
,
Y.
,
Chowdhury
,
K.
,
Pituello
,
F.
and
Gruss
,
P.
(
1994
).
Regulated expression of Brachyury(T), Nkx1.1 and Pax genes in embryoid bodies
.
Biochem. Biophys. Res. Commun.
199
,
552
-
563
.
Yamaguchi
,
T. P.
,
Harpal
,
K.
,
Henkemeyer
,
M.
and
Rossant
,
J.
(
1994
).
fgfr-1 is required for embryonic growth and mesodermal patterning during mouse gastrulation
.
Genes Dev.
8
,
3032
-
3044
.
Yamaguchi
,
T. P.
,
Takada
,
S.
,
Yoshikawa
,
Y.
,
Wu
,
N.
and
McMahon
,
A. P.
(
1999
).
T (Brachyury) is a direct target of Wnt3a during paraxial mesoderm specification
.
Genes Dev.
13
,
3185
-
3190
.
Yang
,
L.
,
Soonpaa
,
M. H.
,
Adler
,
E. D.
,
Roepke
,
T. K.
,
Kattman
,
S. J.
,
Kennedy
,
M.
,
Henckaerts
,
E.
,
Bonham
,
K.
,
Abbott
,
G. W.
,
Linden
,
R. M.
, et al. 
(
2008
).
Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population
.
Nature
453
,
524
-
528
.
Yasuno
,
T.
,
Osafune
,
K.
,
Sakurai
,
H.
,
Asaka
,
I.
,
Tanaka
,
A.
,
Yamaguchi
,
S.
,
Yamada
,
K.
,
Hitomi
,
H.
,
Arai
,
S.
,
Kurose
,
Y.
, et al. 
(
2014
).
Functional analysis of iPSC-derived myocytes from a patient with carnitine palmitoyltransferase II deficiency
.
Biochem. Biophys. Res. Commun.
448
,
175
-
181
.
Yokoyama
,
S.
,
Ito
,
Y.
,
Ueno-Kudoh
,
H.
,
Shimizu
,
H.
,
Uchibe
,
K.
,
Albini
,
S.
,
Mitsuoka
,
K.
,
Miyaki
,
S.
,
Kiso
,
M.
,
Nagai
,
A.
, et al. 
(
2009
).
A systems approach reveals that the myogenesis genome network is regulated by the transcriptional repressor RP58
.
Dev. Cell
17
,
836
-
848
.
Yoon
,
J. K.
and
Wold
,
B.
(
2000
).
The bHLH regulator pMesogenin1 is required for maturation and segmentation of paraxial mesoderm
.
Genes Dev.
14
,
3204
-
3214
.
Yoon
,
J. K.
,
Moon
,
R. T.
and
Wold
,
B.
(
2000
).
The bHLH class protein pMesogenin1 can specify paraxial mesoderm phenotypes
.
Dev. Biol.
222
,
376
-
391
.
Young
,
C. S.
,
Hicks
,
M. R.
,
Ermolova
,
N. V.
,
Nakano
,
H.
,
Jan
,
M.
,
Younesi
,
S.
,
Karumbayaram
,
S.
,
Kumagai-Cresse
,
C.
,
Wang
,
D.
,
Zack
,
J. A.
, et al. 
(
2016
).
A single CRISPR-Cas9 deletion strategy that targets the majority of DMD patients restores dystrophin function in hiPSC-derived muscle cells
.
Cell Stem Cell
18
,
533
-
540
.
Yusuf
,
F.
and
Brand-Saberi
,
B.
(
2006
).
The eventful somite: patterning, fate determination and cell division in the somite
.
Anat. Embryol.
211
Suppl. 1
,
21
-
30
.
Zammit
,
P. S.
,
Carvajal
,
J. J.
,
Golding
,
J. P.
,
Morgan
,
J. E.
,
Summerbell
,
D.
,
Zolnerciks
,
J.
,
Partridge
,
T. A.
,
Rigby
,
P. W. J.
and
Beauchamp
,
J. R.
(
2004
).
Myf5 expression in satellite cells and spindles in adult muscle is controlled by separate genetic elements
.
Dev. Biol.
273
,
454
-
465
.
Zetser
,
A.
,
Gredinger
,
E.
and
Bengal
,
E.
(
1999
).
p38 mitogen-activated protein kinase pathway promotes skeletal muscle differentiation. Participation of the Mef2c transcription factor
.
J. Biol. Chem.
274
,
5193
-
5200
.
Zhang
,
P.
,
Li
,
J.
,
Tan
,
Z.
,
Wang
,
C.
,
Liu
,
T.
,
Chen
,
L.
,
Yong
,
J.
,
Jiang
,
W.
,
Sun
,
X.
,
Du
,
L.
, et al. 
(
2008
).
Short-term BMP-4 treatment initiates mesoderm induction in human embryonic stem cells
.
Blood
111
,
1933
-
1941
.
Zheng
,
J. K.
,
Wang
,
Y.
,
Karandikar
,
A.
,
Wang
,
Q.
,
Gai
,
H.
,
Liu
,
A. L.
,
Peng
,
C.
and
Sheng
,
H. Z.
(
2006
).
Skeletal myogenesis by human embryonic stem cells
.
Cell Res.
16
,
713
-
722
.

Competing interests

O.P. and J.C. are co-founders and share-holders of Anagenesis Biotechnologies, a start-up company specialized in the production of muscle cells in vitro for cell therapy and drug screening.

Supplementary information