Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Keep Your Friends Close: Cell–Cell Contact and Skeletal Myogenesis

Robert S. Krauss, Giselle A. Joseph, and Aviva J. Goel

Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029 Correspondence: [email protected]

Development of skeletal muscle is a multistage process that includes lineage commitment of multipotent progenitor cells, differentiation and fusion of myoblasts into multinucleated myofibers, and maturation of myofibers into distinct types. Lineage-specific transcriptional regulation lies at the core of this process, but myogenesis is also regulated by extracellular cues. Some of these cues are initiated by direct cell–cell contact between muscle precursor cells themselves or between muscle precursors and cells of other lineages. Examples of the latter include interaction of migrating cells with multipotent muscle progenitor cells, muscle interstitial cells with myoblasts, and neurons with myofibers. Among the sig- naling factors involved are Notch ligands and receptors, cadherins, Ig superfamily members, and Ephrins and Eph receptors. In this article we describe recent progress in this area and highlight open questions raised by the findings.

keletal muscle is the most abundant tissue in based on expression of distinct myosin heavy Sthe vertebrate body and necessary for breath- chain (MyHC) isoforms and metabolic capabil- ing, metabolic homeostasis, and locomotion. ities (Schiaffino and Reggiani 2011; Talbot and Development of skeletal muscle occurs in a se- Maves 2016). quential, multistage process that includes spec- Myogenic specification and differentiation ification of multipotent progenitors to lineage- are coordinated by the myogenic basic helix– committed myoblasts, differentiation of myo- loop–helix (bHLH) transcription factors Myf5, blasts into multinucleated myofibers, and mat- MyoD, myogenin, and MRF4. Uncommitted uration of myofibers by innervation and bun- progenitor cells are specified to become line- dling into functional muscles (Biressi et al. age-committed myoblasts through the com- 2007; Comai and Tajbakhsh 2014; Tintignac bined actions of Myf5, MRF4, and MyoD, et al. 2015). Myoblast differentiation is itself a whereas differentiation of myoblasts is regulated complex process that involves both induction of by myogenin, MyoD, and MRF4 (Tapscott2005; the muscle-specific transcriptome and fusion of Biressi et al. 2007; Comai and Tajbakhsh 2014). myoblasts into an elongated syncytium. Simi- Expression of these factors, particularly MyoD, larly, maturation is multifaceted. Mature myo- in many nonmuscle cell types converts such cells fibers are classified as “slow” or “fast” types, to the skeletal muscle program, revealing their

Editors: Carien M. Niessen and Alpha S. Yap Additional Perspectives on Cell–Cell Junctions available at www.cshperspectives.org Copyright # 2017 Cold Spring Harbor Laboratory Press; all rights reserved Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298

1 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al.

ability to drive tissue-specific transcription and dermomyotome, which has epithelial character cell differentiation (Tapscott 2005; Fong and and gives rise to skeletal muscle and dermis, and Tapscott2013). The ability of MyoD to function ventrally into the sclerotome, which has mesen- in this manner occurs in conjunction with non- chymal character and gives rise to the bones and muscle-specific factors such as its heterodimeric cartilage of the axial skeleton (Brand-Saberi and partners, the E ; members of the Mef2 Christ 2000). Some dermomyotomal progenitor family of transcription factors; transcriptional cells undergo an epithelial-mesenchymal transi- coactivators; and chromatin remodeling factors tion (EMT), become committed to the skeletal (Tapscott 2005; Biressi et al. 2007; Sartorelli and muscle lineage, and migrate between the dermo- Juan 2011; Fong and Tapscott 2013; Comai and myotome and sclerotome to form the myotome, Tajbakhsh 2014). These and additional tran- a set of differentiated embryonic muscle cells. scription factors orchestrate development of Subsequent embryonic, fetal, and postnatal the skeletal muscle lineage. stages of myogenesis involve additional muscle Although the importance of lineage-specific progenitorsthat originally migrate from the der- transcriptional regulation in skeletal myogene- momyotome and ultimately establish the trunk sis has long been appreciated, myogenesis is also and limb musculature (Biressi et al. 2007; Comai regulated by extracellular cues that initiate in- and Tajbakhsh 2014). tracellular signaling (Guasconi and Puri 2009). Signals from the adjacent dorsal neural tube Some of these extracellular cues are secreted fac- and surface ectoderm play important roles in tors, such as fibroblast growth factor (Fgf ), Wnt maturation of the dorsal (Munsterberg family ligands, and (Shh). A and Lassar 1995; Stern and Hauschka 1995). growing body of evidence indicates that signal- These tissues secrete Wnt1 and Wnt3a, which ing initiated by direct cell–cell contact also pro- signal via the “canonical” pathway to stimu- vides key regulatory information during devel- late b-catenin/T-cell factor (TCF)–dependent opment of the skeletal muscle lineage. In adult transcription. These Wnt ligands promote myo- skeletal muscle, stable cell–cell junctions (ex- genesis of explanted epithelial in vitro emplified by epithelial, cadherin-based adhe- (Munsterberg et al. 1995; Stern et al. 1995; Taj- rens junctions) do not appear to exist between bakhsh et al. 1998; Borello et al. 2006). Further- myofibers, which are the unit cells of this tissue. more, an enhancer that regulates Myf5 expres- (Cell–cell junctions do exist between myofibers sion in the dorsal somite harbors TCF binding and skeletal muscle stem cells known as satellite sites that are critical for Wnt responsiveness and cells; this is discussed below.) Nevertheless, enhancer function (Borello et al. 2006) (N.B.: cell–cell adhesion, and signaling that derives Myf5 is the first myogenic bHLH factor ex- from cell–cell contact, occurs between various pressed during development, and its role is muscle precursor cells and between these cells solely in lineage determination.) These results and nonmuscle cell types during myogenesis. argued that canonical Wnt ligands, along with Furthermore, such interactions are important Shh and soluble bone morphogenetic for muscle development. In this review, we dis- (BMP) inhibitors (Munsterberg et al. 1995; Re- cuss the roles and mechanisms whereby cell– shef et al. 1998; Borycki et al. 1999), induce cell contact regulates skeletal myogenesis. muscle lineage specification of dermomyotomal progenitor cells via b-catenin/TCF-dependent expression of Myf5. PROGENITOR CELL COMMITMENT TO THE b-catenin is well known for having two dis- SKELETAL MUSCLE CELL LINEAGE tinct cellular functions: (1) as a transcriptional In vertebrates, skeletal muscles of the trunk and regulator that associates with the DNA-binding limbs develop from somites, transiently existing TCF proteins in a Wnt signaling-responsive blocks of columnar epithelial cells that form in manner; and (2) as an adaptor protein that an anterior-to-posterior manner from paraxial binds to the cytoplasmic tail of cadherins and mesoderm. Somites mature dorsally into the promotes linkage to the cytoskeleton (van

2 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Cell–Cell Contact and Skeletal Myogenesis

Amerongen and Nusse 2009; Niessen et al. of the dermomyotome express Notch receptors 2011). Cells of the epithelial dermomyotome (Rios et al. 2011). Transient cell–cell contact express the cell– N-cad- between migrating, Delta1þ neural crest cells herin with an apical localization; its adhesive and Notchþ muscle progenitors in the dermo- junction partner, b-catenin, is localized there myotome leads to Notch signaling in the latter. as well (Cinnamon et al. 2006). Studies with This is, in turn, required for Myf5 induction and chick embryos suggest that changes in N-cad- adoption of the skeletal muscle cell fate (Rios herin expression and subcellular localization et al. 2011). This “kiss and run” mode of cell– play important roles in specification of multi- cell contact-dependent signaling strongly sug- potent dermomyotomal cells to the muscle lin- gests that the concurrent timing of neural crest eage. As these cells divide, N-cadherin is asym- cell migration and myotome formation is mech- metrically localized in cells that are fated to anistically linked. become skeletal muscle cells of the myotome; Canonical Notch signaling involves cleavage conversely, daughter cells that lack N-cadherin of its intracellular domain (NICD), which take on features of early dermal cells (Cinna- translocates to the nucleus and binds RBP-J to mon et al. 2006). Consistent with these obser- stimulate pathway-responsive expression vations, electroporation of wild-type or domi- (Fortini 2009). In exploring how Notch signal- nant negative mutant forms of N-cadherin into ing promotes Myf5 expression in dermomyoto- dermomyotomal cells promoted or diminished mal progenitor cells, it was found that NICD contribution of such cells to the myotome com- inhibited GSK3b activity independently of partment, respectively (Cinnamon et al. 2006). NICD’s transcriptional role in the nucleus During EMTof dermomyotomal cells that were (Sieiro et al. 2016). This, in turn, led to stabili- fated to become muscle, the apical distribution zation of Snai1, which promoted EMT of these of N-cadherin and b-catenin was disrupted, cells. Unexpectedly, NICD also drove TCF-de- consistent with a transient loss of adhesion dur- pendent reporter activity and Myf5 expression, ing migration and acquisition of the myogenic again independently of its canonical transcrip- fate (Cinnamon et al. 2006). tional role. Notch-stimulated, TCF-dependent Based on the results described above, the reporter activity and Myf5 expression were in- adhesive and transcriptional “pools” of b-cate- dependent of Wnt signaling and appeared to nin in dermomyotomal muscle progenitor cells result from liberation of a membrane source might be viewed as distinct; that is, they are of b-catenin (Sieiro et al. 2016). As described involved in either cell–cell adhesion or Wnt sig- above, EMT of dermomyotomal cells was ac- nal-dependent activation of Myf5 expression, companied by loss of N-cadherin and b-catenin but not both. However, nuclear b-catenin sig- from an apical junction location. It may be that naling from adhesion complexes can occur the reduction in cell–cell adhesion that allows (McCrea et al. 2015), and recent results from muscle progenitor cells to migrate out of the the Marcelle group suggest that this process dermomyotome (and which is promoted by may be involved in commitment of dermomyo- the EMT program) allows b-catenin to reloc- tomal progenitor cells to the muscle lineage. alize from the plasma membrane to the nucleus These investigators have studied the role of neu- and specify the myogenic cell fate via TCF-de- ral crest cells in promoting somitic myogenesis. pendent induction of Myf5 (Fig. 1). The mech- Neural crest cells migrate from the dorsal neural anism by which b-catenin is relocalized is not tube to populate various structures of the em- clear, but expression of a phosphorylation-de- bryo. Some of these cells migrate past the dor- ficient b-catenin mutant that is poorly mobi- somedial aspect of somites, a portion of the lized from junctional complexes was ineffective dermomyotome known to give rise to myoblasts in Myf5 induction. Therefore, b-catenin may be early in myotome formation. Neural crest cells phosphorylated during EMT to trigger alter- in close proximity to such dermomyotomal ation of its subcellular localization. Taken to- cells express the Notch ligand, Delta1, and cells gether, these results uncovered a novel mecha-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 3 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al. Notch1 Notch1

N-Cadherin N-Cadherin β-Catenin β-Catenin

Dermomyotomal cell

Myf5 Myf5 TCF TCF

Neural crest cell migration

Neural crest cell Delta1 Notch1 Notch1 GSK- NICD N-Cadherin 3β β-Catenin Snai1

Dermomyotomal cell EMT

N-Cadherin

β-Catenin Myf5 Myf5 TCF TCF

Figure 1. Delta/Notch signaling from migrating neural crest cells to dermomyotomal muscle progenitor cells stimulates translocation of b-catenin from adhesive junctions to the nucleus to promote Myf5 expression and myogenic commitment. Before engagement with a neural crest cell (top), dermomyotomal cells express N- cadherin and b-catenin in an apical location. The Myf5 gene is not expressed and the cells are multipotent. When neural crest cells migrate across the surface of the dorsal somite, Delta1 on their surface binds to Notch1 on the surface of dermomyotomal cells (bottom). This leads to cleavage of the Notch intracellular domain (NICD), which acts in the cytoplasm to inhibit GSK-3b, in turn stabilizing Snai1. Snai1 induces an epithelial–mesen- chymal transition (EMT). The dashed line between Snai1 and EMT indicates that this is a complex event; the multiple dashed lines emanating from EMT indicate that EMT induces multiple profound changes to the cell. Among these, N-cadherin no longer localizes to an apical site and b-catenin translocates from an adhesive junction to the nucleus where it associates with TCF to induce expression of Myf5. Expression of Myf5 commits progenitor cells to the skeletal muscle lineage. Alterations in N-cadherin localization are also associated with asymmetric cell divisions (not shown, but see text for further details).

4 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Cell–Cell Contact and Skeletal Myogenesis

nism whereby contact between migrating neural ly likely that changes in gene expression and cell crest cells and dermomyotomal progenitor cells morphology are coordinated during differenti- triggers a nontranscriptional Notch signaling ation to produce this very specialized cell type. function. This noncanonical Notch signal pro- The mechanisms by which this coordination motes a change in cell adhesion and redirection occurs are not fully clear, but cell–cell contact of b-catenin’s function from the membrane to between myoblasts is likely to play a role. the nucleus, thereby stimulating myogenic cell Myogenic differentiation is amenable to fate acquisition (Sieiro et al. 2016). analysis in vitro, as both primary mouse myo- These findings raise numerous questions. If blasts and myoblast cell lines are easily obtained Notch-dependent b-catenin/TCF activity is key and studied. Myoblasts are generally allowed to to myogenic specification, what is the role of proliferate in mitogen-rich medium until near canonical Wnt signaling—which can be ob- confluence, at which point they are switched to served and is distinguishable from the Notch/ mitogen-poor medium to stimulate differentia- b-catenin activity—in this process? What kinas- tion. Although growth factor deprivation is a esmaybeinvolvedinb-cateninphosphorylation primary differentiation signal in these cultures, andhowistheiractivity regulated?IsN-cadherin high cell density itself is strongly promyogenic. It the b-catenin partner that releases the “pool” of was subsequently showed that cell–cell contact b-catenin in this process or are other cadherins and adhesion regulates promyogenic signal involved? We note that P-cadherin, and perhaps transduction pathways. Multiple mechanisms additional cadherins, is also expressed in the ep- underlie this effect, and we have reviewed the ithelial dermomyotome (Thuault et al. 2013). fieldpreviously(Kraussetal.2005;Krauss2010). How do these new findings relate to the earlier A central factor in cell contact-dependent results on N-cadherin in myogenic specification signaling in myoblasts is the multifunctional via asymmetric cell divisions (Cinnamon et al. cell-surface coreceptor, Cdon (also called Cdo). 2006)? Canonical Notch signaling plays roles Cdon has multiple immunoglobulin (Ig) and later in myogenesis, including inhibition of FNIII repeats in its extracellular region, a single myoblast differentiation such that appropriate transmembrane region, and a long cytoplasmic numbers of myoblasts are produced (Schuster- tail that does not resemble other proteins and Gossler et al. 2007; Vasyutina et al. 2007); how, lacks known catalytic activity (Kang et al. 1997, if at all, are these two Notch functions related? 1998). Cdon binds in cis to several different Finally, these findings are compelling, but have adhesion molecules and signaling receptors, been performed only with electroporated chick and it can serve in ligand binding or signal prop- embryos. It will be interesting and important to agation, depending on the receptor complex in addressthe pathway withconditionalmutants in question. One major binding partner for Cdon mouse embryos also. in skeletal myoblasts is N-cadherin (Kang et al. 2003; Lu and Krauss 2010). Trans-ligation of N-cadherin on adjacent myoblasts stimulates MYOBLAST DIFFERENTIATION assembly of signaling complexes on the Cdon AND FUSION intracellular region (Fig. 2). These include direct Differentiation of skeletal myoblasts into myo- association with (1) Bnip-2, which binds and fibers is a multistep process involving withdraw- brings to the complex the small GTPase, al from the cell cycle, adoption of a muscle-spe- Cdc42 (Kang et al. 2008); and (2) JLP,a scaffold cific transcriptional program, cell elongation, protein for the p38a/b mitogen-associated and cell–cell fusion (Biressi et al. 2007; Comai protein (MAP) kinase pathway (Takaesu et al. and Tajbakhsh 2014). Cell–cell contact is, of 2006). Formation of such complexes stimulates course, a prerequisite for fusion of myoblasts Cdc42-dependent activation of p38a/b (here- with each other or with nascent myofibers, but after, p38) via a cascade of kinases that includes the myoblast differentiation program as a whole Pak1/2, Tak1 and/or Ask1, and MKK3/6(Wu is regulated by direct cell–cell contact. It is high- et al. 2000; Takaesu et al. 2006; Tran et al. 2012;

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 5 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al.

Intracellular

Cdon Extracellular space

N-Cadherin Kir2.1 β-Catenin Bnip-2 J Cdc42 Pak1/2 L Intracellular P TAK1; ASK1 MKK3/6 p38

Kir2.1

E47 MEF2 BAF60c KSRP EZH2

MyoD-dependent, muscle-specific gene expression

Figure 2. N-cadherin ligation between myoblasts stimulates Cdon-dependent activation of p38, which promotes myogenic differentiation. N-cadherin ligation occurs on contact and adhesion of myoblasts and this, in turn, leads to assembly of signaling complexes on the intracellular region of Cdon. The scaffold protein Bnip-2 associates with both Cdon and Cdc42. Activated Cdc42 signals via Pak1/2 kinases to stimulate a p38 MAP kinase cascade bound to the Cdon-associated scaffold protein JLP.Activated p38 phosphorylates substrates that promote MyoD-dependent, muscle-specific gene expression to drive differentiation; among these substrates are E47, MEF2, BAF60c, KSRP, and EZH2. p38 activity also promotes cell-surface localization of the potassium channel, Kir2.1, which associates with Cdon. The dashed lines indicate that the p38 substrate involved in trafficking of Kir2.1 is not identified, and it is not clear whether the association between Cdon and Kir2.1 is a direct one. The dashed circle indicates that Kir2.1 is likely in a vesicular location before p38 activation, but the specific trafficking events that lead to cell-surface localization are not known. See text for further details. MAP, Mitogen-associated protein.

Joseph and Krauss, in prep). Activated p38, in MEF2 proteins; the SWI-SNF chromatin re- turn, promotes myogenic differentiation modeling complex subunit BAF60c; the RNA through phosphorylation of proteins that stim- decay-promoting factor KSRP; and the poly- ulate the muscle-specific transcriptional activity comb repressive complex 2 catalytic subunit, of MyoD. Among these promyogenic p38 sub- EZH2 (Fig. 2) (Wu et al. 2000; Simone et al. strates are the MyoD dimeric partner, E47; 2004; Briata et al. 2005; de Angelis et al. 2005;

6 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Cell–Cell Contact and Skeletal Myogenesis

Lluis et al. 2005; Rampalli et al. 2007; Serra et al. but not to soluble netrin, implying its role is as a 2007; Palacios et al. 2010). coreceptor (Kang et al. 2004), but Cdon’s mech- Kang and colleagues recently showed that, anistic function in netrin signaling is less clear in addition to its transcriptional regulatory here than with cadherin-based signaling. Cdon function, Cdon-dependent p38 activation also also serves to regulate the Hedgehog and Wnt promoted cell-surface expression of the inward- pathways (Zhang et al. 2006; Allen et al. 2011; ly rectifying potassium channel, Kir2.1 (Leem Jeong et al. 2014). However, these are less likely et al. 2016). Furthermore, Cdon and Kir2.1 to be cell–cell contact-dependent events, and formed complexes with each other at the cell Cdon’s specific role in the ability of these path- surface. Importantly, plasma membrane hyper- ways to influence muscle development is not polarization mediated by Kir2.1 promoted clear. Mice lacking Cdon displayed delayed skel- myoblast differentiation (Fischer-Lougheed etal muscle development, and primary myo- et al. 2001; Konig et al. 2004). Cdon2/2 myo- blasts from such mice differentiated defectively blasts displayed both defective differentiation in vitro (Cole et al. 2004). The phenotype asso- and defective Kir2.1 activity, both of which ciated with loss of Cdon may arise from a com- could be rescued by expression of a constitutive- bination of suboptimal signaling via several of ly active form of the p38 upstream activating the pathways it regulates. kinase, MKK6 (Leem et al. 2016). Cdon expres- Myoblast fusion is fundamental to produc- sion is itself up-regulated by MyoD (Cole et al. tion of myofibers and obviously requires cell– 2004). Cell–cell adhesion between myoblasts, cell contact and adhesion. Led by genetic therefore, helps stimulate a complex feedback screens in Drosophila and followed by studies network of signaling and transcriptional regu- in zebrafish and mice, great progress has been lation that reinforces both cell surface and nu- made in understanding mechanisms of myo- clear drivers of myoblast differentiation (Fig. 2). blast fusion. A recent model proposes that three Cdon has multiple additional roles that in- steps underlie myoblast fusion: (1) cell recogni- fluence skeletal myogenesis. Cdon interacts in tion and adhesion, (2) enhancement of cell cis with the netrin receptor, neogenin (Kang proximity via F-actin-propelled membrane et al. 2004). Like Cdon, neogenin is an Ig and protrusions by one fusion partner cell and my- FNIII repeat–containing receptor. Myoblasts osin II-dependent cortical tension in the other, produce both netrin-3 ligand and neogenin, and (3) destabilization of the two apposed plas- suggesting that they function in an autocrine ma membrane lipid bilayers and formation of a manner (Kang et al. 2004). Although netrins fusion pore (Kim et al. 2015). Many intracellu- are secreted molecules, in myoblast cultures lar components that mediate steps 2 and 3 have they are associated with cell membranes and/ been identified in Drosophila and, where tested, or the extracellular matrix, indicating that they appear to be evolutionarily conserved in zebra- signal via short range in these cells, likely con- fish and mice, and cultured cells. In contrast, a tributing to the promyogenic effects of cell–cell clear picture of the factors that mediate cell rec- contact. Stimulation of primary or C2C12 myo- ognition and adhesion in these different models blasts with netrin led to activation of focal has not yet emerged. Comprehensive reviews adhesion kinase (FAK) and the extracellular sig- have recently been published on myoblast fu- nal–regulated kinase (ERK) MAP kinase (Kang sion (Abmayr and Pavlath 2012; Hindi et al. et al. 2004; Bae et al. 2009). ERK, in turn, phos- 2013; Kim et al. 2015), and for the purposes phorylated the calcium sensor Stim1, which of this review, we will focus on open questions promoted activation of the calcium-sensitive, pertaining to the cell–cell adhesion molecules promyogenic transcription factor, NFATc3 involved in this process. (Lee et al. 2012). These netrin-initiated signal- In the Drosophila embryo, fusion takes place ing events occurred in a manner that is depen- between two types of myoblasts: muscle founder dent on both neogenin and Cdon (Kang et al. cells and fusion competent myoblasts. The for- 2004; Bae et al. 2009). Cdon binds to neogenin, mer express the related adhesion receptors

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 7 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al.

Dumbfounded and Roughest, whereas the latter there is little genetic evidence to support a crit- express the related counter-receptors, Sticks- ical role of any single cadherin in this process and-stones, and Hibris. All of these are Ig super- (Charlton et al. 1997; Radice et al. 1997; Holl- family receptors (Kim et al. 2015). Engagement nagel et al. 2002). One possibility is that the of these cell adhesion molecules triggers signal- multiple cadherins that are expressed in myo- ing events, which ultimately promote steps 2 and blasts may have overlapping or compensatory 3 above. The cell–cell contact points at which functions (Charlton et al. 1997; Radice et al. adhesion molecules and actin-rich protrusions 1997; Hollnagel et al. 2002; Marthiens et al. are localized are complex cell–cell junctions and 2002). Consistent with this notion is that forced have been likened to synapses (Sens et al. 2010; expression of the intracellular region of E-cad- Duan et al. 2012; Kim et al. 2015). Many intra- herin in C2C12 myoblasts blocked both fusion cellular signaling proteins and actin regulatory into myotubes and cell-surface localization of factors that are required for myoblast fusion in endogenous N- and M-cadherin (note E-cad- Drosophila are also required for this process in herin is not expressed endogenously in these vertebrates (Krauss 2007; Moore et al. 2007; Sri- cells, but its intracellular region is similar to nivas et al. 2007; Laurin et al. 2008; Vasyutina that of the other cadherins) (Ozawa 2015). We et al. 2009; Gruenbaum-Cohen et al. 2012). have recently constructed mice that lack N- and However, the cell adhesion molecules that are M-cadherin in the muscle lineage. Although critical for this process in zebrafish and mice myoblast fusion was not obviously perturbed may be different from those in Drosophila, and in these animals, primary myoblasts isolated all the critical factors are probably not yet in from them had a specific fusion defect in vitro sight. In zebrafish, a Dumbfounded ortholog (AJ Goel and RS Krauss, in prep). This suggests (Kirrel3l) and a Sticks-and-Stones ortholog that multiple cadherins may indeed display re- (Nephrin) have been assessed for roles in myo- dundant functions in myoblast fusion, but there blast fusion via -mediated knock- may be additional redundancy or requirements down. Kirrel3l morphants had a strong fusion in vivo than in vitro. defect, suggesting a potentially conserved func- It is important to note that although Kirrel, tion, but they also had myofiber attachment de- Nephrin, Jam, and cadherin family proteins are fects (Srinivas et al. 2007). Nephrin morphants sufficient to promote cell–cell adhesion, they had shorter, disorganized myofibers but did not are not sufficient to drive cell–cell fusion (Mar- display overt loss of myoblast fusion (Sohn et tı`n-Padura et al. 1998; Galletta et al. 2004; Shi- al. 2009). Morpholino-dependent knockdown lagardi et al. 2013). Furthermore, their expres- was, however, inefficient and it may be that a sion is not restricted to the skeletal muscle fusion defect would require a greater level of lineage or even to other fusogenic cell types. depletion. In contrast, JAM-B and -C, two dif- The identity of a true fusogen in myoblast ferent Ig superfamilycounter-receptors,were es- fusion is, therefore, of very high interest. One sential for myoblast fusion in zebrafish (Powell possibility here is the recently discovered myo- and Wright 2011). Studies with mice carrying maker, a cell-surface protein with seven mem- individual mutations in the members of the Kir- brane-spanning regions (Millay et al. 2013, rel, Nephrin, and JAM families have not been 2016). Myomaker (also called TMEM8c) is es- studied specifically for muscle development or sential for myoblast fusion in both mice and myoblast fusion, but most mutants survive long zebrafish (Millay et al. 2013, 2014; Landemaine enough that it is not possible for a complete et al. 2014). It is expressed exclusively in the block to myoblast fusion to have occurred in skeletal muscle lineage and at the times during these animals (Donoviel et al. 2001; Putaala muscle development and regeneration when fu- et al. 2001; Gliki et al. 2004; Sakaguchi et al. sion is occurring. Significantly, ectopic expres- 2006; Prince et al. 2013; Yesildag et al. 2015). sion of myomaker in fibroblasts allows them to Cadherins have been suggested to play a role fuse with myoblasts, an activity not seen previ- as adhesion molecules in myoblast fusion, but ously with any other protein (Millay et al. 2013,

8 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Cell–Cell Contact and Skeletal Myogenesis

2016; Landemaine et al. 2014). Whether myo- and 2B. Slow-twitch and Type 2A fibers use ox- maker is the long sought direct fusogen, or idative metabolism. As their name implies, somehow regulates the process in another fun- slow-twitch fibers are the slowest contracting damental way, is not yet clear. However, under- and fatiguing of all the myofiber types, with standing its mechanism of action, how it might 2A fibers being the slowest contracting and fa- interact with adhesion molecules, and what cell- tiguing of the fast-twitch fibers. Type 2X and 2B surface junctional complexes it is a component fibers use glycolytic metabolism, and they con- of are important questions. tract and fatigue faster than do 2A fibers (or, Finally, a recent finding has suggested that naturally, than slow-twitch fibers). cell–cell contact and signaling between myo- Fiber type identity begins to be specified blasts and a nonmyogenic, muscle interstitial during embryonic development. However, late cell stimulates neonatal muscle growth in mice in prenatal development and continuing into by promoting myoblast migration to fusion the postnatal period, interactions between my- sites on myofibers (Gu et al. 2016). Myofiber ofibers and their innervating motor neurons formation is thought to be largely complete by play a key role in stable fiber-type specification. birth, but fibers continue to grow during the Newly formed myofibers can contact axons early postnatal period via fusion of myoblasts from many different motor neurons, but over at the tips of fibers. Specific, muscle-resident time all neuromuscular synapses but one, per cells that expressed the surface marker NG2 fiber, are lost. Mature motor neurons innervate contacted myoblasts to promote their migration many individual fibers, and together these are to these sites. NG2þ cells also had high NF-kB called motor units. Fiber type identity, includ- activity, which directly regulated expression of ing expression of specific isoforms of MyHC, is ephrin-A5. Signaling by ephrins and their Eph plastic until myofibers are innervated by a single receptors is a widely used mechanism of direct- motor neuron and organized into a motor unit ed cell motility and migration (Lisabeth et al. (Sanes and Lichtman 1999). Motor neurons 2013). Genetic removal of the p65 subunit of themselves are also categorized into slow versus NF-kB or ephrin-A5 from NG2þ cells showed fast-resistant versus fast-fatigable, based on, that these factors were important for the ability among other properties, their firing rate. With- of NG2þ cells to promote myoblast motility in in a motor unit, the neuronal firing rate helps vitro and for proper myoblast localization and specify fiber type. These observations raise the fusion in postnatal myofiber growth (Gu et al. critical question of how motor neurons of the 2016). As seen with neural crest cells and so- appropriate type interact stably with fibers that mitic muscle progenitors, these intriguing re- already possess some degree of fiber-type iden- sults provide another example of how cell con- tity. Recent work by Cornelison and colleagues tact–mediated signaling between nonmuscle implicates ephrins and their cognate Eph recep- cell types and muscle precursors regulates spe- tors in this process (Stark et al. 2015). cific events in muscle development. Of eight ephrins screened for expression in adult mouse muscle, only ephrin-A3 was ex- pressed in a nonuniform pattern (Stark et al. MUSCLE FIBER-TYPE PATTERNING 2015). Costaining for the various types of Skeletal myofibers are classified into different MyHC revealed that ephrin-A3 was expressed types by a variety of criteria, including expres- exclusively on Type 1 (slow) fibers and that all sion of specific isoforms of MyHC, use of gly- such fibers were positive for ephrin-A3 (Stark colytic versus oxidative metabolism, and speed et al. 2015). Ephrin-A3 expression on such fi- of contraction (i.e., twitch) and fatigue (Schiaf- bers initiated after expression of MyHC-I (the fino and Reggiani 2011; Talbot and Maves isoform expressed by slow fibers), indicating 2016). A broad classification scheme separates that it is a part of the program that characterizes slow-twitch fibers, called Type 1, from fast- the early, cell-autonomous slow fiber program. twitch fibers, which encompass Types 2A, 2X, To assess a role for ephrin-A3 in fiber-type pat-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 9 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al.

terning, a series of genetic experiments were these fibers (with fast axons outnumbering performed. The number of MyHC-Iþ fibers slow ones), leading to polyinnervation. During was not different between control and ephrin- the 2- to 4-week postnatal period, MyHC-Iþ/ A3-null mice through the first two weeks of ephrin-A3þ fibers become stably monoinner- postnatal life. However, between P14 and adult- vated only by neurons whose Schwann cells hood, the mutant mice underwent a striking lack EphA8; in contrast, neuromuscular junc- diminution in numbers of slow fibers in most tions that harbor EphA8þ Schwann cells are hind limb muscles, without a change in overall found only on fast fibers, which lack ephrin- fiber number (Stark et al. 2015). Taken together, A3 (Stark et al. 2015). It is logical to propose, these results suggested that ephrin-A3 is dispen- therefore, that cell contact-based repulsion be- sable for early specification of the slow fiber tween MyHC-Iþ/ephrin-A3þ fibers and EphA8þ type, but is required for the process whereby presynaptic termini promotes stable monoin- fiber type identity is solidified and maintained nervation of slow fibers byonly EphA82 synaps- by motor neuron engagement. To further test es. In contrast, ephrin-A32 (fast) fibers can be this notion, ephrin-A3 was ectopically ex- monoinnervated by EphA8þ synapses. This pressed via in vivo plasmid electroporation of cell–cell contact-based mechanism can explain the anterior region of the adult Tibialis anterior at least some of the final reciprocal patterns of muscle, which normally contains no slow fibers. myofiber types and their associated motor neu- This was followed by a sciatic nerve crush de- rons, and the overall pattern of slow versus fast nervation protocol, which stimulates reinnerva- fiber types. tion by endogenous repair processes. Electropo- ration of a control plasmid was without effect, SATELLITE CELL–MYOFIBER INTERACTIONS irrespective of whether denervation had oc- curred, and expression of ephrin-A3 in myofib- Adherens junction-like structures are not ers was without effect in the absence of dener- known to exist between skeletal myofibers. (It vation; however, ephrin-A3 expression followed is worth noting that “cardiomyocytes” do have by denervation resulted in production of abun- such structures (Vite and Radice 2014.) How- dant slow fibers in the electroporated area. ever, adult myofibers have stable cell–cell junc- These data are consistent with the conclusion tions in two notable cases. One is the neuro- that ephrin-A3 plays a role in motor neuron– muscular junction. This is a complex synapse dependent fiber-type specification and pattern- essential for nervous control of muscle contrac- ing, both during normal muscle maturation tion. Development and maintenance of the and reinnervation after injury. neuromuscular junction is beyond the scope As Ephrins bind their cognate Eph receptors of this review, which is focused on myogenesis, during cell–cell interactions to provide contact- and readers are directed to a review on this topic based repulsion signals (Lisabeth et al. 2013), (Tintignac et al. 2015). The second is the junc- these investigators searched for Eph receptors tion that exists between myofibers and their as- with a reciprocal expression pattern to ephrin- sociated satellite cells. Satellite cells are adult A3. EphA8 was found to be expressed at the skeletal muscle stem cells located between the neuromuscular junctions of fast, but not slow, myofiber and its surrounding basal lamina, and fibers (Stark et al. 2015). Interestingly, the cell they are the source of skeletal muscle’s remark- type on which synaptic EphA8 is expressed is a able regenerative properties (Brack and Rando nonmyelinating glial cell present at neuromus- 2012; Doles and Olwin 2015; Dumont et al. cular junctions called terminal Schwann cells. 2015). Unlike tissues such as the epidermis or Taken together, these results lead to the follow- hematopoietic system, myofibers are not re- ing model: After early specification during plenished homeostatically, and as such, satellite embryonic development, MyHC-Iþ slow fibers cells do not play a major role in maintenance of express ephrin-A3. Many motor axons of both adult skeletal muscle (Fry et al. 2015; Keefe et al. slow and fast subtypes can make contact with 2015). Therefore, satellite cells exist in a largely

10 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Cell–Cell Contact and Skeletal Myogenesis

quiescent state in adult mice. However, on mus- et al. 2011; Bjornson et al. 2012; Chakkalakal cle injury they are activated to proliferate and et al. 2012; Cheung et al. 2012; Mourikis et al. produce the myoblasts that will ultimately dif- 2012; Gopinath et al. 2014; Dumont et al. 2015). ferentiate to form new myofibers; they also self- In 1990, Bischoff (1990) reported that myofib- renew to replenish the muscle stem cell com- ers themselves are an important component of partment (Brack and Rando 2012; Doles and the quiescence-inducing niche. These results in- Olwin 2015; Dumont et al. 2015). dicated that direct cell–cell interactions be- Within their niche, satellite cells display po- tween myofibers and satellite cells were likely larized adhesive interactions (Fig. 3). On their to regulate this key property of the latter. basal side, they express integrins (e.g., integrin The molecular mechanisms that underlie a7b1) that bind laminins present in the basal the ability of myofibers to promote satellite cell lamina that enwraps each individual myofiber. quiescence are poorly understood. However, On their apical side, satellite cells are directly Notch signaling is critical to the maintenance apposed to the plasma membrane of their asso- of satellite cell quiescence (Bjornson et al. ciated myofiber. M-cadherin, a muscle-specific 2012; Mourikis et al. 2012). A transgenic Notch classical cadherin, is localized exclusively at the reporter construct showed activity in quiescent site of contact between the satellite cell and the satellite cells in vivo, and freshly isolated satellite fiber. Quiescence is the hallmark property of cells expressed high levels of direct Notch path- satellite cells, and it is widely believed that the way target such as Hes1, Hey1, and HeyL. satellite cell niche plays a role in its maintenance. Expression of these genes was sharply di- This is important as in instances in which qui- minished when satellite cells were activated in escence is inappropriately broken, satellite cell response to injury in vivo or cultured under number and/or function is diminished (Fukada activating conditions in vitro. RBP-J is a DNA-

Itgα7β1 Basal lamina

Satellite cell Notch

Cadherins Delta

Myofiber

Figure 3. Quiescent satellite cells are polarized and form cell–cell junctions with myofibers. Satellite cells reside between myofibers and their surrounding basal lamina. Cadherins and Notch receptors are localized to the apical membrane of satellite cells and bind to cadherins and Notch ligands (e.g., Delta) on the myofiber membrane. Integrin a7b1 (Itga7b1) is localized to the basal membrane of satellite cells and binds laminin present in the basal lamina.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 11 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al.

binding factor required for Notch-mediated seems well suited to play a role in fiber-satellite transcriptional responses (Fortini 2009). When cell communication. However, the germline Rbpj was conditionally mutated in adult mouse Cdh15 mouse mutant displayed little to no phe- satellite cells, these cells were depleted as a result notype in skeletal muscle development or re- of spontaneous activation and terminal dif- generation (Hollnagel et al. 2002). Satellite cells ferentiation in the absence of self-renewal express additional classical cadherins, which are (Bjornson et al. 2012; Mourikis et al. 2012). likely to act redundantly with M-cadherin in Consequently, such animals failed to regenerate satellite cell function. For example, we have ob- skeletal muscles in response to injury. Com- served that satellite cells that lack both N- and bined germline knockout of Hey1 and HeyL M-cadherin were prone to activation in the ab- (also called Hesr1 and Hesr3) resulted in a sim- sence of injury, suggesting cadherins play a role ilar, although not identical, phenotype, suggest- in maintenance of quiescence (AJ Goel and RS ing they are relevant Notch target genes (Fukada Krauss, in prep.). This topic warrants further et al. 2011). attention, as does the general subject of adhesive Presumably, Notch ligands on myofibers ac- interactions between myofibers and their asso- tivate Notch receptors on satellite cells to stim- ciated satellite cells. ulate contact-dependent signals that maintain quiescence in the absence of injury. The identi- CONCLUDING REMARKS ty of the relevant Notch receptors expressed by satellite cells and Notch ligands expressed by Most research on the role of cell junctions and myofibers is not known. Notch3 is likely to be cell–cell contact-dependent signaling in skele- involved, as its germline mutation resulted in tal muscle development has been focused on perturbation of satellite cell proliferation (Kita- interactions between myoblasts that promoted moto and Hanaoka 2010), but the phenotype of differentiation and fusion, or formation of neu- these mice was distinct from, and mild relative romuscular junctions (Krauss 2010; Kim et al. to, that seen in Rbpj conditional mutants or 2015; Tintignac et al. 2015). Recent studies have Hey1/HeyL double knockouts. Resting satellite shown that cell–cell contact between various cells also express Notch1 and Notch2 (Bjornson nonmuscle and muscle cell types is important et al. 2012; Mourikis et al. 2012), so signaling by for several steps of myogenesis and provided multiple Notch receptors may be involved in mechanistic information on these processes. maintenance of quiescence. Although Notch/ Commitment of progenitor cells to the skeletal RBP-J signaling is clearly a key regulator of sat- muscle lineage, postnatal growth of fibers by ellite cell quiescence, the transcriptional targets recruitment of myoblasts to sites of fusion, that are responsible for the long-term mainte- and fiber-type patterning are each regulated in nance of this fascinating cellular state are not this manner. Therefore, cell–cell contact-de- yet illuminated. Although Hes/Hey factors are pendent signaling is important at most, if not proximal downstream components, they are all, steps of myogenesis (Table 1). transcriptional regulators themselves, so must Many questions remain. The details of the be acting to help control a quiescence program. downstream signaling events triggered by Notch It is worth noting that Notch signaling is also receptors, cadherins, ephrins, and other adhe- required for satellite cells to occupy their niche sion receptors in most stages of myogenesis are in late development and, in its absence, satellite not well understood. Additionally, it is clear that cells remain in the interstitium between fibers contact between cells in some of these processes (Bro¨hl et al. 2012). The relationship between is transient. How long such cells must be in this function for Notch signaling and Notch’s direct contact for successful signaling, how con- role in maintenance of quiescence in adult sat- tact is broken, and the molecular nature of the ellite cells is unexplored and of high interest. cell junctions that support signaling are all in- Based on its specific expression pattern and teresting questions. Despite tremendous pro- localization, M-cadherin (encoded by Cdh15) gress in understanding mechanisms of myoblast

12 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Cell–Cell Contact and Skeletal Myogenesis

Table 1. Roles of cell–cell contact-dependent mechanisms in various steps of myogenesis Process in myogenesis Cell–cell contact-dependent mechanism involveda Lineage commitment Delta/Notch signaling between neural crest cells and dermomyotomal muscle progenitor cells (þ) Myoblast differentiation Delta/Notch signaling (–) N-cadherin/Cdon ! p38 signaling (þ) Myoblast fusion Species-specific adhesion receptors (þ) Myomakerb (þ) Postnatal myofiber growth via Ephrin-A5/Eph signaling between NG2þ interstitial cells and myoblasts (þ) promotion of myoblast migration to sites of fusion Fiber-type patterning Ephrin-A3/Eph8 signaling between slow fibers and Schwann cells at sites of innervation (þ) Maintenance of the quiescent Delta/Notch signaling between myofibers and satellite cells (þ) satellite cell niche Cadherin-based adhesion between myofibers and satellite cells (þ) aEach mechanism is designated (þ) or (–), depending on whether its role is to promote or inhibit the process, respectively. bMyomaker has not been directly shown to exert its effects via cell–cell contact, but its role as a potential fusogen suggests this may be the case.

fusion, the adhesion receptors required in ver- Allen BL, Song JY, Izzi L, Althaus IW, Kang JS, Charron F, tebrate species are not fully identified. The dis- Krauss RS, McMahon AP. 2011. Overlapping roles and collective requirement for the co-receptors Gas1, Cdo covery of myomaker was groundbreaking, and and Boc in Shh pathway function. Dev Cell 20: 775–787. studies on whether it is the long elusive fusogen, Bae GU, Yang YJ, Jiang G, Hong M, Lee HJ, Tessier-Lavigne what its interacting partners are, and how it M, Kang JS, Krauss RS. 2009. Neogenin regulates skeletal myofiber size and focal adhesion kinase and extracellular works are of high interest. Finally, it is somewhat signal-regulated kinase activities in vivo and in vitro. Mol ironic that the stable cell–cell junction present Biol Cell 20: 4920–4931. between myofibers and their associated satellite Biressi S, Molinaro M, Cossu G. 2007. Cellular heterogeneity cells resembles well-studied junctions in other during vertebrate skeletal muscle development. Dev Biol 308: 281–293. tissues, yet is poorly characterized and how it Bischoff R. 1990. Interaction between satellite cells and skel- regulates satellite cell biology is largely un- etal muscle fibers. Development 109: 943–952. known. Continued study with a variety of mod- Bjornson C, Cheung T,Liu L, Tripathi P,Steeper K, Rando T. el organisms will surely illuminate these ques- 2012. Notch signaling is necessary to maintain quies- tions. Furthermore, the growing emphasis on cence in adult muscle stem cells. Stem Cells 30: 232–242. Borello U, Berarducci B, Murphy P, Bajard L, Buffa V, Pic- understanding regenerative myogenesis (which colo S, Buckingham M, Cossu G. 2006. The Wnt/b-cat- is similar but not identical to developmental enin pathway regulates Gli-mediated Myf5 expression myogenesis) and its implications for muscle during somitogenesis. Development 133: 3723–3732. diseases and aging will bring deserved attention Borycki AG, Brunk B, Tajbakhsh S, Buckingham M, Chiang C, Emerson CP Jr, 1999. Sonic hedgehog controls epaxial to these fundamental questions. muscle determination through Myf5 activation. Develop- ment 126: 4053–4063. Brack AS, Rando TA. 2012. Tissue-specific stem cells: Les- ACKNOWLEDGMENTS sons from the skeletal muscle satellite cell. Cell Stem Cell 10: 504–514. Work in R.S.K.’s laboratory on this subject was Brand-Saberi B, Christ B. 2000. Evolution and development supported by grants from the National Insti- of distinct cell lineages derived from somites. Curr Top tutes of Health (NIH) (AR046207 and Dev Biol 48: 1–42. Briata P,Forcales SV,Ponassi M, Corte G, Chen CY,Karin M, AR070231). Puri PL, Gherzi R. 2005. p38-dependent phosphoryla- tion of the mRNA decay-promoting factor KSRP controls the stability of select myogenic transcripts. Mol Cell 20: REFERENCES 891–903. Bro¨hl D, Vasyutina E, Czajkowski MT, Griger J, Rassek C, Abmayr SM, Pavlath GK. 2012. Myoblast fusion: Lessons Rahn HP,Purfu¨rst B, WendeH, Birchmeier C. 2012. Col- from flies and mice. Development 139: 641–656. onization of the satellite cell niche by skeletal muscle

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 13 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al.

progenitor cells depends on Notch signals. Dev Cell 23: Galletta BJ, Chakravarti M, Banerjee R, Abmayr SM. 2004. 469–481. SNS: Adhesive properties, localization requirements and Chakkalakal JV,Jones KM, Basson MA, Brack AS. 2012. The ectodomain dependence in S2 cells and embryonic myo- aged niche disrupts muscle stem cell quiescence. Nature blasts. Mech Dev 121: 1455–1468. 490: 355–360. Gliki G, Ebnet K, Aurrand-Lions M, Imhof BA, Adams RH. Charlton CA, Mohler WA,Radice GL, Hynes RO, Blau HM. 2004. Spermatid differentiation requires the assembly of a 1997. Fusion competence of myoblasts rendered geneti- cell polarity complex downstream of junctional adhesion cally null for N-cadherin in culture. J Cell Biol 138: 331– molecule-C. Nature 431: 320–324. 336. Gopinath S, Webb A, Brunet A, Rando T. 2014. FOXO3 Cheung TH, Quach NL, Charville GW,Liu L, Park L, Edalati promotes quiescence in adult muscle stem cells during A, Yoo B, Hoang P, Rando TA. 2012. Maintenance of the process of self-renewal. Stem Cell Rep 2: 414–426. muscle stem-cell quiescence by microRNA-489. Nature Gruenbaum-Cohen Y,Harel I, Umansky K, Tzahor E, Snap- 482: 524–528. per SB, Shilo BZ, Schejter ED. 2012. The actin regulator Cinnamon Y, Ben-Yair R, Kalcheim C. 2006. Differential N-WASpis required for muscle-cell fusion in mice. Proc effects of N-cadherin-mediated adhesion on the devel- Nati Acad Sci 109: 11211–11216. opment of myotomal waves. Development 133: 1102– Gu J-M, WangDJ, Peterson JM, Shintaku J, Liyanarachchi S, 1112. Coppola V, Frakes AE, Kaspar BK, Cornelison DD, Gut- Cole F,Zhang W,Geyra A, Kang JS, Krauss RS. 2004. Positive tridge DC. 2016. An NF-kB–ephrinA5-dependent com- þ regulation of myogenic bHLH factors and skeletal muscle munication between NG2 interstitial cells and myo- development by the cell surface receptor CDO. Dev Cell 7: blasts promotes muscle growth in neonates. Dev Cell 843–854. 36: 215–224. Comai G, Tajbakhsh S. 2014. Molecular and cellular regu- Guasconi V, Puri PL. 2009. Chromatin: The interface be- lation of skeletal myogenesis. Curr Top Dev Biol 110: 1– tween extrinsic cues and the epigenetic regulation of 73. muscle regeneration. Trends Cell Biol 19: 286–294. de Angelis L, Zhao J, Andreucci JJ, Olson EN, Cossu G, Hindi SM, Tajrishi MM, Kumar A. 2013. Signaling mecha- McDermott JC. 2005. Regulation of vertebrate myotome nisms in mammalian myoblast fusion. Sci Signal 6: re2. development by the p38 MAP kinase-MEF2 signaling Hollnagel A, Grund C, Franke WW, Arnold HH. 2002. The pathway. Dev Biol 283: 171–179. cell adhesion molecule M-cadherin is not essential for Doles JD, Olwin BB. 2015. Muscle stem cells on the edge. muscle development and regeneration. Mol Cell Biol 22: Curr Opin Genet Dev 34: 24–28. 4760–4770. Donoviel DB, Freed DD, Vogel H, Potter DG, Hawkins E, Jeong MH, Ho SM, Vuong TA, Jo SB, Liu G, Aaronson SA, Barrish JP, Mathur BN, Turner CA, Geske R, Montgom- Leem YE, Kang JS. 2014. Cdo suppresses canonical Wnt ery CA, et al. 2001. Proteinuria and perinatal lethality in signalling via interaction with Lrp6 thereby promoting mice lacking NEPH1, a novel protein with homology to neuronal differentiation. Nat Commun 19: 5455. NEPHRIN. Mol Cell Biol 21: 4829–4836. Joseph GA, Lu M, Radu M, Lee J, Burden SJ, Chernoff J, Duan R, Jin P,Luo F,Zhang G, Anderson N, Chen EH. 2012. Krauss RS. 2016. Group I Paks promote myoblast differ- Group I PAKs function downstream of Rac to promote entiation in vivo and in vitro. Mol Cell Biol doi:10.1128/ podosome invasion during myoblast fusion in vivo. J Cell MCB.00222-16. Biol 199: 169–185. Kang J-S, Gao M, Feinleib JL, Cotter PD, Guadagno SN, Dumont NA, Wang YX, Rudnicki MA. 2015. Intrinsic and Krauss RS. 1997. CDO: An oncogene-, serum-, and an- extrinsic mechanisms regulating satellite cell function. chorage-regulated member of the Ig/fibronectin type III Development 142: 1572–1581. repeat family. J Cell Biol 138: 203–213. Fischer-Lougheed J, Liu JH, Espinos E, Mordasini D, Bader Kang J-S, Mulieri PJ, Miller C, Sassoon DA, Krauss RS. 1998. CR, Belin D, Bernheim L. 2001. Human myoblast fusion CDO, a robo-related cell surface protein that mediates requires expression of functional inward rectifier Kir2.1 myogenic differentiation. J Cell Biol 143: 403–413. channels. J Cell Biol 153: 677–686. Kang J-S, Feinleib JL, Knox S, Ketteringham MA, Krauss RS. Fong AP, Tapscott SJ. 2013. Skeletal muscle programming 2003. Promyogenic members of the Ig and cadherin fam- and re-programming. Curr Opin Genet Dev 23: 568–573. ilies associate to positively regulate differentiation. Proc Fortini ME. 2009. Notch signaling: The core pathway and its Natl Acad Sci 100: 3989–3994. posttranslational regulation. Dev Cell 16: 633–647. Kang J-S, Yi MJ, Zhang W, Feinleib JL, Cole F, Krauss RS. Fry CS, Lee JD, Mula J, Kirby TJ, Jackson JR, Liu F, Yang L, 2004. Netrins and neogenin promote myotube forma- Mendias CL, Dupont-Versteegden EE, McCarthy JJ, et al. tion. J Cell Biol 167: 493–504. 2015. Inducible depletion of satellite cells in adult, sed- Kang J-S, Bae GU, Yi MJ, Yang YJ, Oh JE, Takaesu G, Zhou entary mice impairs muscle regenerative capacity with- YT, Low BC, Krauss RS. 2008. ACdo/Bnip-2/Cdc42 sig- out affecting sarcopenia. Nat Med 21: 76–80. naling pathway regulates p38a/b MAPK activity and Fukada S, Yamaguchi M, Kokubo H, Ogawa R, Uezumi A, myogenic differentiation. J Cell Biol 182: 497–507. YonedaT,Matev MM, Motohashi N, Ito T,Zolkiewska A, Keefe AC, Lawson JA, Flygare SD, Fox ZD, Colasanto MP, et al. 2011. Hesr1 and Hesr3 are essential to generate Mathew SJ, YandellM, Kardon G. 2015. Muscle stem cells undifferentiated quiescent satellite cells and to maintain contribute to myofibres in sedentary adult mice. Nat satellite cell numbers. Development 138: 4609–4619. Commun 6: 7807.

14 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Cell–Cell Contact and Skeletal Myogenesis

Kim JH, Jin P, Duan R, Chen EH. 2015. Mechanisms of is a membrane activator of myoblast fusion and muscle myoblast fusion during muscle development. Curr formation. Nature 499: 301–305. Opin Genet Dev 32: 162–170. Millay DP, Sutherland LB, Bassel-Duby R, Olson EN. 2014. Kitamoto T, Hanaoka K. 2010. Notch3 null mutation in Myomaker is essential for muscle regeneration. Genes mice causes muscle hyperplasia by repetitive muscle re- Dev 28: 1641–1646. generation. Stem Cells 28: 2205–2216. Millay DP,Gamage DG, Quinn ME, Min YL, Mitani Y, Bas- Konig S, Hinard V,Arnaudeau S, Holzer N, Potter G, Bader sel-Duby R, Olson EN. 2016. Structure–function analy- CR, Bernheim L. 2004. Membrane hyperpolarization sis of myomaker domains required for myoblast fusion. triggers myogenin and myocyte enhancer factor-2 ex- Proc Natl Acad Sci 113: 2116–2121. pression during human myoblast differentiation. J Biol Moore CA, Parkin CA, Bidet Y,Ingham PW.2007. A role for Chem 279: 28187–28196. the Myoblast city homologues Dock1 and Dock5 and the Krauss RS. 2007. Evolutionary conservation in myoblast adaptor proteins Crk and Crk-like in zebrafish myoblast fusion. Nat Genet 39: 704–705. fusion. Development 134: 3145–3153. Krauss RS. 2010. Regulation of promyogenic signal trans- Mourikis P, Sambasivan R, Castel D, Rocheteau P, Bizzarro duction by cell–cell contact and adhesion. Exp Cell Res V, Tajbakhsh S. 2012. A critical requirement for Notch 316: 3042–3049. signaling in maintenance of the quiescent skeletal muscle Krauss RS, Cole F, Gaio U, Takaesu G, Zhang W, Kang JS. stem cell state. Stem Cells 30: 243–252. 2005. Close encounters: Regulation of vertebrate skeletal Munsterberg AE, Lassar AB. 1995. Combinatorial signals myogenesis by cell-cell contact. J Cell Sci 118: 2355–2362. from the neural tube, floor plate and notochord induce myogenic bHLH gene expression in the somite. Develop- Landemaine A, Rescan PY, Gabillard J-C. 2014. Myomaker ment 121: 651–660. mediates fusion of fast myocytes in zebrafish embryos. Biochem Biophys Res Commun 451: 480–484. Munsterberg AE, Kitajewski J, Bumcrot DA, McMahon AP, Lassar AB. 1995. Combinatorial signaling by Sonic Laurin M, Fradet N, Blangy A, Hall A, Vuori K, Coˆte´ JF. hedgehog and Wnt family members induces myogenic 2008. The atypical Rac activator Dock180 (Dock1) regu- bHLH gene expression in the somite. Genes Dev 9: 2911– lates myoblast fusion in vivo. Proc Natl Acad Sci 105: 2922. 15446–15451. Niessen CM, Leckband D, YapAS. 2011. Tissue organization Lee HJ, Bae G, Leem YE, Choi HK, Kang TM, Cho H, Kim by cadherin adhesion molecules: Dynamic molecular and ST,Kang JS. 2012. Phosphorylation of Stim1 at serine 575 cellular mechanisms of morphogenetic regulation. Phys- / / via netrin-2 Cdo–activated ERK1 2 is critical for the iol Rev 91: 691–731. promyogenic function of Stim1. Mol Biol Cell 23: 1376–1387. Ozawa M. 2015. E-cadherin cytoplasmic domain inhibits cell surface localization of endogenous cadherins and Leem YE, Jeong HJ, Kim HJ, Koh J, Kang K, Bae GU, Cho H, fusion of C2C12 myoblasts. Biol Open 4: 1427–1435. Kang JS. 2016. Cdo regulates surface expression of Kir2.1 Kþ channel in myoblast differentiation. PLoS ONE 11: Palacios D, Mozzetta C, Consalvi S, Caretti G, Saccone V, Proserpio V, Marquez VE, Valente S, Mai A, Forcales SV, e0158707. et al. 2010. TNF/p38a /polycomb signaling to Pax7 locus Lisabeth EM, Falivelli G, Pasquale EB. 2013. Eph receptor in satellite cells links inflammation to the epigenetic con- signaling and ephrins. Cold Spring Harb Perspect Biol 5: trol of muscle regeneration. Cell Stem Cell 7: 455–469. a009159. Powell GT, Wright GJ. 2011. Jamb and jamc are essential for Lluis F,Ballestar E, Suelves M, Esteller M, Munoz-Canoves P. vertebrate myocyte fusion. PLoS Biol 9: e1001216. 2005. E47 phosphorylation by p38 MAPK promotes Prince JEA, Brignall AC, Cutforth T, Shen K, Cloutier JF. MyoD/E47 association and muscle-specific gene tran- 2013. Kirrel3 is required for the coalescence of vomero- scription. EMBO J 24: 974–984. nasal sensory neuron axons into glomeruli and for male- Lu M, Krauss RS. 2010. N-cadherin ligation, but not Sonic male aggression. Development 140: 2398–2408. a/b hedgehog binding, initiates Cdo-dependent p38 Putaala H, Soininen R, Kilpela¨inen P, Wartiovaara J, Trygg- MAPK signaling in skeletal myoblasts. Proc Natl Acad vason K. 2001. The murine nephrin gene is specifically Sci 107: 4212–4217. expressed in kidney, brain and pancreas: Inactivation of Marthiens V, Gavard J, Lambert M, Me`ge RM. 2002. Cad- the gene leads to massive proteinuria and neonatal death. herin-based cell adhesion in neuromuscular develop- Hum Mol Genet 10: 1–8. ment. Biol Cell 94: 315–326. Radice GL, Rayburn H, Matsunami H, Knudsen KA, Take- Martı`n-Padura I, Lostaglio S, Schneemann M, Williams L, ichi M, Hynes RO. 1997. Developmental defects in mouse Romano M, Fruscella P,Panzeri C, Stoppacciaro A, Ruco embryos lacking N-cadherin. Dev Biol 181: 64–78. Li, Villa A, et al. 1998. Junctional adhesion molecule, a Rampalli S, Li L, Mak E, Ge K, Brand M, Tapscott SJ, Dil- novel member of the immunoglobulin superfamily that worth FJ. 2007. p38 MAPK signaling regulates recruit- distributes at intercellular junctions and modulates ment of Ash2L-containing methyltransferase complexes monocyte transmigration. J Cell Biol 142: 117–127. to specific genes during differentiation. Nat Struct Mol McCrea PD, Maher MT, Gottardi CJ. 2015. Nuclear signal- Biol 14: 1150–1156. ing from cadherin adhesion complexes. Curr TopDev Biol Reshef R, Maroto M, Lassar AB. 1998. Regulation of dorsal 112: 129–196. somitic cell fates: BMPs and Noggin control the timing Millay DP,O’Rourke JR, Sutherland LB, Bezprozvannaya S, and pattern of myogenic regulator expression. Genes Dev Shelton JM, Bassel-Duby R, Olson EN. 2013. Myomaker 12: 290–303.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 15 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

R.S. Krauss et al.

Rios AC, Serralbo O, Salgado D, Marcelle C. 2011. Neural ral tube and by cells expressing Wnt-1. Development 121: crest regulates myogenesis through the transient activa- 3675–3686. tion of NOTCH. Nature 473: 532–535. Tajbakhsh S, Borello U, Vivarelli E, Kelly R, Papkoff J, Du- Sakaguchi T, Nishimoto M, Miyagi S, Iwama A, Morita Y, prez D, Buckingham M, Cossu G. 1998. Differential ac- Iwamori N, Nakauchi H, Kiyonari H, Muramatsu M, tivation of Myf5 and MyoD by different Wnts in explants Okuda A. 2006. Putative “Stemness” gene Jam-B is not of mouse paraxial mesoderm and the later activation of required for maintenance of stem cell state in embryonic, myogenesis in the absence of Myf5. Development 125: neural, or hematopoietic stem cells. Mol Cell Biol 26: 4155–4162. 6557–6570. Takaesu G, Kang JS, Bae GU, Yi MJ, Lee CM, Reddy EP, Sanes JR, Lichtman JW.1999. Development of the vertebrate Krauss RS. 2006. Activation of p38a/b MAPK in myo- neuromuscular junction. Annu Rev Neurosci 22: 389– genesis via binding of the scaffold protein JLP to the cell 442. surface protein Cdo. J Cell Biol 175: 383–388. Sartorelli V,Juan AH. 2011. Sculpting chromatin beyond the Talbot J, Maves L. 2016. Skeletal muscle fiber type: Using double helix: Epigenetic control of skeletal myogenesis. insights from muscle developmental biology to dissect Curr Top Dev Biol 96: 57–83. targets for susceptibility and resistance to muscle disease. Schiaffino S, Reggiani C. 2011. Fiber types in mammalian Wiley Interdiscip Rev Dev Biol 5: 518–534. skeletal muscles. Physiol Rev 91: 1447–1531. Tapscott SJ. 2005. The circuitry of a master switch: Myod and the regulation of skeletal muscle gene transcription. Schuster-Gossler K, Cordes R, Gossler A. 2007. Premature Development 132: 2685–2695. myogenic differentiation and depletion of progenitor cells cause severe muscle hypotrophy in Delta1 mutants. Thuault S, Hayashi S, Lagirand-Cantaloube J, Plutoni C, Proc Natl Acad Sci 104: 537–542. Comunale F, Delattre O, Relaix F, Gauthier-Rouviere C. 2013. P-cadherin is a direct PAX3-FOXO1A target in- Sens KL, Zhang S, Jin P,Duan R, Zhang G, Luo F, Parachini volved in alveolar rhabdomyosarcoma aggressiveness. L, Chen EH. 2010. An invasive podosome-like structure Oncogene 32: 1876–1887. promotes fusion pore formation during myoblast fusion. J Cell Biol 191: 1013–1027. Tintignac LA, Brenner H-R, Ru¨egg MA. 2015. Mechanisms regulating neuromuscular junction development and Serra C, Palacios D, Mozzetta C, Forcales SV, Morantte I, function and causes of muscle wasting. Physiol Rev 95: Ripani M, Jones DR, Du K, Jhala US, Simone C, et al. 809–852. 2007. Functional interdependence at the chromatin level between the MKK6/p38 and IGF1/PI3K/AKT pathways Tran P,Ho SM, Kim BG, Vuong TA,Leem YE, Bae GU, Kang during muscle differentiation. Mol Cell 28: 200213. JS. 2012. TGF-b-activated kinase 1 (TAK1)and apoptosis signal-regulating kinase 1 (ASK1) interact with the pro- Shilagardi K, Li S, Luo F, Marikar F, Duan R, Jin P,Kim JH, myogenic receptor Cdo to promote myogenic differenti- Murnen K, Chen EH. 2013. Actin-propelled invasive ation via activation of p38MAPK pathway. J Biol Chem membrane protrusions promote fusogenic protein en- 287: 11602–11615. gagement during cell-cell fusion. Science 340: 359–363. van Amerongen R, Nusse R. 2009. Towards an integrated Sieiro D, Rios AC, Hirst CE, Marcelle C. 2016. Cytoplasmic view of Wnt signaling in development. Development 136: NOTCH and membrane-derived b-catenin link cell fate 3205–3214. choice to epithelial–mesenchymal transition during Vasyutina E, Lenhard DC, WendeH, Erdmann B, Epstein JA, myogenesis. eLife 24: e14847. Birchmeier C. 2007. RBP-J (Rbpsuh) is essential to main- Simone C, Forcales SV, Hill DA, Imbalzano AN, Latella L, tain muscle progenitor cells and to generate satellite cells. Puri PL. 2004. p38 pathway targets SWI-SNF chromatin- Proc Natl Acad Sci 104: 4443–4448. remodeling complex to muscle-specific loci. Nat Genet Vasyutina E, Martarelli B, Brakebusch C, Wende H, Birch- 36: 738–743. meier C. 2009. The small G-proteins Rac1 and Cdc42 are Sohn RL, Huang P, Kawahara G, Mitchell M, Guyon J, Kal- essential for myoblast fusion in the mouse. Proc Natl Acad luri R, Kunkel LM, Gussoni E. 2009. A role for nephrin, a Sci 106: 8935–8940. renal protein, in vertebrate skeletal muscle cell fusion. Vite A, Radice GL. 2014. N-cadherin/catenin complex as a Proc Natl Acad Sci 106: 9274–9279. master regulator of intercalated disc function. Cell Com- Srinivas BP, Woo J, Leong WY, Roy S. 2007. A conserved mun Adhes 21: 169–179. molecular pathway mediates myoblast fusion in insects Wu Z, Woodring PJ, Bhakta KS, Tamura K, WenF,Feramisco and vertebrates. Nat Genet 39: 781–786. JR, Karin M, Wang JY, Puri PL. 2000. p38 and extracel- Stark DA, Coffey NJ, Pancoast HR, Arnold LL, Walker JPD, lular signal-regulated kinases regulate the myogenic pro- Valle´e J, Robitaille R, Garcia ML, Cornelison DDW.2015. gram at multiple steps. Mol Cell Biol 20: 3951–3964. Ephrin-A3 promotes and maintains slow muscle fiber Yesildag B, Bock T, Herrmanns K, Wollscheid B, Stoffel M. identity during postnatal development and reinnerva- 2015. Kin of IRRE-like protein 2 Is a phosphorylated tion. J Cell Biol 211: 1077–1091. glycoprotein that regulates basal insulin secretion. J Biol Stern HM, Hauschka SD. 1995. Neural tube and notocord Chem 290: 25891–25906. promote in vitro myogenesis in single somite explants. Zhang W, Kang JS, Cole F, Yi MJ, Krauss RS. 2006. Cdo Dev Biol 167: 87–103. functions at multiple points in the Sonic Hedgehog path- Stern HM, Brown AMC, Hauschka SD. 1995. Myogenesis in way, and Cdo-deficient mice accurately model human paraxial mesoderm: Preferential induction by dorsal neu- holoprosencephaly. Dev Cell 10: 657–665.

16 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Keep Your Friends Close: Cell−Cell Contact and Skeletal Myogenesis

Robert S. Krauss, Giselle A. Joseph and Aviva J. Goel

Cold Spring Harb Perspect Biol published online January 6, 2017

Subject Collection Cell-Cell Junctions

Vascular Endothelial (VE)-Cadherin, Endothelial Signaling by Small GTPases at Cell−Cell Adherens Junctions, and Vascular Disease Junctions: Protein Interactions Building Control Maria Grazia Lampugnani, Elisabetta Dejana and and Networks Costanza Giampietro Vania Braga Adherens Junctions and Desmosomes Making Connections: Guidance Cues and Coordinate Mechanics and Signaling to Receptors at Nonneural Cell−Cell Junctions Orchestrate Tissue Morphogenesis and Function: Ian V. Beamish, Lindsay Hinck and Timothy E. An Evolutionary Perspective Kennedy Matthias Rübsam, Joshua A. Broussard, Sara A. Wickström, et al. Cell−Cell Contact and Receptor Tyrosine Kinase The Cadherin Superfamily in Neural Circuit Signaling Assembly Christine Chiasson-MacKenzie and Andrea I. James D. Jontes McClatchey Hold Me, but Not Too Tight−−Endothelial Cell−Cell Mechanosensing and Mechanotransduction at Junctions in Angiogenesis Cell−Cell Junctions Anna Szymborska and Holger Gerhardt Alpha S. Yap, Kinga Duszyc and Virgile Viasnoff Connexins and Disease Beyond Cell−Cell Adhesion: Sensational Mario Delmar, Dale W. Laird, Christian C. Naus, et Cadherins for Hearing and Balance al. Avinash Jaiganesh, Yoshie Narui, Raul Araya-Secchi, et al. Cell Junctions in Hippo Signaling Cell−Cell Junctions Organize Structural and Ruchan Karaman and Georg Halder Signaling Networks Miguel A. Garcia, W. James Nelson and Natalie Chavez Loss of E-Cadherin-Dependent Cell−Cell Adhesion Cell Biology of Tight Junction Barrier Regulation and the Development and Progression of Cancer and Mucosal Disease Heather C. Bruner and Patrick W.B. Derksen Aaron Buckley and Jerrold R. Turner

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press

Desmosomes and Intermediate Filaments: Their Integration of Cadherin Adhesion and Consequences for Tissue Mechanics Cytoskeleton at Adherens Junctions Mechthild Hatzfeld, René Keil and Thomas M. René Marc Mège and Noboru Ishiyama Magin

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved