Developmental Fate of the Mammalian Myotome

Size: px
Start display at page:

Download "Developmental Fate of the Mammalian Myotome"

Transcription

1 a DEVELOPMENTAL DYNAMICS 239: , 2010 RESEARCH ARTICLE Developmental Fate of the Mammalian Myotome Marianne Deries, 1,4 Ronen Schweitzer, 2,3 and Marilyn J. Duxson 1 * The myotome is a segmented paraxial muscle present in all early vertebrate embryos, which in amniotes disappears in mid-embryogenesis, and is replaced by complex epaxial and hypaxial musculature. Little is known about how this transition occurs. Here, we describe the detailed morphogenesis of the epaxial muscles from the epaxial myotome, in rodent embryos. The results show there is no apoptosis of myotomal fibres during the transition, and that the epaxial muscles arise by translocation, re-orientation, and elongation of the myotomal myocytes followed by cleavage of the myotomal masses. Myotomal myocytes transit from a mononucleated to a multinucleated state just before onset of this transformation. Each newly-formed epaxial muscle anlagen includes populations of Pax3- and Pax7-positive muscle progenitors, with different distributions. Using transgenic mouse embryos bearing a GFP marker for Scleraxis, we show that tendon progenitors are tightly associated with the sides and ends of myotomal myocytes as they re-orient and elongate. 239: , VC 2010 Wiley-Liss, Inc. Key words: myotome; rat; mouse; development; morphogenesis; apoptosis; Pax3; Pax7; scleraxis Accepted 16 August 2010 INTRODUCTION This report examines the manner in which the epaxial muscles of the vertebrate (the deep back muscles) arise from the embryonic myotome. The myotome is a metameric muscle that forms within the somite, lying between the lateral dermomyotome and the medial sclerotome, and is the first muscle to develop in every vertebrate. Its mononucleated myocytes span the whole length of each segment and lie parallel to the axis of the embryo (Denetclaw et al., 1997; Kahane et al., 1998; Venters et al., 1999). In birds and mammals, myotomes are transitory muscles and disappear to be replaced in the epaxial domain by the deep muscles of the back. These attach to the vertebrae, where they act as extensor muscles, and receive their innervation from the dorsal ramus of the mixed spinal nerve. The adult epaxial muscles comprise three main muscle groups, the transversospinalis, longissimus, and iliocostalis muscles, which are identifiable all along the axis (Vallois, 1922). At a thoracic level, the levatores costarum muscles form a fourth group attaching to the ribs and elevating them during respiration (Sato, 1973; Smith and Hollyday, 1983). The transition from the simple unisegmental myotome to the complex morphology of the deep back muscles occurs rapidly during midembryogenesis, but remarkably little is known about how it occurs. A key question is whether the original myocytes of the myotome are retained and contribute to the formation of epaxial muscles. Although it has been proposed that they do (Butcher, 1929; Christ et al., 1983; Christ and Ordahl, 1995), the only direct evidence for retention of myotomal fibres within the definitive axial muscles comes from the work of Cinnamon et al. (1999) where hypaxial myotomal myocytes labelled with DiI during early development were found to persist throughout the segmental intercostal muscles. If the original myocytes are retained within all epaxial and hypaxial derivatives of the myotome, then the further processes that lead to the transformation of the 1 Department of Anatomy and Structural Biology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand 2 Shriners Hospital for Children, Research Division, Portland, Oregon 3 Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, Oregon 4 Centro de Biologia Ambiental, Faculdade de Ciencias, Universidade de Lisboa, Lisbon, Portugal *Correspondence to: Marilyn J. Duxson, Department of Anatomy and Structural Biology, Otago School of Medical Sciences, University of Otago, PO Box 913, Dunedin, New Zealand. marilyn.duxson@stonebow.otago.ac.nz DOI /dvdy Published online 23 September 2010 in Wiley Online Library (wileyonlinelibrary.com). VC 2010 Wiley-Liss, Inc.

2 FATE OF MAMMALIAN MYOTOME 2899 simple segmental myotomal scaffold into the complex forms of the adult axial muscles are almost completely unknown. Other possibilities are that the myotomal fibres might undergo programmed death and the axial muscles form de novo from dermomyotomally derived progenitors, or that the myotomal fibres might give rise directly to some muscle groups with simple segmental morphology (e.g., intercostal muscles hypaxially; unisegmental transversospinal muscles epaxially) while muscles with non-segmental forms are formed de novo by migration of precursors from the dorsomedial lip of the dermomyotome. Our current lack of understanding of the fate of the myotome and of the origin of the adult axial muscles is in stark contrast to our detailed knowledge about early myotome formation (Kalcheim et al., 1999; Venters and Ordahl, 2002; Gros et al., 2004; Hollway and Currie, 2005) and the origins of the migratory hypaxial muscles (reviewed in Buckingham, 2001). Transformation of the embryonic myotome into the complex arrays of epaxial and hypaxial muscles of the trunk is unlikely to occur only through the actions of myogenic cells. Muscular, skeletal, and connective tissues develop in close spatial and temporal association and their morphogenesis must be synchronised in order to form functional musculoskeletal units. Stopak and Harris (1982) have shown that connective tissue fibroblasts in vitro can actively organise extracellular matrix so as to exert traction, resulting in movement of dispersed muscle cells into organised muscle masses with functional relations to the forming bones. Similarly, Kieny and Chevallier (1979) and then Chevallier and Kieny (1982) have used chick-quail grafting experiments to demonstrate that the segregation and morphogenesis of limb muscle masses in vivo are dependent on the somatopleural mesenchyme of the limbs. Overall, the consensus from experiments in the limb is that muscle progenitor cells are not obviously committed to any specific muscle or region (Christ et al., 1977; Kardon et al., 2002; Rees et al., 2003); rather, they are instructed by TCF4- expressing mesenchymal cells to differentiate in a specific area and pattern (Kardon, 1998; Kardon et al., 2003). The somitic compartment that gives rise to the connective tissue of axial muscles is the syndetome (Schweitzer et al., 2001; Brent et al., 2003). The progenitor cells of the syndetome specifically express the transcription factor Scx but not MyoD (specific to muscle progenitor cells) or Pax1 (specific to sclerotomal cells) (Brent et al., 2003). At later times, Scx is expressed in a sub-group of developing connective tissues including tendons and aponeuroses. (Schweitzer et al., 2001). Recent lineage studies suggest that internal muscle connective tissues are also derived from Scx-expressing cells (Pryce and Schweitzer, unpublished data), as expression of the ScxGFP transgenic reporter can be detected here, likely due to persistence of the highly expressed GFP transcript and protein. Early interactions between myotome, syndetome, and sclerotome have been studied and induction of Scx found to be dependent on FGF proteins secreted by the myotome (Brent et al., 2003, 2005). However little is known about the later relationship between the Scx-expressing cells and the myotomal muscle fibres over the period when the myotome transforms into the epaxial musculature. A particular question is whether the Scx-expressing cells are appropriately positioned to play a part in the reorganisation of the muscular tissues in the region. After initial formation of the epaxial muscles, their further growth and differentiation requires a continuing source of myogenic progenitors, both to increase fibre number and for addition of nuclei during fibre enlargement. This raises the question of the origin of these progenitors. Epaxial progenitor cells may already be present in the epaxial muscular anlagen as they segregate to form the different muscle groups, as seems likely from the work of Gros et al. (2004, 2005). However, this has never been formally demonstrated. Alternately, progenitors might migrate from the dorsomedial lip of the dermomyotome to colonise the new epaxial muscle masses. This second system would imply a mechanism similar to that of the migration of limb progenitors. The aim of this study was to understand the late development of the mammalian epaxial myotome and exactly how it gives rise to the definitive epaxial muscles. Here we report that there is virtually no death of myotomal myocytes during this transition. Confocal microscopy on wholemount specimens shows that the differentiated fibres of the epaxial myotome cleave into discrete masses, simultaneously altering their orientation and length, to give rise to the anlagen of the epaxial muscles. During this translocation, Scx-expressing connective tissue cells are tightly associated with the ends and sides of the myotomal myocytes, consistent with an active role in shaping the muscles. As the myotomal fibres cleave into the discrete groups that will form each future epaxial muscle, a pool of Pax3- and Pax7-positive myogenic progenitors intermingles with them, providing the source of their future growth and development. RESULTS Epaxial Myotome Undergoes a Progressive Transformation to Form the Epaxial Muscles To understand the morphogenesis of epaxial myotome into epaxial muscles, the morphology of the region was initially studied in rat embryos at the anterior trunk level (just posterior to the forelimb), at half day intervals over the period from E12.5 to E14.5 and at E15.5. This period spans from about a day after the myotome is first established, through until the segmental myotomes have disappeared at forelimb level (E14.5) and the epaxial musculature is well defined (E15.5) (Deries et al., 2008). Myocytes, myofibres, and surrounding connective tissues were visualised in situ and in three dimensions within whole mount preparations dually immunostained for myosin heavy chain (MHC) and tenascin. Tenascin is an extracellular matrix glycoprotein broadly expressed within developing connective tissues including tendons, ligaments, cartilage, and perichondrium (Riou et al., 1992). Images shown are photomontages of multiple (up to 40) confocal Z-stacks. Interpretation of the whole mount preparations was aided by parallel studies of transverse sections and the two are presented together in Figure 1.

3 2900 DERIES ET AL. Fig. 1. Progressive transformation of the myotome into epaxial muscles. Fluorescent images of rat embryos immunostained for MHC (red) and tenascin (green) at anterior trunk level, on transverse sections (A, C, E, I, L, O) or whole mounts viewed in the parasagittal plane (B, D, F H, J, K, M, N, P R). B and D are flattened confocal Z-stacks, all other parasagittal views are 10-mm optical sections. The dotted lines on some transverse sections (E, I, L, O) indicate the approximate plane of the corresponding sagittal sections. A,B: E12.5. C,D: E13.0. E H: E13.5. Arrows in G indicate shortened myocytes in the dorsal region. H is a slightly more advanced specimen, stained only for myosin; the arrow indicates the beginnings of the first cleavage plane. I K: E14.0. Arrows in I and K indicate the first cleavage plane; arrowheads in K indicate the second, more ventral, cleavage plane; asterisks show where fibres from adjacent segments are beginning to blend together. L N: E14.5. The four muscle masses of the thoracic level are almost segregated. Arrows in N indicate some vestiges of segmentation within iliocostalis muscle. O R: E The four epaxial muscle groups are now well defined. MHC, myosin heavy chain, Dm, dermomyotome; Sc, sclerotome; NT, neural tube; DRG, dorsal root ganglia; Ep, epaxial; Hyp, hypaxial; Ve, vertebra; lc, levatores costarum; ts, transversospinalis; long, longissimus; ilio, iliocostalis; HypM, hypaxial muscle; D, dorsal; V, ventral; M, medial; L, lateral; R, rostral; C, caudal.

4 FATE OF MAMMALIAN MYOTOME 2901 At E12.5 and E13.0, the epaxial myotome presents in its classical segmented form with distinct tenascinstained intersegmental boundaries and arrays of axially-oriented parallel myocytes (Fig. 1A D). In cross section, the dermomyotome is visible as an unstained region superficial to the myotome and surrounded by a tenascin-positive extracellular matrix at E12.5 (Fig. 1A), but begins to disperse in its central region by E13.0 (Fig. 1C). All fibres of the myotome span the whole segment, but at E13.0 tenascin expression begins to expand into the myotome in some regions, blurring the intersegmental boundaries (Fig. 1D). E13.5 marks the onset of transformation of the myotome, especially in the dorsal region (Fig. 1E H). While the deeper part of the myotome still has parallel fibres spanning the whole length of the segment (Fig. 1F), the most dorsal and superficial myocytes no longer extend to the segmental boundary (arrows, Fig. 1G). In the most advanced embryos, these dorsalmost myocytes have tilted upwards at the anterior end of each segment, forming a cleavage plane that begins to separate them from their ventral neighbours (arrow, Fig. 1H). Half a day later, the transformation is more apparent (E14.0, Fig. 1I K). In transverse sections, the dorsal cleavage plane starts to become visible as an indentation of the myotomal mass (arrow, Fig. 1I). The deeper muscle masses remain segmented, still recognisably in the pattern of the myotome, but the tenascin-positive intersegmental zone has expanded to form the nascent ribs (Fig. 1J). In contrast, bundles of fibres lying just superficial to the ribs start to elongate and blend together (asterisks, Fig. 1K), marking initiation of the formation of the long back muscles, iliocostalis and longissimus. In this superficial region, two cleavage planes are now visible. Above the first (arrows, Fig. 1K), the dorsalmost fibres of the myotome have continued to tilt dorsally at their rostral ends, segregating a group of fibres that most likely represent the future transversospinalis muscles. A second cleavage plane (arrowheads, Fig. 1K), ventral to the first one, separates the future iliocostalis from longissimus. Within the next 12 hr, further major changes occur, resulting in the clear separation of the four epaxial muscle groups at thoracic level by E14.5. The most medial mass can now be clearly identified as levatores costarum (Fig. 1L, M); the three other adult muscle groups, transversospinalis, longissimus, and iliocostalis, are visible more laterally (Fig. 1L, N). At this stage, iliocostalis muscle still shows the vestiges of its segmental origin (arrows, Fig 1N). The cleavage and re-orientation of the muscles continues over the next 24 hr, so that by E15.5 the four major epaxial muscle masses have largely attained their distinctive features (Fig. 1O R). No aspect of their morphology now resembles the segmented myotome. Myotomal Transformation Does Not Involve Myocyte Apoptosis Transformation of the myotome might occur either by death and replacement of the embryonic myotomal myocytes, or by their morphogenetic re-shaping. To assess whether programmed cell death of myotomal myocytes occurs, we examined the epaxial region in serial frontal sections of the region just behind the forelimb at E12.5, E13.5, and E14.0, using caspase-3 staining to detect the apoptotic cascade (Wolf et al., 1999), TUNEL to detect DNA breaks, and Hoechst nuclear staining to detect nuclear fragmentation and condensation. Detection of apoptosis was combined with immunohistochemistry to detect either muscle precursors and myoblasts (using a combination of Pax3, Pax7 and MyoD monoclonal antibodies) or differentiated muscle masses (MHC antibodies) (Fig. 2). At all three stages, expression of caspase-3, accompanied by nuclear condensation, was seen regularly in the embryos (arrows and arrowheads in Fig. 2A F), but extremely rarely in myogenic cells. After extensive searching (serial sections of three segments just caudal to the forelimb level, examined for 3 embryos at each age), no more than a single caspase-3 positive myogenic cell was seen in any embryo. Detection using TUNEL gave the same result (Fig. 2G L). When apoptosis was found in the myotomal area, it was of non-muscle cells (arrowheads, Fig. 2K, L). As an internal positive control, a dying population of non-myogenic cells was found consistently with both techniques, just lateral to the E12.5 myotome and close to the base of the forelimb (arrows, Fig. 2A, B, G, H). The lack of apoptosis within the transforming myotome is consistent with our morphological observations (in whole mount preparations) that no population of myotomal myocytes or myofibres was ever seen to show fragmentation, nuclear condensation (in Hoechst staining), or other features suggestive of large-scale cell death. Thus, the bulk of the differentiated muscle cells of the myotome survive through the period of myotome-toepaxial transformation, rather than undergoing morphogenetic cell death, as happens in other regions where dramatic re-shaping of tissues takes place. Appearance of Multinucleated Myocytes Myotomal myocytes are mononucleated cells during their early development, whereas the later epaxial musculature is composed of multinucleated muscle fibres. This raises the possibility that addition of nuclei from a different lineage might be the factor that initiates transformation within the myotomal myocytes. We, therefore, examined the timing of the transition from mononucleation to multinucleation in the muscle cells of the epaxial region, using dual immunostaining for MHC and myogenin on parasagittal sections. At the level of the forelimb, occasional binucleate myotomal cells could be found at E13.0 (arrow, Fig. 3A, enlarged in Fig. 3 B), with these becoming common throughout the myotome by E13.5 (arrow, Fig. 3C, enlarged in Fig. 3D). Thus, the transition to multinucleation just precedes initiation of myotomal transformation at the forelimb level. Scleraxis-Positive Cells Are Tightly Associated With Myotomal Myocytes During Mouse Epaxial Morphogenesis The above results suggest mammalian epaxial muscles develop through

5 2902 DERIES ET AL. Fig. 2. Myogenic cells do not undergo apoptosis in the transforming myotome of the rat. Frontal sections of E12.5 (A,B,G,H), E13.5 (C,D,I,J), and E14.0 (E,F,K,L) rat embryos, taken just caudal to the forelimb and immunostained to mark muscle progenitor cells (Pax3, Pax7, and MyoD; red, A,C,E,G,I,K) or myocytes (MHC, red; B,D,F,H,J,L) combined with caspase-3 (green, A F) or TUNEL (green, G L). Nuclear staining is with Hoechst (blue). A dying population is marked by caspase-3 and TUNEL at E12.5 in the region lateral to the myotome (arrows, A,B,G,H). Neither caspase-3 nor TUNEL mark any myogenic cells but a few non-muscle cells are dying within the muscle masses (arrowheads, A E,G,H,K,L). L, lateral; M, medial; R, rostral; C, caudal; MHC, myosin heavy chain. a progressive transformation of the epaxial myotomes. Such a process would require the translocation of differentiated muscle fibres. It is know that tendon primordia form independently of muscles (Chevallier and Kieny, 1982; Kardon, 1998) and that the connective tissues associated with muscle direct myogenic patterning (Kardon et al., 2003). To begin to understand the mechanisms involved in epaxial transformation, morphogenesis of the developing tendon Fig. 3. Fig. 3. Transition from mono- to multi-nucleation in rat epaxial myocytes. Confocal images showing dual immunohistochemistry for MHC (red) and myogenin (green) on 10-mm parasagittal sections at forelimb level. A,B: At E13.0, most myotomal myocytes remain mononucleated, but an occasional binucleated cell can be located (arrow in A, enlarged in B). C,D: E13.5. Binucleated myocytes are now common throughout the epaxial myotome (arrows in C, the lower arrowed cell is enlarged in D). L, lateral; M, medial; R, rostral; C, caudal; MHC, myosin heavy chain.

6 FATE OF MAMMALIAN MYOTOME 2903 primordia and muscle-associated connective tissue was followed alongside that of the muscle fibres, using scleraxis expression to identify these cells (Schweitzer et al., 2001). After failing in many attempts to reliably detect scleraxis mrna or protein in the rat, we used a transgenic ScxGFP mouse line in which Scx gene function is not affected (Pryce et al., 2007) for these experiments. For brevity, we focussed only on development of the most dorsal group of myotomal fibres, which gives rise to the transversospinalis muscle group. First we established that development of epaxial muscles in the mouse occurs similarly to rat, but with all events occurring about 2 days earlier in gestation. We then studied ScxGFP mice at E11.5, E12.5, and E13.5, at both anterior trunk (just behind forelimb) and hindlimb levels. Because myotomal development at hindlimb level lags about half a day behind the forelimb level, this allowed examination of intermediate developmental stages. As described by Brent et al. (2003), Scx-positive cells were first found in a layer covering the medial and ventral sides of the myotome (Fig. 4A, arrow), and in the intersegmental spaces (Fig. 4B, arrows). (Note that scattered yellow staining in Fig. 4B, and to a lesser extent in Fig. 4D, M, is autofluorescence in red blood cells.) As the dorsalmost myotomal myocytes started to translocate (Fig. 4C E; E11.5 forelimb level and Fig. 4F, G; E12.5 hindlimb level), Scx-positive cells invaded the spaces between the myotomal myocytes (arrows, Fig. 4C, D), and were particularly prominent around the dorsally migrating, anterior ends of the myocytes(fig.4d,e,g,arrowheads)and in regions where cleavage planes were forming (arrows, Fig. 4F, G). Over the subsequent stages, dense clusters of Scx-positive cells formed dorsally, marking the positions of the tendon anlagen associated with the spinous process of each vertebra (asterisks in Fig. 4I), and the fibres of the transversospinal group progressively diverged into uni- and multisegmental portions (illustrated by blue highlighted fibres in Fig. 4I, K, M). This latter divergence was first apparent at E12.5, when the anterior end of all fibres in a segment lay anchored within the dorsal Scx-positive cell cluster, but, caudally, the more ventral fibres began to extend so they blended with those of the next posterior segment (longer highlighted fibre in Fig. 4I). A large number of Scx-positive cells were spread widely within all muscle regions at this stage, so it was not possible to say if specific Scx-positive cell clusters were also associated with the caudal ends of individual elongating myocytes. This elongation and divergence into uni- and multi-segmental bundles within the transversospinalis muscle group consolidated over the next two stages, with the longer ventral fibres eventually attaching together onto a continuous lateral tendon expressing Scx (asterisks, Fig. 4K, M). Pax3- and Pax7-Positive Muscle Progenitors Segregate Along With The Epaxial Muscle Masses The above results show that myotomal fibres form the core of the emerging epaxial muscles, but do not define the source of the muscle progenitors that will allow for their growth. A population expressing Pax3 and Pax7, and believed to represent the pool of myogenic progenitors, has been described in the myotome and in limbs, in both chick (Ben-Yair and Kalcheim, 2005; Gros et al., 2005) and mouse (Kassar-Duchossoy et al., 2005; Relaix et al., 2005). We thus examined the distribution of Pax3- and Pax7-expressing cells in relation to the differentiating epaxial muscle masses, as they segregated from the myotome, in rat embryos. Figure 5 shows the extent of differentiation of the myotome and epaxial muscles in the left column, as assessed by expression of MHC (red) and myogenin (green), while the middle- and right-hand columns show Pax3- and Pax7-positive progenitors (red) in relation to myogenin (green). Before the segregation of the epaxial muscles begins (E12.5, Fig. 5A C), the populations of muscle precursors expressing Pax3 and Pax7 are present throughout the myotome and intermingle relatively homogeneously with the differentiated myotomal myocytes expressing myogenin (arrows, Fig. 5B, C). Within the overlying dermomyotome, their distributions are also similar, except that Pax3 is expressed more strongly within the dorsal lip (Dm, Fig. 4B, C). As the myotome starts to cleave into the epaxial masses (E13.5, 14.0 Fig. 4D I), the distributions of Pax3- and Pax7-positive cells within the muscle masses also begin to diverge. Pax3 is now expressed predominantly at the lateral edge of the muscle masses where almost no differentiated cells are present (arrowheads, Fig 5E, H), while Pax7-positive cells lie throughout the cleaving and differentiating epaxial masses (Fig 5F, I). Finally, Pax3 expression becomes restricted to a small ventral region (arrowhead in Fig. 5K) that is largely MHC negative (compare Fig. 5J and K) and so presumed to be the last region to differentiate, and then disappears by E15.5 (Fig. 5N). This leaves the developing epaxial muscle masses populated by progenitors that express solely Pax7 (Fig. 5O). Pax3 and Pax7 Are Associated With Sequential Waves of Epaxial Muscle Growth Given the different distributions of Pax3- and Pax7-positive muscle precursors within the cleaving epaxial muscle masses, it seems likely they are contributing differently to epaxial development. We, therefore, examined the proliferative and differentiative behaviour of the populations through the main period of cleavage of these muscles, from E13.5 to E14.5. Over this period, both populations are in a highly proliferative state, as measured by their ability to incorporate BrDU (Table 1 and Fig. 6E G, L N). Both populations are also transiting into terminal differentiation, as measured by their co-expression of myogenin (Table 1 and Fig. 6A D, H K), but this occurs with distinctively different spatial distributions. As Pax3-positive cells rapidly reduce in number and became restricted to lateral and ventral regions of the epaxial muscle masses (E13.5 E14.5, Fig. 5E, H, K), Pax3/myogenin co-expression is observed in a modest proportion of nuclei within these areas (Fig. 6B D). In contrast, Pax7 and myogenin co-

7 2904 DERIES ET AL. TABLE 1. Proportion of Pax3- or Pax7-Positive Progenitors Co-Expressing BrDU or Myogenin a Pax3/BrDU (%) Pax3/myog (%) Pax7/BrDU (%) Pax7/myog (%) E , 42 (40) 4.4, 2.3 (3.4) 27, 36 (32) 16, 12 (14) E , 35 (40) 15, 12 (14) 36, 31 (34) 18, 16 (17) E , 47 (43) 18, 10 (14) 32, 38 (35) 24, 19 (22) a Numbers are for individual animals, with the average shown in brackets. expression occurs at a similar frequency (Table 1) but throughout the whole of the epaxial muscle masses from E13.5 onwards. (Fig. 6I K). This suggests that the bulk of new myonuclei in the growing muscles at these stages originates from Pax7-positive precursors, while the small number of cells co-expressing Pax3 and myogenin may be involved in completing an earlier stage of myogenesis at the lateral and ventral edges of the masses. One possibility, consistent with the timing and distribution of the two populations, is that Pax3-positive cells are responsible for the initial establishment of new myocytes during expansion of the epaxial masses, while Pax7-positive cells fuse into the initial myocytes to drive their growth and establish them as multinuclear muscle fibres. DISCUSSION Mammalian Epaxial Myotome: A Scaffold for the Epaxial Muscle Masses Our results suggest that the epaxial musculature of the rat and mouse are formed by a process of transformation of the embryonic epaxial myotome, involving translocation, re-orientation, and elongation of existing myotomal myocytes. We found no morphological or other evidence suggesting cell death within the myotome during this transformation process. All three techniques used to detect apoptosis within the myotomal myocytes gave consistent negative results at every stage examined, and the fine morphology similarly showed no myocyte degeneration or fragmentation. Thus, although apoptosis is known to be a feature of some muscles during morphogenesis (e.g., chick biventer cervicis, McClearn et al., 1995; tadpole tail, Watanabe and Sasaki, 1974; rat rectus abdominus, Lynch, 1984), it does not seem to be a feature of myotomal transformation. The myotomal cells instead survive and contribute to epaxial muscle formation. The rat myotome begins as a segmental series of paraxially-positioned, parallel myocytes. However, at E13.5 the dorsal-most myocytes of each segment begin to shorten, and to tilt upwards at their rostral ends, so that they cleave away from the bulk of the myotome. These changes continue at E14.0, by which time the first cleavage is more marked, and a second, more ventral, cleavage plane has opened up. Simultaneously, the myocytes begin to lengthen, particularly in the more ventral regions, so that the intersegmental boundaries of the original myotome are obliterated. A factor in the lengthening of the myotomal fibres may be their transition from a mononucleated to a multinucleated form, which begins at about E13.0 at forelimb level. By E14.5 and E15.5, four muscle masses, corresponding to the transversospinalis, iliocostalis, longissimus, and levatores costarum are distinguishable, divided by cleavage planes. The more ventral epaxial muscles now span multiple segments, with the extension of the muscle mass duebothtofibreelongationandtothe merging of adjacent segments. Thus, the simple myotomal myocytes become transformed into the complex scaffold of the epaxial musculature. This transformation clearly requires movement of differentiated muscle cells, which is in contrast to what we know of limb muscle formation. In the limbs, a pre-pattern of connective tissue influences the orientation of muscle fibres, which is appropriate for each muscle from the time of their first appearance (Kardon, 1998; Kardon et al., 2002, 2003). However, movement of differentiated fibres occurs in other systems too. For example, intercostal muscles, are known to derive from the parallel myocytes of the hypaxial myotome (Cinnamon et al., 1999) yet the adult intercostal muscles are formed of three layers, the internal, the middle, and the external intercostal muscles, all with different orientations. The hypaxial myotome also forms some of the abdominal muscles (Bardeen, 1900; Rizk and Adieb, 1982; Christ et al., 1983). In this case again, differentiated cells end in very different places and with different orientations to those of the myotome. Finally, the translocation of differentiated muscle fibres has been observed in the head during migration of extraocular muscles to their final destination (Noden and Francis-West, 2006) and in the body during the formation of perineal muscles (Evans et al., 2006). Mechanism for Morphogenesis of Epaxial Muscles The movement of differentiated myotomal myocytes might occur by their active migration over the extracellular matrix or passively, when the structures to which they are anchored migrate or change position. Migrating cells, finding their way across a surface, characteristically exhibit branching lamellipodia and filopodia (Bray, 1992). Indeed, newly differentiated myocytes in the rat limb bud briefly exhibit a complex branched form as they search for their correct orientation (personal observation, M.J. Duxson). At the time when the myotomal fibres are altering their position and orientation, however, they do not show any kind of branching, as seen in our whole mount preparations. For example, as the dorsal-most fibres tilt upwards, the only visible change of morphology is a shortening of the myocytes accompanied by an expansion of the intersegmental territory occupied by Scx-positive cells. These observations suggest that active myocyte migration is unlikely to be involved in the morphogenesis of the epaxial muscles. Similarly, during translocation of muscle fibres in the head and despite the presence of Lbx1,

8 FATE OF MAMMALIAN MYOTOME 2905 Fig. 4. Morphogenesis of transversospinal muscles in ScxGFP mouse embryos. Immunohistochemistry on 20-mm transverse sections for tenascin (blue) and MHC (red) combined with intrinsic GFP signal (green) (A, C, F, H, J, L) and on whole mount specimens for MHC (red) and GFP (green) (B, D, E, G, I, K, M). B is a flattened confocal Z-stack, all other sagittal views are single 10-mm optical sections. Dotted lines in the transverse sections indicate the approximate plane of the accompanying sagittal sections. A,B: E11.5, hindlimb level. Scx-positive cells line the medial and ventral myotome and fill the intersegmental spaces (arrows). C E: E11.5, interlimb level. As the dorsalmost myocytes tilt upwards, Scx-positive cells invade between the myocytes (arrows) and cluster around their ends (arrowheads). F,G: E12.5, hindlimb level. Scx-positive cells remain prominent at the ends of the tilting fibres (arrowhead in G) and also fill the forming cleavage planes (arrows in F, G). H,I: E12.5, interlimb level. Dense clusters of Scx-positive cells lie at the rostral ends of the transversospinal muscle bundles (asterisks), overlying the spinous process. Long and short fibre bundles are apparent (blue highlighted fibres). J,K: E13.5, hindlimb level. The fibre bundles are more clearly demarcated. The long bundles attach to a Scx-positive lateral tendon sheath (asterisks). L,M: E13.5, interlimb level. MHC, myosin heavy chain; Scx, scleraxis; Dm, dermomyotome; Sc, sclerotome; Ep, epaxial; Hyp, hypaxial; Ve, vertebra; ts, transversospinalis; long, longissimus; ilio, iliocostalis; D, dorsal; V, ventral; M, medial; L, lateral; R, rostral; C, caudal. Fig. 6. Fig. 6. Differentiation and proliferation within Pax3 and Pax7 precursor populations. Serial sections of the epaxial muscle mass in an E14 rat embryo, at forelimb level, stained to show Pax 3 or Pax7 alongside markers for proliferation or differentiation. A,H: Low-magnification images showing the overall pattern of staining of the myotome with Pax3 and myogenin (A) or with Pax7 and myogenin (H). B G: Highmagnification panels showing details taken from an area indicated approximately by the box in A showing co-expression of Pax3 and myogenin (B D) or Pax3 and BrDU (E G). I N: High-magnification panels showing details of the boxed area in H, showing coexpression of Pax7 and myogenin (I K) or Pax7 and BrDU (L N). Many cells within both Pax3 and Pax7 populations have incorporated BrDU, indicating they are in an active proliferative state. Both populations also show coexpression of myogenin, a marker for terminal myogenic differentiation. Note that nuclei of two distinct cross-sectional sizes stain for myogenin: large nuclei are presumed to be those of myogenic precursors, and some of these co-stain for Pax3 or Pax7; small nuclei are those of differentiated fibres, and these never co-stain for Pax3 or Pax7.

9 2906 DERIES ET AL. Fig. 5. Association of Pax3- and Pax7-expressing muscle precursors with myotome and emerging epaxial muscle masses. Adjacent 10-mm transverse sections of rat embryos at forelimb level at stages from E12.5 to E15.5 to detect MHC and myogenin (left column), Pax3 and myogenin (middle column), or Pax7 and myogenin (right column). A C: E12.5. Arrows in B and C indicate intermingling of Pax3- or Pax7-positive cells with differentiated cells within the myotome. D F: E13.5. The expression of Pax3 has decreased within the myotome but persists laterally, where few differentiated cells are present (arrowhead in E). Pax7-positive cells are scattered throughout the muscle mass (F). G I: E14.0. Pax3-positive cells now lie more towards the lateral edge of the muscles, where differentiation is less advanced (arrowheads in H), whereas Pax7-positive progenitors are widespread. Arrows indicate forming cleavage planes. J O: E14.5 and E15.5. As segregation of the epaxial muscles continues, Pax3 becomes restricted to the least differentiated, ventral areas (arrowhead in K), while Pax7-positive cells are prominent throughout all muscle masses. MHC, myosin heavy chain; NT, neural tube; DRG, dorsal root ganglia; Dm, dermomyotome; M, medial; L, lateral; R, rostral; C, caudal.

10 FATE OF MAMMALIAN MYOTOME 2907 a migratory marker for precursor muscles in the trunk (Mootoosamy and Dietrich, 2002), the main hypothesis remains that the differentiated cells are moved by their environment, rather than by their own active migration (Noden and Trainor, 2005; Noden and Francis-West, 2006). If the muscle fibres are not actively moving, then what is the motor that drives them? The Scx-positive cells, which will form muscle tendons and connective tissues, are one possible candidate. Our observations show that these cells are tightly associated with the myotomal myocytes, and particularly with their rostral and caudal ends (Fig. 4). As the dorsalmost fibres start to move, separate clusters of Scx-positive cells move in association with them, and, at any one boundary, different clusters move in different directions. These observations are consistent with the connective tissues acting as a motor for the movement of differentiated muscle fibres, although they do not prove that this is the mechanism. Alternatively, the Scx-positive cells may simply form the attachment points to cartilaginous structures such as the vertebral processes, which are enlarging by appositional growth and carrying the myocytes along with them. To further investigate the role of Scx expressing connective tissue cells in muscle morphogenesis, it would be informative to disrupt them. Murchison et al. (2007) recently created an Scx null mutant mouse in which the early tendon progenitors seemed normal, but there was failure of differentiation of many tendons at the time when they normally begin to individuate and condense. Failure of tendon formation was particularly severe for the longer tendons that transmit force across joints, such as at the distal ends of many limb muscles and for the long tendons of the tail. Interestingly, function of the back muscles was particularly badly affected in Scx / animals, but this aspect of the phenotype was not investigated in detail. A tantalizing possibility is that normal morphogenesis of the epaxial muscles may have failed in the absence of maturation of the tendon progenitors, resulting in dysfunctional muscular anatomy. Alternately, Scx itself may not be critical for epaxial muscle morphogenesis, in which case it might be necessary to entirely delete the Scx-expressing cells at critical windows of time, perhaps using an inducible Scx-DTA construct, or utilizing some of the TGFbeta mutants in which Scx-expressing cells initially form, but fail to develop into tendons (Pryce et al., 2009). Epaxial Muscle Progenitor Populations Our results show that myogenic precursors expressing Pax3 and Pax7 are intermingled with the differentiated muscle fibres of the late myotome, as expected from previous reports (Gros et al., 2005; Relaix et al., 2005) and segregate along with each epaxial muscle mass as it cleaves. Both populations are actively proliferating throughout this period, as shown here by their incorporation of BrDU (Fig. 6E G, L N). Interestingly, although the patterns of Pax3 and Pax7 distribution are initially similar within the myotome (see Fig. 5B,C), once transformation towards the epaxial musculature begins, their distribution diverges. Pax3-positive cells steadily decline in frequency within the muscle, despite their proliferative activity, and become progressively restricted to the lateral and ventral edges of the differentiated muscle mass (Fig. 5E, H, K, N), where a proportion of cells co-express myogenin (Fig. 6B D). In contrast, from E13.5 onwards, Pax7-expressing progenitors are found in large numbers throughout the muscle mass, also showing coexpression with myogenin. The different spatial domains and dynamics of the two progenitor populations suggest they may be playing different roles in epaxial muscle development, as in the limb. In limb muscles, Pax3-positive muscle progenitors are associated with the embryonic phase of muscle development, when initial formation of primary myotubes occurs, while Pax7-positive progenitors drive later, foetal stages of muscle growth including secondary myogenesis (Biressi et al., 2007; Hutcheson et al., 2009). Our results in the epaxial muscles of the rat are broadly consistent with this pattern. Pax3-positive precursors are widespread throughout the myotome during its initial formation and growth and until E13.0, but then contract towards the edge of the epaxial muscle masses, where perhaps the last primary myotubes/myocytes are forming, just as the masses start to cleave. Loss of the highly proliferative Pax3- positive population at this stage (E ) seems likely to result from a majority of the daughter cells transiting into expression of Pax7 (Hutcheson et al., 2009). Secondary myogenesis in rat epaxial muscles does not start until E16.5 (E14.5 in the mouse, Auda-Boucher et al., 1997), well after the time when we observe that the Pax3-positive population of progenitors has disappeared. Solely Pax7-positive progenitors must, therefore, be responsible for the secondary phase of myogenesis. There is, however, a crucial intermediate phase from E13.5 until E16.5, before secondary myogenesis commences, when many Pax7-positive nuclei co-express myogenin throughout the epaxial masses, in regions where no Pax3 expression remains. In this lag between primary and secondary myogenesis, few new myotubes form, suggesting that differentiating Pax7 nuclei are most likely to be fusing with existing primary myocytes, contributing to the switch from a mononuclear form to a multinuclear one and perhaps also driving changes in phenotype. Despite the parallels with limb muscle development, the origins and formation of the epaxial muscles remain unique in that at least a proportion of their primary myotubes are built upon the transformed scaffold of the larval myotomal fibres, rather than forming de novo from a migratory population of precursors. EXPERIMENTAL PROCEDURES Collection of Dated Embryos Wistar rat couples were mated overnight, with the morning on which a copulation plug was found considered embryonic day 0.5 (E0.5). At the required stages (between E11.5 and E15.5), pregnant females were deeply anaesthetised by intraperitoneal injection of xylazine (20 mg/kg; Wyeth- Ayerst Veterinary Laboratories, New York, NY) and ketamine (100 mg/ kg;

11 2908 DERIES ET AL. Monarch Pharmaceuticals, Bristol, TN). In some cases, the dams were injected intraperitoneally with bromodeoxyuridine (BrDU, 25 mg/kg) 1 hr before administration of the anaesthetic. Embryos were individually extracted from the uterus, then quickly beheaded and eviscerated before processing as below. All procedures were approved by the University of Otago Committee on the Ethical Use of Animals. Homozygous ScxGFP male mice (Pryce et al., 2007) were crossed to Swiss Webster females to produce ScxGFP-positive embryos at three different stages: E11.5, E12.5, and E13.5. Embryos were fixed in 2% PFA/PB overnightat4 C before processing for whole mount immunohistochemistry or frozen section immunohistochemistry as below. Whole Mount Fluorescent Immunohistochemistry All embryos were processed as described in Deries et al, (2008). In brief, tissues were fixed in 4% buffered paraformaldehyde, washed, dehydrated into methanol, then stored in a 1:4 mixture of DMSO:methanol for 3 weeks to permeabilise tissues. Subsequently, they were washed in methanol, rehydrated, and passaged through a standard immunohistochemistry protocol, using an extended (overnight 2 day, 4 C) incubation in the primary antibodies and overnight incubation (4 C) in the secondary antibodies, with 20% DMSO present in the diluent. Embryos were finally washed extensively, dehydrated in methanol, then cleared and mounted in BA:BB (1:2, benzyl alcohol:benzyl benzoate) on concave well slides. Fluorescent Immunohistochemistry on Frozen Sections Embryos for frozen sectioning were prepared exactly as described in Bajanca et al. (2004). Briefly, embryos were fixed overnight in 0.2% paraformaldehyde, then progressively processed into a sucrose/gelatine mixture, which was then used as the cryoembedding medium. Sections were either 10 or 20 mm in thickness and were fixed on the slides with 0.2% paraformaldehyde in phosphate buffer (PFA/PB) before processing through a standard immunohistochemical protocol. For immunodetection of incorporated BrDU, sections were pre-incubated in 4M hydrochloric acid at 37 C for 10 min to denature DNA, then washed in 0.1M sodium tetraborate to neutralise, before immunostaining. TUNEL Combined With Immunohistochemistry TUNEL (In Situ Cell Death Detection Kit, Fluorescein, Roche Applied Science, Nutley, NJ) was used to detect apoptotic cells. It was combined with fluorescent immunohistochemistry to detect muscle cells using a method adapted from Hurst et al (2006). Embryos prepared for immunohistochemistry were cryosectioned at 8 mm and sections dried then fixed on the slide with 0.2% PFA/PB. After incubation with primary antibodies, the TUNEL reaction was performed according to the manufacturer s protocol. The final step was the secondary antibody incubation. Antibodies Differentiated muscle cells were detected with a mix of mouse monoclonal antibodies to embryonic myosin heavy chain (MHC) (F59; 1:20; provided by F. Stockdale) and pan-mhc (MF20; 1:20; provided by D. Fischman). Nuclei of muscle progenitor cells were detected with mouse monoclonal anti-pax3 (Developmental Studies Hybridoma Bank [DSHB]; 1:100) and anti-pax7 (DSHB; 1:20) antibodies; myoblast nuclei were detected with a mouse monoclonal antibody against MyoD (BD Pharmigen TM ; San Jose, CA, 1:100); and nuclei of differentiated muscle cells with a rabbit polyclonal antibody to myogenin (Santa Cruz Biotechnology, Inc., Santa Cruz, CA; 1:200). Connective tissues in rat embryos were detected with a rabbit polyclonal antibody to tenascin (antibody 2873 provided by H Erickson; 1:500); while developing tendons and muscle connective tissue in ScxGFP mice were detected with a rabbit polyclonal anti- GFP (Molecular Probes, Eugene, OR; 1:5,000). Caspase-3 detection used a rabbit polyclonal antibody against active forms of caspase-3 (R&D Systems, Minneapolis, MN; 1:200). BrDu detection used a sheep polyclonal antibody (Abcam, Cambridge, MA, ab1893; 1:50). Primary antibodies were detected with goat anti-mouse, goat anti-rabbit, or goat anti-sheep F(ab 0 )2 fragment secondary antibodies conjugated to Alexa fluor VR 568/ 488/ 633 as appropriate (Molecular Probes; all at 1:2,000). Cell Counting Dual labelling of Pax3 or Pax7 with BrDU or myogenin was assessed in at least 4 sections from each of 2 animals, with sections selected in a random systematic fashion from a serial set through the forelimb level of the myotome. A counting grid was placed over the image of each selected section and all Pax-positive cells falling within every fourth grid square (in the red channel) were assessed for dual staining with myogenin or BrDU in the green channel. A minimum of 145 and a maximum of 385 Pax-positive cells were assessed for each animal and staining protocol. Imaging Specimens (sections and whole mount) were viewed with a Zeiss laser scanning confocal microscope and digital image stacks manipulated and flattened using ImageJ software (Rasband, W.S., ImageJ, U.S. National Institutes of Health, nih.gov/ij/, ). For whole mount specimens, up to 50 separate Z- stacks were taken in register to cover the whole surface of the specimen. Each stack was then split into individual 10-mm optical sections and sections from the same layer montaged to obtain an image of the whole specimen at each level. Montages were then recombined into stacks to produce the final Z-series that were then manipulated with the ImageJ software. Figures were produced with Adobe Photoshop and Adobe Illustrator (VC Adobe Systems Incorporated, San Jose, CA) software packages. ACKNOWLEDGMENTS The authors acknowledge the support of the Department of Anatomy and Structural Biology in providing a PhD bursary to Marianne Deries, and to

12 FATE OF MAMMALIAN MYOTOME 2909 the University of Otago for internal grants that supported other aspects of the work. The MF20 monoclonal antibody was a gift to the authors from Professor Don Fischman, the F59 antibody a gift from Professor Frank Stockdale, while the antibodies against Pax3 and Pax7 came from the Developmental Studies Hybridoma Bank, developed under the auspices of the NICHD, and maintained by the University of Iowa. We also thank our colleagues Lorryn Fisher for untiring technical assistance, Andrew McNaughton for confocal support, Jon Cornwall for help in anatomical interpretation, and Philip Sheard and John Harris for useful discussions. REFERENCES Auda-Boucher G, Jarno V, Fournier-Thibault C, Butler-Browne G, Fontaine- Perus J Acetylcholine receptor formation in mouse-chick chimera. Exp Cell Res 236: Bajanca F, Luz M, Duxson MJ, Thorsteinsdottir S Integrins in the mouse myotome: developmental changes and differences between the epaxial and hypaxial lineage. Dev Dyn 231: Bardeen CR The development of the musculature of the body wall in the pig, including its histogenesis and its relations to the myotomes and to the skeletal and nervous appartatus. Johns Hopkins Hosp Rep 9: Ben-Yair R, Kalcheim C Lineage analysis of the avian dermomyotome sheet reveals the existence of single cells with both dermal and muscle progenitor fates. Development 132: Biressi S, Tagliafico E, Lamorte G, Monteverde S, Tenedini E, Roncaglia E, Ferrari S, Cusella-De Angelis MG, Tajbakhsh S, Cossu G Intrinsic phenotypic diversity of embryonic and fetal myoblasts is revealed by genomewide gene expression analysis on purified cells. Dev Biol 304: Bray D Cell movements. New York: Garland Publishing Inc. Brent AE, Schweitzer R, Tabin CJ A somitic compartment of tendon progenitors. Cell 113: Brent AE, Braun T, Tabin CJ Genetic analysis of interactions between the somitic muscle, cartilage and tendon cell lineages during mouse development. Development 132: Buckingham M Skeletal muscle formation in vertebrates. Curr Opin Genet Dev 11: Butcher EO The development of the somites in the white rat (mus norvegicus albinus) and the fate of the myotomes, neural tubes and gut in the tail. Am J Anat 44: Chevallier A, Kieny M On the role of the connective tissue in the patterning of the chick limb musculature. Wilhelm Roux s Arch 191: Christ B, Jacob HJ, Jacob M Experimental analysis of the origin of the wing musculature in avian embryos. Anat Embryol (Berl) 150: Christ B, Jacob M, Jacob HJ On the origin and development of the ventrolateral abdominal muscles in the avian embryo. An experimental and ultrastructural study. Anat Embryol (Berl) 166: Christ B, Ordahl CP Early stages of chick somite development. Anat Embryol (Berl) 191: Cinnamon Y, Kahane N, Kalcheim C Characterization of the early development of specific hypaxial muscles from the ventrolateral myotome. Development 126: Denetclaw WF, Jr., Christ B, Ordahl CP Location and growth of epaxial myotome precursor cells. Development 124: Deries M, Collins JJ, Duxson MJ The mammalian myotome: a muscle with no innervation. Evol Dev 10: Evans DJ, Valasek P, Schmidt C, Patel K Skeletal muscle translocation in vertebrates. Anat Embryol (Berl) 211 Suppl 1: Gros J, Scaal M, Marcelle C A twostep mechanism for myotome formation in chick. Dev Cell 6: Gros J, Manceau M, Thome V, Marcelle C A common somitic origin for embryonic muscle progenitors and satellite cells. Nature 435: Hollway G, Currie P Vertebrate myotome development. Birth Defects Res C Embryo Today 75: Hurst PR, Mora JM, Fenwick MA Caspase-3, TUNEL and ultrastructural studies of small follicles in adult human ovarian biopsies. Hum Reprod 21: Hutcheson DA, Zhao J, Merrell A, Haldar M, Kardon G Embryonic and fetal limb myogenic cells are derived from developmentally distinct progenitors and have different requirements for beta-catenin. Genes Dev 23: Kahane N, Cinnamon Y, Kalcheim C The origin and fate of pioneer myotomal cells in the avian embryo. Mech Dev 74: Kalcheim C, Cinnamon Y, Kahane N Myotome formation: a multistage process. Cell Tissue Res 296: Kardon G Muscle and tendon morphogenesis in the avian hind limb. Development 125: Kardon G, Campbell JK, Tabin CJ Local extrinsic signals determine muscle and endothelial cell fate and patterning in the vertebrate limb. Dev Cell 3: Kardon G, Harfe BD, Tabin CJ A Tcf4-positive mesodermal population provides a prepattern for vertebrate limb muscle patterning. Dev Cell 5: Kassar-Duchossoy L, Giacone E, Gayraud-Morel B, Jory A, Gomes D, Tajbakhsh S Pax3/Pax7 mark a novel population of primitive myogenic cells during development. Genes Dev 19: Kieny M, Chevallier A Autonomy of tendon development in the embryonic chick wing. J Embryol Exp Morphol 49: Lynch K Growth and metamorphosis of the rectus abdominis muscle in Rana pipiens. J. Morph. 182: McClearn D, Medville R, Noden D Muscle cell death during the development of head and neck muscles in the chick embryo. Dev Dyn 202: Mootoosamy RC, Dietrich S Distinct regulatory cascades for head and trunk myogenesis. Development 129: Murchison ND, Pryce BA, Conner DA, Keene DR, Olson EN, Tabin CJ, Schweitzer R Regulation of tendon differentiation by scleraxis distinguishes force-transmitting tendons from muscle-anchoring tendons. Development 134: Noden DM, Francis-West P The differentiation and morphogenesis of craniofacial muscles. Dev Dyn 235: Noden DM, Trainor PA Relations and interactions between cranial mesoderm and neural crest populations. J Anat 207: Pryce BA, Brent AE, Murchison ND, Tabin CJ, Schweitzer R Generation of transgenic tendon reporters, ScxGFP and ScxAP, using regulatory elements of the scleraxis gene. Dev Dyn 236: Pryce BA, Watson SS, Murchison ND, Staverosky JA, Dunker N, Schweitzer R Recruitment and maintenance of tendon progenitors by TGFbeta signaling are essential for tendon formation. Development 136: Rees E, Young RD, Evans DJ Spatial and temporal contribution of somitic myoblasts to avian hind limb muscles. Dev Biol 253: Relaix F, Rocancourt D, Mansouri A, Buckingham M A Pax3/Pax7- dependent population of skeletal muscle progenitor cells. Nature 435: Riou JF, Umbhauer M, Shi DL, Boucaut JC Tenascin: a potential modulator of cell-extracellular matrix interactions during vertebrate embryogenesis. Biol Cell 75:1 9. Rizk NN, Adieb N The development of the anterior abdominal wall in the rat in the light of a new anatomical description. J Anat 134: Sato T Innervation and morphology of the musculi levatores costarum longi. Proc Jpn Acad 49: Schweitzer R, Chyung JH, Murtaugh LC, Brent AE, Rosen V, Olson EN, Lassar A, Tabin CJ Analysis of the tendon cell fate using Scleraxis, a specific marker for tendons and ligaments. Development 128:

Life Sciences For NET & SLET Exams Of UGC-CSIR. Section B and C. Volume-08. Contents A. BASIC CONCEPT OF DEVELOPMENT 1

Life Sciences For NET & SLET Exams Of UGC-CSIR. Section B and C. Volume-08. Contents A. BASIC CONCEPT OF DEVELOPMENT 1 Section B and C Volume-08 Contents 5. DEVELOPMENTAL BIOLOGY A. BASIC CONCEPT OF DEVELOPMENT 1 B. GAMETOGENESIS, FERTILIZATION AND EARLY DEVELOPMENT 23 C. MORPHOGENESIS AND ORGANOGENESIS IN ANIMALS 91 0

More information

Paraxial and Intermediate Mesoderm

Paraxial and Intermediate Mesoderm Biology 4361 Paraxial and Intermediate Mesoderm December 7, 2006 Major Mesoderm Lineages Mesodermal subdivisions are specified along a mediolateral axis by increasing amounts of BMPs more lateral mesoderm

More information

Paraxial and Intermediate Mesoderm

Paraxial and Intermediate Mesoderm Biology 4361 Paraxial and Intermediate Mesoderm December 6, 2007 Mesoderm Formation Chick Major Mesoderm Lineages Mesodermal subdivisions are specified along a mediolateral axis by increasing amounts of

More information

Paraxial and Intermediate Mesoderm

Paraxial and Intermediate Mesoderm Biology 4361 Paraxial and Intermediate Mesoderm December 6, 2007 Mesoderm Formation Chick Major Mesoderm Lineages Mesodermal subdivisions are specified along a mediolateral axis by increasing amounts of

More information

Characterization of the early development of specific hypaxial muscles from the ventrolateral myotome

Characterization of the early development of specific hypaxial muscles from the ventrolateral myotome Development 126, 4305-4315 (1999) Printed in Great Britain The Company of Biologists Limited 1999 DEV1451 4305 Characterization of the early development of specific hypaxial muscles from the ventrolateral

More information

Differential regulation of epaxial and hypaxial muscle development by Paraxis

Differential regulation of epaxial and hypaxial muscle development by Paraxis Development 126, 5217-5229 (1999) Printed in Great Britain The Company of Biologists Limited 1999 DEV1449 5217 Differential regulation of epaxial and hypaxial muscle development by Paraxis Jeanne Wilson-Rawls*,

More information

Mediolateral somitic origin of ribs and dermis determined by quail-chick chimeras

Mediolateral somitic origin of ribs and dermis determined by quail-chick chimeras Development 127, 4611-4617 (2000) Printed in Great Britain The Company of Biologists Limited 2000 DEV2597 4611 Mediolateral somitic origin of ribs and dermis determined by quail-chick chimeras Isabel Olivera-Martinez

More information

Integrins in the Mouse Myotome: Developmental Changes and Differences Between the Epaxial and Hypaxial Lineage

Integrins in the Mouse Myotome: Developmental Changes and Differences Between the Epaxial and Hypaxial Lineage DEVELOPMENTAL DYNAMICS 231:402 415, 2004 PATTERNS & PHENOTYPES Integrins in the Mouse Myotome: Developmental Changes and Differences Between the Epaxial and Hypaxial Lineage Fernanda Bajanca, 1,2 Marta

More information

Nitza Kahane, Yuval Cinnamon, Ido Bachelet and Chaya Kalcheim* SUMMARY

Nitza Kahane, Yuval Cinnamon, Ido Bachelet and Chaya Kalcheim* SUMMARY Development 128, 2187-2198 (2001) Printed in Great Britain The Company of Biologists Limited 2001 DEV1674 2187 The third wave of myotome colonization by mitotically competent progenitors: regulating the

More information

Smooth muscle of the dorsal aorta shares a common clonal origin with skeletal muscle of the myotome

Smooth muscle of the dorsal aorta shares a common clonal origin with skeletal muscle of the myotome RESEARCH ARTICLE 3 Development 133, 3-49 doi:10.1242/dev.02226 Smooth muscle of the dorsal aorta shares a common clonal origin with skeletal muscle of the myotome Milan Esner 1, Sigolène M. Meilhac 1,,

More information

2/23/09. Regional differentiation of mesoderm. Morphological changes at early postgastrulation. Segments organize the body plan during embryogenesis

2/23/09. Regional differentiation of mesoderm. Morphological changes at early postgastrulation. Segments organize the body plan during embryogenesis Regional differentiation of mesoderm Axial Paraxial Intermediate Somatic Splanchnic Chick embryo Morphological changes at early postgastrulation stages Segments organize the body plan during embryogenesis

More information

Results and Problems in Cell Differentiation. Series Editor: Hennig

Results and Problems in Cell Differentiation. Series Editor: Hennig Results and Problems in Cell Differentiation Series Editor: w. 38 Hennig Springer-Verlag Berlin Heidelberg GmbH Beate Brand-Saberi (Ed.) Vertebrate Myogenesis With 31 Figures Springer Professor Dr. Beate

More information

Paraxial and Intermediate Mesoderm

Paraxial and Intermediate Mesoderm Biology 4361 Paraxial and Intermediate Mesoderm July 28, 2008 Paraxial and Intermediate Mesoderm Overview Development of major mesodermal lineages Somites: formation specification and differentiation Mesodermal

More information

Relationship between Neural Crest Cells and Cranial Mesoderm during Head Muscle Development

Relationship between Neural Crest Cells and Cranial Mesoderm during Head Muscle Development Relationship between Neural Crest Cells and Cranial Mesoderm during Head Muscle Development Julien Grenier, Marie-Aimée Teillet, Raphaëlle Grifone, Robert G Kelly, Delphine Duprez To cite this version:

More information

18.4 Embryonic development involves cell division, cell differentiation, and morphogenesis

18.4 Embryonic development involves cell division, cell differentiation, and morphogenesis 18.4 Embryonic development involves cell division, cell differentiation, and morphogenesis An organism arises from a fertilized egg cell as the result of three interrelated processes: cell division, cell

More information

Role of Organizer Chages in Late Frog Embryos

Role of Organizer Chages in Late Frog Embryos Ectoderm Germ Layer Frog Fate Map Frog Fate Map Role of Organizer Chages in Late Frog Embryos Organizer forms three distinct regions Notochord formation in chick Beta-catenin localization How does beta-catenin

More information

Sonic hedgehog (Shh) signalling in the rabbit embryo

Sonic hedgehog (Shh) signalling in the rabbit embryo Sonic hedgehog (Shh) signalling in the rabbit embryo In the first part of this thesis work the physical properties of cilia-driven leftward flow were characterised in the rabbit embryo. Since its discovery

More information

Mesoderm Development

Mesoderm Development Quiz rules: Spread out across available tables No phones, text books, or (lecture) notes on your desks No consultation with your colleagues No websites open other than the Quiz page No screen snap shots

More information

inhibition of embryonic myogenesis

inhibition of embryonic myogenesis Dermomyotome development and myotome patterning require Tbx6 inhibition of embryonic myogenesis Stefanie E. Windner 1,2, Nathan C. Bird 1, Sara Elizabeth Patterson 1, Rosemarie Doris 1, and Stephen Henri

More information

Limb Development Involving the development of the appendicular skeleton and muscles

Limb Development Involving the development of the appendicular skeleton and muscles Limb Development Involving the development of the appendicular skeleton and muscles 1 Objectives Timing and location of limb bud development The tissues from which limb buds are made Determining the position

More information

1. What are the three general areas of the developing vertebrate limb? 2. What embryonic regions contribute to the developing limb bud?

1. What are the three general areas of the developing vertebrate limb? 2. What embryonic regions contribute to the developing limb bud? Study Questions - Lecture 17 & 18 1. What are the three general areas of the developing vertebrate limb? The three general areas of the developing vertebrate limb are the proximal stylopod, zeugopod, and

More information

Emergence of determined myotome precursor cells in the somite

Emergence of determined myotome precursor cells in the somite Development 124, 4983-4997 (1997) Printed in Great Britain The Company of Biologists Limited 1997 DEV3690 4983 Emergence of determined myotome precursor cells in the somite Brian A. Williams and Charles

More information

Axis Specification in Drosophila

Axis Specification in Drosophila Developmental Biology Biology 4361 Axis Specification in Drosophila November 2, 2006 Axis Specification in Drosophila Fertilization Superficial cleavage Gastrulation Drosophila body plan Oocyte formation

More information

A distinct development programme for the cranial paraxial mesoderm

A distinct development programme for the cranial paraxial mesoderm A distinct development programme for the cranial paraxial mesoderm Article (Published Version) Hacker, A and Guthrie, S (1998) A distinct development programme for the cranial paraxial mesoderm. Development

More information

Activation of different myogenic pathways: myf-5 is induced by the neural tube and MyoD by the dorsal ectoderm in mouse paraxial mesoderm

Activation of different myogenic pathways: myf-5 is induced by the neural tube and MyoD by the dorsal ectoderm in mouse paraxial mesoderm Development 122, 429-437 (1996) Printed in Great Britain The Company of Biologists Limited 1996 DEV2041 429 Activation of different myogenic pathways: myf-5 is induced by the neural tube and MyoD by the

More information

Developmental Biology 3230 Midterm Exam 1 March 2006

Developmental Biology 3230 Midterm Exam 1 March 2006 Name Developmental Biology 3230 Midterm Exam 1 March 2006 1. (20pts) Regeneration occurs to some degree to most metazoans. When you remove the head of a hydra a new one regenerates. Graph the inhibitor

More information

Developmental Zoology. Ectodermal derivatives (ZOO ) Developmental Stages. Developmental Stages

Developmental Zoology. Ectodermal derivatives (ZOO ) Developmental Stages. Developmental Stages Developmental Zoology (ZOO 228.1.0) Ectodermal derivatives 1 Developmental Stages Ø Early Development Fertilization Cleavage Gastrulation Neurulation Ø Later Development Organogenesis Larval molts Metamorphosis

More information

Axis Specification in Drosophila

Axis Specification in Drosophila Developmental Biology Biology 4361 Axis Specification in Drosophila November 6, 2007 Axis Specification in Drosophila Fertilization Superficial cleavage Gastrulation Drosophila body plan Oocyte formation

More information

Initial steps of myogenesis in somites are independent of influence from axial structures

Initial steps of myogenesis in somites are independent of influence from axial structures Development 120, 3073-3082 (1994) Printed in Great Britain The Company of Biologists Limited 1994 3073 Initial steps of myogenesis in somites are independent of influence from axial structures Eva Bober

More information

Morphogenesis and specification of the muscle lineage during Xenopus laevis embryo development. Armbien F. Sabillo

Morphogenesis and specification of the muscle lineage during Xenopus laevis embryo development. Armbien F. Sabillo Morphogenesis and specification of the muscle lineage during Xenopus laevis embryo development by Armbien F. Sabillo A dissertation submitted in partial satisfaction of the requirements for the degree

More information

Chapter 11. Development: Differentiation and Determination

Chapter 11. Development: Differentiation and Determination KAP Biology Dept Kenyon College Differential gene expression and development Mechanisms of cellular determination Induction Pattern formation Chapter 11. Development: Differentiation and Determination

More information

Nature Neuroscience: doi: /nn.2662

Nature Neuroscience: doi: /nn.2662 Supplementary Figure 1 Atlastin phylogeny and homology. (a) Maximum likelihood phylogenetic tree based on 18 Atlastin-1 sequences using the program Quicktree. Numbers at internal nodes correspond to bootstrap

More information

Chick muscle development

Chick muscle development Int. J. Dev. Biol. 62: 127-136 (2018) https://doi.org/10.1387/ijdb.170312cm www.intjdevbiol.com Chick muscle development MARTIN SCAAL 1 and CHRISTOPHE MARCELLE*,2,3 1 Institute of Anatomy II, University

More information

Introduction to Embryology. He who sees things grow from the beginning will have the finest view of them.

Introduction to Embryology. He who sees things grow from the beginning will have the finest view of them. He who sees things grow from the beginning will have the finest view of them. Aristotle 384 322 B.C. Introduction to Embryology This lecture will introduce you to the science of developmental biology or

More information

Developmental Biology Lecture Outlines

Developmental Biology Lecture Outlines Developmental Biology Lecture Outlines Lecture 01: Introduction Course content Developmental Biology Obsolete hypotheses Current theory Lecture 02: Gametogenesis Spermatozoa Spermatozoon function Spermatozoon

More information

S. Tajbakhsh 1, U. Borello 2, E. Vivarelli 2, R. Kelly 1, J. Papkoff 3, D. Duprez 4, M. Buckingham 1 and G. Cossu 2, * SUMMARY

S. Tajbakhsh 1, U. Borello 2, E. Vivarelli 2, R. Kelly 1, J. Papkoff 3, D. Duprez 4, M. Buckingham 1 and G. Cossu 2, * SUMMARY Development 125, 4155-4162 (1998) Printed in Great Britain The Company of Biologists Limited 1998 DEV2314 4155 Differential activation of Myf5 and MyoD by different Wnts in explants of mouse paraxial mesoderm

More information

Biology 218, practise Exam 2, 2011

Biology 218, practise Exam 2, 2011 Figure 3 The long-range effect of Sqt does not depend on the induction of the endogenous cyc or sqt genes. a, Design and predictions for the experiments shown in b-e. b-e, Single-cell injection of 4 pg

More information

Dynamic Somite Cell Rearrangements Lead to Distinct Waves of Myotome Growth

Dynamic Somite Cell Rearrangements Lead to Distinct Waves of Myotome Growth Wesleyan University WesScholar Faculty Scholarship Biology 2007 Dynamic Somite Cell Rearrangements Lead to Distinct Waves of Myotome Growth F. Stellabotte B. Dobbs-McAuliffe D. A. Fernandez X. Feng Stephen

More information

Hox genes and morphological identity: axial versus lateral patterning in the

Hox genes and morphological identity: axial versus lateral patterning in the Development 127, 4265-4275 (2000) Printed in Great Britain The Company of Biologists Limited 2000 DEV2574 4265 Hox genes and morphological identity: axial versus lateral patterning in the vertebrate mesoderm

More information

Publisher Rights Statement: Publisher's Version/PDF: green tick author can archive publisher's version/pdf

Publisher Rights Statement: Publisher's Version/PDF: green tick author can archive publisher's version/pdf Edinburgh Research Explorer Notch and bone morphogenetic protein differentially act on dermomyotome cells to generate endothelium, smooth, and striated muscle Citation for published version: Ben-Yair,

More information

Cellular Neurobiology BIPN 140 Fall 2016 Problem Set #8

Cellular Neurobiology BIPN 140 Fall 2016 Problem Set #8 Cellular Neurobiology BIPN 140 Fall 2016 Problem Set #8 1. Inductive signaling is a hallmark of vertebrate and mammalian development. In early neural development, there are multiple signaling pathways

More information

Lesson 10. References: Chapter 8: Reading for Next Lesson: Chapter 8:

Lesson 10. References: Chapter 8: Reading for Next Lesson: Chapter 8: Lesson 10 Lesson Outline: General Features of the Skeletal System Axial Skeleton (Vertebral Column) Ontogenetic Origins (Morphogenesis) of the Skeletal System Functions of the Axial Skeleton Phylogenetic

More information

Bio Section III Organogenesis. The Neural Crest and Axonal Specification. Student Learning Objectives. Student Learning Objectives

Bio Section III Organogenesis. The Neural Crest and Axonal Specification. Student Learning Objectives. Student Learning Objectives Bio 127 - Section III Organogenesis The Neural Crest and Axonal Specification Gilbert 9e Chapter 10 Student Learning Objectives 1. You should understand that the neural crest is an evolutionary advancement

More information

Chapter 18 Lecture. Concepts of Genetics. Tenth Edition. Developmental Genetics

Chapter 18 Lecture. Concepts of Genetics. Tenth Edition. Developmental Genetics Chapter 18 Lecture Concepts of Genetics Tenth Edition Developmental Genetics Chapter Contents 18.1 Differentiated States Develop from Coordinated Programs of Gene Expression 18.2 Evolutionary Conservation

More information

Hedgehog Acts Directly on the Zebrafish Dermomyotome to Promote Myogenic Differentiation

Hedgehog Acts Directly on the Zebrafish Dermomyotome to Promote Myogenic Differentiation Wesleyan University WesScholar Division III Faculty Publications Natural Sciences and Mathematics 8-30-2006 Hedgehog Acts Directly on the Zebrafish Dermomyotome to Promote Myogenic Differentiation Stephen

More information

Pax3 and Dach2 Positive Regulation in the Developing Somite

Pax3 and Dach2 Positive Regulation in the Developing Somite DEVELOPMENTAL DYNAMICS 224:350 355 (2002) BRIEF COMMUNICATIONS Pax3 and Dach2 Positive Regulation in the Developing Somite G. KARDON, T.A. HEANUE, AND C.J. TABIN* Department of Genetics, Harvard Medical

More information

The growth rate of sensory nerve fibres in the mammalian embryo

The growth rate of sensory nerve fibres in the mammalian embryo Development 00, 307-3 (987) Printed in Great Britain The Company of Biologists Limited 987 307 The growth rate of sensory nerve fibres in the mammalian embryo ALUN M. DAVIES Department of Anatomy, Si George's

More information

Questions in developmental biology. Differentiation Morphogenesis Growth/apoptosis Reproduction Evolution Environmental integration

Questions in developmental biology. Differentiation Morphogenesis Growth/apoptosis Reproduction Evolution Environmental integration Questions in developmental biology Differentiation Morphogenesis Growth/apoptosis Reproduction Evolution Environmental integration Representative cell types of a vertebrate zygote => embryo => adult differentiation

More information

The origin of secondary myotubes in mammalian skeletal muscles: ultrastructural studies

The origin of secondary myotubes in mammalian skeletal muscles: ultrastructural studies Development 107, 743-750 (1989) Printed in Great Britain The Company of Biologists Limited 1989 743 The origin of secondary myotubes in mammalian skeletal muscles: ultrastructural studies M. J. DUXSON

More information

Nature Biotechnology: doi: /nbt Supplementary Figure 1. Overexpression of YFP::GPR-1 in the germline.

Nature Biotechnology: doi: /nbt Supplementary Figure 1. Overexpression of YFP::GPR-1 in the germline. Supplementary Figure 1 Overexpression of YFP::GPR-1 in the germline. The pie-1 promoter and 3 utr were used to express yfp::gpr-1 in the germline. Expression levels from the yfp::gpr-1(cai 1.0)-expressing

More information

Wnt signaling from the dorsal neural tube is required for the formation of the medial dermomyotome

Wnt signaling from the dorsal neural tube is required for the formation of the medial dermomyotome Development 125, 4969-4976 (1998) Printed in Great Britain The Company of Biologists Limited 1998 DEV4080 4969 Wnt signaling from the dorsal neural tube is required for the formation of the medial dermomyotome

More information

Distinct regulatory cascades for head and trunk myogenesis

Distinct regulatory cascades for head and trunk myogenesis Development 129, 573-583 (2002) Printed in Great Britain The Company of Biologists Limited 2002 DEV2806 573 Distinct regulatory cascades for head and trunk myogenesis Roy C. Mootoosamy and Susanne Dietrich*

More information

Determination of epithelial half-somites in skeletal morphogenesis

Determination of epithelial half-somites in skeletal morphogenesis Development 116, 441-445 (1992) Printed in Great Britain The Company of Biologists Limited 1992 441 Determination of epithelial half-somites in skeletal morphogenesis RONALD S. GOLDSTEIN * and CHAYA KALCHEIM

More information

Axis Specification in Drosophila

Axis Specification in Drosophila Developmental Biology Biology 4361 Axis Specification in Drosophila July 9, 2008 Drosophila Development Overview Fertilization Cleavage Gastrulation Drosophila body plan Oocyte formation Genetic control

More information

Name. Biology Developmental Biology Winter Quarter 2013 KEY. Midterm 3

Name. Biology Developmental Biology Winter Quarter 2013 KEY. Midterm 3 Name 100 Total Points Open Book Biology 411 - Developmental Biology Winter Quarter 2013 KEY Midterm 3 Read the Following Instructions: * Answer 20 questions (5 points each) out of the available 25 questions

More information

Lectures 3, 9, 10, 11: Prenatal and

Lectures 3, 9, 10, 11: Prenatal and Animal growth and development 2 nd review Lectures 3, 9, 10, 11: Prenatal and Muscle Growth and Development Development Principles similarity among species progressive causal and irreversible; ing complexity

More information

MOLECULAR CONTROL OF EMBRYONIC PATTERN FORMATION

MOLECULAR CONTROL OF EMBRYONIC PATTERN FORMATION MOLECULAR CONTROL OF EMBRYONIC PATTERN FORMATION Drosophila is the best understood of all developmental systems, especially at the genetic level, and although it is an invertebrate it has had an enormous

More information

3/8/ Complex adaptations. 2. often a novel trait

3/8/ Complex adaptations. 2. often a novel trait Chapter 10 Adaptation: from genes to traits p. 302 10.1 Cascades of Genes (p. 304) 1. Complex adaptations A. Coexpressed traits selected for a common function, 2. often a novel trait A. not inherited from

More information

In ovo time-lapse analysis after dorsal neural tube ablation shows rerouting of chick hindbrain neural crest

In ovo time-lapse analysis after dorsal neural tube ablation shows rerouting of chick hindbrain neural crest In ovo time-lapse analysis after dorsal neural tube ablation shows rerouting of chick hindbrain neural crest Paul Kulesa, Marianne Bronner-Fraser and Scott Fraser (2000) Presented by Diandra Lucia Background

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature11589 Supplementary Figure 1 Ciona intestinalis and Petromyzon marinus neural crest expression domain comparison. Cartoon shows dorsal views of Ciona mid gastrula (left) and Petromyzon

More information

CELL REPLICATION. Fluorescent light microscopy showing mitosis, especially immunolabelled cytoskeleton and tubulin

CELL REPLICATION. Fluorescent light microscopy showing mitosis, especially immunolabelled cytoskeleton and tubulin CELL REPLICATION Fluorescent light microscopy showing mitosis, especially immunolabelled cytoskeleton and tubulin Cell REPLICATION PROLIFERATION MUTIPLICATION DIVISION CELL REPLICATION Fluorescent light

More information

Segment boundary formation in Drosophila embryos

Segment boundary formation in Drosophila embryos Segment boundary formation in Drosophila embryos Development 130, August 2003 Camilla W. Larsen, Elizabeth Hirst, Cyrille Alexandre and Jean Paul Vincent 1. Introduction: - Segment boundary formation:

More information

N-Cadherin/Catenin-Mediated Morphoregulation of Somite Formation

N-Cadherin/Catenin-Mediated Morphoregulation of Somite Formation DEVELOPMENTAL BIOLOGY 202, 85 102 (1998) ARTICLE NO. DB989025 N-Cadherin/Catenin-Mediated Morphoregulation of Somite Formation Kersti K. Linask,*,1 Cheryl Ludwig,* Ming-Da Han,* Xiao Liu,* Glenn L. Radice,

More information

Growth in the Larval Zebrafish Pectoral Fin and Trunk Musculature

Growth in the Larval Zebrafish Pectoral Fin and Trunk Musculature Wesleyan University WesScholar Division III Faculty Publications Natural Sciences and Mathematics 10-26-2007 Growth in the Larval Zebrafish Pectoral Fin and Trunk Musculature Stephen Devoto Wesleyan University,

More information

Supplementary Figure 1: Mechanism of Lbx2 action on the Wnt/ -catenin signalling pathway. (a) The Wnt/ -catenin signalling pathway and its

Supplementary Figure 1: Mechanism of Lbx2 action on the Wnt/ -catenin signalling pathway. (a) The Wnt/ -catenin signalling pathway and its Supplementary Figure 1: Mechanism of Lbx2 action on the Wnt/ -catenin signalling pathway. (a) The Wnt/ -catenin signalling pathway and its transcriptional activity in wild-type embryo. A gradient of canonical

More information

10/2/2015. Chapter 4. Determination and Differentiation. Neuroanatomical Diversity

10/2/2015. Chapter 4. Determination and Differentiation. Neuroanatomical Diversity Chapter 4 Determination and Differentiation Neuroanatomical Diversity 1 Neurochemical diversity: another important aspect of neuronal fate Neurotransmitters and their receptors Excitatory Glutamate Acetylcholine

More information

Chapter 4 Evaluating a potential interaction between deltex and git in Drosophila: genetic interaction, gene overexpression and cell biology assays.

Chapter 4 Evaluating a potential interaction between deltex and git in Drosophila: genetic interaction, gene overexpression and cell biology assays. Evaluating a potential interaction between deltex and git in Drosophila: genetic interaction, gene overexpression and cell biology assays. The data described in chapter 3 presented evidence that endogenous

More information

Formation of the Cortex

Formation of the Cortex Formation of the Cortex Neuronal Birthdating with 3 H-thymidine 3H-thymidine is incorporated into the DNA during the S-phase (replication of DNA). It marks all mitotic cells Quantitative technique. (you

More information

Name KEY. Biology Developmental Biology Winter Quarter Midterm 3 KEY

Name KEY. Biology Developmental Biology Winter Quarter Midterm 3 KEY Name KEY 100 Total Points Open Book Biology 411 - Developmental Biology Winter Quarter 2009 Midterm 3 KEY All of the 25 multi-choice questions are single-answer. Choose the best answer. (4 pts each) Place

More information

Intravital Imaging Reveals Ghost Fibers as Architectural Units Guiding Myogenic Progenitors during Regeneration

Intravital Imaging Reveals Ghost Fibers as Architectural Units Guiding Myogenic Progenitors during Regeneration Cell Stem Cell Supplemental Information Intravital Imaging Reveals Ghost Fibers as Architectural Units Guiding Myogenic Progenitors during Regeneration Micah T. Webster, Uri Manor, Jennifer Lippincott-Schwartz,

More information

4. Neural tube cells are specified by opposing dorsal-ventral gradients of a. Wnts and Nodal. b. FGF and Shh. c. BMPs and Wnts. d. BMPs and Shh.

4. Neural tube cells are specified by opposing dorsal-ventral gradients of a. Wnts and Nodal. b. FGF and Shh. c. BMPs and Wnts. d. BMPs and Shh. Biology 4361 Name: KEY Exam 4 ID#: August 1, 2008 Multiple choice (one point each; indicate the best answer) 1. Neural tube closure is accomplished by movement of the a. medial hinge point cells. b. medial

More information

A Crucial Role for Pax3 in the Development of the Hypaxial Musculature and the Long-Range Migration of Muscle Precursors

A Crucial Role for Pax3 in the Development of the Hypaxial Musculature and the Long-Range Migration of Muscle Precursors DEVELOPMENTAL BIOLOGY 203, 49 61 (1998) ARTICLE NO. DB989041 A Crucial Role for Pax3 in the Development of the Hypaxial Musculature and the Long-Range Migration of Muscle Precursors Patrick Tremblay,*,1,2

More information

Origin of muscle satellite cells in the Xenopus embryo

Origin of muscle satellite cells in the Xenopus embryo AND STEM CELLS RESEARCH ARTICLE 821 Development 138, 821-830 (2011) doi:10.1242/dev.056481 2011. Published by The Company of Biologists Ltd Origin of muscle satellite cells in the Xenopus embryo Randall

More information

!!!!!!!! DB3230 Midterm 2 12/13/2013 Name:

!!!!!!!! DB3230 Midterm 2 12/13/2013 Name: 1. (10 pts) Draw or describe the fate map of a late blastula stage sea urchin embryo. Draw or describe the corresponding fate map of the pluteus stage larva. Describe the sequence of gastrulation events

More information

Influence of the neural tube/notochord complex on MyoD expression and cellular proliferation in chicken embryos

Influence of the neural tube/notochord complex on MyoD expression and cellular proliferation in chicken embryos Brazilian Journal of Medical and Biological Research (2003) 36: 191-197 Influence of neural tube/notochord on MyoD expression ISSN 0100-879X 191 Influence of the neural tube/notochord complex on MyoD expression

More information

Redefining the Genetic Hierarchies Controlling Skeletal Myogenesis: Pax-3 and Myf-5 Act Upstream of MyoD

Redefining the Genetic Hierarchies Controlling Skeletal Myogenesis: Pax-3 and Myf-5 Act Upstream of MyoD Cell, Vol. 89, 127 138, April 4, 1997, Copyright 1997 by Cell Press Redefining the Genetic Hierarchies Controlling Skeletal Myogenesis: Pax-3 and Myf-5 Act Upstream of MyoD Shahragim Tajbakhsh,* Didier

More information

Homeotic Genes and Body Patterns

Homeotic Genes and Body Patterns Homeotic Genes and Body Patterns Every organism has a unique body pattern. Although specialized body structures, such as arms and legs, may be similar in makeup (both are made of muscle and bone), their

More information

Identification of separate slow and fast muscle precursor cells in vivo, prior to somite formation

Identification of separate slow and fast muscle precursor cells in vivo, prior to somite formation Development 122, 3371-3380 (1996) Printed in Great Britain The Company of Biologists Limited 1996 DEV9467 3371 Identification of separate slow and fast muscle precursor cells in vivo, prior to somite formation

More information

SIGNIFICANCE OF EMBRYOLOGY

SIGNIFICANCE OF EMBRYOLOGY This lecture will discuss the following topics : Definition of Embryology Significance of Embryology Old and New Frontiers Introduction to Molecular Regulation and Signaling Descriptive terms in Embryology

More information

The role of FGF2 in craniofacial skeletogenesis

The role of FGF2 in craniofacial skeletogenesis The role of FGF2 in craniofacial skeletogenesis P. Ferretti, S. Sarkar, R. Moore, A. Petiot, C. J. Chan and A. Copp Summary E vidence that the major craniosynostosis syndromes are caused by mutations in

More information

10/15/09. Tetrapod Limb Development & Pattern Formation. Developing limb region is an example of a morphogenetic field

10/15/09. Tetrapod Limb Development & Pattern Formation. Developing limb region is an example of a morphogenetic field Tetrapod Limb Development & Pattern Formation Figure 16.5(1) Limb Bud Formation derived from lateral plate (somatic) & paraxial (myotome) Fig. 16.2 Prospective Forelimb Field of Salamander Ambystoma maculatum

More information

Six1 and Six4 homeoproteins are required for Pax3 and Mrf expression during myogenesis in the mouse embryo

Six1 and Six4 homeoproteins are required for Pax3 and Mrf expression during myogenesis in the mouse embryo Research article and disease 2235 Six1 and Six4 homeoproteins are required for Pax3 and Mrf expression during myogenesis in the mouse embryo Raphaelle Grifone 1, Josiane Demignon 1, *, Christophe Houbron

More information

Dennis Summerbell*, Peter R. Ashby, Oliver Coutelle, David Cox*, Siu-Pok Yee and Peter W. J. Rigby*, SUMMARY

Dennis Summerbell*, Peter R. Ashby, Oliver Coutelle, David Cox*, Siu-Pok Yee and Peter W. J. Rigby*, SUMMARY Development 127, 3745-3757 (2000) Printed in Great Britain The Company of Biologists Limited 2000 DEV2592 3745 The expression of Myf5 in the developing mouse embryo is controlled by discrete and dispersed

More information

Mesoderm Divided into three main types - Paraxial (somite) - Intermediate - Lateral (somatic and splanchnic)

Mesoderm Divided into three main types - Paraxial (somite) - Intermediate - Lateral (somatic and splanchnic) Mesoderm Divided into three main types - Paraxial (somite) - Intermediate - Lateral (somatic and splanchnic) Fates of Mesoderm Paraxial - Dermis of skin - Axial Skeleton - Axial and limb muscles/tendons

More information

178 Part 3.2 SUMMARY INTRODUCTION

178 Part 3.2 SUMMARY INTRODUCTION 178 Part 3.2 Chapter # DYNAMIC FILTRATION OF VARIABILITY WITHIN EXPRESSION PATTERNS OF ZYGOTIC SEGMENTATION GENES IN DROSOPHILA Surkova S.Yu. *, Samsonova M.G. St. Petersburg State Polytechnical University,

More information

Chapter 10 Development and Differentiation

Chapter 10 Development and Differentiation Part III Organization of Cell Populations Chapter Since ancient times, people have wondered how organisms are formed during the developmental process, and many researchers have worked tirelessly in search

More information

Developmental genetics: finding the genes that regulate development

Developmental genetics: finding the genes that regulate development Developmental Biology BY1101 P. Murphy Lecture 9 Developmental genetics: finding the genes that regulate development Introduction The application of genetic analysis and DNA technology to the study of

More information

Cell-Cell Communication in Development

Cell-Cell Communication in Development Biology 4361 - Developmental Biology Cell-Cell Communication in Development October 2, 2007 Cell-Cell Communication - Topics Induction and competence Paracrine factors inducer molecules Signal transduction

More information

Inhibition of cranial neural crest cell development by vitamin A in the cultured chick embryo

Inhibition of cranial neural crest cell development by vitamin A in the cultured chick embryo /. Embryol. exp. Morph. Vol. 39, pp. 267-27J, 1977 267 Printed in Great Britain Inhibition of cranial neural crest cell development by vitamin A in the cultured chick embryo JOHN R. HASSELL, 1 JUDITH H.

More information

Where Do Bat Wings Come From?

Where Do Bat Wings Come From? Where o at Wings ome From? 1 ats are the only mammals that have evolved the power of flight. They can avoid obstacles and slip through tight spaces. Many species are nocturnal and use echolocation to guide

More information

Chapter 9. Benefits of Being Large. Levels of Organization in Organismal Complexity. Hierarchical Organization of Animal Complexity. Fig. 9.

Chapter 9. Benefits of Being Large. Levels of Organization in Organismal Complexity. Hierarchical Organization of Animal Complexity. Fig. 9. Copyright The McGraw-Hill Companies, Inc. Permission required for reproduction or display. Chapter 9 Architectural Pattern of an Animal Levels of Organization in Organismal Complexity Zoologists recognize

More information

Sonic hedgehog is required for survival of both myogenic and chondrogenic somitic lineages

Sonic hedgehog is required for survival of both myogenic and chondrogenic somitic lineages Development 125, 2019-2030 (1998) Printed in Great Britain The Company of Biologists Limited 1998 DEV2281 2019 Sonic hedgehog is required for survival of both myogenic and chondrogenic somitic lineages

More information

Bi 117 Final (60 pts) DUE by 11:00 am on March 15, 2012 Box by Beckman Institute B9 or to a TA

Bi 117 Final (60 pts) DUE by 11:00 am on March 15, 2012 Box by Beckman Institute B9 or to a TA Bi 117 Final (60 pts) DUE by 11:00 am on March 15, 2012 Box by Beckman Institute B9 or to a TA Instructor: Marianne Bronner Exam Length: 6 hours plus one 30-minute break at your discretion. It should take

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature09414 Supplementary Figure 1 FACS-isolated 8c hepatocytes are highly pure. a, Gating strategy for identifying hepatocyte populations based on DNA content. b, Detection of mchry and mchr9

More information

1/30/2009. Copyright The McGraw Hill Companies, Inc. Permission required for reproduction or display.

1/30/2009. Copyright The McGraw Hill Companies, Inc. Permission required for reproduction or display. CHAPTER 9 Architectural Pattern of an Animal New Designs for Living Zoologists recognize 34 major phyla of living multicellular animals Survivors of around 100 phyla that appeared 600 million years ago

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION DOI: 10.1038/ncb3267 Supplementary Figure 1 A group of genes required for formation or orientation of annular F-actin bundles and aecm ridges: RNAi phenotypes and their validation by standard mutations.

More information

Unit 4 Evaluation Question 1:

Unit 4 Evaluation Question 1: Name: Unit 4 Evaluation Question 1: /7 points A naturally occurring dominant mutant in mice is the Doublefoot (Dbf) mutant. Below is an image of the bones from a wildtype (wt) and Doublefoot mutant mouse.

More information

Supplementary Materials for

Supplementary Materials for www.sciencesignaling.org/cgi/content/full/6/301/ra98/dc1 Supplementary Materials for Regulation of Epithelial Morphogenesis by the G Protein Coupled Receptor Mist and Its Ligand Fog Alyssa J. Manning,

More information

presumptiv e germ layers during Gastrulatio n and neurulation Somites

presumptiv e germ layers during Gastrulatio n and neurulation Somites Vertebrate embryos are similar at the phylotypic stage Patterning the Vertebrate Body Plan II: Mesoderm & Early Nervous System Wolpert L, Beddington R, Jessell T, Lawrence P, Meyerowitz E, Smith J. (2001)

More information

Question Set # 4 Answer Key 7.22 Nov. 2002

Question Set # 4 Answer Key 7.22 Nov. 2002 Question Set # 4 Answer Key 7.22 Nov. 2002 1) A variety of reagents and approaches are frequently used by developmental biologists to understand the tissue interactions and molecular signaling pathways

More information