Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A Twist2-dependent progenitor cell contributes to adult skeletal muscle

Abstract

Skeletal muscle possesses remarkable regenerative potential due to satellite cells, an injury-responsive stem cell population located beneath the muscle basal lamina that expresses Pax7. By lineage tracing of progenitor cells expressing the Twist2 (Tw2) transcription factor in mice, we discovered a myogenic lineage that resides outside the basal lamina of adult skeletal muscle. Tw2+ progenitors are molecularly and anatomically distinct from satellite cells, are highly myogenic in vitro, and can fuse with themselves and with satellite cells. Tw2+ progenitors contribute specifically to type IIb/x myofibres during adulthood and muscle regeneration, and their genetic ablation causes wasting of type IIb myofibres. We show that Tw2 expression maintains progenitor cells in an undifferentiated state that is poised to initiate myogenesis in response to appropriate cues that extinguish Tw2 expression. Tw2-expressing myogenic progenitors represent a previously unrecognized, fibre-type-specific stem cell involved in postnatal muscle growth and regeneration.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Tw2-expressing and Pax7-expressing cells are distinct cell types in skeletal muscle.
Figure 2: Progressive contribution of Tw2+ cells to adult skeletal muscle.
Figure 3: Ablation of the Tw2+ lineage causes type-IIb-myofibre atrophy.
Figure 4: Tw2+ cells contribute to skeletal muscle regeneration.
Figure 5: Molecular profiling of the Tw2+ lineage in skeletal muscle.
Figure 6: Myogenic potential of Tw2+ cells in culture.
Figure 7: Myogenic and osteogenic potential of tdTO+/CD34 cells in vitro.
Figure 8: Overexpression of Tw2 blocks myogenesis in vitro.

Similar content being viewed by others

References

  1. Chang, N. C. & Rudnicki, M. A. Satellite cells: the architects of skeletal muscle. Curr. Top. Dev. Biol. 107, 161–181 (2014).

    CAS  PubMed  Google Scholar 

  2. Brack, A. S. & Rando, T. A. Tissue-specific stem cells: lessons from the skeletal muscle satellite cell. Cell Stem Cell 10, 504–514 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Sacco, A., Doyonnas, R., Kraft, P., Vitorovic, S. & Blau, H. M. Self-renewal and expansion of single transplanted muscle stem cells. Nature 456, 502–506 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Fry, C. S. et al. Inducible depletion of satellite cells in adult, sedentary mice impairs muscle regenerative capacity without affecting sarcopenia. Nat. Med. 21, 76–80 (2015).

    CAS  PubMed  Google Scholar 

  5. Keefe, A. C. et al. Muscle stem cells contribute to myofibres in sedentary adult mice. Nat. Commun. 6, 7087 (2015).

    CAS  PubMed  Google Scholar 

  6. McCarthy, J. J. et al. Effective fiber hypertrophy in satellite cell-depleted skeletal muscle. Development 138, 3657–3666 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Schiaffino, S. & Reggiani, C. Fiber types in mammalian skeletal muscles. Physiol. Rev. 91, 1447–1531 (2011).

    CAS  PubMed  Google Scholar 

  8. Wang, Y. & Pessin, J. E. Mechanisms for fiber-type specificity of skeletal muscle atrophy. Curr. Opin. Clin. Nutr. Metab. Care 16, 243–250 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Tonkin, J., Villarroya, F., Puri, P. L. & Vinciguerra, M. SIRT1 signaling as potential modulator of skeletal muscle diseases. Curr. Opin. Pharmacol. 12, 372–376 (2012).

    CAS  PubMed  Google Scholar 

  10. Arany, Z. PGC-1 coactivators and skeletal muscle adaptations in health and disease. Curr. Opin. Genet. Dev. 18, 426–434 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Pawlikowski, B., Pulliam, C., Betta, N. D., Kardon, G. & Olwin, B. B. Pervasive satellite cell contribution to uninjured adult muscle fibers. Skeletal Muscle 5, 42 (2015).

    PubMed  PubMed Central  Google Scholar 

  12. Barnes, R. M. & Firulli, A. B. A twist of insight—the role of Twist-family bHLH factors in development. Int. J. Dev. Biol. 53, 909–924 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Baylies, M. K. & Bate, M Twist: a myogenic switch in Drosophila. Science 272, 1481–1484 (1996).

    CAS  PubMed  Google Scholar 

  14. Cripps, R. M. et al. The myogenic regulatory gene Mef2 is a direct target for transcriptional activation by Twist during Drosophila myogenesis. Genes Dev. 12, 422–434 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Bate, M., Rushton, E. & Currie, D. A. Cells with persistent twist expression are the embryonic precursors of adult muscles in Drosophila. Development 113, 79–89 (1991).

    CAS  PubMed  Google Scholar 

  16. Cripps, R. M. & Olson, E. N. Twist is required for muscle template splitting during adult Drosophila myogenesis. Dev. Biol. 203, 106–115 (1998).

    CAS  PubMed  Google Scholar 

  17. Currie, D. A. & Bate, M. The development of adult abdominal muscles in Drosophila: myoblasts express twist and are associated with nerves. Development 113, 91–102 (1991).

    CAS  PubMed  Google Scholar 

  18. Fuchtbauer, E. M. Expression of M-twist during postimplantation development of the mouse. Dev. Dyn. 204, 316–322 (1995).

    CAS  PubMed  Google Scholar 

  19. Li, L., Cserjesi, P. & Olson, E. N. Dermo-1: a novel twist-related bHLH protein expressed in the developing dermis. Dev. Biol. 172, 280–292 (1995).

    CAS  PubMed  Google Scholar 

  20. Rohwedel, J., Horak, V., Hebrok, M., Fuchtbauer, E. M. & Wobus, A. M. M-twist expression inhibits mouse embryonic stem cell-derived myogenic differentiation in vitro. Exp. Cell Res. 220, 92–100 (1995).

    CAS  PubMed  Google Scholar 

  21. Hebrok, M., Wertz, K. & Fuchtbauer, E. M. M-twist is an inhibitor of muscle differentiation. Dev. Biol. 165, 537–544 (1994).

    CAS  PubMed  Google Scholar 

  22. Spicer, D. B., Rhee, J., Cheung, W. L. & Lassar, A. B. Inhibition of myogenic bHLH and MEF2 transcription factors by the bHLH protein Twist. Science 272, 1476–1480 (1996).

    CAS  PubMed  Google Scholar 

  23. Hjiantoniou, E. et al. Twist induces reversal of myotube formation. Differentiation 76, 182–192 (2008).

    CAS  PubMed  Google Scholar 

  24. Lepper, C., Conway, S. J. & Fan, C. M. Adult satellite cells and embryonic muscle progenitors have distinct genetic requirements. Nature 460, 627–631 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Sosic, D., Richardson, J. A., Yu, K., Ornitz, D. M. & Olson, E. N. Twist regulates cytokine gene expression through a negative feedback loop that represses NF-κB activity. Cell 112, 169–180 (2003).

    CAS  PubMed  Google Scholar 

  26. Yu, K. et al. Conditional inactivation of FGF receptor 2 reveals an essential role for FGF signaling in the regulation of osteoblast function and bone growth. Development 130, 3063–3074 (2003).

    CAS  PubMed  Google Scholar 

  27. Voehringer, D., Liang, H. E. & Locksley, R. M. Homeostasis and effector function of lymphopenia-induced ‘memory-like’ T cells in constitutively T cell-depleted mice. J. Immunol. 180, 4742–4753 (2008).

    CAS  PubMed  Google Scholar 

  28. Goebel, H. H. Desmin-related neuromuscular disorders. Muscle Nerve 18, 1306–1320 (1995).

    CAS  PubMed  Google Scholar 

  29. Helliwell, T. R. Lectin binding and desmin staining during bupivicaine-induced necrosis and regeneration in rat skeletal muscle. J. Pathol. 155, 317–326 (1988).

    CAS  PubMed  Google Scholar 

  30. Muzumdar, M. D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double-fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

    CAS  PubMed  Google Scholar 

  31. Mitchell, K. J. et al. Identification and characterization of a non-satellite cell muscle resident progenitor during postnatal development. Nat. Cell Biol. 12, 257–266 (2010).

    CAS  PubMed  Google Scholar 

  32. Dellavalle, A. et al. Pericytes resident in postnatal skeletal muscle differentiate into muscle fibres and generate satellite cells. Nat. Commun. 2, 499 (2011).

    CAS  PubMed  Google Scholar 

  33. Doyle, M. J. et al. Abcg2 labels multiple cell types in skeletal muscle and participates in muscle regeneration. J. Cell Biol. 195, 147–163 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. LaBarge, M. A. & Blau, H. M. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell 111, 589–601 (2002).

    CAS  PubMed  Google Scholar 

  35. Minasi, M. G. et al. The meso-angioblast: a multipotent, self-renewing cell that originates from the dorsal aorta and differentiates into most mesodermal tissues. Development 129, 2773–2783 (2002).

    CAS  PubMed  Google Scholar 

  36. Qu-Petersen, Z. et al. Identification of a novel population of muscle stem cells in mice: potential for muscle regeneration. J. Cell Biol. 157, 851–864 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Dellavalle, A. et al. Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat. Cell Biol. 9, 255–267 (2007).

    CAS  PubMed  Google Scholar 

  38. Joe, A. W. et al. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat. Cell Biol. 12, 153–163 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Kuang, S., Charge, S. B., Seale, P., Huh, M. & Rudnicki, M. A. Distinct roles for Pax7 and Pax3 in adult regenerative myogenesis. J. Cell Biol. 172, 103–113 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lepper, C., Partridge, T. A. & Fan, C. M. An absolute requirement for Pax7-positive satellite cells in acute injury-induced skeletal muscle regeneration. Development 138, 3639–3646 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Murphy, M. M., Lawson, J. A., Mathew, S. J., Hutcheson, D. A. & Kardon, G. Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration. Development 138, 3625–3637 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Sambasivan, R. et al. Pax7-expressing satellite cells are indispensable for adult skeletal muscle regeneration. Development 138, 3647–3656 (2011).

    CAS  PubMed  Google Scholar 

  43. Beck, B. et al. Different levels of Twist1 regulate skin tumor initiation, stemness, and progression. Cell Stem Cell 16, 67–79 (2015).

    CAS  PubMed  Google Scholar 

  44. Isenmann, S. et al. TWIST family of basic helix-loop-helix transcription factors mediate human mesenchymal stem cell growth and commitment. Stem Cells 27, 2457–2468 (2009).

    CAS  PubMed  Google Scholar 

  45. Schmidt, J. M. et al. Stem-cell-like properties and epithelial plasticity arise as stable traits after transient Twist1 activation. Cell Rep. 10, 131–139 (2015).

    CAS  PubMed  Google Scholar 

  46. Yang, J. et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117, 927–939 (2004).

    CAS  PubMed  Google Scholar 

  47. Michailovici, I., Eigler, T. & Tzahor, E. Craniofacial muscle development. Curr. Top. Dev. Biol. 115, 3–30 (2015).

    PubMed  Google Scholar 

  48. Webster, C., Silberstein, L., Hays, A. P. & Blau, H. M. Fast muscle fibers are preferentially affected in Duchenne muscular dystrophy. Cell 52, 503–513 (1988).

    CAS  PubMed  Google Scholar 

  49. Scott, W., Stevens, J. & Binder-Macleod, S. A. Human skeletal muscle fiber type classifications. Phys. Ther. 81, 1810–1816 (2001).

    CAS  PubMed  Google Scholar 

  50. Liu, N. et al. microRNA-206 promotes skeletal muscle regeneration and delays progression of Duchenne muscular dystrophy in mice. J. Clin. Invest. 122, 2054–2065 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Patel, R. K. & Jain, M. NGS QC Toolkit: a toolkit for quality control of next generation sequencing data. PLoS ONE 7, e30619 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  53. Liao, Y., Smyth, G. K. & Shi, W. FeatureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  54. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. EdgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to UT Southwestern Flow Cytometry Core Facility, the Moody Foundation Flow Cytometry Facility, and UTSW Genomics and Microarray Core Facility for technical help and service. We thank C. Nolen for technical assistance and D. Garry, M. Rudnicki, M. Buckingham and H. Sadek for constructive suggestions. This work was supported by grants from the NIH (HL-077439, AR-067294, HL-130253, DK-099653 and U01-HL-100401) and the Robert A. Welch Foundation (grant 1-0025 to E.N.O.). N.L. was supported by a Beginning-Grant-In-Aid (13BGIA17150004) from the American Heart Association. G.A.G. is a research Fellow supported by the Sarnoff Cardiovascular Research Foundation.

Author information

Authors and Affiliations

Authors

Contributions

Experiments were designed by N.L., G.A.G. and S.L. Experiments were performed by N.L., G.A.G., S.L., S.B., E.S.-O., J.M.S. and P.J. Data were interpreted by N.L., G.A.G., S.L., B.C., R.B.-D. and E.N.O. The paper was written by N.L. and E.N.O.

Corresponding authors

Correspondence to Ning Liu or Eric N. Olson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Analysis of Tw2 expression in skeletal muscle.

(a) CPM (counts per million) of Tw2, MyoD and Myh4 expression by RNA-seq in G/P muscles of WT mice at 1, 2 and 4 months of age. Data are mean ± s.e.m.N = 3 mice for each time point. (b) Real-time RT-PCR demonstrates Tw2 mRNA is enriched in mononuclear cells of the adult muscle (mono) compared to whole quadriceps muscle (quad). Data are mean ± s.e.m.; N = 3 independent experiments. (c) Immunostaining of Tw2 (red) and Pax7 (green) on transverse sections of G/P muscle of 12-month old WT mice. Myofibers were co-stained with wheat germ agglutinin (white) and DAPI (blue). Arrows indicate Pax7+ cells and arrowheads indicate Tw2+ cells. Scale bar, 50 μm. (d) Quantification of the number of Pax7+ (tdTO+), Tw2+, and Pax7+/Tw2+ double positive cells per field in CTL TA muscles of Pax7-CreERT2; R26-tdTO mice. For each muscle section, at least 6 different fields were quantified and averaged. Data are mean ± S.E.M; N = 3 mice. Statistic source data for a, b, d are provided in Supplementary Table 3.

Supplementary Figure 2 Generation of Tw2-CreERT2 mice and analysis of the Tw2 lineage tracing.

(a) Targeting strategy of the Tw2-CreERT2 allele. Coding region of the Tw2 gene was replaced by a CreERT2-Frt-Neo-Frt cassette by homologous recombination in ES cells. Out of 500 ES cell clones 10% were correctly targeted. (b) Southern blot analysis of ES cells using 5′ probe and 3′ probes to demonstrate correct targeting. Unprocessed original scans of blots are shown in Supplementary Fig. 9. (c) In the absence of TMX treatment, tdTO is not detected in muscles of Tw2-CreERT2; R26-tdTO mice at 9 months of age. Scale bar, 100 μm. (d) Whole mount images showing tdTO+ muscles at 10 days (left) and 5 months (right) post-TMX. At 5 months, all muscles examined showed strong tdTO signals except for the tongue muscle. (e) Transverse-sections of tongue and masseter muscles from Tw2-CreERT2; R26- tdTO mice at 4 months post-TMX were co-stained with My32 (green) and DAPI (blue). Scale bar, 100 μm. (f) Longitudinal-sections of TA muscle from Tw2-CreERT2; R26-tdTO mice at 10 days and 4 months post-TMX were co-stained with My32 (green), Laminin (white) and DAPI (blue). tdTO signal expands through the entire myofibers. Scale bar, 100 μm. (g) Transverse sections of Tw2-CreERT2; R26-tdTO mice at indicated days post TMX were co-stained with wCD31 (green; left panel), an endothelial cell marker and vimentin (green; right panel), a fibroblast marker. Tw2+ cells do not give rise to endothelial cells or fibroblasts in adult skeletal muscle. Scale bar, 100 μm. (h) Type II myofiber specificity after prolonged labeling of Tw2-CreERT2; R26-tdTO mice. Transverse sections of indicated muscles of Tw2-CreERT2; R26-tdTO mice were obtained at 18 months post-TMX. Myosin staining using a type II specific antibody (My32, green) showed only a subset of Type II fibers were labeled by tdTO. Myofibers in tongue muscle were excluded from tdTO expression. Bottom panels show co-staining for type IIa myofibers. Scale bar, 100 μm. (i) Quantification of the percentage of tdTO+ myofibers among all myofibers in each field in G/P muscle at 18 months post-TMX. Data are mean ± S.E.M; N = 3 mice. (j) Seven-month old Tw2-CreERT2; R26-tdTO/+ mice were injected with 3 doses of TMX as described in Fig. 2a. Three months later, muscles were harvested and stained for type II myofibers. Tw2+ cells strongly labeled a subset of type II fibers in G/P, TA, masseter muscles, but not in tongue. Scale bar, 100 μm. (k) Soleus muscle of the same mice described in panel (b) were co-stained with antibodies against type I and type II myofibers. A subset of type II myofibers but not type I myofibers are labeled by tdTO. Scale bar, 100 μm. (l) Whole mount images showed intense tdTO signals in G/P, soleus, masseter and tongue of Pax7-CreERT2; R26-tdTO mice (right) compared to muscles of R26-tdTO mice (left) 8 weeks post-TMX. Mice were treated with the same regimen as shown in Fig. 2a. (m) Myosin staining using a type II specific antibody (My32, green) showed tdTO signals in the majority of myofibers of soleus, tongue and masseter muscle from Pax7-CreERT2; R26-tdTO mice at 8 weeks post-TMX. Scale bar, 100 μm. (n) Pax7-CreERT2; R26-tdTO mice were subjected to CTX injury and transverse sections of TA muscles were analyzed 7 and 14 days later. Contralateral uninjured muscle served as control (CTL). The results showed that Pax7+ cells contribute to all regenerating myofibers (indicated by centralized nuclei) on days 7 and 14 after CTX injury. Sections were co-stained with Laminin (green) and DAPI (blue). Scale bar, 100 μm. (o) Tw2+ cells do not contribute to embryonic myogenesis during development. Sections of Tw2-Cre; R26-tdTO embryos at E10.5, E15.5 and P1 were stained with α-actinin to detect somites (s) and muscle cells (m). Body wall muscles were shown for E15.5 and P1. Scale bar, 100 μm. Statistic source data for i are provided in Supplementary Table 3.

Supplementary Figure 3 Effect of Tw2+ cell ablation on non-muscle tissues.

(a) Cre+; DTA mice were smaller than control DTA mice at 9 months post-TMX. The difference in coat color is due to the mixed genetic background. (b) Whole mount images of G/P, Quad and heart of Cre+; DTA and control DTA mice at 9 months post-TMX. (c) Hematoxylin and eosin staining of liver, kidney, heart, white adipose tissue (WAT) and brown adipose tissue (BAT) from DTA and Cre+; DTA mice at 9 months post TMX. Black scale bar, 1 mm; White scale bar, 100 μm. (d) Quantification of type IIb myofibers per field and total myofibers per field in G/P, quad, mass and soleus muscles of DTA and Cre+; DTA mice. Data are mean ± S.E.M; N = 5 mice for each genotype. Two sample t-test;*: P < 0.05. Statistic source data for d are provided in Supplementary Table 3.

Supplementary Figure 4 Analysis of Tw2+ cells during regeneration following CTX injury and their engraftment capacity.

(a) Co-staining for type IIa myosin revealed regenerated tdTO+ myofibers are not type IIa myofibers. Scale bar, 100 μm. (b) Sections of TA muscle from Tw2-CreERT2; R26-tdTO mice were co-stained with desmin (green) on day 3 after CTX. The majority of tdTO+ cells are negative for desmin. Scale bar, 100 μm. (c) Sections of TA muscle from Tw2-CreERT2; R26-tdTO mice were co-stained with Pax7 (green) on day 7 after CTX. tdTO+ cells are negative for Pax7. Scale bar, 100 μm. (d) Quantification of the number of Pax7+, Tw2+ (tdTO+) and Pax7+/Tw2+ double positive cells per field in Tw2-CreERT2; R26-tdTO on days 7 and 14 post-CTX injury. CTL: contralateral TA muscle. For each muscle section, at least 6 different fields were quantified and averaged. Data are mean ± S.E.M; N = 3 mice. (e) Schematic of lineage tracing with Tw2-CreERT2; R26-mT/mG/+ mice. (f) Transverse-sections of G/P and masseter muscles from Tw2-CreERT2; R26-mT/mG mice at 4 months post-TMX. Tw2+ cells are labeled by GFP expression. All GFP+ myofibers remained TdTO+ in G/P and master, indicating Tw2+ cells fuse with existing myofibers. Scale bar, 100 μm. (g) Freshly isolated Tw2+ cells can engraft and form myofibers when transplanted into the TA muscle of mdx mice. 60,000 freshly isolated Tw2+ cells from Tw2-CreERT2; R26-tdTO mice at 10 days post-TMX were injected into TA muscle of 4 month-old mdx mice, which were injected with CTX 1 day before engraftment. TA muscles were harvested at 4 weeks post-injection and stained with laminin (green) and DAPI (blue) to visualize engrafted tdTO+ myofibers. Scale bar, 100 μm. (h) Quantification of the number of tdTO+ myofibers in the engraftment experiemnts. For each mouse, at least 6 fields of G/P muscle sections were quantified and averaged. Data are mean ± S.E.M; N = 3 mice. Statistic source data for d,h are provided in Supplementary Table 3.

Supplementary Figure 5 FACS analysis of Tw2+ cells.

(a) Representative FACS plots of Tw2+ (tdTO+) cells. Mononuclear cells from Tw2-CreERT2; R26-tdTO mice at 10 days post-TMX were sorted based on expression of tdTO. Approximately 3.3% of all mononuclear cells were positive for tdTO+. Sorting gates were drawn as indicated for both tdTO+ and tdTO- cell populations. (b) FACS plots of cell surface marker expression of tdTO+ cells from the Tw2-CreERT2; R26-tdTO mice at 10 days post-TMX.

Supplementary Figure 6 Properties of Tw2+ cells in culture.

(a) Tw2+ cells isolated by FACS sorting proliferated efficiently in growth medium (GM) and differentiated into multinucleated myotubes in differentiation medium (DM). These cells remain tdTO+ in GM and DM. Bright field images are presented for comparison. Scale bar, 20 μm. (b) Western blotting analysis for Tw2 protein of Tw2+ cells and Pax7+ cells in GM and DM. Ad-Ctl represents protein samples from neonatal rat cardiomyocytes infected with adenoviruses expressing either Twist1 (Tw1) or Twist2 (Tw2). The upper band present in all samples represents a non-specific band. GAPDH protein is detected as loading control. Unprocessed original scans of blots are shown in Supplementary Fig. 9. (c) Heat map of genes expressed in Pax7+ cells and Tw2+ cells in GM and DM identified by RNA-seq analysis (left panel). Heat map of the top 39 genes enriched in Pax7-DM versus Pax7-GM are shown in the middle panel, and the heat map of top 35 genes enriched in Pax7-GM versus Pax7-DM are list on the right panel. Importantly, these genes showed the same trend of enrichment and repression in Tw2-DM versus T w2-GM samples. (d) Tw2+ cells and SCs can fuse with each other to form multinucleated myotubes. Tw2+ cells were isolated by FACS sorting from adult Tw2-CreERT2; R26-tdTO mice 10 days post-TMX, which are labeled by tdTO expression. SCs, which are labeled by GFP expression, were isolated by FACS sorting from CAG-eGFP mice. Equal numbers of Tw2+ cells and SCs were mixed and grown in GM, followed by differentiation in DM. Cells were visualized by direct fluorescence. (e) Myosin immunostaining (My32) revealed formation of multi-nucleated myotubes that express both GFP and tdTO. Scale bar, 100 μm.

Supplementary Figure 7 Growth and myogenesis of tdTO+/CD34− cells in culture.

Freshly sorted tdTO+/CD34− cells were grown in GM for 48 h before being switched to DM to induce myogenesis. Scale bar, 20 μm.

Supplementary Figure 8 Model of Tw2 maintains stemness and blocks myogenesis.

Pax7 expression is not detectable in Tw2+ cells in vivo. However, when removed from their native milieu, Tw2+ cells rapidly down-regulate Twist expression and enter a Pax7+ state en route to a myogenic pathway.

Supplementary Figure 9 Unprocessed original scans of for Fig. 6e, Supplementary Fig. 2b, and Supplementary Fig. 6b.

Supplementary information

Supplementary Information

Supplementary Information (PDF 6565 kb)

Supplementary Table 1

Supplementary Information (XLSX 40 kb)

Supplementary Table 2

Supplementary Information (XLSX 36 kb)

Supplementary Table 3

Supplementary Information (XLSX 55 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, N., Garry, G., Li, S. et al. A Twist2-dependent progenitor cell contributes to adult skeletal muscle. Nat Cell Biol 19, 202–213 (2017). https://doi.org/10.1038/ncb3477

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3477

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing