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.

  • Letter
  • Published:

The endothelial Dll4–muscular Notch2 axis regulates skeletal muscle mass

A Publisher Correction to this article was published on 16 March 2022

This article has been updated

Abstract

Adult skeletal muscle is a highly plastic tissue that readily reduces or gains its mass in response to mechanical and metabolic stimulation; however, the upstream mechanisms that control muscle mass remain unclear. Notch signalling is highly conserved, and regulates many cellular events, including proliferation and differentiation of various types of tissue stem cell via cell–cell contact. Here we reveal that multinucleated myofibres express Notch2, which plays a crucial role in disuse- or diabetes-induced muscle atrophy. Mechanistically, in both atrophic conditions, the microvascular endothelium upregulates and releases the Notch ligand, Dll4, which then activates muscular Notch2 without direct cell–cell contact. Inhibition of the Dll4–Notch2 axis substantively prevents these muscle atrophy and promotes mechanical overloading-induced muscle hypertrophy in mice. Our results illuminate a tissue-specific function of the endothelium in controlling tissue plasticity and highlight the endothelial Dll4–muscular Notch2 axis as a central upstream mechanism that regulates catabolic signals from mechanical and metabolic stimulation, providing a therapeutic target for muscle-wasting diseases.

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

Fig. 1: Notch2 inactivation prevents muscle atrophy.
Fig. 2: Dll4 induces myofibre atrophy through Notch2.
Fig. 3: Dll4 derived from ECs induces muscle atrophy.
Fig. 4: Blocking Dll4 is a therapeutic target for muscle weakness.

Similar content being viewed by others

Data availability

The RNA-seq data have been deposited in the Gene Expression Omnibus repository under accession codes GSE161693 (RNA-seq) and GSE185678 (snRNA-seq). Uncropped immunoblot images are provided as source data. Other raw data are available on reasonable requests to the corresponding author. Source data are provided with this paper.

Change history

References

  1. Sjoqvist, M. & Andersson, E. R. Do as I say, Not(ch) as I do: lateral control of cell fate. Dev. Biol. 447, 58–70 (2019).

    Article  PubMed  Google Scholar 

  2. Kovall, R. A., Gebelein, B., Sprinzak, D. & Kopan, R. The canonical notch signaling pathway: structural and biochemical insights into shape, sugar, and force. Dev. Cell 41, 228–241 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Koch, U., Lehal, R. & Radtke, F. Stem cells living with a Notch. Development 140, 689–704 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Baghdadi, M. B. et al. Reciprocal signalling by Notch-Collagen V-CALCR retains muscle stem cells in their niche. Nature 557, 714–718 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Fukada, S. et al. Hesr1 and Hesr3 are essential to generate undifferentiated quiescent satellite cells and to maintain satellite cell numbers. Development 138, 4609–4619 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bjornson, C. R. et al. Notch signaling is necessary to maintain quiescence in adult muscle stem cells. Stem Cells 30, 232–242 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mourikis, P. et al. A critical requirement for notch signaling in maintenance of the quiescent skeletal muscle stem cell state. Stem Cells 30, 243–252 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Vasyutina, E. et al. RBP-J (Rbpsuh) is essential to maintain muscle progenitor cells and to generate satellite cells. Proc. Natl Acad. Sci. USA 104, 4443–4448 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bi, P. et al. Stage-specific effects of Notch activation during skeletal myogenesis. eLife 5, e17355 (2016).

  10. Fujimaki, S. et al. Notch1 and Notch2 coordinately regulate stem cell function in the quiescent and activated states of muscle satellite cells. Stem Cells 36, 278–285 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Lahmann, I. et al. Oscillations of MyoD and Hes1 proteins regulate the maintenance of activated muscle stem cells. Genes Dev. 33, 524–535 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zalc, A. et al. Antagonistic regulation of p57kip2 by Hes/Hey downstream of Notch signaling and muscle regulatory factors regulates skeletal muscle growth arrest. Development 141, 2780–2790 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Schuster-Gossler, K., Cordes, R. & Gossler, A. Premature myogenic differentiation and depletion of progenitor cells cause severe muscle hypotrophy in Delta1 mutants. Proc. Natl Acad. Sci. USA 104, 537–542 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Kalyani, R. R., Corriere, M. & Ferrucci, L. Age-related and disease-related muscle loss: the effect of diabetes, obesity, and other diseases. Lancet Diabetes Endocrinol. 2, 819–829 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Park, S. W. et al. Excessive loss of skeletal muscle mass in older adults with type 2 diabetes. Diabetes Care 32, 1993–1997 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Deeds, M. C. et al. Single dose streptozotocin-induced diabetes: considerations for study design in islet transplantation models. Lab. Anim. 45, 131–140 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hirata, Y. et al. Hyperglycemia induces skeletal muscle atrophy via a WWP1/KLF15 axis. JCI Insight 4, e124952 (2019).

  18. Yoshioka, M., Kayo, T., Ikeda, T. & Koizumi, A. A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes 46, 887–894 (1997).

    Article  CAS  PubMed  Google Scholar 

  19. Yang, H. H. et al. Notch1 gain of function in skeletal muscles leads to neuromuscular junction formation defects and neonatal death. CNS Neurosci. Ther. 24, 456–459 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. O’Neill, B. T. et al. FoxO transcription factors are critical regulators of diabetes-related muscle atrophy. Diabetes 68, 556–570 (2019).

    Article  PubMed  Google Scholar 

  21. O’Neill, B. T. et al. Insulin and IGF-1 receptors regulate FoxO-mediated signaling in muscle proteostasis. J. Clin. Invest. 126, 3433–3446 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zhang, X., Tang, N., Hadden, T. J. & Rishi, A. K. Akt, FoxO and regulation of apoptosis. Biochim. Biophys. Acta 1813, 1978–1986 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Milan, G. et al. Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy. Nat. Commun. 6, 6670 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Brocca, L. et al. FoxO-dependent atrogenes vary among catabolic conditions and play a key role in muscle atrophy induced by hindlimb suspension. J. Physiol. 595, 1143–1158 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Yang, W. L. et al. The E3 ligase TRAF6 regulates Akt ubiquitination and activation. Science 325, 1134–1138 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zou, Y. et al. NOTCH2 negatively regulates metastasis and epithelial-mesenchymal transition via TRAF6/AKT in nasopharyngeal carcinoma. J. Exp. Clin. Cancer Res. 38, 456 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Low, S., Barnes, J. L., Zammit, P. S. & Beauchamp, J. R. Delta-like 4 activates notch 3 to regulate self-renewal in skeletal muscle stem cells. Stem Cells 36, 458–466 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Verma, M. et al. Muscle satellite cell cross-talk with a vascular niche maintains quiescence via VEGF and notch signaling. Cell Stem Cell 23, 530–543 e539 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Miloudi, K. et al. NOTCH1 signaling induces pathological vascular permeability in diabetic retinopathy. Proc. Natl Acad. Sci. USA 116, 4538–4547 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lu, J. et al. Endothelial cells promote the colorectal cancer stem cell phenotype through a soluble form of Jagged-1. Cancer Cell 23, 171–185 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Billiard, F. et al. Delta-like ligand-4-notch signaling inhibition regulates pancreatic islet function and insulin secretion. Cell Rep. 22, 895–904 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Trindade, A. et al. Low-dosage inhibition of Dll4 signaling promotes wound healing by inducing functional neo-angiogenesis. PLoS ONE 7, e29863 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gilbert, R. E., Connelly, K., Kelly, D. J., Pollock, C. A. & Krum, H. Heart failure and nephropathy: catastrophic and interrelated complications of diabetes. Clin. J. Am. Soc. Nephrol. 1, 193–208 (2006).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wu, S., Wu, Y. & Capecchi, M. R. Motoneurons and oligodendrocytes are sequentially generated from neural stem cells but do not appear to share common lineage-restricted progenitors in vivo. Development 133, 581–590 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Rao, P. & Monks, D. A. A tetracycline-inducible and skeletal muscle-specific Cre recombinase transgenic mouse. Dev. Neurobiol. 69, 401–406 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. McCright, B., Lozier, J. & Gridley, T. Generation of new Notch2 mutant alleles. Genesis 44, 29–33 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Han, H. et al. Inducible gene knockout of transcription factor recombination signal binding protein-J reveals its essential role in T versus B lineage decision. Int. Immunol. 14, 637–645 (2002).

    Article  CAS  PubMed  Google Scholar 

  39. Imayoshi, I., Shimogori, T., Ohtsuka, T. & Kageyama, R. Hes genes and neurogenin regulate non-neural versus neural fate specification in the dorsal telencephalic midline. Development 135, 2531–2541 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Fujimura, S., Jiang, Q., Kobayashi, C. & Nishinakamura, R. Notch2 activation in the embryonic kidney depletes nephron progenitors. J. Am. Soc. Nephrol. 21, 803–810 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bothe, G. W., Haspel, J. A., Smith, C. L., Wiener, H. H. & Burden, S. J. Selective expression of Cre recombinase in skeletal muscle fibers. Genesis 26, 165–166 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Hozumi, K. et al. Delta-like 4 is indispensable in thymic environment specific for T cell development. J. Exp. Med. 205, 2507–2513 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Okabe, K. et al. Neurons limit angiogenesis by titrating VEGF in retina. Cell 159, 584–596 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Ono, Y. et al. Muscle stem cell fate is controlled by the cell-polarity protein Scrib. Cell Rep. 10, 1135–1148 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Sakai, H. et al. Notch ligands regulate the muscle stem-like state ex vivo but are not sufficient for retaining regenerative capacity. PLoS ONE 12, e0177516 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Yoshioka, K. et al. Hoxa10 mediates positional memory to govern stem cell function in adult skeletal muscle. Sci. Adv. 7, eabd7924 (2021).

  48. Yoshioka, K. et al. A modified pre-plating method for high-yield and high-purity muscle stem cell isolation from human/mouse skeletal muscle tissues. Front. Cell Dev. Biol. 8, 793 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Muramatsu, M. et al. Loss of syndrome critical region-1 mediated-hypercholesterolemia accelerates corneal opacity via pathological neovessel formation. Arterioscler. Thromb. Vasc. Biol. 40, 2425–2439 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ong, C. T. et al. Target selectivity of vertebrate notch proteins. Collaboration between discrete domains and CSL-binding site architecture determines activation probability. J. Biol. Chem. 281, 5106–5119 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Yamazaki, Y. et al. The cathepsin L gene is a direct target of FOXO1 in skeletal muscle. Biochemical J. 427, 171–178 (2010).

    Article  CAS  Google Scholar 

  52. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Huang da, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  PubMed  Google Scholar 

  54. Huang da, W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  PubMed  Google Scholar 

  55. Verma, M. et al. Inhibition of FLT1 ameliorates muscular dystrophy phenotype by increased vasculature in a mouse model of Duchenne muscular dystrophy. PLoS Genet. 15, e1008468 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Dimauro, I., Pearson, T., Caporossi, D. & Jackson, M. J. A simple protocol for the subcellular fractionation of skeletal muscle cells and tissue. BMC Res. Notes 5, 513 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank R. Fujita, S. Ogawa, Y. Tsuchiya, K. Yoshioka, Y. Kitajima, D. T. Shima, Y. Kubota and N. Suzuki for sharing materials and technical assistance. We also thank S. Burden for providing Mlc1f-Cre mice; I. Imayoshi and R. Kageyama for providing Hes1-flox mice and T. Honjo for providing RBPJ-flox mice. The Pax7 and MF20 antibodies, developed by A. Kawakami and D.A. Fischman, respectively, were obtained from the Developmental Studies Hybridoma Bank developed under the auspices of the NICHD and maintained by the University of Iowa. This work was supported by the FOREST program of the Japan Science and Technology Agency (JST, grant no. JPMJFR205C to Y.O.), the Japan Agency for Medical Research and Development (AMED, grant no. JP19bm0704036 to Y.O.) and Grants-in-Aid for Scientific Research KAKENHI (grant nos. 25882024 to S.M.; 15H05368, 18H03193 and 20K21763 to Y.O.; 20K19641 to S.F. and 20J01669 to T.Ma.). This work was also supported, in part, by the Center for Metabolic Regulation of Healthy Aging (to Y.O. and S.F.), the Takeda Science Foundation (to Y.O. and S.F.), the Naito Foundation (to Y.O.), the Uehara Memorial Foundation (to S.F.), NIH R21 (grant no. AR078400 to A.A.) and Regenerative Medicine Minnesota (grant no. 092319 TR010 to A.A.). T.Ma., H.N. and N.H. were funded by a JSPS Research Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

S.F. conducted and performed the experiments, interpreted the data, assembled the input data and wrote the paper. T.Ma., H.N., N.H., D.S. and S.M. performed cell culture and animal experiments. M.M. and T.Mi. performed EC-culture and immunohistochemical experiments, interpreted and analysed the data, and wrote the paper. Y.T., Y.M. and T.F. performed electron microscopy and immunohistochemical experiments, interpreted the data and wrote the paper. X.W., Y.A. and A.A. performed the animal experiments, interpreted the data and wrote the paper. S.U. and K.Y. prepared data library for snRNA-seq and analysed the data. Y.K and R.N. generated key materials and interpreted the data. Y.O. designed and performed the experiments, interpreted the data, assembled the input data and wrote the paper. All authors reviewed the paper and approved the final version.

Corresponding author

Correspondence to Yusuke Ono.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Brian O’Neill and the other, anonymous, reviewers for their contribution to the peer review of this work. Isabella Samuelson was the Primary Handling Editor.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Notch2 is expressed in adult myofibres.

a, Representative images of primary cultures of satellite cells maintained in GM or Diff. M for 4 days (Diff. M 4d). b, qPCR analysis for gene expression of Notch1 and Notch2 during myogenic progression (up to day 5 in Diff. M) (n = 3). Myh3 served as a marker of cell differentiation. c, Immunoblotting analysis for protein levels of Notch1 and Notch2 during myogenic progression (up to day 5 in Diff. M). Upper and lower bands in the images of Notch1 and Notch2 indicate the full-length proteins (FL) and the transmembrane/intracellular region (NTM), respectively. Myosin heavy chain (MyHC) and β-tubulin served as a marker of cell differentiation and an internal control, respectively. d, To determine the expression of Notch1 and Notch2 in terminally differentiated adult myofibres with or without satellite cells, Pax7CreERT2/+;Rosa26LSL-DTA/+ mice that express tamoxifen (TMX)-inducible diphtheria toxin-A (DTA) under the control of the Pax7 promoter were used. TMX was intraperitoneally injected five times into Pax7CreERT2/+;Rosa26LSL-DTA/+ mice to deplete the satellite cell population, and individual myofibres were isolated from EDL muscle (Pax7-DTA myofibre). Non-TMX treated mice were used as a control (Control myofibre). e, qPCR analysis for gene expression of Notch1 and Notch2 in Pax7-DTA myofibres. Pax7 served as a marker of satellite cell depletion in Pax7-DTA myofibres (n = 3). fj, Satellite cells were isolated from EDL muscle in Pax7CreERT2/+;Rosa26LSL-DTA mice, cultured in Diff. M, and treated with 4OH TMX in order to deplete the self-renewed and undifferentiated satellite cells. f, Time course of culture conditions. g, Representative images of immunostaining for Pax7 (scale bar, 50 µm). Yellow arrows indicate Pax7+ cells. h, Percentage of Pax7+ nuclei in the total DAPI+ nuclei (n = 6). i, qPCR analysis for gene expression of Notch2 in Cont and DTA myotubes (n = 6). j, Immunoblotting analysis for protein levels of Notch2 and Pax7. All qPCR data were normalised to Tbp expression. Data represent the mean ± s.e.m. and points are individual mice.

Source data

Extended Data Fig. 2 Constitutive activation of Notch2 induces muscle atrophy.

ag, To examine the effect of constitutive activation of Notch2 on adult muscle in vivo, HSArtTA/+;TRECre/+;Rosa26LSL-N2ICD/+ mice (N2-mTg) were generated. HSArtTA/+;TRECre/+ or Rosa26LSL-N2ICD/+ mice were used as a control (Cont). a, N2-mTg and Cont mice were treated with DOX (2 mg ml−1 in drinking water) to induce constitutive expression of N2ICD. b, qPCR analysis for confirming the overexpression of Notch2 in GAS (n = 3). c,d, Body weight (c) and muscle weights of GAS, TA, and PLA normalised to body weight (d, n = 3 for Cont and n = 5 for N2-mTg). e, Representative images of immunostaining for Laminin on TA cross-sections (scale bar, 100 µm). f, Myofibre CSA of TA (n = 3). g, Grip strength (n = 4 for Cont and n = 5 for N2-mTg). qPCR data were normalised to Tbp expression. Data represent the means ± s.e.m., and points indicate individual mice. *P < 0.05, **P < 0.01, NS as determined by Student’s two-tailed unpaired t-test (c,d,f,g).

Extended Data Fig. 3 Effects of Notch2 ablation on mouse growth and Akita diabetic mice.

a, qPCR analysis for confirming the deletion of the Notch2 gene in individual myofibres isolated from EDL muscles of Notch2flox/flox (N2f/f) mice and Mlc1fCre/+;Notch2flox/flox (N2−/−) mice (normalised to Tbp expression). be, Profiles of N2–/– adult mice with regard to body weight (b), muscle weights normalised to body weight (c), the number of nuclei per myofibre (d), and the number of Pax7+ cells (satellite cells) per myofibre (e) (n = 5). f,g, Akita mice were used as a diabetic hyperglycemic model. Thirteen-week-old male littermates were used. Blood glucose levels (f) and muscle weights normalised to body weight (g) (n = 3). Data represent the means ± s.e.m. and points indicate individual mice. **P < 0.01, ***P < 0.001, NS (not significant) as determined by: two-way ANOVA followed by Tukey’s multiple comparisons post-hoc test (f,g); Student’s two-tailed unpaired t-test (be).

Extended Data Fig. 4 Notch2-induced muscle atrophy is independent of RBPJ and Hes1.

ae, Effects of HU or DM on muscles in muscle-specific Rbpj-ablated mice. HSArtTA/+;TRECre/+;Rbpjflox/flox mice were treated with DOX (2 mg ml−1 in drinking water) for 3 weeks to induce deletion of Rbpj in myofibres (Rbpj–/–). Rbpjflox/flox mice were used as control (Rbpjf/f). a, qPCR analysis for confirming the deletion of Rbpj expression in myofibres isolated from EDL (n = 3). b, Rbpjf/f and Rbpj–/– mice started 7 day-HU after 2 week-DOX treatment. Mice maintained in the ground condition were used as ground control (GC). c, Muscle weight of SOL, PLA, and GAS muscles normalised to body weight of Rbpjf/f and Rbpj–/– mice under GC and HU conditions (n = 3). d, Rbpjf/f and Rbpj–/– mice were intraperitoneally injected with STZ (150 mg kg−1 body weight; DM) or vehicle (Veh; citrate buffer) 2 weeks after starting DOX treatment and sacrificed 14 days following STZ infusion. e, Muscle weight of TA, PLA, and GAS muscles normalised to body weight of Rbpjf/f and Rbpj–/– mice under Veh and DM conditions (n = 3). fj, Effects of HU or DM on muscles in muscle-specific Hes1-ablated mice. Hes1flox/flox (Hes1f/f) and Mlc1fCre/+;Hes1flox/flox (Hes1–/–) mice were used. f, qPCR analysis for confirming the deletion of Hes1 expression in myofibres isolated from EDL (n = 4). g, Time course of the HU experiment. h, Muscle weight of SOL and GAS muscles normalised to body weight of Hes1f/f and Hes1–/– mice (n = 3 for GC and n = 5 for HU). i, Time course of the diabetes experiment. j, Muscle weight of TA muscle normalised to body weight in Hes1f/f and Hes1–/– mice (n = 4, except n = 6 for Hes1–/– DM). ks, Effects of constitutive activation of Notch2 on muscles of muscle-specific Rbpj–/– mice. k, HSArtTA/+;TRECre/+;Rosa26LSL-N2ICD/+;Rbpjflox/flox mice were treated with DOX for 3 weeks to induce constitutive activation of Notch2 and Rbpj deletion in myofibres (N2-mTg;Rbpj–/–). Rbpjflox/flox mice were used as control (Rbpjf/f). l, qPCR analysis for confirming the overexpression of Notch2 and the deletion of Rbpj in myofirers isolated from EDL (n = 4 for Rbpjf/f and n = 5 for N2-mTg;Rbpj–/–). m,n, Body weight (m) and muscle weights normalised to body weight (GAS, TA, and PLA; n) (n = 6 for Rbpjf/f and n = 7 for N2-mTg;Rbpj–/–). o,p, the number of nuclei per myofibre (o), and the number of Pax7+ cells (satellite cells) per myofibre (p) (n = 4). qs, Neuromuscular junctions (NMJs) of EDL myofibres isolated from Rbpj–/– and N2-mTg;Rbpj–/– mice were analysed. q, Representative phase-contrast images of NMJs on myofibres (scale bar, 50 µm). r,s, Distribution of myonuclear numbers in NMJ region (r, n = 3) and the ratio of fragmented NMJs (s, n = 3). All qPCR data were normalised to Tbp. Data represent the means ± s.e.m., and points indicate individual mice. *P < 0.05, **P < 0.01, ***P < 0.001, NS (not significant) as determined by: two-way ANOVA followed by Tukey’s multiple comparisons post-hoc test (c,e,h,j); Student’s two-tailed unpaired t-test (mp,r,s).

Extended Data Fig. 5 Notch2-induced muscle atrophy is mediated through FoxO signalling pathway.

ad, Pathway analysis using RNA-Seq data sets that were significantly upregulated in N2f/f muscle but not in N2–/– muscle, under hindlimb unloading or diabetic conditions, corresponding to Fig. 1o-q. a, Venn diagram showing DEGs obtained from the comparison between the HU and Cont conditions in GAS muscles from N2f/f (red circle) and N2–/– mice (blue circle) (genes with fold change >2.0, q < 0.05, and TPM > 500 were analysed). b, Upregulated pathways obtained from KEGG pathway analysis using genes upregulated in unloaded GAS muscle of N2f/f but not N2–/– mice. c, Venn diagram showing DEGs obtained from the comparison between the DM and Cont conditions in GAS muscles from N2f/f mice (red circle) and N2–/– mice (blue circle) (genes with fold change >2.0, q < 0.05, and TPM > 500 were analysed). d, Upregulated pathways obtained from KEGG pathway analysis using genes upregulated in diabetic GAS muscle of N2f/f but not N2–/– mice. eg, Effect of constitutive activation of Notch2 with Rbpj ablation on the expressions of atrogenes and FoxO target genes in myofibres. e, HSArtTA/+;TRECre/+;Rosa26LSL-N2ICD/+;Rbpjflox/flox mice were treated with DOX for 1 week to induce constitutive activation of Notch2 and Rbpj deletion in myofibres (N2-mTg;Rbpj–/–). Rbpjflox/flox mice were used as control (Rbpjf/f). f, qPCR analysis for confirming the overexpression of Notch2 and its target gene Hes1, and the deletion of Rbpj in GAS muscles (n = 4). g. Expressions of FoxO target genes and atrogenes in GAS muscles (n = 4). h,i, Assessment of direct interaction between Notch2 and FoxO transcription factors. h, Satellite cells isolated from EDL muscles of WT mice were transfected with 3xFLAG-N2ICD plasmid by electroporation. Cells were induced to undergo myogenic differentiation for 5 days and harvested as IP samples. i, Immunoblotting analysis for Notch2 (NTM), FoxO1, FoxO3a, and Akt using plasmid-transfected (TF) and non-transfected (Cont) samples immunoprecipitated using FLAG antibody. Total cell lysates were used as input controls. jl, Effect of Notch2 deficiency on FoxO phosphorylation and localization under atrophic conditions. j, Protein accumulation of phospho-Akt (Ser473), total-Akt, phospho-FoxO1/3a (Thr24/Thr32), total-FoxO3a in GAS muscle of N2f/f and N2−/− mice under control, HU, and DM conditions. k,l, Protein accumulation of FoxO1 and FoxO3a in nuclear fractions of GAS muscles from N2f/f and N2−/− mice under control and DM conditions. Representative blot patterns (k) and quantitative data (l) were shown. Histone H3 served as an internal control. mr, Effects of Notch2 deficiency on FoxO1-induced muscle atrophy. m, Satellite cells were isolated from N2f/f or N2−/− and transfected with plasmid encoding constitutively active form of human FOXO1 (caFOXO1) or empty vector (Cont) by electroporation. Cells were induced to undergo myogenic differentiation for 4 days and then harvested. n, Gene expressions of Notch2, FoxO1, and FoxO1-target genes including Ccng2 and Gadd45 in myotubes (n = 4). o, Representative images of MyHC immunostaining (scale bars, 50 µm). p,q, Quantitative data of the myotube diameter (p) and the fusion index, which is a proportion of the number of nuclei within MyHC+ myotubes (containing more than two nuclei) relative to the total number of nuclei (q) (n = 4). r, Gene expressions of atrogenes. All qPCR data were normalised to Tbp (n = 4). Data represent the means ± s.e.m., and points indicate individual mice. *P < 0.05, **P < 0.01, ***P < 0.001, NS (not significant) as determined by: two-way ANOVA followed by Tukey’s multiple comparisons post-hoc test (l,n,pr); Student’s two-tailed unpaired t-test (g).

Source data

Extended Data Fig. 6 Inhibition of γ-secretase activity attenuates muscle atrophy in unloading and diabetic conditions.

a,b, Wild-type mice were intraperitoneally injected with a γ-secretase inhibitor DAPT (10 mg kg−1 body weight) or corn oil (Cont) every other day during the HU period. a, Time course. b, Muscle weight of SOL, PLA, and GAS (n = 4 for GC and n = 7 for HU). ce, Wild-type mice were intraperitoneally injected with STZ (150 mg kg−1 body weight; DM) or vehicle (Veh). Mice were then injected with DAPT or corn oil (Cont) every other day from day 3 following STZ infusion. c, Time course. d,e, Blood glucose levels 14 days after STZ infusion (d) and muscle weight of TA, PLA, and GAS (e) (n = 6 for Veh and n = 8 for DM). Muscle weights were normalised to body weight. Data represent the means ± s.e.m. and points indicate individual mice. *P < 0.05, **P < 0.01, ***P < 0.001, NS (not significant) as determined by: two-way ANOVA followed by Tukey’s multiple comparisons post-hoc test (b,d,e).

Extended Data Fig. 7 Dll4 expression is restricted to ECs in adult muscle tissue.

ae, snRNA-Seq of mouse GAS and PLA muscles at 8 weeks of age. a, Schematic of nuclei isolation and sequencing. b, UMAP plot from unbiased clustering of 16,159 single nuclei isolated from GAS and PLA muscles. c, UMAP plots showing marker gene expression for nuclear populations, including Ttn (myonuclei), Colq (neuromuscular junctions, NMJs), Tigd4 (myotendinous junctions, MTJs), Pax7 (satellite cells, SCs), Pecam1 (endothelial cells, ECs), Ptprc (immune cells), Myh11 (smooth muscle cells), Pdgfra (Fibro-adipogenic progenitors, FAPs), Mkx (tenocytes), and Plin1 (adipocytes). d, UMAP plot showing Dll4 expression with magnified plot of the EC cluster. e, UMAP plots showing Notch1 and Notch2 expressions. f, Immunoblotting for Dll4 protein using GAS muscle from EC-specific homozygous Dll4 knockout mice (Cdh5CreERT2/+;Dll4flox/flox, Dll4−/−), heterozygous Dll4 knockout mice (Cdh5CreERT2/+;Dll4flox/+, Dll4+/−), and control mice (Dll4flox/flox, Dll4+/+). To induce Cre recombination, mice were intraperitoneally injected with tamoxifen (75 mg kg−1 body weight, 4 times) 1 week before analysis. GAPDH served as the internal control. g, Gene expression of Notch ligands (Dll1, Dll4, Jag1, and Jag2) normalised to Tbp expression in myofibres isolated from wild-type PLA or EDL muscle in the HU or DM conditions, respectively (n = 4 mice per condition). h, Representative TEM images of immunogold labelling for Dll4 protein in GAS muscle isolated from diabetic mice. Gold particles were localized on the surface of myofibres (left), collagen-like fibres (middle), and extracellular vesicles (right) in the interstitial spaces between ECs and myofibres (scale bar, 200 nm). Data represent the means ± s.e.m. and points indicate individual mice. *P < 0.05, ***P < 0.001 as determined by two-way ANOVA followed by Tukey’s multiple comparisons post-hoc test (g).

Source data

Extended Data Fig. 8 DLL4 blockade prevents atrophy of human skeletal myotubes.

ai, Effect of coculture of human skeletal muscle myotubes with human umbilical vein endothelial cells (HUVECs) in the transwell culture system. a, Schematic of the transwell culture system. HUVECs were transfected with control siRNA (si-Cont) or siRNA against DLL4 (si-DLL4) 1 day before coculture with myotubes. b, DLL4 protein expression in si-Cont- and si-DLL4-transfected HUVEC lysates and supernatants (Sup) of those culture media. Immunodetection for β-tubulin and ponceau staining served as internal controls. c, Representative images of MyHC-positive myotubes (scale bar, 50 µm). d,e, Quantitative data of myotube diameters (d) and fusion index (e) (n = 4 well per condition, myotubes in more than 3 field of view (10×)/well were measured). f, Effects of treatment with 500 ng ml−1 anti-DLL4 neutralizing antibody (αDLL4) or control antibody (αCont) on human myotubes–HUVECs co-culture. g, Representative images of MyHC-positive myotubes (scale bar, 50 µm). h,i, Quantitative data of myotube diameters (h) and fusion index (i) (n = 4 well per condition, myotubes in more than 3 field of view (10×)/well were measured). Data represent the mean ± s.e.m., and points are individual mice. ***P < 0.001, NS (not significant) as determined by two-way repeated-measures ANOVA followed by Tukey’s multiple comparisons post-hoc test (d,h).

Source data

Extended Data Fig. 9 Effects of αDll4 or Fc-fused Dll4 recombinant protein on muscle atrophy.

ac, Effect of treatment with EC-conditioned media (EC-CM) on myofibres. a, Primary ECs were isolated from hindlimb muscle tissues. Concentrated EC-CM were added into culture media containing wild-type EDL myofibres. Myofibres were then cultured with the addition of 500 ng ml−1 anti-Dll4 neutralising antibody (αDll4) or control antibody (αCont) for 48 hours. b, Representative images of myofibres (scale bar, 50 µm). c, Quantitative data of myofibre diameters (n = 4 mice per condition, >15 individual myofibres per mouse were measured). df, Effect of treatment with Fc-fused Dll4 recombinant protein (rDll4-Fc) on myofibres co-cultured with primary ECs. d, Wild-type EDL myofibres were co-cultured with ECs isolated from hindlimb muscle tissues with the addition of 500 ng ml−1 rDll4-Fc for 48 hours. e, Representative images of myofibres (scale bar, 50 µm). f, Quantitative data of myofibre diameters (n = 4 mice per condition, >15 individual myofibres per mouse were measured). gi, Effect of rDll4-Fc administration on diabetic muscle atrophy in vivo. g, Wild-type mice were intraperitoneally injected with STZ (150 mg kg−1 body weight; DM) or vehicle (Veh) and analysed 14 days after the injection. Mice were administered with rDll4-Fc at day 7, 9, 11, and 13 following the STZ infusion. PBS was used as a control. h, Blood glucose levels 14 days after STZ infusion (n = 5). i, Muscle weight normalised to body weight. Data represent the means ± s.e.m., and points indicate individual mice. *P < 0.05, **P < 0.01, ***P < 0.001 NS (not significant) as determined by two-way ANOVA followed by Tukey’s multiple comparisons post-hoc test (c,f,h,i).

Extended Data Fig. 10 Inhibition of the Dll4-Notch2 axis promotes hypertrophic response.

a, Gene expression of Notch ligands (Dll1, Dll4, Jag1, and Jag2) in CD31+ ECs and myofibres isolated from overloaded or control PLA muscle (n = 4 mice per condition). qPCR data of ECs and myofibres were normalised to Pecam1 and GAPDH, respectively. bd, To assess the hypertrophic response of muscle, N2f/f and N2–/– mice were overloaded (OL) by tenotomy for 14 days. b, Time course. c,d, Muscle weight (c) and OL-induced weight gain of PLA muscle (d), PLA muscle weight/body weight of OL vs. sedentary control (n = 6-8). eg, Wild-type mice were intraperitoneally administered with DAPT (10 mg kg−1 body weight) following tenotomy. e, Time course. f,g, Muscle weight (f) and OL-induced PLA weight gain (g) (n = 6). hj, Effects of EC-specific conditional deletion of Dll4 on muscle hypertrophy. h, Experimental scheme for assessing the hypertrophic response of EC-specific Dll4-deficient mice after overloading. Dll4flox/flox (Dll4+/+) and Cdh5CreERT2/+;Dll4f/f (Dll4–/–) mice were overloaded (OL) after TMX was intraperitoneally injected four times. i,j, Muscle weight (i) and OL-induced weight gain of PLA muscle (j) (n = 5). Muscle weights were normalised to body weight. Data represent the means ± s.e.m. and points represent individual mice. *P < 0.05, **P < 0.01, ***P < 0.001 as determined by: two-way ANOVA followed by Tukey’s multiple comparisons post-hoc test (c,f,i); Student’s two-tailed unpaired t-test (a,d,g,j).

Supplementary information

Supplementary Information

Supplementary Fig. 1 Gating strategy. Supplementary Table 1 Antibody list. Supplementary Table 2 Primer sequences.

Reporting Summary

Source data

Source Data Fig. 3

Uncropped immunoblots.

Source Data Extended Data Fig. 1

Uncropped immunoblots.

Source Data Extended Data Fig. 5

Uncropped immunoblots.

Source Data Extended Data Fig. 7

Uncropped immunoblots.

Source Data Extended Data Fig. 8

Uncropped immunoblots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fujimaki, S., Matsumoto, T., Muramatsu, M. et al. The endothelial Dll4–muscular Notch2 axis regulates skeletal muscle mass. Nat Metab 4, 180–189 (2022). https://doi.org/10.1038/s42255-022-00533-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00533-9

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