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:

Denervation-activated STAT3–IL-6 signalling in fibro-adipogenic progenitors promotes myofibres atrophy and fibrosis

An Author Correction to this article was published on 07 February 2024

This article has been updated

Abstract

Fibro-adipogenic progenitors (FAPs) are typically activated in response to muscle injury, and establish functional interactions with inflammatory and muscle stem cells (MuSCs) to promote muscle repair. We found that denervation causes progressive accumulation of FAPs, without concomitant infiltration of macrophages and MuSC-mediated regeneration. Denervation-activated FAPs exhibited persistent STAT3 activation and secreted elevated levels of IL-6, which promoted muscle atrophy and fibrosis. FAPs with aberrant activation of STAT3–IL-6 signalling were also found in mouse models of spinal cord injury, spinal muscular atrophy, amyotrophic lateral sclerosis (ALS) and in muscles of ALS patients. Inactivation of STAT3–IL-6 signalling in FAPs effectively countered muscle atrophy and fibrosis in mouse models of acute denervation and ALS (SODG93A mice). Activation of pathogenic FAPs following loss of integrity of neuromuscular junctions further illustrates the functional versatility of FAPs in response to homeostatic perturbations and suggests their potential contribution to the pathogenesis of neuromuscular diseases.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Abnormal accumulation of FAPs during muscle atrophy by acute denervation.
Fig. 2: FAPs from denervated muscles exhibit distinct functional properties and gene expression profiles.
Fig. 3: Persistent activation of IL-6–STAT3 pathway in FAPs and myofibres from denervated muscles and prevention of myofibre atrophy and fibrosis by IL-6 blockade.
Fig. 4: STAT3 inhibition prevents the atrophic effect of denervation-derived FAPs.
Fig. 5: Selective STAT3 inhibition in FAPs is sufficient to counter myofibre atrophy.
Fig. 6: FAPs with aberrant IL-6–STAT3 activation accumulate in skeletal muscles from SCI.
Fig. 7: FAPs with aberrant IL-6–STAT3 activation accumulate in ALS skeletal muscles.

Similar content being viewed by others

Change history

References

  1. Pannérec, A., Marazzi, G. & Sassoon, D. Stem cells in the hood: the skeletal muscle niche. Trends Mol. Med.10, 599–606 (2012).

    Article  Google Scholar 

  2. Farup, J., Madaro, L., Puri, P. L. & Mikkelsen, U. R. Interactions between muscle stem cells, mesenchymal-derived cells and immune cells in muscle homeostasis, regeneration and disease. Cell Death Dis.6, e1830 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bentzinger, C. F., Wang, Y. X., Dumont, N. A. & Rudnicki, M. A. Cellular dynamics in the muscle satellite cell niche. EMBO Rep.14, 1062–1072 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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).

    Article  CAS  PubMed  Google Scholar 

  5. Collins, C. A. et al. Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell122, 289–301 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Kang, J.-S. & Krauss, R. S. Muscle stem cells in developmental and regenerative myogenesis. Curr. Opin. Clin. Nutr. Metab. Care13, 243–248 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tidball, J. G. Mechanisms of muscle injury, repair, and regeneration. Compr. Physiol.1, 2029–2062 (2011).

    Article  PubMed  Google Scholar 

  8. Serrano, A. L. & Muñoz-Cánoves, P. Fibrosis development in early-onset muscular dystrophies: mechanisms and translational implications. Semin. Cell Dev. Biol.64, 181–190 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Uezumi, A., Fukada, S., Yamamoto, N., Takeda, S. & Tsuchida, K. Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle. Nat. Cell Biol.12, 143–152 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Gonzalez, D. et al. ALS skeletal muscle shows enhanced TGF-β signaling, fibrosis and induction of fibro/adipogenic progenitor markers. PloS One12, e0177649 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kharraz, Y., Guerra, J., Mann, C. J., Serrano, A. L. & Muñoz-Cánoves, P. Macrophage plasticity and the role of inflammation in skeletal muscle repair. Mediat. Inflamm.2013, 491497 (2013).

    Article  Google Scholar 

  13. Tidball, J. G. Regulation of muscle growth and regeneration by the immune system. Nat. Rev. Immunol.17, 165–178 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Heredia, J. E. et al. Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell153, 376–388 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lemos, D. R. et al. Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF-mediated apoptosis of fibro/adipogenic progenitors. Nat. Med.21, 786–794 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Mozzetta, C. et al. Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old Mdx mice. EMBO Mol. Med.5, 626–639 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Uezumi, A. et al. Identification and characterization of PDGFRα+ mesenchymal progenitors in human skeletal muscle. Cell Death Dis.5, e1186 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Saccone, V. et al. HDAC-regulated myomiRs control BAF60 variant exchange and direct the functional phenotype of fibro-adipogenic progenitors in dystrophic muscles. Genes Dev.28, 841–857 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sala, D., & Sacco, A. Signal transducer and activator of transcription 3 signaling as a potential target to treat muscle wasting diseases. Curr. Opin. Clin. Nutr. Metab. Care.9, 171–176 (2016).

    Google Scholar 

  20. Bonetto, A. et al. STAT3 activation in skeletal muscle links muscle wasting and the acute phase response in cancer cachexia. PloS ONE6, e22538 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bonetto, A. et al. JAK/STAT3 pathway inhibition blocks skeletal muscle wasting downstream of IL-6 and in experimental cancer cachexia. Am. J. Physiol. Endocrinol. Metab.303, E410–421 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Haddad, F., Zaldivar, F., Cooper, D. M. & Adams, G. R. IL-6-induced skeletal muscle atrophy. J. Appl. Physiol.98, 911–917 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Tang, H. et al. mTORC1 promotes denervation-induced muscle atrophy through a mechanism involving the activation of FoxO and E3 ubiquitin ligases. Sci. Signal.7, ra18 (2014).

    Article  PubMed  Google Scholar 

  24. Moresi, V. et al. Myogenin and class II HDACs control neurogenic muscle atrophy by inducing E3 ubiquitin ligases. Cell1, 35–45 (2010).

    Article  Google Scholar 

  25. Joassard, O. R. et al. HuR mediates changes in the stability of AChR β-subunit mRNAs after skeletal muscle denervation. J. Neurosci.35, 10949–10962 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Le, T. T. et al. SMNDelta7, the major product of the centromeric survival motor neuron (SMN2) gene, extends survival in mice with spinal muscular atrophy and associates with full-length SMN. Hum. Mol. Genet.14, 845–857 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Mead, R. J. et al. Optimised and rapid pre-clinical screening in the SOD1G93A transgenic mouse model of amyotrophic lateral sclerosis (ALS). PLoS ONE6, e23244 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Olivan, S. et al. Comparative study of behavioural tests in the SOD1G93A mouse model of amyotrophic lateral sclerosis. Exp. Anim.64, 147–153 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Tierney, M. T. et al. STAT3 signaling controls satellite cell expansion and skeletal muscle repair. Nat. Med.20, 1182–1186 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Muñoz-Cánoves, P., Scheele, C., Pedersen, B. K. & Serrano, A. L. Interleukin-6 myokine signaling in skeletal muscle: a double-edged sword? FEBS J.280, 4131–48 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Zimmers, T. A., Fishel, M. L. & Bonetto, A. STAT3 in the systemic inflammation of cancer cachexia. Semin. Cell Dev. Biol.54, 28–41 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Serrano, A. L., Baeza-Raja, B., Perdiguero, E., Jardí, M. & Muñoz-Cánoves, P. Interleukin-6 is an essential regulator of satellite cell-mediated skeletal muscle hypertrophy. Cell Metab.1, 33–44 (2008).

    Article  Google Scholar 

  33. Rossi, J.-F. et al. A phase I/II study of siltuximab (CNTO 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Br. J. Cancer103, 1154–1162 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. van Rhee, F. et al. Siltuximab, a novel anti-interleukin-6 monoclonal antibody, for Castleman’s disease. J. Clin. Oncol.28, 3701–3708 (2010).

    Article  PubMed  Google Scholar 

  35. Johnson, D. E., O’Keefe, R. A. & Grandis, J. R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol.15, 234–248 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Takeda, K. et al. Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: generation and characterization of T cell-specific Stat3-deficient mice. J. Immunol.161, 4652–4660 (1998).

    Article  CAS  PubMed  Google Scholar 

  37. White, A. T., LaBarge, S. A., McCurdy, C. E. & Schenk, S. Knockout of STAT3 in skeletal muscle does not prevent high-fat diet-induced insulin resistance. Mol. Metab.4, 569–575 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Sala, D. et al. Autophagy-regulating TP53INP2 mediates muscle wasting and is repressed in diabetes. J. Clin. Invest.124, 1914–1927 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Pasut, A., Jones, A. E. & Rudnicki, M. A. Isolation and culture of individual myofibers and their satellite cells from adult skeletal muscle. J. Vis. Exp.73, e50074 (2013).

    Google Scholar 

Download references

Acknowledgements

This work was supported by the Italian Ministry of Health (grant no. GR-2013-02356592) to L.M., NIH grants R01AR056712, R01AR052779, P30 AR061303, an MDA grant and EPIGEN grant to P.L.P., NIH grants R01 AR064873 and P30 AR061303 and MDA grant to A.S., California Institute for Regenerative Medicine (CIRM) training grant TG2-01162 and AFM-Téléthon Postdoctoral fellowship (no. 21084) to D.S. The authors thank E. Aleo at the Institute of Applied Genomics in Udine, Italy, for RNA–seq library preparation and sequencing, L. Battistini and G. Borsellino for flow cytometry related discussions and advice, L. Berghella for preliminary analysis, and R. Rizzi for support in in vivo treatments. The authors also thank D. Guttridge, S. Akira and S. Schenk for sharing mice generated in their laboratories, and C. Heil for help with graphic formatting. D.S. and U.E. contributed equally to this work.

Author information

Authors and Affiliations

Authors

Contributions

Project conception and design was carried out by L.M. and P.L.P., denervation procedures and related experiments in vivo and ex vivo, data generation and analysis by L.M., M.P., F.L., D.P. and M.V.A., RNA–seq data analysis and statistics by C.N. and S.G., experiments with STAT3 fl/fl mice, ex vivo STAT3 deletion/blockade and related transplantation assays by D.S., U.E., L.M. and A.S., flow cytometry by L.M. and M.d.B., human ALS sample sourcing by R.R.-G., CD90 expression analysis in mouse and human FAPs by L.G. and L.M., experiments with SODG93A mice and ALS sample analysis by L.M. and M.P., experiments with SCI by S.M. and L.M., and experiments with SMA by V.P., C.S. and L.M. The manuscript was written by P.L.P., and funding was sourced by P.L.P. and L.M.

Corresponding author

Correspondence to Pier Lorenzo Puri.

Ethics declarations

Competing interests.

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Collagen deposition but not regeneration characterize denervated muscles.

(a) Representative immunofluorescence of Collagen1 (Green) in TA muscle cryosection. Collagen area values represent mean ± s.d. **P < 0.01; by one-way ANOVA (n=3 animals/group), 3 independent experiments were performed. Scale bar 100 µm. (b) Representative immunofluorescence of embryonic Myosin Heavy Chain (eMyHC) (Red) in TA muscle cryosections. eMyHC-positive fibers were detected at 7days post-injury only. Scale bar 200 µm. Data shown represent results from 3 animals/group, 3 independent experiments were performed. (c) General gating strategy is shown (d) Representative EdU (Red), CD90 (Green), Laminin (Cyan) and Dapi (White) immunofluorescence in muscle cryosections of control (Ct) and 15 days denervated (Den) TA muscle. Quantification is shown in the graph (left panel). Values represent mean ± s.d. **P<0.01; by student t-test (n=3 animals/group), 3 independent experiments were performed.

Supplementary Figure 2 Increased number of FAPs but not Satellite Cells and Macrophages in denervated muscles.

(a) Pax7 (Red) immunofluorescence in control, injured and denervated TA muscle cryosections at the indicated time-point. Laminin (green) and Dapi (white) counterstain is shown in the bottom merged panel. (b) F4/80 (Green) immunofluorescence analysis is shown in control (CTR), injured (CTX) and denervated (DEN) TA muscle cryosections at the indicated time-points. Laminin (Cyan) and Dapi (white) counterstain is shown in the bottom merged panel. (c) Sca-1 (Red) immunofluorescence analysis is shown in control (CTR), injured (CTX) and denervated (DEN) TA muscle cryosections at the indicated time-points. Laminin (Cyan) and Dapi (white) counterstain is shown in the bottom merged panel. In a-c, Scale bar 100 µm. Data shown in a-c represent results from 3 animals/group, 3 independent experiments were performed. Quantification was shown in (d-e-f). Values represent mean ±s.d. *P<0.05, **P<0.01; by one-way ANOVA (n=3 animals/group).

Supplementary Figure 3 Pathway analysis of RNA-seq from FAP DEN and FAP CTX.

(a) MA-plots showing the fold change and normalized counts for FAPs from denervated (DEN 7 and 15 days) and injured (CTX) muscles, compared to control mice. Data show results from the mean of 2 animals/group (b) Top enriched pathways identified with IPA software, for differentially expressed genes unique to injury (CTX; left), denervation day 15 (DEN15d; right) or common to the two conditions (down). For each plot, x-axis represents the –log of the P-value (right-tailed Fisher’s exact test) and the orange dots on each pathway bar correspond to the ratio of the number of genes in a given pathway that meets the cutoff criteria, divided by the total number of genes belonging to that pathway.

Supplementary Figure 4 RNA-seq analysis from denervated myofibres vs control.

(a) MA-plots showing the fold change and normalized counts for myofibers from 7 days denervated (DEN) muscles compared to control myofibers. Data show results from the mean of 2 animals/group (b) Expression heatmap of genes differentially expressed in myofibers derived from denervated (DEN) muscle compared to myofibers derived from Control (CTR) mice. Gene expression is represented as z-score calculated across the rows. Data show results from the mean of 2 animals/group (c) Upstream Regulators predicted to be differentially activated or inhibited in myofibers from denervated (DEN) myofibers compared to control (CTR) myofibers, by IPA analysis. (d) Top Canonical Pathways altered in myofibers from 7 days Denervated (DEN) muscles, compared to control myofibers. For each plot, x-axis represents the –log of the P-value (right-tailed Fisher’s exact test) and the orange dots on each pathway bar correspond to the ratio of the number of genes in a given pathway that meet the cutoff criteria, divided by the total number of genes belonging to that pathway. (e) Top Metabolic Pathways altered in Myofibers from 7 days Denervated (DEN) muscle compared to control myofibers (see Supp.Fig.3b for further details). For each plot, x-axis represents the –log of the P-value (right-tailed Fisher’s exact test) and the orange dots on each pathway bar correspond to the ratio of the number of genes in a given pathway that meets the cutoff criteria, divided by the total number of genes belonging to that pathway.

Supplementary Figure 5 Myofiber- or satellite cell-specific genetic deletion of STAT3 does not prevent denervation induced muscle atrophy.

(a) Representative immunofluorescence of Laminin (Red) in TA sections from WT, MCK-STAT3-/-, Denervated (WT Den) and MCK_STAT3-/- mice, either unperturbed or 15 days after denervation. (n=4 WT and 5 KO animals per group, from two independent experiments). Scale bar 100 µm (b) Normalized TA muscle weight. (n=4 WT and 5 KO animals per group, from two independent experiments). (c) Frequency distribution of fiber CSA in denervated (WT-DEN) and MCK-STAT3-/- TA muscles. Values represent mean ± S.E.M. (n=4 WT and 5 KO animals per group, from two independent experiments). (d) Western Blot analysis of phopsho- and total-STAT3 with whole muscle extracts. (e) Representative Western Blot of total STAT3, from extract of muscle fibers. (2 independent experiments were performed) (f) Representative immunofluorescence of Laminin (Red) in TA muscle from WT and Pax7-STAT3-/-, mice, either unperturbed or 15 days after denervation. (n=4–5 animals per group, two independent experiments). Scale bar 100 µm (g) Frequency distribution of fiber CSA in WT and Pax7-STAT3-/-, mice, either unperturbed or 15 days after denervation. Values represent mean ± S.E.M. (n=4 WT and 5 KO animals per group, from two independent experiments). (h) Left panel: Western Blot analysis of phosphor-STAT3 from protein extracts of freshly isolated FAPs pooled from from denervated TA muscles of mice (n=3 animals) treated or not with aIL6 neutralizing antibodies or STAT3i. Right panel: Summary of ELISA quantification of secreted IL-6 by FAPs from denervated TA muscles of mice treated or not with aIL6 neutralizing antibodies or STAT3i. Values represent mean of two independent assays.

Supplementary Figure 6 Alteration of FAPs during aging and in a mouse model of SMA.

(a) Frequency distribution of fiber CSA in TA of 2month (Young) and ~ 24 month-old mice (Old). Values represent mean ± s.d. *P < 0.05, **P < 0.01, ***P < 0.001; by two side student t-test (n=4 animals/group). (b) Representative Sirius red staining in Young and Old muscle cryosections. Scale bar 200 μm. (c) Quantification of collagen staining shown in j. Values represent mean ± s.d., **P = 0.006; by two side student t-test (n=4 animals/group). (d) Cyto-fluorimetric count of FAPs in Young and Old mice. Values represent mean ± s.d. (n=3 CTR and n=4 Old animals) (e) Relative expression of indicated genes in freshly isolated FAPs from Young or Old mice. Values represent mean ± s.d. (n=3 CTR and n=4 Old animals) (f) Representative images of 9-day old WT and SMAD7 mice. (g) Muscle weight of 9-day old WT and SMAΔ7 mice. Values represent mean ± s.d. ***P = 0.0001; by two side student t-test (n=5 animals). (h) Cyto-fluorimetric counts of FAPs from 9-day old WT or SMAΔ7 mice. Values represent mean ± s.d., ***P = 0.0001; by two side student t-test. (n=4 animals/group) (i) FAP’s number normalization over muscle weight. Values represent mean ± s.d., **P = 0.001; by two side student t-test. (n=4 animals/group) (j) qPCR analysis of indicated genes in WT and SMAΔ7 freshly isolated FAPs. Values represent mean ± s.d. *P / 0.05; by two side student t-test (n=4 animals/group).

Supplementary Figure 7 Activation of STAT3 in CD90 positive FAPs from human ALS muscles.

(a) Graph showing the Cytofluorimetric profile of surface markers, CD90 and SCA1, expressed in FAPs derived from WT mice. Data shown represent 2 independent experiments) (b) Relative expression of the indicated genes in SC, FAPs Sca1 and FAPs CD90 isolated from WT muscle by qPCR. (n=2 independent experiments) (c) Representative Images of Laminin (Cyan), DAPI (White), STAT3tyr705(Green) and CD90 (purple) immunolocalization in ALS muscle cryosection from SODG93A. Arrows indicate STAT3tyr705 myonuclei while stars indicate STAT3tyr705 positive CD90 interstitial cells. Data shown represent 3 human samples.

Supplementary Figure 8 Unprocessed scan of blots: Part1.

Unprocessed scan of blots was shown.

Supplementary Figure 8 Unprocessed scan of blots: Part2.

Unprocessed scan of blots was shown.

Supplementary information

Supplementary Information

Supplementary Figures 1–8 and legend for Supplementary Table 1.

Reporting Summary

Supplementary Table 1

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Madaro, L., Passafaro, M., Sala, D. et al. Denervation-activated STAT3–IL-6 signalling in fibro-adipogenic progenitors promotes myofibres atrophy and fibrosis. Nat Cell Biol 20, 917–927 (2018). https://doi.org/10.1038/s41556-018-0151-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0151-y

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