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 serum-free media formulation for cultured meat production supports bovine satellite cell differentiation in the absence of serum starvation

Abstract

Cultured meat production requires the robust differentiation of satellite cells into mature muscle fibres without the use of animal-derived components. Current protocols induce myogenic differentiation in vitro through serum starvation, that is, an abrupt reduction in serum concentration. Here we used RNA sequencing to investigate the transcriptomic remodelling of bovine satellite cells during myogenic differentiation induced by serum starvation. We characterized canonical myogenic gene expression, and identified surface receptors upregulated during the early phase of differentiation, including IGF1R, TFRC and LPAR1. Supplementation of ligands to these receptors enabled the formulation of a chemically defined media that induced differentiation in the absence of serum starvation and/or transgene expression. Serum-free myogenic differentiation was of similar extent to that induced by serum starvation, as evaluated by transcriptome analysis, protein expression and the presence of a functional contractile apparatus. Moreover, the serum-free differentiation media supported the fabrication of three-dimensional bioartificial muscle constructs, demonstrating its suitability for cultured beef production.

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: Bovine SCs undergo extensive transcriptomic changes upon serum starvation.
Fig. 2: Differential expression analysis identifies surface receptors upregulated upon serum starvation.
Fig. 3: Serum-free differentiation is induced by supplementation of ligands to upregulated receptors.
Fig. 4: Serum-free differentiation is of comparable extent to serum starvation.
Fig. 5: Transcriptional remodelling during serum-free differentiation is similar to serum starvation.
Fig. 6: Serum-free differentiation medium enables fabrication of bioartificial muscles.

Similar content being viewed by others

Data availability

RNA-seq data has been deposited to the GEO (accession number GSE173199). Source data are provided with this paper. Further data supporting the findings of this study are available from the authors upon request.

Code availability

Analysis code is available from the authors upon request.

References

  1. Parodi, A. et al. The potential of future foods for sustainable and healthy diets. Nat. Sustain. 1, 782–789 (2018).

    Article  Google Scholar 

  2. Post, M. J. et al. Scientific, sustainability and regulatory challenges of cultured meat. Nat. Food 1, 403–415 (2020).

    Article  Google Scholar 

  3. Bischoff, R. Regeneration of single skeletal muscle fibers in vitro. Anat. Rec. 182, 215–235 (1975).

    Article  CAS  PubMed  Google Scholar 

  4. Collins, C. A. et al. Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell 122, 289–301 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Vandenburgh, H. et al. Tissue-engineered skeletal muscle organoids for reversible gene therapy. Hum. Gene Ther. 7, 2195–2200 (1996).

    Article  CAS  PubMed  Google Scholar 

  6. Ben-Arye, T. et al. Textured soy protein scaffolds enable the generation of three-dimensional bovine skeletal muscle tissue for cell-based meat. Nat. Food 1, 210–220 (2020).

  7. Pirkmajer, S. & Chibalin, A. V. Serum starvation: caveat emptor. Am. J. Physiol. Cell Physiol. 301, C272–C279 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Das, M. et al. Developing a novel serum-free cell culture model of skeletal muscle differentiation by systematically studying the role of different growth factors in myotube formation. In Vitro Cell. Dev. Biol. Anim. 45, 378–387 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Guo, X. et al. In vitro differentiation of functional human skeletal myotubes in a defined system. Biomater. Sci. 2, 131–138 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Chal, J. et al. Differentiation of pluripotent stem cells to muscle fiber to model Duchenne muscular dystrophy. Nat. Biotechnol. 33, 962–969 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Akiyama, T. et al. Efficient differentiation of human pluripotent stem cells into skeletal muscle cells by combining RNA-based MYOD1-expression and POU5F1-silencing. Sci. Rep. 8, 1189 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  12. Chal, J. & Pourquié, O. Making muscle: skeletal myogenesis in vivo and in vitro. Development 144, 2104–2122 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Millay, D. P. et al. Myomaker: a membrane activator of myoblast fusion and muscle formation. Nature 499, 301–305 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bryson-Richardson, R. J. & Currie, P. D. The genetics of vertebrate myogenesis. Nat. Rev. Genet. 9, 632–646 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Tsumagari, K. et al. Gene expression during normal and FSHD myogenesis. BMC Med. Genomics 4, 67 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. He, H. & Liu, X. Characterization of transcriptional complexity during longissimus muscle development in bovines using high-throughput sequencing. PLoS ONE 8, e64356 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tripathi, A. K. et al. Transcriptomic dissection of myogenic differentiation signature in caprine by RNA-Seq. Mech. Dev. 132, 79–92 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Lee, E. J. et al. Identification of genes differentially expressed in myogenin knock-down bovine muscle satellite cells during differentiation through RNA sequencing analysis. PLoS ONE 9, e92447 (2014).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  19. Nishiyama, T., Kii, I. & Kudo, A. Inactivation of Rho/ROCK signaling is crucial for the nuclear accumulation of FKHR and myoblast fusion. J. Biol. Chem. 279, 47311–47319 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Capkovic, K. L., Stevenson, S., Johnson, M. C., Thelen, J. J. & Cornelison, D. Neural cell adhesion molecule (NCAM) marks adult myogenic cells committed to differentiation. Exp. Cell. Res. 314, 1553–1565 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tang, Z. et al. Molecular cloning of caveolin-3, a novel member of the caveolin gene family expressed predominantly in muscle. J. Biol. Chem. 271, 2255–2261 (1996).

    Article  CAS  PubMed  Google Scholar 

  22. Seale, P. et al. Pax7 is required for the specification of myogenic satellite cells. Cell 102, 777–786 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Weiss, A. & Leinwand, L. A. The mammalian myosin heavy chain gene family. Annu. Rev. Cell Dev. Biol. 12, 417–439 (1996).

    Article  CAS  PubMed  Google Scholar 

  24. Pardee, A. B. A restriction point for control of normal animal cell proliferation. Proc. Natl Acad. Sci. USA 71, 1286–1290 (1974).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. He, K. et al. A transcriptomic study of myogenic differentiation under the overexpression of PPARÎł by RNA-Seq. Sci. Rep. 7, 15308 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  26. Massoner, P., Ladurner-Rennau, M., Eder, I. E. & Klocker, H. Insulin-like growth factors and insulin control a multifunctional signalling network of significant importance in cancer. Br. J. Cancer 103, 1479–1484 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, G. et al. Chemically defined conditions for human iPSC derivation and culture. Nat. Methods 8, 424–429 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Breton, C. et al. Presence of functional oxytocin receptors in cultured human myoblasts. J. Clin. Endocrinol. Metab. 87, 1415–1418 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Nikolić, N. et al. Electrical pulse stimulation of cultured skeletal muscle cells as a model for in vitro exercise—possibilities and limitations. Acta Physiol. 220, 310–331 (2017).

    Article  Google Scholar 

  30. Gawlitta, D., Boonen, K. J. M., Oomens, C. W. J., Baaijens, F. P. T. & Bouten, C. V. C. The influence of serum-free culture conditions on skeletal muscle differentiation in a tissue-engineered model. Tissue Eng. A 14, 161–171 (2008).

    Article  CAS  Google Scholar 

  31. Lawson, M. A. & Purslow, P. P. Differentiation of myoblasts in serum-free media: effects of modified media are cell line-specific. Cells Tissues Organs 167, 130–137 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Shoji, E., Woltjen, K. & Sakurai, H. Directed myogenic differentiation of human induced pluripotent stem cells. Methods Mol. Biol. Clifton NJ 1353, 89–99 (2016).

    Article  CAS  Google Scholar 

  33. Tong, H. L. et al. Transcriptional profiling of bovine muscle-derived satellite cells during differentiation in vitro by high throughput RNA sequencing. Cell. Mol. Biol. Lett. 20, 351–373 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Brunetti, A., Maddux, B. A., Wong, K. Y. & Goldfine, I. D. Muscle cell differentiation is associated with increased insulin receptor biosynthesis and messenger RNA levels. J. Clin. Invest. 83, 192–198 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ozawa, E. Transferrin as a muscle trophic factor. Rev. Physiol., Biochem. Pharmacol. 113, 89–141 (1989).

    Article  CAS  Google Scholar 

  36. Cox, R. D., Garner, I. & Buckingham, M. E. Transcriptional regulation of actin and myosin genes during differentiation of a mouse muscle cell line. Differ. Res. Biol. Divers. 43, 183–191 (1990).

    CAS  Google Scholar 

  37. Hansen, L. H., Abrahamsen, N. & Nishimura, E. Glucagon receptor mRNA distribution in rat tissues. Peptides 16, 1163–1166 (1995).

    Article  CAS  PubMed  Google Scholar 

  38. Uhlén, M. et al. Proteomics. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  Google Scholar 

  39. Cencetti, F. et al. Lysophosphatidic acid stimulates cell migration of satellite cells. A role for the sphingosine kinase/sphingosine 1-phosphate axis. FEBS J. 281, 4467–4478 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Yoshida, S., Fujisawa-Sehara, A., Taki, T., Arai, K. & Nabeshima, Y. Lysophosphatidic acid and bFGF control different modes in proliferating myoblasts. J. Cell Biol. 132, 181–193 (1996).

    Article  CAS  PubMed  Google Scholar 

  41. Zhang, Z. et al. Oxytocin is involved in steroid hormone-stimulated bovine satellite cell proliferation and differentiation in vitro. Domest. Anim. Endocrinol. 66, 1–13 (2019).

    Article  PubMed  Google Scholar 

  42. Denes, L. T. et al. Culturing C2C12 myotubes on micromolded gelatin hydrogels accelerates myotube maturation. Skelet. Muscle 9, 17 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Stephens, N. et al. Bringing cultured meat to market: technical, socio-political, and regulatory challenges in cellular agriculture. Trends Food Sci. Technol. 78, 155–166 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Furuhashi, M. et al. Formation of contractile 3D bovine muscle tissue for construction of millimetre-thick cultured steak. NPJ Sci. Food 5, 6 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Costantini, M. et al. Microfluidic-enhanced 3D bioprinting of aligned myoblast-laden hydrogels leads to functionally organized myofibers in vitro and in vivo. Biomaterials 131, 98–110 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Lk, L. et al. A pilot study on pain and the upregulation of myoglobin through low-frequency and high-amplitude electrical stimulation-induced muscle contraction. J. Phys. Ther. Sci. 26, 985–988 (2014).

    Article  Google Scholar 

  47. Melzener, L., Verzijden, K., Buijs, A., Post, M. & Flack, J. Cultured beef: from small biopsy to substantial quantity. J. Sci. Food Agric. 101, 7–14 (2020).

  48. Ding, S. et al. Maintaining bovine satellite cells stemness through p38 pathway. Sci. Rep. 8, 10808 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  49. 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).

    Article  CAS  PubMed  Google Scholar 

  50. Yates, A. D. et al. Ensembl 2020. Nucleic Acids Res. 48, D682–D688 (2020).

    CAS  PubMed  Google Scholar 

  51. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  53. The Gene Ontology Consortium. The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 47, D330–D338 (2019).

    Article  Google Scholar 

  54. Zhang, X. et al. Proteome-wide identification of ubiquitin interactions using UbIA-MS. Nat. Protoc. 13, 530–550 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Derks and D. Tserpelis (Genome Services Maastricht UMC+) for their support in the acquisition and analysis of RNA-seq data, R. Mohren (Imaging Mass Spectrometry, M4I, Maastricht University) for proteomics data, and H. Duimel and C. LĂłpez Iglesia (Microscopy CORE Lab, M4I, Maastricht University) for SEM. We also thank J. Melke and D. Remmers (Mosa Meat BV) for their assistance with confocal microscopy.

Author information

Authors and Affiliations

Authors

Contributions

T.M., I.K., C.F., E.O., A.D. and J.E.F. performed experiments and analysis. A.D., H.C., M.J.P. and J.E.F. supervised the study. T.M., M.J.P. and J.E.F. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Joshua E. Flack.

Ethics declarations

Competing interests

T.M., I.K., C.F., E.O., A.D., H.C. and J.E.F. are employees of Mosa Meat BV. M.J.P. is co-founder and stakeholder of Mosa Meat BV. The study was funded by Mosa Meat BV. Mosa Meat BV has patents pending on serum-free proliferation medium (PCT/P125933PC00) and serum-free differentiation medium (JBB/P126144NL00). All authors declare no other competing interests.

Additional information

Peer review information Nature Food thanks Deepak Choudhury, Laura Domigan and Min Du for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Gene expression during myogenic differentiation induced by serum starvation (related to Fig. 1).

a, RNA-seq-derived median fold changes of selected muscle-related genes during serum starvation compared to 0 h. b, Mean fold changes of genes shown in a), determined via RT–qPCR; error bars indicate SD, n = 3. c, Median fold changes of selected strongly upregulated myogenic genes compared to 0 h, determined via RNA-seq. d, Median fold changes of selected downregulated genes compared to 0 h, determined via RNA-seq; boxes indicate IQR, whiskers show 1.5 × IQR.

Extended Data Fig. 2 Transcriptomic and proteomic characterisation of serum starvation (related to Fig. 2).

a, Volcano plot showing differentially expressed genes between 0 h (yellow) and 96 h (red) of serum starvation. Selected differentially expressed muscle, stem cell or cell cycle-related genes are indicated. b, Scatter plot showing correlation of log2-fold changes of overlapping genes from bovine (x-axis) with C2C12 (y-axis) with indicated Pearson correlation coefficient (R). Colours indicate upregulation (red) or downregulation (yellow) in bovine gene expression, shapes indicate whether differentially expressed genes are simultaneously up/downregulated in both species (squares) or significantly up/downregulated in one species while inversely regulated in the other (triangles). c, Median fold changes of muscle-related protein levels from 0 h to 72 h post serum starvation, normalised against 0 h; boxes indicate IQR, whiskers show 1.5 × IQR. d, Scatter plot showing the Pearson correlation of log2-fold changes of genes from RNA-seq (y-axis) and corresponding proteins from mass spectrometry (x-axis) upon serum starvation with indicated correlation coefficient (R). Colours indicate upregulation (red) or downregulation (yellow) while shapes indicate significantly regulated proteins (points), genes (triangles), or both (squares). e, Mean fold gene expression changes of differentially regulated surface receptors, determined by RT–qPCR; error bars indicate SD, n = 3.

Extended Data Fig. 3 Serum-free differentiation and serum starvation are similar with respect to cell age and coating (related to Fig. 4).

a, Normalised nuclei counts of SCs differentiating on indicated coatings after 72 h in SFB, SFDM and serum starvation as percentage against SFB; statistical significance is indicated for each media against respective Matrigel control, error bars indicate SD, n = 3. b, Normalised nuclei counts of SCs after 72 h of SFB, SFDM or serum starvation at early (left), medium (centre), and late (right) passages with indicated PDs, as percentage of low PDs in SFB; asterisks directly above bars indicate statistical significance against SFB; error bars indicate SD, n = 4. c, Representative fluorescence images of differentiating SCs at early (top), medium (middle) or late (bottom row) passages after 72 h in SFB (left), SFDM (centre) or serum starvation (right), corresponding to Fig. 4e, Extended Data Fig. 3b; green, desmin; blue, Hoechst. Scale bar, 500 µm. *P < 0.05, **P < 0.005, ***P < 0.001.

Extended Data Fig. 4 Extent of serum-free differentiation varies between donor animals (related to Fig. 4).

a, Normalised nuclei counts of SCs from different donor animals after 72 h of myogenic differentiation as percentage of SFB with statistical significance indicated between SFDM and serum starvation respectively for each donor; error bars indicate SD, n = 4. b, Mean fusion indices of SCs from different donor animals after 72 h of serum-free or serum starvation induced differentiation, normalised against respective SFB condition. Statistical significance is indicated between SFDM and serum starvation respectively for each donor; error bars indicate SD, n = 4. c, Representative fluorescence images of myogenic differentiation of SCs from different donor animals after 72 h in SFB (top), SFDM (middle) and serum starvation (bottom row); green, desmin; blue, Hoechst. Scale bar, 500 µm. *P < 0.05, **P < 0.005, ***P < 0.001.

Extended Data Fig. 5 2D serum-free differentiation can be achieved with different basal media (related to Fig. 6).

a, Representative fluorescence images of SCs after 72 h in SFB, SFDM with DMEM/F-12 and DMEM base, and upon serum starvation; green, desmin; blue, Hoechst. Scale bar, 500 µM. b, Normalised nuclei counts from a) as percentage of SFB with statistical significance indicated against SFDM (DMEM/F-12); error bars indicate SD, n = 4. c, Mean fusion indices derived from a) with statistical significance performed against SFDM (DMEM/F-12); error bars indicate SD, n = 4. d, Scatter plot indicating correlation of log2-fold changes between SFGM and DMEM/F-12-based SFDM (x-axis) against log2-fold changes between SFGM and DMEM-based SFDM (y-axis) with Pearson correlation coefficient indicated. *P < 0.05, **P < 0.005, ***P < 0.001.

Extended Data Fig. 6 Acetylcholine supplementation improves myogenic fusion in bioartificial muscles (related to Fig. 6).

a, Ultrastructure of BAMs after 192 h in SFB or SFDM (with DMEM/F-12 or DMEM basal media) or serum starvation. Scale bar, 100 µm. b, Representative fluorescence images of BAMs after 192 h in DMEM-based SFDM without (left) and with (right) 10 µM acetylcholine (ACh); pink, desmin; red, 𝛂-actin; green, myosinHC; blue, Hoechst. Scale bars, 100 µm. c, Ultrastructure (top), wide (middle) and close-up (bottom) scanning electron microscopy images of BAMs after 192 h in DMEM-based SFDM with (right) and without (left) 10 µM acetylcholine. Scale bars, 100 µm.

Extended Data Table 1 Media Formulations
Extended Data Table 2 RT–qPCR primers

Supplementary information

Reporting Summary

Supplementary Video 1

Electrical pulse stimulation of serum-free myotubes (related to Fig. 4). SCs were differentiated in SFDM for 192 h before electrical pulse stimulation by a C-PACE EP stimulator at 12 V, 1.0 ms pulse width and indicated frequencies.

Supplementary Table 1

Differential gene expression analysis between 0 and 96 h of serum starvation.

Supplementary Table 2

Proteomic changes during serum starvation.

Source data

Source Data Fig. 1

Uncropped blots corresponding to Fig. 1.

Source Data Fig. 4

Uncropped blots corresponding to Fig. 4.

Source Data Fig. 6

Uncropped blots corresponding to Fig. 6.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Messmer, T., Klevernic, I., Furquim, C. et al. A serum-free media formulation for cultured meat production supports bovine satellite cell differentiation in the absence of serum starvation. Nat Food 3, 74–85 (2022). https://doi.org/10.1038/s43016-021-00419-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43016-021-00419-1

This article is cited by

Search

Quick links

Nature Briefing Anthropocene

Sign up for the Nature Briefing: Anthropocene newsletter — what matters in anthropocene research, free to your inbox weekly.

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