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.

  • Protocol
  • Published:

3D human induced pluripotent stem cell–derived bioengineered skeletal muscles for tissue, disease and therapy modeling

Abstract

Skeletal muscle is a complex tissue composed of multinucleated myofibers responsible for force generation that are supported by multiple cell types. Many severe and lethal disorders affect skeletal muscle; therefore, engineering models to reproduce such cellular complexity and function are instrumental for investigating muscle pathophysiology and developing therapies. Here, we detail the modular 3D bioengineering of multilineage skeletal muscles from human induced pluripotent stem cells, which are first differentiated into myogenic, neural and vascular progenitor cells and then combined within 3D hydrogels under tension to generate an aligned myofiber scaffold containing vascular networks and motor neurons. 3D bioengineered muscles recapitulate morphological and functional features of human skeletal muscle, including establishment of a pool of cells expressing muscle stem cell markers. Importantly, bioengineered muscles provide a high-fidelity platform to study muscle pathology, such as emergence of dysmorphic nuclei in muscular dystrophies caused by mutant lamins. The protocol is easy to follow for operators with cell culture experience and takes between 9 and 30 d, depending on the number of cell lineages in the construct. We also provide examples of applications of this advanced platform for testing gene and cell therapies in vitro, as well as for in vivo studies, providing proof of principle of its potential as a tool to develop next-generation neuromuscular or musculoskeletal therapies.

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: Workflow to generate and analyse 3D skeletal muscle constructs using hiPSC-derived progenitors.
Fig. 2: Pilot testing and quality control for biomaterials and cells.
Fig. 3: Characterization of hiPSC-derived 3D bioengineered muscles.
Fig. 4: Modeling muscular dystrophies caused by abnormal nuclear envelope by using hiPSC-derived 3D muscles: stepwise 3D nuclear reconstruction of hiPSC-derived healthy control and LMNA-mutant skeletal myotubes in monolayer versus 3D (single-lineage) artificial muscles.
Fig. 5: Examples of in vitro and in vivo applications of hiPSC-derived 3D artificial muscles: testing gene therapy vectors, assessing cell migration capacity and studying biocompatibility.

Similar content being viewed by others

Data availability

Key raw data linked to the original publications30,31,54 reported in this protocol have been deposited in Mendeley Data and are available at https://doi.org/10.17632/d826fxhr3b.1. Raw data relative to Figs. 3c and 5b,c are available at https://doi.org/10.17632/hzmx5nkyff.1.

References

  1. Tedesco, F. S., Dellavalle, A., Diaz-Manera, J., Messina, G. & Cossu, G. Repairing skeletal muscle: regenerative potential of skeletal muscle stem cells. J. Clin. Invest. 120, 11–19 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Wosczyna, M. N. & Rando, T. A. A muscle stem cell support group: coordinated cellular responses in muscle regeneration. Dev. Cell. 46, 135–143 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Tedesco, F. S., Moyle, L. A. & Perdiguero, E. Muscle interstitial cells: a brief field guide to non-satellite cell populations in skeletal muscle. Methods Mol. Biol. 1556, 129–147 (2017).

    Article  PubMed  Google Scholar 

  4. Mercuri, E., Bonnemann, C. G. & Muntoni, F. Muscular dystrophies. Lancet 394, 2025–2038 (2019).

    Article  PubMed  Google Scholar 

  5. Jalal, S., Dastidar, S. & Tedesco, F. S. Advanced models of human skeletal muscle differentiation, development and disease: Three-dimensional cultures, organoids and beyond. Curr. Opin. Cell Biol. 73, 92–104 (2021).

  6. Fatehullah, A., Tan, S. H. & Barker, N. Organoids as an in vitro model of human development and disease. Nat. Cell Biol. 18, 246–254 (2016).

    Article  PubMed  Google Scholar 

  7. Lancaster, M. A. & Knoblich, J. A. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345, 1247125 (2014).

    Article  PubMed  Google Scholar 

  8. Wang, L., Wu, Y., Guo, B. & Ma, P. X. Nanofiber yarn/hydrogel core-shell scaffolds mimicking native skeletal muscle tissue for guiding 3D myoblast alignment, elongation, and differentiation. ACS Nano 9, 9167–9179 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Shimizu, K. et al. Microfluidic devices for construction of contractile skeletal muscle microtissues. J. Biosci. Bioeng. 119, 212–216 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Sakar, M. S. et al. Formation and optogenetic control of engineered 3D skeletal muscle bioactuators. Lab Chip 12, 4976–4985 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Pruller, J., Mannhardt, I., Eschenhagen, T., Zammit, P. S. & Figeac, N. Satellite cells delivered in their niche efficiently generate functional myotubes in three-dimensional cell culture. PLoS One 13, e0202574 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Morimoto, Y., Kato-Negishi, M., Onoe, H. & Takeuchi, S. Three-dimensional neuron-muscle constructs with neuromuscular junctions. Biomaterials 34, 9413–9419 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Juhas, M., Engelmayr, G. C. Jr., Fontanella, A. N., Palmer, G. M. & Bursac, N. Biomimetic engineered muscle with capacity for vascular integration and functional maturation in vivo. Proc. Natl. Acad. Sci. USA 111, 5508–5513 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Heher, P. et al. A novel bioreactor for the generation of highly aligned 3D skeletal muscle-like constructs through orientation of fibrin via application of static strain. Acta Biomater. 24, 251–265 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Cvetkovic, C. et al. Three-dimensionally printed biological machines powered by skeletal muscle. Proc. Natl. Acad. Sci. USA 111, 10125–10130 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Corona, B. T., Ward, C. L., Baker, H. B., Walters, T. J. & Christ, G. J. Implantation of in vitro tissue engineered muscle repair constructs and bladder acellular matrices partially restore in vivo skeletal muscle function in a rat model of volumetric muscle loss injury. Tissue Eng. Part A 20, 705–715 (2014).

    CAS  PubMed  Google Scholar 

  17. Chen, H. et al. Enhanced growth and differentiation of myoblast cells grown on E-jet 3D printed platforms. Int. J. Nanomedicine 14, 937–950 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Capel, A. J. et al. Scalable 3D printed molds for human tissue engineered skeletal muscle. Front. Bioeng. Biotechnol. 7, 20 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Fusto, A., Moyle, L. A., Gilbert, P. M. & Pegoraro, E. Cored in the act: the use of models to understand core myopathies. Dis. Model. Mech. 12, dmm.041368 (2019).

    Article  Google Scholar 

  20. Trevisan, C. et al. Generation of a functioning and self-renewing diaphragmatic muscle construct. Stem Cells Transl. Med. 8, 858–869 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tchao, J. et al. Engineered human muscle tissue from skeletal muscle derived stem cells and induced pluripotent stem cell derived cardiac cells. Int. J. Tissue Eng. 2013, 198762 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Quarta, M. et al. Bioengineered constructs combined with exercise enhance stem cell-mediated treatment of volumetric muscle loss. Nat. Commun. 8, 15613 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Powell, C. A., Smiley, B. L., Mills, J. & Vandenburgh, H. H. Mechanical stimulation improves tissue-engineered human skeletal muscle. Am. J. Physiol. Cell Physiol. 283, C1557–C1565 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Passipieri, J. A. et al. Keratin hydrogel enhances in vivo skeletal muscle function in a rat model of volumetric muscle loss. Tissue Eng. Part A 23, 556–571 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Madden, L., Juhas, M., Kraus, W. E., Truskey, G. A. & Bursac, N. Bioengineered human myobundles mimic clinical responses of skeletal muscle to drugs. Elife 4, e04885 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Fuoco, C. et al. In vivo generation of a mature and functional artificial skeletal muscle. EMBO Mol. Med. 7, 411–422 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chiron, S. et al. Complex interactions between human myoblasts and the surrounding 3D fibrin-based matrix. PLoS One 7, e36173 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bersini, S. et al. Engineering an environment for the study of fibrosis: a 3D human muscle model with endothelium specificity and endomysium. Cell Rep. 25, 3858–3868.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Afshar Bakooshli, M. et al. A 3D culture model of innervated human skeletal muscle enables studies of the adult neuromuscular junction. Elife 8, e44530 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Steele-Stallard, H. B. et al. Modeling skeletal muscle laminopathies using human induced pluripotent stem cells carrying pathogenic LMNA mutations. Front. Physiol. 9, 1332 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Maffioletti, S. M. et al. Three-dimensional human iPSC-derived artificial skeletal muscles model muscular dystrophies and enable multilineage tissue engineering. Cell Rep. 23, 899–908 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rao, L., Qian, Y., Khodabukus, A., Ribar, T. & Bursac, N. Engineering human pluripotent stem cells into a functional skeletal muscle tissue. Nat. Commun. 9, 126 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Osaki, T., Uzel, S. G. M. & Kamm, R. D. Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells and optogenetic motor neurons. Sci. Adv. 4, eaat5847 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Faustino Martins, J. M. et al. Self-organizing 3D human trunk neuromuscular organoids. Cell Stem Cell 26, 172–186.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Relaix, F. & Zammit, P. S. Satellite cells are essential for skeletal muscle regeneration: the cell on the edge returns centre stage. Development 139, 2845–2856 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Worman, H. J. Nuclear lamins and laminopathies. J. Pathol. 226, 316–325 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Maffioletti, S. M. et al. Efficient derivation and inducible differentiation of expandable skeletal myogenic cells from human ES and patient-specific iPS cells. Nat. Protoc. 10, 941–958 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Caron, L. et al. A human pluripotent stem cell model of facioscapulohumeral muscular dystrophy-affected skeletal muscles. Stem Cells Transl. Med. 5, 1145–1161 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Orlova, V. V. et al. Generation, expansion and functional analysis of endothelial cells and pericytes derived from human pluripotent stem cells. Nat. Protoc. 9, 1514–1531 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Stacpoole, S. R. et al. Efficient derivation of NPCs, spinal motor neurons and midbrain dopaminergic neurons from hESCs at 3% oxygen. Nat. Protoc. 6, 1229–1240 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hall, C. E. et al. Progressive motor neuron pathology and the role of astrocytes in a human stem cell model of VCP-related ALS. Cell Rep. 19, 1739–1749 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Machado, C. B. et al. In vitro modelling of nerve-muscle connectivity in a compartmentalised tissue culture device. Adv. Biosyst. 3, 1800307 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Paredes-Redondo, A. et al. Optogenetic modeling of human neuromuscular circuits in Duchenne muscular dystrophy with CRISPR and pharmacological corrections. Sci. Adv. 7, eabi8787 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Martin, N. R. et al. Neuromuscular junction formation in tissue-engineered skeletal muscle augments contractile function and improves cytoskeletal organization. Tissue Eng. Part A 21, 2595–2604 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mills, R. J. et al. Development of a human skeletal micro muscle platform with pacing capabilities. Biomaterials 198, 217–227 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Afshar, M. E. et al. A 96-well culture platform enables longitudinal analyses of engineered human skeletal muscle microtissue strength. Sci. Rep. 10, 6918 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Grebenyuk, S. & Ranga, A. Engineering organoid vascularization. Front. Bioeng. Biotechnol. 7, 39 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  48. de la Puente, P. & Ludena, D. Cell culture in autologous fibrin scaffolds for applications in tissue engineering. Exp. Cell Res. 322, 1–11 (2014).

    Article  PubMed  Google Scholar 

  49. Ben Yaou, R. et al. International retrospective natural history study of LMNA-related congenital muscular dystrophy. Brain Commun. 3, fcab075 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Rose, N., et al. Bioengineering a miniaturized in vitro 3D myotube contraction monitoring chip for modelization of muscular dystrophies. Biomater. 293, 121935 (2023).

  51. Tedesco, F. S. et al. Stem cell–mediated transfer of a human artificial chromosome ameliorates muscular dystrophy. Sci. Transl. Med. 3, 96ra78 (2011).

  52. Mendell, J. R. et al. Single-dose gene-replacement therapy for spinal muscular atrophy. New England J. Med. 377, 1713–1722 (2017).

  53. Choi, S., Ferrari, G. & Tedesco, F. S. Cellular dynamics of myogenic cell migration: molecular mechanisms and implications for skeletal muscle cell therapies. EMBO Mol. Med. 12, e12357 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Choi, S. et al. Assessing and enhancing migration of human myogenic progenitors using directed iPS cell differentiation and advanced tissue modelling. EMBO Mol. Med. 14, e14526 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hagemann, C. et al. Soft-lithography on SLA 3D printed moulds for fast, versatile, and accessible high-resolution fabrication of customised multiscale cell culture devices with complex designs Preprint at bioRxiv https://doi.org/10.1101/2022.02.22.481424 (2022).

  56. Loperfido, M., Steele-Stallard, H. B., Tedesco, F. S. & VandenDriessche, T. Pluripotent stem cells for gene therapy of degenerative muscle diseases. Curr. Gene Ther. 15, 364–380 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. Selvaraj, S., Kyba, M. & Perlingeiro, R. C. R. Pluripotent stem cell-based therapeutics for muscular dystrophies. Trends Mol. Med. 25, 803–816 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Darabi, R. et al. Human ES- and iPS-derived myogenic progenitors restore DYSTROPHIN and improve contractility upon transplantation in dystrophic mice. Cell Stem Cell 10, 610–619 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tedesco, F. S. et al. Transplantation of genetically corrected human iPSC-derived progenitors in mice with limb-girdle muscular dystrophy. Sci. Transl. Med. 4, 140ra189 (2012).

    Article  Google Scholar 

  60. Goudenege, S. et al. Myoblasts derived from normal hESCs and dystrophic hiPSCs efficiently fuse with existing muscle fibers following transplantation. Mol. Ther. 20, 2153–2167 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Shoji, E. et al. Early pathogenesis of Duchenne muscular dystrophy modelled in patient-derived human induced pluripotent stem cells. Sci. Rep. 5, 12831 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kim, H. et al. Genomic safe harbor expression of PAX7 for the generation of engraftable myogenic progenitors. Stem Cell Rep. 16, 10–19 (2021).

    Article  CAS  Google Scholar 

  63. Albini, S. et al. Epigenetic reprogramming of human embryonic stem cells into skeletal muscle cells and generation of contractile myospheres. Cell Rep. 3, 661–670 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hicks, M. R. et al. ERBB3 and NGFR mark a distinct skeletal muscle progenitor cell in human development and hPSCs. Nat. Cell Biol. 20, 46–57 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Chal, J. et al. Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro. Nat. Protoc. 11, 1833–1850 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Borchin, B., Chen, J. & Barberi, T. Derivation and FACS-mediated purification of PAX3+/PAX7+ skeletal muscle precursors from human pluripotent stem cells. Stem Cell Rep. 1, 620–631 (2013).

    Article  CAS  Google Scholar 

  67. Hansen, A. et al. Development of a drug screening platform based on engineered heart tissue. Circ. Res. 107, 35–44 (2010).

    Article  CAS  PubMed  Google Scholar 

  68. Sala, L. et al. MUSCLEMOTION: a versatile open software tool to quantify cardiomyocyte and cardiac muscle contraction in vitro and in vivo. Circ. Res. 122, e5–e16 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all co-authors of the original articles describing this technology, the EU FP7 projects Biodesign (262948) and Plurimes (602423) and G. Cossu, H. Redl and T. Eschenaghen for their initial support and contribution. We are grateful to CureCMD, Cellular Dynamics International, Inc., M. Oshimura and T. VandenDriessche for cell lines used in our original papers; to J. Ng for AAV9 vector particles; to the DSHB for providing MF20, F5D and titin antibodies; and to all laboratory members for helpful discussions. This work was supported by the European Research Council (759108–HISTOID); MDUK (19GRO-PS48-0188 and 17GRO-PS48-0093-1); the BBSRC London Interdisciplinary Biosciences Consortium (LIDo; BB/M009513/1 to F.S.T., P.S.Z., L.P. and N.K.); AFM-Telethon (21687 and 23782); Duchenne Parent Project NL (19.005); and the Francis Crick Institute, which receives its core funding from Cancer Research UK, the UK Medical Research Council (MRC) and the Wellcome Trust (CC0102). F.S.T. also acknowledges funding by the NIHR (CL-2018-18-008; the views expressed are those of the authors and not necessarily those of the National Health Service (NHS), the NIHR or the Department of Health). E.N. and A.S.B. acknowledge support from the Company of Biologists (DMMTF1905206) and Wellcome Trust (210987/Z/18/Z), respectively. The Zammit laboratory acknowledges the support of the MRC (MR/P023215/1 and MR/S002472/1), MDUK (19GRO-PG12-0493) and the FSHD Society. Work with human cells was performed under approval of the NHS Health Research Authority Research Ethics Committee (reference no. 13/LO/1826) and Integrated Research Application System (IRAS) project (ID No. 141100).

Author information

Authors and Affiliations

Authors

Contributions

L.P., M.K. and F.S.T. wrote the paper, assembled the figures and performed the protocolization and optimization of all the steps listed. V.M.L. and S.S. contributed equally to this work. L.P. performed disease modeling experiments and analysis. M.K. performed calcium experiments and advanced imaging. M.K. and S.S. performed AAV experiments with support from J.R.C. V.M.L. performed contractility and calcium experiments with support from A.S.B. S.D. and S.W.C. performed (Fig. 5a,c) experiments, respectively. S.M.M. and S.S. developed the 3D skeletal muscle platform with F.S.T. and discussed results. N.K. derived neural progenitor cells. E.N. performed work on immortalized myoblasts provided by A.B. P.S.Z. co-supervised L.P. with F.S.T. during his PhD research, contributed to his funding and reviewed manuscript drafts. F.S.T. provided funding, coordinated the work, discussed and analyzed results, wrote and reviewed manuscript drafts with all co-authors and finalized the paper.

Corresponding author

Correspondence to Francesco Saverio Tedesco.

Ethics declarations

Competing interests

F.S.T. has received speaker and consultancy honoraria from Takeda, Sanofi Genzyme and Aleph Farms (via UCL Consultants). All other authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Pier Puri and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Related links

Key references using this protocol

Maffioletti, S. M. et al. Cell Rep. 23, 899–908 (2018): https://doi.org/10.1016/j.celrep.2018.03.091

Steele-Stallard, H. B. et al. Front. Physiol. 9, 1332 (2018): https://doi.org/10.3389/fphys.2018.01332

Choi, S. et al. EMBO Mol. Med. 14, e14526 (2022): https://doi.org/10.15252/emmm.202114526

Supplementary information

Supplementary Information

Supplementary Table 1 and Protocol 1

Supplementary Video 1

Real-time video of DMD hiPSC-derived 3D skeletal muscle construct contractions in response to 10-V electrical stimulation at 1 Hz

Supplementary Video 2

Calcium transients in DMD hiPSC-derived 3D artificial muscles incubated with the fluorescence Ca2+ indicator Fluo-4AM upon electrical stimulation. During the first 10 s, no electrical stimuli were applied to the muscle. At second 10, the 3D muscle was stimulated with 10 V at 0.33 Hz, and at second 20, the 3D muscle was stimulated with 20 V at 0.33 Hz

Supplementary Video 3

Maximum-intensity projection time-lapse video of 5-chloromethylfluorescein diacetate (CMFDA)-stained HIDEMs (green) deposited on hiPSC-derived 3D muscles (red). 100-µm stacks of muscles were imaged every 12 min for 4 h by using a spinning disk microscope. Scale bar, 100 µm

Supplementary Video 4

3D reconstruction of a 3D artificial muscle imaged with a light-sheet microscope immunostained as detailed in the advanced imaging section of Fig. 2

Supplementary Video 5

3D reconstruction nuclear morphology (lamin A/C in red) and myotubes (eMyHC in green) of a healthy control hiPSC-derived 3D muscle construct shown in Fig. 3. The 3D reconstruction was done by using Imaris software and confocal z-stacks of immunostained artificial muscles. Video reproduced from ref. 29 under Creative Commons license CC BY 4.0

Supplementary Video 6

3D reconstruction nuclear morphology (lamin A/C in red) and myotubes (eMyHC in green) of an LMNA-mutant (R249W) hiPSC-derived 3D muscle construct shown in Fig. 3. The 3D reconstruction was done by using Imaris software and confocal z-stacks of immunostained artificial muscles. Video reproduced from ref. 29 under Creative Commons license CC BY 4.0

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pinton, L., Khedr, M., Lionello, V.M. et al. 3D human induced pluripotent stem cell–derived bioengineered skeletal muscles for tissue, disease and therapy modeling. Nat Protoc 18, 1337–1376 (2023). https://doi.org/10.1038/s41596-022-00790-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-022-00790-8

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research