リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Grafting of iPS cell-derived tenocytes promotes motor function recovery after Achilles tendon rupture」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Grafting of iPS cell-derived tenocytes promotes motor function recovery after Achilles tendon rupture

Nakajima, Taiki Nakahata, Akihiro Yamada, Naoki Yoshizawa, Keiko Kato, M, Tomoaki Iwasaki, Mio Zhao, Chengzhu Kuroki, Hiroshi Ikeya, Makoto 京都大学 DOI:10.1038/s41467-021-25328-6

2021

概要

Tendon self-renewal is a rare occurrence because of the poor vascularization of this tissue; therefore, reconstructive surgery using autologous tendon is often performed in severe injury cases. However, the post-surgery re-injury rate is relatively high, and the collection of autologous tendons leads to muscle weakness, resulting in prolonged rehabilitation. Here, we introduce an induced pluripotent stem cell (iPSC)-based technology to develop a therapeutic option for tendon injury. First, we derived tenocytes from human iPSCs by recapitulating the normal progression of step-wise narrowing fate decisions in vertebrate embryos. We used single-cell RNA sequencing to analyze the developmental trajectory of iPSC-derived tenocytes. We demonstrated that iPSC-tenocyte grafting contributed to motor function recovery after Achilles tendon injury in rats via engraftment and paracrine effects. The biomechanical strength of regenerated tendons was comparable to that of healthy tendons. We suggest that iPSC-tenocytes will provide a therapeutic option for tendon injury.

この論文で使われている画像

参考文献

1.

Webster, K. E., Feller, J. A., Leigh, W. B. & Richmond, A. K. Younger patients

are at increased risk for graft rupture and contralateral injury after anterior

cruciate ligament reconstruction. Am. J. Sports Med. 42, 641–647 (2014).

2. Gifstad, T. et al. Long-term follow-up of patellar tendon grafts or hamstring

tendon grafts in endoscopic ACL reconstructions. Knee Surg. Sports

Traumatol. Arthrosc. 21, 576–583 (2013).

3. Alshomer, F., Chaves, C. & Kalaskar, D. M. Advances in tendon and ligament

tissue engineering: materials perspective. J. Mater. 2018, 9868151 (2018).

4. Huang, T. F. et al. Mesenchymal stem cells from a hypoxic culture improve

and engraft achilles tendon repair. Am. J. Sports Med. 41, 1117–1125 (2013).

5. Chamberlain, C. S., Saether, E. E., Aktas, E. & Vanderby, R. Mesenchymal

stem cell therapy on tendon/ligament healing. J. cytokine Biol. 2, 112 (2017).

6. Tang, Q.-M. et al. Fetal and adult fibroblasts display intrinsic differences in

tendon tissue engineering and regeneration. Sci. Rep. 4, 5515 (2014).

7. Okamoto, N. et al. Treating Achilles tendon rupture in rats with bone-marrowcell transplantation therapy. J. Bone Jt. Surg. Am. 92, 2776–2784 (2010).

8. Yang, G., Rothrauff, B. B. & Tuan, R. S. Tendon and ligament regeneration

and repair: clinical relevance and developmental paradigm. Birth Defects Res.

C. Embryo Today 99, 203–222 (2013).

9. Ni, M. et al. Tendon-derived stem cells (TDSCs) promote tendon repair in a

rat patellar tendon window defect model. J. Orthop. Res. 30, 613–619 (2012).

10. Ho, J. O., Sawadkar, P. & Mudera, V. A review on the use of cell therapy in the

treatment of tendon disease and injuries. J. Tissue Eng. 5,

2041731414549678–2041731414549678 (2014).

11. Lui, P. P. Y. Stem cell technology for tendon regeneration: current status,

challenges, and future research directions. Stem Cells Cloning 8, 163–174 (2015).

ARTICLE

12. Dominici, M. et al. Minimal criteria for defining multipotent mesenchymal

stromal cells. The International Society for Cellular Therapy position

statement. Cytotherapy 8, 315–317 (2006).

13. Nakajima, T. et al. Modeling human somite development and fibrodysplasia

ossificans progressiva with induced pluripotent stem cells. Development 145

(2018).

14. Nakajima, T., Sakurai, H. & Ikeya, M. In vitro generation of somite derivatives

from human induced pluripotent stem cells. J. Vis. Exp. https://doi.org/

10.3791/59359 (2019).

15. Nakajima, T. & Ikeya, M. Development of pluripotent stem cell-based human

tenocytes. Dev. Growth Differ. 63, 38–46 (2021).

16. Li, Y. et al. The role of scleraxis in fate determination of mesenchymal stem

cells for tenocyte differentiation. Sci. Rep. 5, 13149 (2015).

17. Chen, X. et al. Stepwise differentiation of human embryonic stem cells

promotes tendon regeneration by secreting fetal tendon matrix and

differentiation factors. Stem Cells 27, 1276–1287 (2009).

18. Lee, J. Y. et al. BMP-12 treatment of adult mesenchymal stem cells in vitro

augments tendon-like tissue formation and defect repair in vivo. PLoS ONE 6,

e17531 (2011).

19. Sassoon, A. A. et al. Skeletal muscle and bone marrow derived stromal cells: a

comparison of tenocyte differentiation capabilities. J. Orthop. Res. 30,

1710–1718 (2012).

20. Komura, S. et al. Induced pluripotent stem cell-derived tenocyte-like cells

promote the regeneration of injured tendons in mice. Sci. Rep. 10, 3992

(2020).

21. Rajpar, I. & Barrett, J. G. Optimizing growth factor induction of tenogenesis in

three-dimensional culture of mesenchymal stem cells. J. Tissue Eng. 10,

2041731419848776–2041731419848776 (2019).

22. Chen, X. et al. Force and scleraxis synergistically promote the commitment of

human ES cells derived MSCs to tenocytes. Sci. Rep. 2, 977 (2012).

23. Chen, X. et al. Scleraxis-overexpressed human embryonic stem cell-derived

mesenchymal stem cells for tendon tissue engineering with knitted silkcollagen scaffold. Tissue Eng. Part A 20, 1583–1592 (2014).

24. Brent, A. E., Schweitzer, R. & Tabin, C. J. A somitic compartment of tendon

progenitors. Cell 113, 235–248 (2003).

25. Brent, A. E. & Tabin, C. J. Developmental regulation of somite derivatives:

muscle, cartilage and tendon. Curr. Opin. Genet. Dev. 12, 548–557 (2002).

26. Benazeraf, B. & Pourquie, O. Formation and segmentation of the vertebrate

body axis. Annu. Rev. Cell Dev. Biol. 29, 1–26 (2013).

27. Ikeya, M. & Takada, S. Wnt signaling from the dorsal neural tube is required

for the formation of the medial dermomyotome. Development 125, 4969–4976

(1998).

28. Pourquié, O., Coltey, M., Teillet, M. A., Ordahl, C. & Le Douarin, N. M.

Control of dorsoventral patterning of somitic derivatives by notochord and

floor plate. Proc. Natl Acad. Sci. USA 90, 5242–5246 (1993).

29. Dubrulle, J. & Pourquie, O. Welcome to syndetome: a new somitic

compartment. Dev. Cell 4, 611–612 (2003).

30. Sakurai, H. et al. In vitro modeling of paraxial mesodermal progenitors derived

from induced pluripotent stem cells. PLoS ONE 7, e47078–e47078 (2012).

31. Chal, J. et al. Differentiation of pluripotent stem cells to muscle fiber to model

Duchenne muscular dystrophy. Nat. Biotechnol. 33, 962–969 (2015).

32. Umeda, K. et al. Human chondrogenic paraxial mesoderm, directed

specification and prospective isolation from pluripotent stem cells. Sci. Rep. 2,

455 (2012).

33. Xi, H. et al. In vivo human somitogenesis guides somite development from

hPSCs. Cell Rep. 18, 1573–1585 (2017).

34. Loh, K. M. M. et al. Mapping the pairwise choices leading from pluripotency

to human bone, heart, and other mesoderm cell types. Cell 166, 451–468

(2016).

35. Nakagawa, M. et al. A novel efficient feeder-free culture system for the

derivation of human induced pluripotent stem cells. Sci. Rep. 4, 3594 (2014).

36. Zhao, J. et al. Small molecule-directed specification of sclerotome-like

chondroprogenitors and induction of a somitic chondrogenesis program from

embryonic stem cells. Development 141, 3848–3858 (2014).

37. Pryce, B. A. et al. Recruitment and maintenance of tendon progenitors by

TGFbeta signaling are essential for tendon formation. Development 136,

1351–1361 (2009).

38. Schwarting, T. et al. Bone morphogenetic protein 7 (BMP-7) influences

tendon-bone integration in vitro. PLoS ONE 10, e0116833 (2015).

39. Schaeffer, J., Tannahill, D., Cioni, J.-M., Rowlands, D. & Keynes, R.

Identification of the extracellular matrix protein Fibulin-2 as a regulator of

spinal nerve organization. Dev. Biol. 442, 101–114 (2018).

40. Murrell, G. A. et al. The achilles functional index. J. Orthop. Res. 10, 398–404

(1992).

41. Molloy, T., Wang, Y. & Murrell, G. The roles of growth factors in tendon and

ligament healing. Sports Med. 33, 381–394 (2003).

42. Hwa, V., Oh, Y. & Rosenfeld, R. G. The insulin-like growth factor-binding

protein (IGFBP) superfamily. Endocr. Rev. 20, 761–787 (1999).

NATURE COMMUNICATIONS | (2021)12:5012 | https://doi.org/10.1038/s41467-021-25328-6 | www.nature.com/naturecommunications

11

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

ARTICLE

NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-25328-6

43. Robertson, I. B. et al. Latent TGF-beta-binding proteins. Matrix Biol. 47,

44–53 (2015).

44. Kurtz, C. A., Loebig, T. G., Anderson, D. D., DeMeo, P. J. & Campbell, P. G.

Insulin-like growth factor I accelerates functional recovery from achilles

tendon injury in a rat model. Am. J. Sports Med. 27, 363–369 (1999).

45. Bortz, W. M. 2nd The disuse syndrome. West. J. Med. 141, 691–694 (1984).

46. Lee, J., Smeriglio, P., Chu, C. R. & Bhutani, N. Human iPSC-derived chondrocytes

mimic juvenile chondrocyte function for the dual advantage of increased

proliferation and resistance to IL-1beta. Stem Cell Res. Ther. 8, 244 (2017).

47. Mandai, M. et al. iPSC-derived retina transplants improve vision in rd1 endstage retinal-degeneration mice. Stem cell Rep. 8, 69–83 (2017).

48. Schweitzer, R. et al. Analysis of the tendon cell fate using Scleraxis, a specific

marker for tendons and ligaments. Development 128, 3855–3866 (2001).

49. Fertin, C. et al. Interleukin-4 stimulates collagen synthesis by normal and

scleroderma fibroblasts in dermal equivalents. Cell. Mol. Biol. 37, 823–829 (1991).

50. Sempowski, G. D., Derdak, S. & Phipps, R. P. Interleukin-4 and interferongamma discordantly regulate collagen biosynthesis by functionally distinct

lung fibroblast subsets. J. Cell. Physiol. 167, 290–296 (1996).

51. Mahboubi, S., Glaser, D. L., Shore, E. M. & Kaplan, F. S. Fibrodysplasia

ossificans progressiva. Pediatr. Radiol. 31, 307–314 (2001).

52. Smith, Z. A., Buchanan, C. C., Raphael, D. & Khoo, L. T. Ossification of the

posterior longitudinal ligament: pathogenesis, management, and current

surgical approaches. A review. Neurosurg. Focus 30, E10 (2011).

53. Chan, K.-M. & Fu, S.-C. Anti-inflammatory management for tendon injuries friends or foes? Sports Med. Arthrosc. Rehabil. Ther. Technol. 1, 23 (2009).

54. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma

by single-cell RNA-seq. Science 352, 189–196 (2016).

55. Wang, T. et al. Functional evaluation outcomes correlate with

histomorphometric changes in the rat sciatic nerve crush injury model: a

comparison between sciatic functional index and kinematic analysis. PLoS

ONE 13, e0208985 (2018).

56. Yamana, R. et al. Rapid and deep profiling of human induced pluripotent stem

cell proteome by one-shot NanoLC-MS/MS analysis with meter-scale

monolithic silica columns. J. Proteome Res. 12, 214–221 (2013).

57. Rappsilber, J., Ishihama, Y. & Mann, M. Stop And Go Extraction tips for

matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS

sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).

58. Hahne, H. et al. DMSO enhances electrospray response, boosting sensitivity of

proteomic experiments. Nat. Methods 10, 989–991 (2013).

59. Nesvizhskii, A. I. & Aebersold, R. Interpretation of shotgun proteomic data.

Mol. Cell. Proteom. 4, 1419–1440 (2005).

60. Iwasaki, M., Tabata, T., Kawahara, Y., Ishihama, Y. & Nakagawa, M. Removal

of interference MS/MS spectra for accurate quantification in isobaric tagbased proteomics. J. Proteome Res. 18, 2535–2544 (2019).

61. Moriya, Y. et al. The jPOST environment: an integrated proteomics data

repository and database. Nucleic Acids Res. 47, D1218–D1224 (2018).

Acknowledgements

We thank Drs. D. Murata, D. Kamiya, Y. Toyooka, and N. Boyd-Gibbins for invaluable

discussions, comments, and critical reading of the manuscript, M. Terashima, and Y.

Inada for technical support, Dr. M. Nomura, K. Ohnishi, and J. Kuwahara for single-cell

12

RNA sequencing, and all members of the Ikeya laboratory for their support in this study.

This work was supported by the Japan Society for the Promotion of Science (JSPS)

KAKENHI grant number 20H03803 (to M.Ik.), the Core Center for iPS Cell Research of

the Research Center Network for Realization of Regenerative Medicine

(20bm0104001h0008) and the Projects for Technical Development, which is a program

of the Research Center Network for Realization of Regenerative Medicine

(20bm0404066h0001), from the Japan Agency for Medical Research and Development

(AMED) (to M.Ik.), a grant from the iPS Cell Research Fund (to M.Ik.), and a research

grant from the Fujiwara Memorial Foundation (to T.N.).

Author contributions

T.N.: Conception and design, data collection and assembly, data analysis and interpretation, manuscript writing, final approval of the manuscript; A.N., N.Y., K.Y., C.Z.:

Data collection and assembly; T.K.: Analysis and interpretation of single-cell RNA

sequencing data; M.Iw.: experimental design and proteome data interpretation; H.K.:

administrative support; M.Ik.: Conception and design, financial support, administrative

support, data interpretation, manuscript writing, final approval of the manuscript.

Competing interests

The authors declare no competing interests.

Additional information

Supplementary information The online version contains supplementary material

available at https://doi.org/10.1038/s41467-021-25328-6.

Correspondence and requests for materials should be addressed to T.N. or M.I.

Peer review information Nature Communications thanks the anonymous reviewer(s) for

their contribution to the peer review of this work. Peer reviewer reports are available.

Reprints and permission information is available at http://www.nature.com/reprints

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in

published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons

Attribution 4.0 International License, which permits use, sharing,

adaptation, distribution and reproduction in any medium or format, as long as you give

appropriate credit to the original author(s) and the source, provide a link to the Creative

Commons license, and indicate if changes were made. The images or other third party

material in this article are included in the article’s Creative Commons license, unless

indicated otherwise in a credit line to the material. If material is not included in the

article’s Creative Commons license and your intended use is not permitted by statutory

regulation or exceeds the permitted use, you will need to obtain permission directly from

the copyright holder. To view a copy of this license, visit http://creativecommons.org/

licenses/by/4.0/.

© The Author(s) 2021

NATURE COMMUNICATIONS | (2021)12:5012 | https://doi.org/10.1038/s41467-021-25328-6 | www.nature.com/naturecommunications

...

参考文献をもっと見る